1
|
Zhang SH, Peng LL, Chen YF, Xu Y, Moradi V. Focusing on exosomes to overcome the existing bottlenecks of CAR-T cell therapy. Inflamm Regen 2024; 44:45. [PMID: 39490997 PMCID: PMC11533312 DOI: 10.1186/s41232-024-00358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Since chimeric antigen receptor T (CAR-T) cells were introduced three decades ago, the treatment using these cells has led to outstanding outcomes, and at the moment, CAR-T cell therapy is a well-established mainstay for treating CD19 + malignancies and multiple myeloma. Despite the astonishing results of CAR-T cell therapy in B-cell-derived malignancies, several bottlenecks must be overcome to promote its safety and efficacy and broaden its applicability. These bottlenecks include cumbersome production process, safety concerns of viral vectors, poor efficacy in treating solid tumors, life-threatening side effects, and dysfunctionality of infused CAR-T cells over time. Exosomes are nano-sized vesicles that are secreted by all living cells and play an essential role in cellular crosstalk by bridging between cells. In this review, we discuss how the existing bottlenecks of CAR-T cell therapy can be overcome by focusing on exosomes. First, we delve into the effect of tumor-derived exosomes on the CAR-T cell function and discuss how inhibiting their secretion can enhance the efficacy of CAR-T cell therapy. Afterward, the application of exosomes to the manufacturing of CAR-T cells in a non-viral approach is discussed. We also review the latest advancements in ex vivo activation and cultivation of CAR-T cells using exosomes, as well as the potential of engineered exosomes to in vivo induction or boost the in vivo proliferation of CAR-T cells. Finally, we discuss how CAR-engineered exosomes can be used as a versatile tool for the direct killing of tumor cells or delivering intended therapeutic payloads in a targeted manner.
Collapse
Affiliation(s)
- Si-Heng Zhang
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China
| | - Ling-Long Peng
- Wuhu Hospital, East China Normal University (The Second People's Hospital of Wuhu), Wuhu, 241000, China
| | - Yi-Fei Chen
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Yan Xu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China.
| | - Vahid Moradi
- Hematology and Bood Transfusion Science Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Zenere G, Wu C, Midkiff CC, Johnson NM, Grice CP, Wimley WC, Kaur A, Braun SE. Extracellular domain, hinge, and transmembrane determinants affecting surface CD4 expression of a novel anti-HIV chimeric antigen receptor (CAR) construct. PLoS One 2024; 19:e0293990. [PMID: 39133676 PMCID: PMC11318886 DOI: 10.1371/journal.pone.0293990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 07/03/2024] [Indexed: 08/15/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cells have demonstrated clinical potential, but current receptors still need improvements to be successful against chronic HIV infection. In this study, we address some requirements of CAR motifs for strong surface expression of a novel anti-HIV CAR by evaluating important elements in the extracellular, hinge, and transmembrane (TM) domains. When combining a truncated CD4 extracellular domain and CD8α hinge/TM, the novel CAR did not express extracellularly but was detectable intracellularly. By shortening the CD8α hinge, CD4-CAR surface expression was partially recovered and addition of the LYC motif at the end of the CD8α TM fully recovered both intracellular and extracellular CAR expression. Mutation of LYC to TTA or TTC showed severe abrogation of CAR expression by flow cytometry and confocal microscopy. Additionally, we determined that CD4-CAR surface expression could be maximized by the removal of FQKAS motif at the junction of the extracellular domain and the hinge region. CD4-CAR surface expression also resulted in cytotoxic CAR T cell killing of HIV Env+ target cells. In this study, we identified elements that are crucial for optimal CAR surface expression, highlighting the need for structural analysis studies to establish fundamental guidelines of CAR designs.
Collapse
Affiliation(s)
- Giorgio Zenere
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Chengxiang Wu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Cecily C. Midkiff
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
| | - Nathan M. Johnson
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Christopher P. Grice
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- BioMedical Sciences Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - William C. Wimley
- Department of BioChemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Stephen E. Braun
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, Louisiana, United States of America
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
3
|
Su H, Mueller A, Goldstein H. Recent advances on anti-HIV chimeric antigen receptor-T-cell treatment to provide sustained HIV remission. Curr Opin HIV AIDS 2024; 19:169-178. [PMID: 38695148 DOI: 10.1097/coh.0000000000000858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
PURPOSE OF REVIEW Successful sustained remission of HIV infection has been achieved after CCR5Δ32/Δ32 allogeneic hematopoietic stem cell transplantation for treatment of leukemia in a small cohort of people living with HIV (PLWH). This breakthrough demonstrated that the goal of curing HIV was achievable. However, the high morbidity and mortality associated with bone marrow transplantation limits the routine application of this approach and provides a strong rationale for pursuing alternative strategies for sustained long-term antiretroviral therapy (ART)-free HIV remission. Notably, long-term immune-mediated control of HIV replication observed in elite controllers and posttreatment controllers suggests that potent HIV-specific immune responses could provide sustained ART-free remission in PLWH. The capacity of chimeric antigen receptor (CAR)-T cells engineered to target malignant cells to induce remission and cure in cancer patients made this an attractive approach to provide PLWH with a potent HIV-specific immune response. Here, we review the recent advances in the design and application of anti-HIV CAR-T-cell therapy to provide a functional HIV cure. RECENT FINDINGS HIV reservoirs are established days after infection and persist through clonal expansion of infected cells. The continuous interaction between latently infected cells and the immune system shapes the landscape of HIV latency and likely contributes to ART-free viral control in elite controllers. CAR-T cells can exhibit superior antiviral activity as compared with native HIV-specific T cells, particularly because they can be engineered to have multiple HIV specificities, resistance to HIV infection, dual costimulatory signaling, immune checkpoint inhibitors, stem cell derivation, CMV TCR coexpression, and tissue homing ligands. These modifications can significantly improve the capacities of anti-HIV CAR-T cells to prevent viral escape, resist HIV infection, and enhance cytotoxicity, persistence, and tissue penetration. Collectively, these novel modifications of anti-HIV CAR-T cell design have increased their capacity to control HIV infection. SUMMARY Anti-HIV CAR-T cells can be engineered to provide potent and sustained in-vitro and in-vivo antiviral function. The combination of anti-HIV CAR-T cells with other immunotherapeutics may contribute to long-term HIV remission in PLWH.
