1
|
Saiprayong K, Chupradit K, Sasithong P, Suwanpitak S, Muneekaew S, Thongsin N, Srisantitham J, Wattanapanitch M. Development of 2LTRZFP-expressing induced pluripotent stem cells as a potential anti-HIV-1 gene therapy against viral integration. J Leukoc Biol 2025; 117:qiaf018. [PMID: 39946247 DOI: 10.1093/jleuko/qiaf018] [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: 06/28/2024] [Revised: 11/29/2024] [Accepted: 02/12/2025] [Indexed: 04/26/2025] Open
Abstract
Highly active antiretroviral drug is the standard treatment for HIV-1 infection to suppress the viral load. However, this treatment does not completely eradicate the virus; it simply decreases the viral load to undetectable levels. The development of a novel therapy to cure the disease is essential. Previously, we developed an engineered zinc finger protein (ZFP) that specifically binds to the 2-LTR-circle junction (2LTRZFP), the target site for viral integrase, preventing HIV-1 integration in human CD34+ hematopoietic stem/progenitor cells (HSPCs) and macrophages. Although the transduction efficiency of 2LTRZFP was ∼50%, purifying and expanding the 2LTRZFP-expressing HSPCs proved difficult. In addition, the batch-to-batch variability in transduction efficiency could have a major impact on the therapeutic efficacy. In this study, we introduced the 2LTRZFP into human induced pluripotent stem cells (iPSCs) followed by clonal isolation and functional validation of the 2LTRZFP. Upon the HIV-1 challenge, the 2LTRZFP protein was found to inhibit the viral integration in iPSCs, iPSC-derived HSPCs, and macrophages. The engineered iPSC clone could be differentiated into functional macrophages, as evidenced by M1 and M2 polarization, and phagocytosis. Our finding revealed that the 2LTRZFP did not perturb the macrophage differentiation process. Therefore, the 2LTRZFP-expressing iPSCs could provide an unlimited supply of HIV-1-resistant HSPCs for transplantation, potentially leading to HIV-1-resistant blood cells. The knowledge obtained from this study will provide a cornerstone for HIV-1 gene therapy using HSPC transplantation as a sustainable HIV-1 treatment in the future.
Collapse
Affiliation(s)
- Kritayaporn Saiprayong
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Koollawat Chupradit
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Pasut Sasithong
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Biomedical Sciences Graduate Program, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Siriwal Suwanpitak
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Saitong Muneekaew
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Nontaphat Thongsin
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Jakkrapatra Srisantitham
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| | - Methichit Wattanapanitch
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok 10700, Thailand
| |
Collapse
|
2
|
Feist WN, Luna SE, Ben-Efraim K, Filsinger Interrante MV, Amorin A, Johnston NM, Bruun TUJ, Utz A, Ghanim HY, Lesch BJ, McLaughlin TM, Dudek AM, Porteus MH. Multilayered HIV-1 resistance in HSPCs through CCR5 Knockout and B cell secretion of HIV-inhibiting antibodies. Nat Commun 2025; 16:3103. [PMID: 40164595 PMCID: PMC11958643 DOI: 10.1038/s41467-025-58371-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Allogeneic transplantation of CCR5 null hematopoietic stem and progenitor cells (HSPCs) is the only known cure for HIV-1 infection. However, this treatment is limited because of the rarity of CCR5-null matched donors, the morbidities associated with allogeneic transplantation, and the prevalence of HIV-1 strains resistant to CCR5 knockout (KO) alone. Here, we propose a one-time therapy through autologous transplantation of HSPCs genetically engineered ex vivo to produce both CCR5 KO cells and long-term secretion of potent HIV-1 inhibiting antibodies from B cell progeny. CRISPR-Cas9-engineered HSPCs engraft and reconstitute multiple hematopoietic lineages in vivo and can be engineered to express multiple antibodies simultaneously (in pre-clinical models). Human B cells engineered to express each antibody secrete neutralizing concentrations capable of inhibiting HIV-1 pseudovirus infection in vitro. This work lays the foundation for a potential one-time functional cure for HIV-1 through combining the long-term delivery of therapeutic antibodies against HIV-1 and the known efficacy of CCR5 KO HSPC transplantation.
Collapse
Affiliation(s)
- William N Feist
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sofia E Luna
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Kaya Ben-Efraim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria V Filsinger Interrante
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford ChEM-H, Stanford University, Stanford, CA, USA
| | - Alvaro Amorin
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicole M Johnston
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Theodora U J Bruun
- Stanford ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Ashley Utz
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Hana Y Ghanim
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin J Lesch
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Amanda M Dudek
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Matthew H Porteus
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
3
|
Tomer S, Pan L, Harding J, Rezek V, Krawczyk M, Cook E, Kedia N, Wang L, Le J, Martin H, Gramajo E, Li X, Zhang Y, Mu W, Wen J, Kitchen S, Zhang Y, Zhen A. A Novel Glia-immune humanized mouse model for investigation of HIV CNS infection and neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641678. [PMID: 40161608 PMCID: PMC11952312 DOI: 10.1101/2025.03.05.641678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Although combined antiretroviral therapy (ART) is successful in suppressing viral replication, HIV persists in anatomic reservoirs, including the central nervous system (CNS). Current models, such as HSC-reconstituted humanized mice, lack matched human glia in the brain, limiting insights into HIV CNS infection and pathogenesis. We developed a novel glia-immune humanized mouse model integrating human glial cells in the brain with donor-matched human immune reconstitution in peripheral blood and lymphoid tissues. Neonatal NSG mice were injected intrahepatically with Hematopoietic Stem Cells (HSCs) and intracranially with donor-matched glia. We observed extensive engraftment of all three types of human glial cells (astrocytes, oligodendroglia, and microglia-like cells) in key CNS regions, including the cerebral cortex and hippocampus. These glia-immune mice supported robust HIV-1 replication in the peripheral blood, lymphoid tissues and CNS. HIV-infected mice exhibited heightened inflammation and elevated expression of type I interferon-stimulated genes (ISGs) in peripheral and brain tissues. Bulk RNAseq revealed significant transcriptional changes in human glia, such as upregulation of ISGs, inflammasome-associated genes, and downregulation of transcriptional regulators implicated in metabolic regulations and epigenetic controls. Overall, this model allows interrogation of glial transcriptomic changes and neuron-immune interactions, offering insights into HIV CNS infection, pathology, and therapeutic strategies targeting CNS HIV reservoirs and neuroinflammatory pathways.
Collapse
|
4
|
Guo Q, Parikh K, Zhang J, Brinkley A, Chen G, Jakramonpreeya N, Zhen A, An DS. Anti-HIV-1 HSPC-based gene therapy with safety kill switch to defend against and attack HIV-1 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623476. [PMID: 39605384 PMCID: PMC11601352 DOI: 10.1101/2024.11.13.623476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hematopoietic stem/progenitor cell (HSPC)-based anti-HIV-1 gene therapy holds promise to provide life-long remission following a single treatment. Here we report a multi-pronged anti-HIV-1 HSPC-based gene therapy designed to defend against and attack HIV-1 infection. We developed a lentiviral vector capable of co-expressing three anti-HIV-1 genes. Two are designed to prevent infection, including a short-hairpin RNA (CCR5sh1005) to knock down HIV-1 co-receptor CCR5 and a membrane anchored HIV-1 fusion inhibitor (C46). The third gene is a CD4-based chimeric antigen receptor (CAR) designed to attack HIV-1 infected cells. Our vector also includes a non-signaling truncated human epidermal growth factor receptor (huEGFRt) which acts as a negative selection-based safety kill switch against transduced cells. Anti-HIV-1 vector-transduced human CD34+ HSPC efficiently reconstituted multi-lineage human hematopoietic cells in humanized bone marrow/liver/thymus (huBLT) mice. HIV-1 viral load was significantly reduced (1-log fold reduction, p <0.001) in transplanted huBLT mice. Anti-huEGFR monoclonal antibody Cetuximab (CTX) administration significantly reduced huEGFRt+ vector-modified cells (>4-fold reduction, p <0.01) in huBLT mice. These results demonstrate that our strategy is highly effective for HIV-1 inhibition, and that CTX-mediated negative selection can deplete anti-HIV-1 vector-modified cells in the event of unwanted adverse effects in huBLT mice.
Collapse
Affiliation(s)
- Qi Guo
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
- Shanghai Key Laboratory of Tumor System Regulation and Clinical Translation, Jiading Branch, Renji Hospital, Shanghai Cancer Institute, Shanghai, China, 201800
| | - Keval Parikh
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| | - Jian Zhang
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| | - Alexander Brinkley
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| | - Grace Chen
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA, USA, 90095
| | - Natnicha Jakramonpreeya
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Anjie Zhen
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA, 90095
| | - Dong Sung An
- UCLA AIDS Institute, UCLA, Los Angeles, CA, USA, 90024
- UCLA School of Nursing, UCLA, Los Angeles, CA, USA, 90095
| |
Collapse
|
5
|
Dudek AM, Feist WN, Sasu EJ, Luna SE, Ben-Efraim K, Bak RO, Cepika AM, Porteus MH. A simultaneous knockout knockin genome editing strategy in HSPCs potently inhibits CCR5- and CXCR4-tropic HIV-1 infection. Cell Stem Cell 2024; 31:499-518.e6. [PMID: 38579682 PMCID: PMC11212398 DOI: 10.1016/j.stem.2024.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 12/29/2023] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
Allogeneic hematopoietic stem and progenitor cell transplant (HSCT) of CCR5 null (CCR5Δ32) cells can be curative for HIV-1-infected patients. However, because allogeneic HSCT poses significant risk, CCR5Δ32 matched bone marrow donors are rare, and CCR5Δ32 transplant does not confer resistance to the CXCR4-tropic virus, it is not a viable option for most patients. We describe a targeted Cas9/AAV6-based genome editing strategy for autologous HSCT resulting in both CCR5- and CXCR4-tropic HIV-1 resistance. Edited human hematopoietic stem and progenitor cells (HSPCs) maintain multi-lineage repopulation capacity in vivo, and edited primary human T cells potently inhibit infection by both CCR5-tropic and CXCR4-tropic HIV-1. Modification rates facilitated complete loss of CCR5-tropic replication and up to a 2,000-fold decrease in CXCR4-tropic replication without CXCR4 locus disruption. This multi-factor editing strategy in HSPCs could provide a broad approach for autologous HSCT as a functional cure for both CCR5-tropic and CXCR4-tropic HIV-1 infections.
Collapse
Affiliation(s)
- Amanda M Dudek
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William N Feist
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena J Sasu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sofia E Luna
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kaya Ben-Efraim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rasmus O Bak
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, 8000 Aarhus, Denmark
| | - Alma-Martina Cepika
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
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
|
7
|
Feist WN, Luna SE, Ben-Efraim K, Filsinger Interrante MV, Amorin NA, Johnston NM, Bruun TUJ, Ghanim HY, Lesch BJ, Dudek AM, Porteus MH. Combining Cell-Intrinsic and -Extrinsic Resistance to HIV-1 By Engineering Hematopoietic Stem Cells for CCR5 Knockout and B Cell Secretion of Therapeutic Antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.583956. [PMID: 38496600 PMCID: PMC10942466 DOI: 10.1101/2024.03.08.583956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Autologous transplantation of CCR5 null hematopoietic stem and progenitor cells (HSPCs) is the only known cure for HIV-1 infection. However, this treatment is limited because of the rarity of CCR5 -null matched donors, the morbidities associated with allogeneic transplantation, and the prevalence of HIV-1 strains resistant to CCR5 knockout (KO) alone. Here, we propose a one-time therapy through autologous transplantation of HSPCs genetically engineered ex vivo to produce both CCR5 KO cells and long-term secretion of potent HIV-1 inhibiting antibodies from B cell progeny. CRISPR-Cas9-engineered HSPCs maintain engraftment capacity and multi-lineage potential in vivo and can be engineered to express multiple antibodies simultaneously. Human B cells engineered to express each antibody secrete neutralizing concentrations capable of inhibiting HIV-1 pseudovirus infection in vitro . This work lays the groundwork for a potential one-time functional cure for HIV-1 through combining the long-term delivery of therapeutic antibodies against HIV-1 and the known efficacy of CCR5 KO HSPC transplantation.
Collapse
|
8
|
Guo Q, Zhang J, Parikh K, Brinkley A, Lin S, Zakarian C, Pernet O, Shimizu S, Khamaikawin W, Hacke K, Kasahara N, An DS. In vivo selection of anti-HIV-1 gene-modified human hematopoietic stem/progenitor cells to enhance engraftment and HIV-1 inhibition. Mol Ther 2024; 32:384-394. [PMID: 38087779 PMCID: PMC10862071 DOI: 10.1016/j.ymthe.2023.12.007] [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: 08/28/2023] [Revised: 11/17/2023] [Accepted: 12/08/2023] [Indexed: 12/26/2023] Open
Abstract
Hematopoietic stem/progenitor cell (HSPC)-based anti-HIV-1 gene therapy holds great promise to eradicate HIV-1 or to provide long-term remission through a continuous supply of anti-HIV-1 gene-modified cells without ongoing antiretroviral therapy. However, achieving sufficient engraftment levels of anti-HIV gene-modified HSPC to provide therapeutic efficacy has been a major limitation. Here, we report an in vivo selection strategy for anti-HIV-1 gene-modified HSPC by introducing 6-thioguanine (6TG) chemoresistance through knocking down hypoxanthine-guanine phosphoribosyl transferase (HPRT) expression using RNA interference (RNAi). We developed a lentiviral vector capable of co-expressing short hairpin RNA (shRNA) against HPRT alongside two anti-HIV-1 genes: shRNA targeting HIV-1 co-receptor CCR5 and a membrane-anchored HIV-1 fusion inhibitor, C46, for efficient in vivo selection of anti-HIV-1 gene-modified human HSPC. 6TG-mediated preconditioning and in vivo selection significantly enhanced engraftment of HPRT-knockdown anti-HIV-1 gene-modified cells (>2-fold, p < 0.0001) in humanized bone marrow/liver/thymus (huBLT) mice. Viral load was significantly reduced (>1 log fold, p < 0.001) in 6TG-treated HIV-1-infected huBLT mice compared to 6TG-untreated mice. We demonstrated that 6TG-mediated preconditioning and in vivo selection considerably improved engraftment of HPRT-knockdown anti-HIV-1 gene-modified HSPC and repopulation of anti-HIV-1 gene-modified hematopoietic cells in huBLT mice, allowing for efficient HIV-1 inhibition.
Collapse
Affiliation(s)
- Qi Guo
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Jian Zhang
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Keval Parikh
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Alexander Brinkley
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Samantha Lin
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Christina Zakarian
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Olivier Pernet
- Maternal, Child, and Adolescent Center for Infectious Diseases, University of Southern California, Los Angeles, CA 90089, USA
| | - Saki Shimizu
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA
| | - Wannisa Khamaikawin
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA; Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - Katrin Hacke
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Phoenix, AZ 85054, USA
| | - Noriyuki Kasahara
- UCSF, Neurological Surgery, Radiation Oncology, San Francisco, CA 94158, USA
| | - Dong Sung An
- UCLA AIDS Institute, UCLA, Los Angeles, CA 90024, USA; UCLA School of Nursing, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
9
|
Mu W, Patankar V, Kitchen S, Zhen A. Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection. Viruses 2024; 16:219. [PMID: 38399994 PMCID: PMC10893210 DOI: 10.3390/v16020219] [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: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Chronic Human Immunodeficiency Virus (HIV) infection remains a significant challenge to global public health. Despite advances in antiretroviral therapy (ART), which has transformed HIV infection from a fatal disease into a manageable chronic condition, a definitive cure remains elusive. One of the key features of HIV infection is chronic immune activation and inflammation, which are strongly associated with, and predictive of, HIV disease progression, even in patients successfully treated with suppressive ART. Chronic inflammation is characterized by persistent inflammation, immune cell metabolic dysregulation, and cellular exhaustion and dysfunction. This review aims to summarize current knowledge of the interplay between chronic inflammation, immune metabolism, and T cell dysfunction in HIV infection, and also discusses the use of humanized mice models to study HIV immune pathogenesis and develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Wenli Mu
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Vaibhavi Patankar
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Scott Kitchen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Anjie Zhen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
10
|
Baroncini L, Bredl S, Nicole KP, Speck RF. The Humanized Mouse Model: What Added Value Does It Offer for HIV Research? Pathogens 2023; 12:pathogens12040608. [PMID: 37111494 PMCID: PMC10142098 DOI: 10.3390/pathogens12040608] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
In the early 2000s, novel humanized mouse models based on the transplantation of human hematopoietic stem and progenitor cells (HSPCs) into immunocompromised mice were introduced (hu mice). The human HSPCs gave rise to a lymphoid system of human origin. The HIV research community has greatly benefitted from these hu mice. Since human immunodeficiency virus (HIV) type 1 infection results in a high-titer disseminated HIV infection, hu mice have been of great value for all types of HIV research from pathogenesis to novel therapies. Since the first description of this new generation of hu mice, great efforts have been expended to improve humanization by creating other immunodeficient mouse models or supplementing mice with human transgenes to improve human engraftment. Many labs have their own customized hu mouse models, making comparisons quite difficult. Here, we discuss the different hu mouse models in the context of specific research questions in order to define which characteristics should be considered when determining which hu mouse model is appropriate for the question posed. We strongly believe that researchers must first define their research question and then determine whether a hu mouse model exists, allowing the research question to be studied.
Collapse
Affiliation(s)
- Luca Baroncini
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Simon Bredl
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Kadzioch P Nicole
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Roberto F Speck
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
11
|
Rothemejer FH, Lauritsen NP, Juhl AK, Schleimann MH, König S, Søgaard OS, Bak RO, Tolstrup M. Development of HIV-Resistant CAR T Cells by CRISPR/Cas-Mediated CAR Integration into the CCR5 Locus. Viruses 2023; 15:202. [PMID: 36680242 PMCID: PMC9862650 DOI: 10.3390/v15010202] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
Adoptive immunotherapy using chimeric antigen receptor (CAR) T cells has been highly successful in treating B cell malignancies and holds great potential as a curative strategy for HIV infection. Recent advances in the use of anti-HIV broadly neutralizing antibodies (bNAbs) have provided vital information for optimal antigen targeting of CAR T cells. However, CD4+ CAR T cells are susceptible to HIV infection, limiting their therapeutic potential. In the current study, we engineered HIV-resistant CAR T cells using CRISPR/Cas9-mediated integration of a CAR cassette into the CCR5 locus. We used a single chain variable fragment (scFv) of the clinically potent bNAb 10-1074 as the antigen-targeting domain in our anti-HIV CAR T cells. Our anti-HIV CAR T cells showed specific lysis of HIV-infected cells in vitro. In a PBMC humanized mouse model of HIV infection, the anti-HIV CAR T cells expanded and transiently limited HIV infection. In conclusion, this study provides proof-of-concept for developing HIV-resistant CAR T cells using CRISPR/Cas9 targeted integration.
Collapse
Affiliation(s)
- Frederik Holm Rothemejer
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Nanna Pi Lauritsen
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Anna Karina Juhl
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Mariane Høgsbjerg Schleimann
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Saskia König
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
| | - Ole Schmeltz Søgaard
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Rasmus O. Bak
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
- Aarhus Institute of Advanced Studies, Aarhus University, 8200 Aarhus, Denmark
| | - Martin Tolstrup
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
12
|
Estep BK, Kuhlmann CJ, Osuka S, Suryavanshi GW, Nagaoka-Kamata Y, Samuel CN, Blucas MT, Jepson CE, Goepfert PA, Kamata M. Skewed fate and hematopoiesis of CD34 + HSPCs in umbilical cord blood amid the COVID-19 pandemic. iScience 2022; 25:105544. [PMID: 36406860 PMCID: PMC9650991 DOI: 10.1016/j.isci.2022.105544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/11/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Umbilical cord blood (UCB) is an irreplaceable source for hematopoietic stem progenitor cells (HSPCs). However, the effects of SARS-CoV-2 infection and COVID-19 vaccination on UCB phenotype, specifically the HSPCs therein, are currently unknown. We thus evaluated any effects of SARS-CoV-2 infection and/or COVID-19 vaccination from the mother on the fate and functionalities of HSPCs in the UCB. The numbers and frequencies of HSPCs in the UCB decreased significantly in donors with previous SARS-CoV-2 infection and more so with COVID-19 vaccination via the induction of apoptosis, likely mediated by IFN-γ-dependent pathways. Two independent hematopoiesis assays, a colony forming unit assay and a mouse humanization assay, revealed skewed hematopoiesis of HSPCs obtained from donors delivered from mothers with SARS-CoV-2 infection history. These results indicate that SARS-CoV-2 infection and COVID-19 vaccination impair the functionalities and survivability of HSPCs in the UCB, which would make unprecedented concerns on the future of HSPC-based therapies.
Collapse
Affiliation(s)
- Benjamin K. Estep
- Department of Microbiology, University of Alabama at Birmingham, 845 19 Street South, Birmingham, AL 35205, USA
| | - Charles J. Kuhlmann
- Department of Microbiology, University of Alabama at Birmingham, 845 19 Street South, Birmingham, AL 35205, USA
| | - Satoru Osuka
- Department of Neurosurgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Gajendra W. Suryavanshi
- Division of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | - Ciearria N. Samuel
- Department of Microbiology, University of Alabama at Birmingham, 845 19 Street South, Birmingham, AL 35205, USA
| | - Madison T. Blucas
- Department of Microbiology, University of Alabama at Birmingham, 845 19 Street South, Birmingham, AL 35205, USA
| | - Chloe E. Jepson
- Department of Microbiology, University of Alabama at Birmingham, 845 19 Street South, Birmingham, AL 35205, USA
| | - Paul A. Goepfert
- Department of Medicine and Division of Infectious Diseases, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Masakazu Kamata
- Department of Microbiology, University of Alabama at Birmingham, 845 19 Street South, Birmingham, AL 35205, USA
| |
Collapse
|
13
|
Kranz E, Kuhlmann CJ, Chan J, Kim PY, Chen ISY, Kamata M. Efficient derivation of chimeric-antigen receptor-modified TSCM cells. Front Immunol 2022; 13:877682. [PMID: 35967430 PMCID: PMC9366550 DOI: 10.3389/fimmu.2022.877682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Chimeric-antigen receptor (CAR) T-cell immunotherapy employs autologous-T cells modified with an antigen-specific CAR. Current CAR-T manufacturing processes tend to yield products dominated by effector T cells and relatively small proportions of long-lived memory T cells. Those few cells are a so-called stem cell memory T (TSCM) subset, which express naïve T-cell markers and are capable of self-renewal and oligopotent differentiation into effector phenotypes. Increasing the proportion of this subset may lead to more effective therapies by improving CAR-T persistence; however, there is currently no standardized protocol for the effective generation of CAR-TSCM cells. Here we present a simplified protocol enabling efficient derivation of gene-modified TSCM cells: Stimulation of naïve CD8+ T cells with only soluble anti-CD3 antibody and culture with IL-7 and IL-15 was sufficient for derivation of CD8+ T cells harboring TSCM phenotypes and oligopotent capabilities. These in-vitro expanded TSCM cells were engineered with CARs targeting the HIV-1 envelope protein as well as the CD19 molecule and demonstrated effector activity both in vitro and in a xenograft mouse model. This simple protocol for the derivation of CAR-TSCM cells may facilitate improved adoptive immunotherapy.
Collapse
Affiliation(s)
- Emiko Kranz
- Division of Hematology-Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Charles J. Kuhlmann
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joshua Chan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Patrick Y. Kim
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Irvin S. Y. Chen
- Division of Hematology-Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Masakazu Kamata
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Masakazu Kamata,
| |
Collapse
|
14
|
Choudhary MC, Cyktor JC, Riddler SA. Advances in HIV-1-specific chimeric antigen receptor cells to target the HIV-1 reservoir. J Virus Erad 2022; 8:100073. [PMID: 35784676 PMCID: PMC9241028 DOI: 10.1016/j.jve.2022.100073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/15/2022] [Indexed: 01/09/2023] Open
Abstract
Antiretroviral therapy (ART) for HIV-1 has dramatically improved outcomes for people living with HIV-1 but requires life-long adherence and can be associated with short and long-term toxicity. Numerous pre-clinical and clinical investigations are underway to develop therapies for immune control of HIV-1 in the absence of ART. The success of chimeric antigen receptor (CAR) cell therapy for hematological malignancy has renewed efforts to develop and investigate CAR cells as strategies to enhance HIV-1 immunity, enable virus control or elimination, and allow ART-free HIV-1 remission. Here, we review the improvements in anti-HIV-1 CAR cell therapy in the two decades since their initial clinical trials were conducted, describe the additional engineering required to protect CAR cells from HIV-1 infection, and preview the current landscape of CAR cell therapies advancing to HIV-1 clinical trials.
Collapse
Affiliation(s)
- Madhu C. Choudhary
- Corresponding author. Division of Infectious Diseases, University of Pittsburgh, Suite 510, 3601 5Th Ave., Pittsburgh, PA, 15213, USA.
| | | | | |
Collapse
|
15
|
York J, Gowrishankar K, Micklethwaite K, Palmer S, Cunningham AL, Nasr N. Evolving Strategies to Eliminate the CD4 T Cells HIV Viral Reservoir via CAR T Cell Immunotherapy. Front Immunol 2022; 13:873701. [PMID: 35572509 PMCID: PMC9098815 DOI: 10.3389/fimmu.2022.873701] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Although the advent of ART has significantly reduced the morbidity and mortality associated with HIV infection, the stable pool of HIV in latently infected cells requires lifelong treatment adherence, with the cessation of ART resulting in rapid reactivation of the virus and productive HIV infection. Therefore, these few cells containing replication-competent HIV, known as the latent HIV reservoir, act as the main barrier to immune clearance and HIV cure. While several strategies involving HIV silencing or its reactivation in latently infected cells for elimination by immune responses have been explored, exciting cell based immune therapies involving genetically engineered T cells expressing synthetic chimeric receptors (CAR T cells) are highly appealing and promising. CAR T cells, in contrast to endogenous cytotoxic T cells, can function independently of MHC to target HIV-infected cells, are efficacious and have demonstrated acceptable safety profiles and long-term persistence in peripheral blood. In this review, we present a comprehensive picture of the current efforts to target the HIV latent reservoir, with a focus on CAR T cell therapies. We highlight the current challenges and advances in this field, while discussing the importance of novel CAR designs in the efforts to find a HIV cure.
Collapse
Affiliation(s)
- Jarrod York
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Kavitha Gowrishankar
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Children’s Cancer Research Unit, Kids Research, The Children’s Hospital at Westmead, Sydney Children’s Hospitals Network, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Kenneth Micklethwaite
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- NSW Health Pathology Blood Transplant and Cell Therapies Laboratory – Institute of Clinical Pathology and Medical Research (ICPMR) Westmead, Sydney, NSW, Australia
| | - Sarah Palmer
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
16
|
Xun J, Zhang X, Guo S, Lu H, Chen J. Editing out HIV: application of gene editing technology to achieve functional cure. Retrovirology 2021; 18:39. [PMID: 34922576 PMCID: PMC8684261 DOI: 10.1186/s12977-021-00581-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 11/05/2021] [Indexed: 03/01/2023] Open
Abstract
Highly active antiretroviral therapy (HAART) successfully suppresses human immunodeficiency virus (HIV) replication and improves the quality of life of patients living with HIV. However, current HAART does not eradicate HIV infection because an HIV reservoir is established in latently infected cells and is not recognized by the immune system. The successful curative treatment of the Berlin and London patients following bone marrow transplantation inspired researchers to identify an approach for the functional cure of HIV. As a promising technology, gene editing-based strategies have attracted considerable attention and sparked much debate. Herein, we discuss the development of different gene editing strategies in the functional cure of HIV and highlight the potential for clinical applications prospects. ![]()
Collapse
Affiliation(s)
- Jingna Xun
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China.,State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Xinyu Zhang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China
| | - Shuyan Guo
- Shanghai Foreign Language School, Shanghai International Studies University, Shanghai, China
| | - Hongzhou Lu
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China
| | - Jun Chen
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China.
| |
Collapse
|
17
|
Sawant N, Singh H, Appukuttan D. Overview of the Cellular Stress Responses Involved in Fatty Acid Overproduction in E. coli. Mol Biotechnol 2021; 64:373-387. [PMID: 34796451 DOI: 10.1007/s12033-021-00426-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/10/2021] [Indexed: 12/29/2022]
Abstract
Research on microbial fatty acid metabolism started in the late 1960s, and till date, various developments have aided in elucidating the fatty acid metabolism in great depth. Over the years, synthesis of microbial fatty acid has drawn industrial attention due to its diverse applications. However, fatty acid overproduction imparts various stresses on its metabolic pathways causing a bottleneck to further increase the fatty acid yields. Numerous strategies to increase fatty acid titres in Escherichia coli by pathway modulation have already been published, but the stress generated during fatty acid overproduction is relatively less studied. Stresses like pH, osmolarity and oxidative stress, not only lower fatty acid titres, but also alter the cell membrane composition, protein expression and membrane fluidity. This review discusses an overview of fatty acid synthesis pathway and presents a panoramic view of various stresses caused due to fatty acid overproduction in E. coli. It also addresses how certain stresses like high temperature and nitrogen limitation can boost fatty acid production. This review paper also highlights the interconnections that exist between these stresses.
Collapse
Affiliation(s)
- Neha Sawant
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS Deemed to be University, Vile Parle (West), Mumbai, 400056, India
| | - Harinder Singh
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS Deemed to be University, Vile Parle (West), Mumbai, 400056, India.
| | - Deepti Appukuttan
- Biosystems Engineering Lab, Department of Chemical Engineering, IIT Bombay, Powai, Mumbai, 400076, India.
| |
Collapse
|
18
|
Mota SI, Pita I, Águas R, Tagorti S, Virmani A, Pereira FC, Rego AC. Mechanistic perspectives on differential mitochondrial-based neuroprotective effects of several carnitine forms in Alzheimer's disease in vitro model. Arch Toxicol 2021; 95:2769-2784. [PMID: 34164711 DOI: 10.1007/s00204-021-03104-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/15/2021] [Indexed: 11/29/2022]
Abstract
Mitochondrial deregulation has emerged as one of the earliest pathological events in Alzheimer's disease (AD), the most common age-related neurodegenerative disorder. Improvement of mitochondrial function in AD has been considered a relevant therapeutic approach. L-carnitine (LC), an amino acid derivative involved in the transport of long-chain fatty acids into mitochondria, was previously demonstrated to improve mitochondrial function, having beneficial effects in neurological disorders; moreover, acetyl-L-carnitine (ALC) is currently under phase 4 clinical trial for AD (ClinicalTrials.gov NCT01320527). Thus, in the present study, we investigated the impact of different forms of carnitines, namely LC, ALC and propionyl-L-carnitine (PLC) on mitochondrial toxicity induced by amyloid-beta peptide 1-42 oligomers (AβO; 1 μM) in mature rat hippocampal neurons. Our results indicate that 5 mM LC, ALC and PLC totally rescued the mitochondrial membrane potential and alleviated both the decrease in oxygen consumption rates and the increase in mitochondrial fragmentation induced by AβO. These could contribute to the prevention of neuronal death by apoptosis. Moreover, only ALC ameliorated AβO-evoked changes in mitochondrial movement by reducing the number of stationary mitochondria and promoting reversal mitochondrial movement. Data suggest that carnitines (LC, ALC and PLC) may act differentially to counteract changes in mitochondrial function and movement in neurons subjected to AβO, thus counteracting AD-related pathological phenotypes.
Collapse
Affiliation(s)
- Sandra I Mota
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
| | - Inês Pita
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Rodolfo Águas
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Slah Tagorti
- Alfasigma B.V, 3528 BG, Utrecht, The Netherlands
| | | | - Frederico C Pereira
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal.
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal.
| | - A Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal.
| |
Collapse
|
19
|
Garcia-Beltran WF, Claiborne DT, Maldini CR, Phelps M, Vrbanac V, Karpel ME, Krupp KL, Power KA, Boutwell CL, Balazs AB, Tager AM, Altfeld M, Allen TM. Innate Immune Reconstitution in Humanized Bone Marrow-Liver-Thymus (HuBLT) Mice Governs Adaptive Cellular Immune Function and Responses to HIV-1 Infection. Front Immunol 2021; 12:667393. [PMID: 34122425 PMCID: PMC8189152 DOI: 10.3389/fimmu.2021.667393] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/28/2021] [Indexed: 01/11/2023] Open
Abstract
Humanized bone marrow-liver-thymus (HuBLT) mice are a revolutionary small-animal model that has facilitated the study of human immune function and human-restricted pathogens, including human immunodeficiency virus type 1 (HIV-1). These mice recapitulate many aspects of acute and chronic HIV-1 infection, but exhibit weak and variable T-cell responses when challenged with HIV-1, hindering our ability to confidently detect HIV-1-specific responses or vaccine effects. To identify the cause of this, we comprehensively analyzed T-cell development, diversity, and function in HuBLT mice. We found that virtually all HuBLT were well-reconstituted with T cells and had intact TCRβ sequence diversity, thymic development, and differentiation to memory and effector cells. However, there was poor CD4+ and CD8+ T-cell responsiveness to physiologic stimuli and decreased TH1 polarization that correlated with deficient reconstitution of innate immune cells, in particular monocytes. HIV-1 infection of HuBLT mice showed that mice with higher monocyte reconstitution exhibited greater CD8+ T cells responses and HIV-1 viral evolution within predicted HLA-restricted epitopes. Thus, T-cell responses to immune challenges are blunted in HuBLT mice due to a deficiency of innate immune cells, and future efforts to improve the model for HIV-1 immune response and vaccine studies need to be aimed at restoring innate immune reconstitution.
Collapse
Affiliation(s)
| | - Daniel T. Claiborne
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| | - Colby R. Maldini
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| | - Meredith Phelps
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| | - Vladimir Vrbanac
- Human Immune System Mouse Program, Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Marshall E. Karpel
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
- Division of Medical Sciences, Harvard University, Boston, MA, United States
| | - Katharine L. Krupp
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| | - Karen A. Power
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| | - Christian L. Boutwell
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| | - Alejandro B. Balazs
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| | - Andrew M. Tager
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
| | - Marcus Altfeld
- Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Todd M. Allen
- Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital, Cambridge, MA, United States
| |
Collapse
|
20
|
Moranguinho I, Valente ST. Block-And-Lock: New Horizons for a Cure for HIV-1. Viruses 2020; 12:v12121443. [PMID: 33334019 PMCID: PMC7765451 DOI: 10.3390/v12121443] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/01/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
HIV-1/AIDS remains a global public health problem. The world health organization (WHO) reported at the end of 2019 that 38 million people were living with HIV-1 worldwide, of which only 67% were accessing antiretroviral therapy (ART). Despite great success in the clinical management of HIV-1 infection, ART does not eliminate the virus from the host genome. Instead, HIV-1 remains latent as a viral reservoir in any tissue containing resting memory CD4+ T cells. The elimination of these residual proviruses that can reseed full-blown infection upon treatment interruption remains the major barrier towards curing HIV-1. Novel approaches have recently been developed to excise or disrupt the virus from the host cells (e.g., gene editing with the CRISPR-Cas system) to permanently shut off transcription of the virus (block-and-lock and RNA interference strategies), or to reactivate the virus from cell reservoirs so that it can be eliminated by the immune system or cytopathic effects (shock-and-kill strategy). Here, we will review each of these approaches, with the major focus placed on the block-and-lock strategy.
Collapse
|
21
|
Ahlenstiel CL, Symonds G, Kent SJ, Kelleher AD. Block and Lock HIV Cure Strategies to Control the Latent Reservoir. Front Cell Infect Microbiol 2020; 10:424. [PMID: 32923412 PMCID: PMC7457024 DOI: 10.3389/fcimb.2020.00424] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
The HIV latent reservoir represents the major challenge to cure development. Residing in resting CD4+ T cells and myeloid cells at multiple locations in the body, including sanctuary sites such as the brain, the latent reservoir is not eliminated by ART and has the ability to reactivate virus replication to pre-therapy levels when ART is ceased. There are four broad areas of HIV cure research. The only successful cure strategy, thus far, is stem cell transplantation using naturally HIV resistant CCR5Δ32 stem cells. A second potential cure approach uses gene editing technology, such as zinc-finger nucleases and CRISPR/Cas9. Another two cure strategies aim to control the HIV reservoir, with polar opposite concepts; The "shock and kill" approach, which aims to "shock" or reactivate the latent virus and then "kill" infected cells via targeted immune responses. Lastly, the "block and lock" approach, which aims to enhance the latent virus state by "blocking" HIV transcription and "locking" the HIV promoter in a deep latent state via epigenetic modifications. "Shock and kill" approaches are a major focus of cure studies, however we predict that the increased specificity of "block and lock" approaches will be required for the successful development of a sustained HIV clinical remission in the absence of ART. This review focuses on the current research of novel "block and lock" approaches being explored to generate an HIV cure via induction of epigenetic silencing. We will also discuss potential future therapeutic delivery and the challenges associated with progressing "block and lock" cure approaches as these move toward clinical trials.
Collapse
Affiliation(s)
| | | | - Stephen J. Kent
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
- ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Parkville, VIC, Australia
| | | |
Collapse
|
22
|
Mu W, Carrillo MA, Kitchen SG. Engineering CAR T Cells to Target the HIV Reservoir. Front Cell Infect Microbiol 2020; 10:410. [PMID: 32903563 PMCID: PMC7438537 DOI: 10.3389/fcimb.2020.00410] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022] Open
Abstract
The HIV reservoir remains to be a difficult barrier to overcome in order to achieve a therapeutic cure for HIV. Several strategies have been developed to purge the reservoir, including the “kick and kill” approach, which is based on the notion that reactivating the latent reservoir will allow subsequent elimination by the host anti-HIV immune cells. However, clinical trials testing certain classes of latency reactivating agents (LRAs) have so far revealed the minimal impact on reducing the viral reservoir. A robust immune response to reactivated HIV expressing cells is critical for this strategy to work. A current focus to enhance anti-HIV immunity is through the use of chimeric antigen receptors (CARs). Currently, HIV-specific CARs are being applied to peripheral T cells, NK cells, and stem cells to boost recognition and killing of HIV infected cells. In this review, we summarize current developments in engineering HIV directed CAR-expressing cells to facilitate HIV elimination. We also summarize current LRAs that enhance the “kick” strategy and how new generation and combinations of LRAs with HIV specific CAR T cell therapies could provide an optimal strategy to target the viral reservoir and achieve HIV clearance from the body.
Collapse
Affiliation(s)
- Wenli Mu
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayra A Carrillo
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Scott G Kitchen
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
23
|
Suryawanshi GW, Khamaikawin W, Wen J, Shimizu S, Arokium H, Xie Y, Wang E, Kim S, Choi H, Zhang C, Yu H, Presson AP, Kim N, An DS, Chen ISY, Kim S. The clonal repopulation of HSPC gene modified with anti-HIV-1 RNAi is not affected by preexisting HIV-1 infection. SCIENCE ADVANCES 2020; 6:eaay9206. [PMID: 32766447 PMCID: PMC7385479 DOI: 10.1126/sciadv.aay9206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 06/09/2020] [Indexed: 05/11/2023]
Abstract
Despite advances in hematopoietic stem/progenitor cell (HSPC) transplant for HIV-1-infected patients, the impact of a preexisting HIV-1 infection on the engraftment and clonal repopulation of HSPCs remains poorly understood. We have developed a long terminal repeat indexing-mediated integration site sequencing (LTRi-Seq) method that provides a multiplexed clonal quantitation of both anti-HIV-1 RNAi (RNA interference) gene-modified and control vector-modified cell populations, together with HIV-1-infected cells-all within the same animal. In our HIV-1-preinfected humanized mice, both therapeutic and control HSPCs repopulated efficiently without abnormalities. Although the HIV-1-mediated selection of anti-HIV-1 RNAi-modified clones was evident in HIV-1-infected mice, the organ-to-organ and intra-organ clonal distributions in infected mice were indistinguishable from those in uninfected mice. HIV-1-infected cells showed clonal patterns distinct from those of HSPCs. Our data demonstrate that, despite the substantial impact of HIV-1 infection on CD4+ T cells, HSPC repopulation remains polyclonal, thus supporting the use of HSPC transplant for anti-HIV treatment.
Collapse
Affiliation(s)
- Gajendra W. Suryawanshi
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Wannisa Khamaikawin
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
- School of Nursing, University of California, Los Angeles, CA 90095, USA
| | - Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Saki Shimizu
- School of Nursing, University of California, Los Angeles, CA 90095, USA
| | - Hubert Arokium
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Yiming Xie
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Eugene Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shihyoung Kim
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Hyewon Choi
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Chong Zhang
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84108, USA
| | - Hannah Yu
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Angela P. Presson
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84108, USA
- Department of Biostatistics, University of California, Los Angeles, CA 90095, USA
| | - Namshin Kim
- Genome Editing Research Center, Korea Research Institute of Biosciences and Biotechnology, Daejeon 34141, Republic of Korea
| | - Dong-Sung An
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
- School of Nursing, University of California, Los Angeles, CA 90095, USA
| | - Irvin S. Y. Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
- Division of Hematology-Oncology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Sanggu Kim
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
24
|
Agarwal Y, Beatty C, Biradar S, Castronova I, Ho S, Melody K, Bility MT. Moving beyond the mousetrap: current and emerging humanized mouse and rat models for investigating prevention and cure strategies against HIV infection and associated pathologies. Retrovirology 2020; 17:8. [PMID: 32276640 PMCID: PMC7149862 DOI: 10.1186/s12977-020-00515-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/31/2020] [Indexed: 12/22/2022] Open
Abstract
The development of safe and effective combination antiretroviral therapies for human immunodeficiency virus (HIV) infection over the past several decades has significantly reduced HIV-associated morbidity and mortality. Additionally, antiretroviral drugs have provided an effective means of protection against HIV transmission. Despite these advances, significant limitations exist; namely, the inability to eliminate HIV reservoirs, the inability to reverse lymphoid tissues damage, and the lack of an effective vaccine for preventing HIV transmission. Evaluation of the safety and efficacy of therapeutics and vaccines for eliminating HIV reservoirs and preventing HIV transmission requires robust in vivo models. Since HIV is a human-specific pathogen, that targets hematopoietic lineage cells and lymphoid tissues, in vivo animal models for HIV-host interactions require incorporation of human hematopoietic lineage cells and lymphoid tissues. In this review, we will discuss the construction of mouse models with human lymphoid tissues and/or hematopoietic lineage cells, termed, human immune system (HIS)-humanized mice. These HIS-humanized mouse models can support the development of functional human innate and adaptive immune cells, along with primary (thymus) and secondary (spleen) lymphoid tissues. We will discuss applications of HIS-humanized mouse models in evaluating the safety and efficacy of therapeutics against HIV reservoirs and associated immunopathology, and delineate the human immune response elicited by candidate HIV vaccines. In addition to focusing on how these HIS-humanized mouse models have already furthered our understanding of HIV and contributed to HIV therapeutics development, we discuss how emerging HIS-humanized rat models could address the limitations of HIS-mouse models.
Collapse
Affiliation(s)
- Yash Agarwal
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cole Beatty
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shivkumar Biradar
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Isabella Castronova
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sara Ho
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin Melody
- Galveston National Laboratory and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Moses Turkle Bility
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
25
|
Sustained delivery and molecular targeting of a therapeutic monoclonal antibody to metastases in the central nervous system of mice. Nat Biomed Eng 2019; 3:706-716. [PMID: 31384008 PMCID: PMC6736720 DOI: 10.1038/s41551-019-0434-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 06/26/2019] [Indexed: 11/24/2022]
Abstract
Approximately 15–40% of all cancers develop metastases in the
central nervous system (CNS), yet few therapeutic options exist to treat them.
Cancer therapies based on monoclonal antibodies are widely successful, yet have
limited efficacy against CNS metastases, owing to the low levels of the drug
reaching the tumour site. Here, we show that the encapsulation of rituximab
within a crosslinked zwitterionic polymer layer leads to the sustained release
of rituximab as the crosslinkers are gradually hydrolyzed, enhancing by
approximately 10-fold the CNS levels of the antibody with respect to the
administration of naked rituximab. When the nanocapsules are functionalized with
CXCL13, the ligand for the chemokine receptor CXCR5 frequently found on B-cell
lymphoma, a single dose led to improved control of CXCR5-expressing metastases
in a murine xenograft model of non-Hodgkin lymphoma, and eliminated lymphoma in
a xenografted humanized bone-marrow–liver–thymus mouse model.
Encapsulation and molecular targeting of therapeutic antibodies could become an
option for the treatment of cancers with CNS metastases.
Collapse
|
26
|
Abstract
In the wake of a breakthrough in biotechnology providing realistic application of recombinant expressed proteins as drugs in the 1990s, gene therapy emerged as the potential approach for providing medicines of the future [...].
Collapse
|
27
|
RNA Viruses as Tools in Gene Therapy and Vaccine Development. Genes (Basel) 2019; 10:genes10030189. [PMID: 30832256 PMCID: PMC6471356 DOI: 10.3390/genes10030189] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/19/2019] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
RNA viruses have been subjected to substantial engineering efforts to support gene therapy applications and vaccine development. Typically, retroviruses, lentiviruses, alphaviruses, flaviviruses rhabdoviruses, measles viruses, Newcastle disease viruses, and picornaviruses have been employed as expression vectors for treatment of various diseases including different types of cancers, hemophilia, and infectious diseases. Moreover, vaccination with viral vectors has evaluated immunogenicity against infectious agents and protection against challenges with pathogenic organisms. Several preclinical studies in animal models have confirmed both immune responses and protection against lethal challenges. Similarly, administration of RNA viral vectors in animals implanted with tumor xenografts resulted in tumor regression and prolonged survival, and in some cases complete tumor clearance. Based on preclinical results, clinical trials have been conducted to establish the safety of RNA virus delivery. Moreover, stem cell-based lentiviral therapy provided life-long production of factor VIII potentially generating a cure for hemophilia A. Several clinical trials on cancer patients have generated anti-tumor activity, prolonged survival, and even progression-free survival.
Collapse
|
28
|
Skinner PJ. Targeting reservoirs of HIV replication in lymphoid follicles with cellular therapies to cure HIV. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/acg2.27] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Pamela J. Skinner
- Microbiology Research Facility; Veterinary and Biomedical Sciences Department; University of Minnesota; Minneapolis Minnesota
| |
Collapse
|
29
|
Lundstrom K. Viral Vectors in Gene Therapy. Diseases 2018; 6:diseases6020042. [PMID: 29883422 PMCID: PMC6023384 DOI: 10.3390/diseases6020042] [Citation(s) in RCA: 310] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 01/02/2023] Open
Abstract
Applications of viral vectors have found an encouraging new beginning in gene therapy in recent years. Significant improvements in vector engineering, delivery, and safety have placed viral vector-based therapy at the forefront of modern medicine. Viral vectors have been employed for the treatment of various diseases such as metabolic, cardiovascular, muscular, hematologic, ophthalmologic, and infectious diseases and different types of cancer. Recent development in the area of immunotherapy has provided both preventive and therapeutic approaches. Furthermore, gene silencing generating a reversible effect has become an interesting alternative, and is well-suited for delivery by viral vectors. A number of preclinical studies have demonstrated therapeutic and prophylactic efficacy in animal models and furthermore in clinical trials. Several viral vector-based drugs have also been globally approved.
Collapse
|
30
|
Carrillo MA, Zhen A, Kitchen SG. The Use of the Humanized Mouse Model in Gene Therapy and Immunotherapy for HIV and Cancer. Front Immunol 2018; 9:746. [PMID: 29755454 PMCID: PMC5932400 DOI: 10.3389/fimmu.2018.00746] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 03/26/2018] [Indexed: 12/31/2022] Open
Abstract
HIV and cancer remain prevailing sources of morbidity and mortality worldwide. There are current efforts to discover novel therapeutic strategies for the treatment or cure of these diseases. Humanized mouse models provide the investigative tool to study the interaction between HIV or cancer and the human immune system in vivo. These humanized models consist of immunodeficient mice transplanted with human cells, tissues, or hematopoietic stem cells that result in reconstitution with a nearly full human immune system. In this review, we discuss preclinical studies evaluating therapeutic approaches in stem cell-based gene therapy and T cell-based immunotherapies for HIV and cancer using a humanized mouse model and some recent advances in using checkpoint inhibitors to improve antiviral or antitumor responses.
Collapse
Affiliation(s)
- Mayra A Carrillo
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA, United States
| | - Anjie Zhen
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA, United States
| | - Scott G Kitchen
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
31
|
Dorner M, de Jong YP. Modeling Unique Patients in Humanized Mice: Toward a Curative Strategy for HIV. Mol Ther 2018; 26:329-330. [DOI: 10.1016/j.ymthe.2018.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
32
|
Scarborough RJ, Gatignol A. RNA Interference Therapies for an HIV-1 Functional Cure. Viruses 2017; 10:E8. [PMID: 29280961 PMCID: PMC5795421 DOI: 10.3390/v10010008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 12/31/2022] Open
Abstract
HIV-1 drug therapies can prevent disease progression but cannot eliminate HIV-1 viruses from an infected individual. While there is hope that elimination of HIV-1 can be achieved, several approaches to reach a functional cure (control of HIV-1 replication in the absence of drug therapy) are also under investigation. One of these approaches is the transplant of HIV-1 resistant cells expressing anti-HIV-1 RNAs, proteins or peptides. Small RNAs that use RNA interference pathways to target HIV-1 replication have emerged as competitive candidates for cell transplant therapy and have been included in all gene combinations that have so far entered clinical trials. Here, we review RNA interference pathways in mammalian cells and the design of therapeutic small RNAs that use these pathways to target pathogenic RNA sequences. Studies that have been performed to identify anti-HIV-1 RNA interference therapeutics are also reviewed and perspectives on their use in combination gene therapy to functionally cure HIV-1 infection are provided.
Collapse
Affiliation(s)
- Robert J Scarborough
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada.
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A0G4, Canada.
| | - Anne Gatignol
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada.
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A0G4, Canada.
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC H3A0G4, Canada.
| |
Collapse
|
33
|
Khamaikawin W, Shimizu S, Kamata M, Cortado R, Jung Y, Lam J, Wen J, Kim P, Xie Y, Kim S, Arokium H, Presson AP, Chen ISY, An DS. Modeling Anti-HIV-1 HSPC-Based Gene Therapy in Humanized Mice Previously Infected with HIV-1. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 9:23-32. [PMID: 29322065 PMCID: PMC5751878 DOI: 10.1016/j.omtm.2017.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 11/26/2017] [Indexed: 01/09/2023]
Abstract
Investigations of anti-HIV-1 human hematopoietic stem/progenitor cell (HSPC)-based gene therapy have been performed by HIV-1 challenge after the engraftment of gene-modified HSPCs in humanized mouse models. However, the clinical application of gene therapy is to treat HIV-1-infected patients. Here, we developed a new method to investigate an anti-HIV-1 HSPC-based gene therapy in humanized mice previously infected with HIV-1. First, humanized mice were infected with HIV-1. When plasma viremia reached >107 copies/mL 3 weeks after HIV-1 infection, the mice were myeloablated with busulfan and transplanted with anti-HIV-1 gene-modified CD34+ HSPCs transduced with a lentiviral vector expressing two short hairpin RNAs (shRNAs) against CCR5 and HIV-1 long terminal repeat (LTR), along with human thymus tissue under the kidney capsule. Anti-HIV-1 vector-modified human CD34+ HSPCs successfully repopulated peripheral blood and lymphoid tissues in HIV-1 previously infected humanized mice. Anti-HIV-1 shRNA vector-modified CD4+ T lymphocytes showed selective advantage in HIV-1 previously infected humanized mice. This new method will be useful for investigations of anti-HIV-1 gene therapy when testing in a more clinically relevant experimental setting.
Collapse
Affiliation(s)
- Wannisa Khamaikawin
- School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Saki Shimizu
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.,School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Masakazu Kamata
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ruth Cortado
- School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Yujin Jung
- School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Jennifer Lam
- School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Patrick Kim
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.,School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Yiming Xie
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Sanggu Kim
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Hubert Arokium
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Angela P Presson
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Division of Epidemiology, University of Utah, Salt Lake City, UT 84132, USA
| | - Irvin S Y Chen
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| | - Dong Sung An
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.,School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA.,UCLA AIDS Institute, Los Angeles, CA 90095, USA
| |
Collapse
|
34
|
New approaches for the enhancement of chimeric antigen receptors for the treatment of HIV. Transl Res 2017; 187:83-92. [PMID: 28755872 DOI: 10.1016/j.trsl.2017.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/30/2017] [Accepted: 07/08/2017] [Indexed: 12/20/2022]
Abstract
HIV infection continues to be a life-long chronic disease in spite of the success of antiretroviral therapy (ART) in controlling viral replication and preventing disease progression. However, because of the high cost of treatment, severe side effects, and inefficiency in curing the disease with ART, there is a call for alternative therapies that will provide a functional cure for HIV. Cytotoxic T lymphocytes (CTLs) are vital in the control and clearance of viral infections and therefore immune-based therapies have attempted to engineer HIV-specific CTLs that would be able to clear the infection from the body. The development of chimeric antigen receptors (CARs) provides an opportunity to engineer superior HIV-specific CTLs that will be independent of the major histocompatibility complex for target recognition. A CD4-based CAR has been previously tested in clinical trials to test the antiviral efficacy of peripheral T cells armed with this CD4-based CAR. The results from these clinical trials showed the safety and feasibility of CAR T cell therapy for HIV infection; however, minimal antiviral efficacy was seen. In this review, we will discuss the various strategies being developed to enhance the therapeutic potency of anti-HIV CARs with the goal of generating superior antiviral responses that will lead to life-long HIV immunity and clearance of the virus from the body.
Collapse
|
35
|
Zhen A, Carrillo MA, Kitchen SG. Chimeric antigen receptor engineered stem cells: a novel HIV therapy. Immunotherapy 2017; 9:401-410. [PMID: 28357916 DOI: 10.2217/imt-2016-0121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite the success of combination antiretroviral therapy (cART) for suppressing HIV and improving patients' quality of life, HIV persists in cART-treated patients and remains an incurable disease. Financial burdens and health consequences of lifelong cART treatment call for novel HIV therapies that result in a permanent cure. Cellular immunity is central in controlling HIV replication. However, HIV adopts numerous strategies to evade immune surveillance. Engineered immunity via genetic manipulation could offer a functional cure by generating cells that have enhanced antiviral activity and are resistant to HIV infection. Recently, encouraging reports from several human clinical trials using an anti-CD19 chimeric antigen receptor (CAR) modified T-cell therapy for treating B-cell malignancies have provided valuable insights and generated remarkable enthusiasm in engineered T-cell therapy. In this review, we discuss the development of HIV-specific chimeric antigen receptors and the use of stem cell based therapies to generate lifelong anti-HIV immunity.
Collapse
Affiliation(s)
- Anjie Zhen
- Division of Hematology & Oncology, Department of Medicine; UCLA AIDS Institute, David Geffen School of Medicine University of California, Los Angeles, CA 90095, USA
| | - Mayra A Carrillo
- Division of Hematology & Oncology, Department of Medicine; UCLA AIDS Institute, David Geffen School of Medicine University of California, Los Angeles, CA 90095, USA
| | - Scott G Kitchen
- Division of Hematology & Oncology, Department of Medicine; UCLA AIDS Institute, David Geffen School of Medicine University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
36
|
Crawford LB, Tempel R, Streblow DN, Kreklywich C, Smith P, Picker LJ, Nelson JA, Caposio P. Human Cytomegalovirus Induces Cellular and Humoral Virus-specific Immune Responses in Humanized BLT Mice. Sci Rep 2017; 7:937. [PMID: 28428537 PMCID: PMC5430540 DOI: 10.1038/s41598-017-01051-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/20/2017] [Indexed: 12/22/2022] Open
Abstract
The strict species specificity of Human Cytomegalovirus (HCMV) has impeded our understanding of antiviral adaptive immune responses in the context of a human immune system. We have previously shown that HCMV infection of human hematopoietic progenitor cells engrafted in immune deficient mice (huNSG) results in viral latency that can be reactivated following G-CSF treatment. In this study, we characterized the functional human adaptive immune responses in HCMV latently-infected huBLT (humanized Bone marrow-Liver-Thymus) mice. Following infection, huBLT mice generate human effector and central memory CD4+ and CD8+ T-cell responses reactive to peptides corresponding to both IE and pp65 proteins. Additionally, both HCMV specific IgM and IgG B-cell responses with the ability to neutralize virus were detected. These results indicate that the HCMV huBLT mouse model may provide a valuable tool to study viral latency and reactivation as well as evaluate HCMV vaccines and immune responses in the context of a functional human immune system.
Collapse
Affiliation(s)
- Lindsey B Crawford
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Rebecca Tempel
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Craig Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Patricia Smith
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Jay A Nelson
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA.
| |
Collapse
|
37
|
Symonds G, Bartlett JS, Kiem HP, Tsie M, Breton L. Cell-Delivered Entry Inhibitors for HIV-1: CCR5 Downregulation and Blocking Virus/Membrane Fusion in Defending the Host Cell Population. AIDS Patient Care STDS 2016; 30:545-550. [PMID: 27905841 DOI: 10.1089/apc.2016.0245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
HIV-1 infection requires the presence of the CD4 receptor on the target cell surface and a coreceptor, predominantly CC-chemokine receptor 5 (CCR5). It has been shown that individuals who are homozygous for a defective CCR5 gene are protected from HIV-1 infection. A novel self-inactivating lentiviral vector LVsh5/C46 (Cal-1) has been engineered to block HIV-1 infection with two viral entry inhibitors, conferring resistance to HIV-1 infection from both CCR5 and CXCR4 tropic strains. Cal-1 encodes a short hairpin RNA (sh5) to downregulate CCR5 and C46, an HIV-1 fusion inhibitor. Gene therapy by Cal-1 is aimed at transducing CD4+ T cells and CD34+ hematopoietic stem/progenitor cells in an autologous transplant setting. Pre-clinical safety and efficacy studies in vitro and in vivo (humanized mouse model and nonhuman primates) have shown that Cal-1 is safe with no indication of any toxicity risk and acts to decrease viral load and increase CD4 counts. Two clinical trials are underway using Cal-1: a phase I/II study to assess safety and feasibility in an adult HIV-1-positive population not on antiretroviral therapy (ART); and a second Fred Hutchinson Investigator Initiated phase I study to assess safety and feasibility in adults with HIV-1-associated non-Hodgkin or Hodgkin lymphoma.
Collapse
|
38
|
Swamy MN, Wu H, Shankar P. Recent advances in RNAi-based strategies for therapy and prevention of HIV-1/AIDS. Adv Drug Deliv Rev 2016; 103:174-186. [PMID: 27013255 PMCID: PMC4935623 DOI: 10.1016/j.addr.2016.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/15/2022]
Abstract
RNA interference (RNAi) provides a powerful tool to silence specific gene expression and has been widely used to suppress host factors such as CCR5 and/or viral genes involved in HIV-1 replication. Newer nuclease-based gene-editing technologies, such as zinc finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN) and the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system, also provide powerful tools to ablate specific genes. Because of differences in co-receptor usage and the high mutability of the HIV-1 genome, a combination of host factors and viral genes needs to be suppressed for effective prevention and treatment of HIV-1 infection. Whereas the continued presence of small interfering/short hairpin RNA (si/shRNA) mediators is needed for RNAi to be effective, the continued expression of nucleases in the gene-editing systems is undesirable. Thus, RNAi provides the only practical way for expression of multiple silencers in infected and uninfected cells, which is needed for effective prevention/treatment of infection. There have been several advances in the RNAi field in terms of si/shRNA design, targeted delivery to HIV-1 susceptible cells, and testing for efficacy in preclinical humanized mouse models. Here, we comprehensively review the latest advances in RNAi technology towards prevention and treatment of HIV-1.
Collapse
Affiliation(s)
- Manjunath N Swamy
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA.
| | - Haoquan Wu
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Premlata Shankar
- Center of Emphasis in Infectious Disease, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA.
| |
Collapse
|
39
|
Pernet O, Yadav SS, An DS. Stem cell-based therapies for HIV/AIDS. Adv Drug Deliv Rev 2016; 103:187-201. [PMID: 27151309 PMCID: PMC4935568 DOI: 10.1016/j.addr.2016.04.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 04/21/2016] [Accepted: 04/25/2016] [Indexed: 12/26/2022]
Abstract
One of the current focuses in HIV/AIDS research is to develop a novel therapeutic strategy that can provide a life-long remission of HIV/AIDS without daily drug treatment and, ultimately, a cure for HIV/AIDS. Hematopoietic stem cell-based anti-HIV gene therapy aims to reconstitute the patient immune system by transplantation of genetically engineered hematopoietic stem cells with anti-HIV genes. Hematopoietic stem cells can self-renew, proliferate and differentiate into mature immune cells. In theory, anti-HIV gene-modified hematopoietic stem cells can continuously provide HIV-resistant immune cells throughout the life of a patient. Therefore, hematopoietic stem cell-based anti-HIV gene therapy has a great potential to provide a life-long remission of HIV/AIDS by a single treatment. Here, we provide a comprehensive review of the recent progress of developing anti-HIV genes, genetic modification of hematopoietic stem progenitor cells, engraftment and reconstitution of anti-HIV gene-modified immune cells, HIV inhibition in in vitro and in vivo animal models, and in human clinical trials.
Collapse
Affiliation(s)
- Olivier Pernet
- School of Nursing, University of California Los Angeles, 188 BSRB, 615 Charles E. Young Dr. South, Los Angeles, CA 90095, USA; UCLA AIDS Institute, 188 BSRB, 615 Charles E. Young Dr. South, Los Angeles, CA 90095, USA.
| | - Swati Seth Yadav
- School of Nursing, University of California Los Angeles, 188 BSRB, 615 Charles E. Young Dr. South, Los Angeles, CA 90095, USA; UCLA AIDS Institute, 188 BSRB, 615 Charles E. Young Dr. South, Los Angeles, CA 90095, USA.
| | - Dong Sung An
- School of Nursing, University of California Los Angeles, 188 BSRB, 615 Charles E. Young Dr. South, Los Angeles, CA 90095, USA; UCLA AIDS Institute, 188 BSRB, 615 Charles E. Young Dr. South, Los Angeles, CA 90095, USA; Hematology-Oncology, The Department of Medicine, David Geffen School of Medicine at UCLA, 188 BSRB, 615 Charles E. Young Dr. South, Los Angeles, CA 90095, USA.
| |
Collapse
|
40
|
DiGiusto DL. Stem cell gene therapy for HIV: strategies to inhibit viral entry and replication. Curr HIV/AIDS Rep 2016; 12:79-87. [PMID: 25578054 DOI: 10.1007/s11904-014-0242-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Since the demonstration of a cure of an HIV+ patient with an allogeneic stem cell transplant using naturally HIV-resistant cells, significant interest in creating similar autologous products has fueled the development of a variety of "cell engineering" approaches to stem cell therapy for HIV. Among the more well-studied strategies is the inhibition of viral entry through disruption of expression of viral co-receptors or through competitive inhibitors of viral fusion with the cell membrane. Preclinical evaluation of these approaches often starts in vitro but ultimately is tested in animal models prior to clinical implementation. In this review, we trace the development of several key approaches (meganucleases, short hairpin RNA (shRNA), and fusion inhibitors) to modification of hematopoietic stem cells designed to impart resistance to HIV to their T-cell and monocytic progeny. The basic evolution of technologies through in vitro and in vivo testing is discussed as well as the pros and cons of each approach and how the addition of postentry inhibitors may enhance the overall antiviral efficacy of these approaches.
Collapse
Affiliation(s)
- David L DiGiusto
- Department of Stem Cell and Cell Therapeutic Operations, Stanford Hospital and Clinics, 300 Pasteur Drive, Stanford, CA, 94305, USA,
| |
Collapse
|
41
|
Llewellyn GN, Exline CM, Holt N, Cannon PM. Using Engineered Nucleases to Create HIV-Resistant Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016. [DOI: 10.1007/978-1-4939-3509-3_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
42
|
Khamaikawin W, Saoin S, Nangola S, Chupradit K, Sakkhachornphop S, Hadpech S, Onlamoon N, Ansari AA, Byrareddy SN, Boulanger P, Hong SS, Torbett BE, Tayapiwatana C. Combined Antiviral Therapy Using Designed Molecular Scaffolds Targeting Two Distinct Viral Functions, HIV-1 Genome Integration and Capsid Assembly. MOLECULAR THERAPY-NUCLEIC ACIDS 2015; 4:e249. [PMID: 26305555 PMCID: PMC4560793 DOI: 10.1038/mtna.2015.22] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/14/2015] [Indexed: 01/06/2023]
Abstract
Designed molecular scaffolds have been proposed as alternative therapeutic agents against HIV-1. The ankyrin repeat protein (Ank(GAG)1D4) and the zinc finger protein (2LTRZFP) have recently been characterized as intracellular antivirals, but these molecules, used individually, do not completely block HIV-1 replication and propagation. The capsid-binder Ank(GAG)1D4, which inhibits HIV-1 assembly, does not prevent the genome integration of newly incoming viruses. 2LTRZFP, designed to target the 2-LTR-circle junction of HIV-1 cDNA and block HIV-1 integration, would have no antiviral effect on HIV-1-infected cells. However, simultaneous expression of these two molecules should combine the advantage of preventive and curative treatments. To test this hypothesis, the genes encoding the N-myristoylated Myr(+)Ank(GAG)1D4 protein and the 2LTRZFP were introduced into human T-cells, using a third-generation lentiviral vector. SupT1 cells stably expressing 2LTRZFP alone or with Myr(+)Ank(GAG)1D4 showed a complete resistance to HIV-1 in viral challenge. Administration of the Myr(+)Ank(GAG)1D4 vector to HIV-1-preinfected SupT1 cells resulted in a significant antiviral effect. Resistance to viral infection was also observed in primary human CD4+ T-cells stably expressing Myr(+)Ank(GAG)1D4, and challenged with HIV-1, SIVmac, or SHIV. Our data suggest that our two anti-HIV-1 molecular scaffold prototypes are promising antiviral agents for anti-HIV-1 gene therapy.
Collapse
Affiliation(s)
- Wannisa Khamaikawin
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Somphot Saoin
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sawitree Nangola
- Division of Clinical Immunology and Transfusion Sciences, School of Allied Health Sciences, University of Phayao, Phayao, Thailand
| | - Koollawat Chupradit
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | | | - Sudarat Hadpech
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nattawat Onlamoon
- Division of Instruments for Research, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Aftab A Ansari
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Siddappa N Byrareddy
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Pierre Boulanger
- University Lyon 1 & INRA UMR-754, Retrovirus and Comparative Pathology, Lyon, France
| | - Saw-See Hong
- University Lyon 1 & INRA UMR-754, Retrovirus and Comparative Pathology, Lyon, France
| | - Bruce E Torbett
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Chatchai Tayapiwatana
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
43
|
Hematopoietic Stem and Immune Cells in Chronic HIV Infection. Stem Cells Int 2015; 2015:148064. [PMID: 26300920 PMCID: PMC4537765 DOI: 10.1155/2015/148064] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 07/15/2015] [Accepted: 07/21/2015] [Indexed: 02/07/2023] Open
Abstract
Hematopoietic stem cell (HSC) belongs to multipotent adult somatic stem cells. A single HSC can reconstitute the entire blood system via self-renewal, differentiation into all lineages of blood cells, and replenishment of cells lost due to attrition or disease in a person's lifetime. Although all blood and immune cells derive from HSC, immune cells, specifically immune memory cells, have the properties of HSC on self-renewal and differentiation into lineage effector cells responding to the invading pathogens. Moreover, the interplay between immune memory cell and viral pathogen determines the course of a viral infection. Here, we state our point of view on the role of blood stem and progenitor cell in chronic HIV infection, with a focus on memory CD4 T-cell in the context of HIV/AIDS eradication and cure.
Collapse
|
44
|
Abstract
During the past decade, the development of humanized mouse models and their general applications in biomedical research greatly accelerated the translation of outcomes obtained from basic research into potential diagnostic and therapeutic strategies in clinic. In this chapter, we firstly present an overview on the history and current progress of diverse humanized mouse models and then focus on those equipped with reconstituted human immune system. The update advancement in the establishment of humanized immune system mice and their applications in the studies of the development of human immune system and the pathogenesis of multiple human immune-related diseases are intensively reviewed here, while the shortcoming and perspective of these potent tools are discussed as well. As a valuable bridge across the gap between bench work and clinical trial, progressive humanized mouse models will undoubtedly continue to play an indispensable role in the wide area of biomedical research.
Collapse
|
45
|
Dey R, Pillai B. Cell-based gene therapy against HIV. Gene Ther 2015; 22:851-5. [PMID: 26079406 DOI: 10.1038/gt.2015.58] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/18/2015] [Accepted: 06/05/2015] [Indexed: 11/09/2022]
Abstract
The ability to integrate inside the host genome lays a strong foundation for HIV to play hide and seek with the host's immune surveillance mechanisms. Present anti-viral therapies, although successful in suppressing the virus to a certain level, fail to wipe it out completely. However, recent approaches in modifying stem cells and enabling them to give rise to potent/resistant T-cells against HIV holds immense hope for eradication of the virus from the host. In this review, we will briefly discuss previous landmark studies on engineering stem cells or T-cells that have been explored for therapeutic efficacy against HIV. We will also analyze potential benefits and pitfalls of some studies done recently and will share our opinion on emerging trends.
Collapse
Affiliation(s)
- R Dey
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - B Pillai
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| |
Collapse
|
46
|
Karpel ME, Boutwell CL, Allen TM. BLT humanized mice as a small animal model of HIV infection. Curr Opin Virol 2015; 13:75-80. [PMID: 26083316 DOI: 10.1016/j.coviro.2015.05.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 11/25/2022]
Abstract
Humanized mice are valuable models for the research and development of vaccine strategies and therapeutic interventions to control or eradicate HIV. The BLT humanized mouse model is particularly promising because the combination of transplantation of human fetal pluripotent hematopoietic stem cells with surgical engraftment of human fetal thymic tissue results in improved T cell reconstitution, maturation, and selection. To date, the BLT humanized mouse model has been used to study many aspects of HIV infection including prevention, mucosal transmission, HIV-specific innate and adaptive immunity, viral latency, and novel antiretroviral and immune-based therapies for suppression and reservoir eradication. Here we describe recent advances and applications of the BLT humanized mouse model of HIV infection and discuss opportunities to further improve this valuable small animal model.
Collapse
Affiliation(s)
- Marshall E Karpel
- Ragon Institute of MGH, MIT and Harvard, Cambridge , MA, United States
| | | | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge , MA, United States.
| |
Collapse
|
47
|
HIV-specific Immunity Derived From Chimeric Antigen Receptor-engineered Stem Cells. Mol Ther 2015; 23:1358-1367. [PMID: 26050990 DOI: 10.1038/mt.2015.102] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 05/25/2015] [Indexed: 12/17/2022] Open
Abstract
The human immunodeficiency virus (HIV)-specific cytotoxic T lymphocyte (CTL) response is critical in controlling HIV infection. Since the immune response does not eliminate HIV, it would be beneficial to develop ways to enhance the HIV-specific CTL response to allow long-term viral suppression or clearance. Here, we report the use of a protective chimeric antigen receptor (CAR) in a hematopoietic stem/progenitor cell (HSPC)-based approach to engineer HIV immunity. We determined that CAR-modified HSPCs differentiate into functional T cells as well as natural killer (NK) cells in vivo in humanized mice and these cells are resistant to HIV infection and suppress HIV replication. These results strongly suggest that stem cell-based gene therapy with a CAR may be feasible and effective in treating chronic HIV infection and other morbidities.
Collapse
|
48
|
Kamata M, Kim PY, Ng HL, Ringpis GEE, Kranz E, Chan J, O'Connor S, Yang OO, Chen ISY. Ectopic expression of anti-HIV-1 shRNAs protects CD8(+) T cells modified with CD4ζ CAR from HIV-1 infection and alleviates impairment of cell proliferation. Biochem Biophys Res Commun 2015; 463:216-21. [PMID: 25998390 DOI: 10.1016/j.bbrc.2015.05.026] [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] [Received: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 12/31/2022]
Abstract
Chimeric antigen receptors (CARs) are artificially engineered receptors that confer a desired specificity to immune effector T cells. As an HIV-1-specific CAR, CD4ζ CAR has been extensively tested in vitro as well as in clinical trials. T cells modified with this CAR mediated highly potent anti-HIV-1 activities in vitro and were well-tolerated in vivo, but exerted limited effects on viral load and reservoir size due to poor survival and/or functionality of the transduced cells in patients. We hypothesize that ectopic expression of CD4ζ on CD8(+) T cells renders them susceptible to HIV-1 infection, resulting in poor survival of those cells. To test this possibility, highly purified CD8(+) T cells were genetically modified with a CD4ζ-encoding lentiviral vector and infected with HIV-1. CD8(+) T cells were vulnerable to HIV-1 infection upon expression of CD4ζ as evidenced by elevated levels of p24(Gag) in cells and culture supernatants. Concurrently, the number of CD4ζ-modified CD8(+) T cells was reduced relative to control cells upon HIV-1 infection. To protect these cells from HIV-1 infection, we co-expressed two anti-HIV-1 shRNAs previously developed by our group together with CD4ζ. This combination vector was able to suppress HIV-1 infection without impairing HIV-1-dependent effector activities of CD4ζ. In addition, the number of CD4ζ-modified CD8(+) T cells maintained similar levels to that of the control even under HIV-1 infection. These results suggest that protecting CD4ζ-modified CD8(+) T cells from HIV-1 infection is required for prolonged HIV-1-specific immune surveillance.
Collapse
Affiliation(s)
- Masakazu Kamata
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Patrick Y Kim
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hwee L Ng
- Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Gene-Errol E Ringpis
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Emiko Kranz
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joshua Chan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sean O'Connor
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Otto O Yang
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; UCLA AIDS Institute, Los Angeles, CA, USA; AIDS Healthcare Foundation, Los Angeles, CA, USA
| | - Irvin S Y Chen
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; UCLA AIDS Institute, Los Angeles, CA, USA
| |
Collapse
|
49
|
Lentivector Knockdown of CCR5 in Hematopoietic Stem and Progenitor Cells Confers Functional and Persistent HIV-1 Resistance in Humanized Mice. J Virol 2015; 89:6761-72. [PMID: 25903342 DOI: 10.1128/jvi.00277-15] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/30/2015] [Indexed: 01/04/2023] Open
Abstract
UNLABELLED Gene-engineered CD34(+) hematopoietic stem and progenitor cells (HSPCs) can be used to generate an HIV-1-resistant immune system. However, a certain threshold of transduced HSPCs might be required for transplantation into mice for creating an HIV-resistant immune system. In this study, we combined CCR5 knockdown by a highly efficient microRNA (miRNA) lentivector with pretransplantation selection of transduced HSPCs to obtain a rather pure population of gene engineered CD34(+) cells. Low-level transduction of HSPCs and subsequent sorting by flow cytometry yielded >70% transduced cells. Mice transplanted with these cells showed functional and persistent resistance to a CCR5-tropic HIV strain: viral load was significantly decreased over months, and human CD4(+) T cells were preserved. In one mouse, viral mutations, resulting presumably in a CXCR4-tropic strain, overcame HIV resistance. Our results suggest that HSPC-based CCR5 knockdown may lead to efficient control of HIV in vivo. We overcame a major limitation of previous HIV gene therapy in humanized mice in which only a proportion of the cells in chimeric mice in vivo are anti-HIV engineered. Our strategy underlines the promising future of gene engineering HIV-resistant CD34(+) cells that produce a constant supply of HIV-resistant progeny. IMPORTANCE Major issues in experimental long-term in vivo HIV gene therapy have been (i) low efficacy of cell transduction at the time of transplantation and (ii) transduction resulting in multiple copies of heterologous DNA in target cells. In this study, we demonstrated the efficacy of a transplantation approach with a selection step for transduced cells that allows transplantation of an enriched population of HSPCs expressing a single (low) copy of a CCR5 miRNA. Efficient maintenance of CD4(+) T cells and a low viral titer resulted only when at least 70% of the HIV target cells were genetically modified. These findings imply that clinical protocols of HIV gene therapy require a selective enrichment of genetically targeted cells because positive selection of modified cells is likely to be insufficient below this threshold. This selection approach may be beneficial not only for HIV patients but also for other patients requiring transplantation of genetically modified cells.
Collapse
|
50
|
Shimizu S, Ringpis GE, Marsden MD, Cortado RV, Wilhalme HM, Elashoff D, Zack JA, Chen ISY, An DS. RNAi-Mediated CCR5 Knockdown Provides HIV-1 Resistance to Memory T Cells in Humanized BLT Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e227. [PMID: 25689223 PMCID: PMC4345313 DOI: 10.1038/mtna.2015.3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 01/06/2015] [Indexed: 11/20/2022]
Abstract
Transplantation of hematopoietic stem/progenitor cells (HSPC) modified with a lentiviral vector bearing a potent nontoxic short hairpin RNA (sh1005) directed to the HIV coreceptor CCR5 is capable of continuously producing CCR5 downregulated CD4+ T lymphocytes. Here, we characterized HIV-1 resistance of the sh1005-modified CD4+ T lymphocytes in vivo in humanized bone marrow/liver/thymus (hu BLT) mice. The sh1005-modified CD4+ T lymphocytes were positively selected in CCR5-tropic HIV-1-challenged mice. The sh1005-modified memory CD4+ T lymphocytes (the primary target of CCR5-tropic HIV-1) expressing sh1005 were maintained in lymphoid tissues in CCR5-tropic HIV-1-challenged mice. Frequencies of HIV-1 p24 expressing cells were significantly reduced in the sh1005-modified splenocytes by ex vivo cell stimulation confirming that CCR5 downregulated sh1005 modified cells are protected from viral infection. These results demonstrate that stable CCR5 downregulation through genetic modification of human HSPC by lentivirally delivered sh1005 is highly effective in providing HIV-1 resistance. Our results provide in vivo evidence in a relevant small animal model that sh1005 is a potent early-step anti-HIV reagent that has potential as a novel anti-HIV-1 HSPC gene therapeutic reagent for human applications.
Collapse
Affiliation(s)
- Saki Shimizu
- Hematology-Oncology, the Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- School of Nursing, University of California, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Gene-Errol Ringpis
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Matthew D Marsden
- Hematology-Oncology, the Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Ruth V Cortado
- School of Nursing, University of California, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Holly M Wilhalme
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - David Elashoff
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jerome A Zack
- Hematology-Oncology, the Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Irvin S Y Chen
- Hematology-Oncology, the Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Dong Sung An
- Hematology-Oncology, the Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- School of Nursing, University of California, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| |
Collapse
|