Collapse
Affiliation(s)
- Hang Su
- Department of Microbiology & Immunology
| | | | - Harris Goldstein
- Department of Microbiology & Immunology
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
4
|
Bui JK, Starke CE, Poole NH, Rust BJ, Jerome KR, Kiem HP, Peterson CW. CD20 CAR T cells safely and reversibly ablate B cell follicles in a non-human primate model of HIV persistence. Mol Ther 2024; 32:1238-1251. [PMID: 38414244 PMCID: PMC11081808 DOI: 10.1016/j.ymthe.2024.02.030] [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: 09/13/2023] [Revised: 01/30/2024] [Accepted: 02/24/2024] [Indexed: 02/29/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapies have demonstrated immense clinical success for B cell and plasma cell malignancies. We tested their impact on the viral reservoir in a macaque model of HIV persistence, comparing the functions of CD20 CAR T cells between animals infected with simian/human immunodeficiency virus (SHIV) and uninfected controls. We focused on the potential of this approach to disrupt B cell follicles (BCFs), exposing infected cells for immune clearance. In SHIV-infected animals, CAR T cells were highly functional, with rapid expansion and trafficking to tissue-associated viral sanctuaries, including BCFs and gut-associated lymphoid tissue (GALT). CD20 CAR T cells potently ablated BCFs and depleted lymph-node-associated follicular helper T (TFH) cells, with complete restoration of BCF architecture and TFH cells following CAR T cell contraction. BCF ablation decreased the splenic SHIV reservoir but was insufficient for effective reductions in systemic viral reservoirs. Although associated with moderate hematologic toxicity, CD20 CAR T cells were well tolerated in SHIV-infected and control animals, supporting the feasibility of this therapy in people living with HIV with underlying B cell malignancies. Our findings highlight the unique ability of CD20 CAR T cells to safely and reversibly unmask TFH cells within BCF sanctuaries, informing future combinatorial HIV cure strategies designed to augment antiviral efficacy.
Collapse
Affiliation(s)
- John K Bui
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Allergy and Infection Diseases, University of Washington, Seattle, WA, USA
| | - Carly E Starke
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nikhita H Poole
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Blake J Rust
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Keith R Jerome
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Allergy and Infection Diseases, University of Washington, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Christopher W Peterson
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
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
|
6
|
Eichholz K, Fukazawa Y, Peterson CW, Haeseleer F, Medina M, Hoffmeister S, Duell DM, Varco-Merth BD, Dross S, Park H, Labriola CS, Axthelm MK, Murnane RD, Smedley JV, Jin L, Gong J, Rust BJ, Fuller DH, Kiem HP, Picker LJ, Okoye AA, Corey L. Anti-PD-1 chimeric antigen receptor T cells efficiently target SIV-infected CD4+ T cells in germinal centers. J Clin Invest 2024; 134:e169309. [PMID: 38557496 PMCID: PMC10977982 DOI: 10.1172/jci169309] [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: 01/31/2023] [Accepted: 02/09/2024] [Indexed: 04/04/2024] Open
Abstract
Programmed cell death protein 1 (PD-1) is an immune checkpoint marker commonly expressed on memory T cells and enriched in latently HIV-infected CD4+ T cells. We engineered an anti-PD-1 chimeric antigen receptor (CAR) to assess the impact of PD-1 depletion on viral reservoirs and rebound dynamics in SIVmac239-infected rhesus macaques (RMs). Adoptive transfer of anti-PD-1 CAR T cells was done in 2 SIV-naive and 4 SIV-infected RMs on antiretroviral therapy (ART). In 3 of 6 RMs, anti-PD-1 CAR T cells expanded and persisted for up to 100 days concomitant with the depletion of PD-1+ memory T cells in blood and tissues, including lymph node CD4+ follicular helper T (TFH) cells. Loss of TFH cells was associated with depletion of detectable SIV RNA from the germinal center (GC). However, following CAR T infusion and ART interruption, there was a marked increase in SIV replication in extrafollicular portions of lymph nodes, a 2-log higher plasma viremia relative to controls, and accelerated disease progression associated with the depletion of CD8+ memory T cells. These data indicate anti-PD-1 CAR T cells depleted PD-1+ T cells, including GC TFH cells, and eradicated SIV from this immunological sanctuary.
Collapse
Affiliation(s)
- Karsten Eichholz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Yoshinori Fukazawa
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Christopher W. Peterson
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and
| | - Francoise Haeseleer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Manuel Medina
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Shelby Hoffmeister
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Derick M. Duell
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Benjamin D. Varco-Merth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Sandra Dross
- Washington National Primate Research Center (WaNPRC), Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Haesun Park
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Caralyn S. Labriola
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Robert D. Murnane
- Washington National Primate Research Center (WaNPRC), Seattle, Washington, USA
| | - Jeremy V. Smedley
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Lei Jin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jiaxin Gong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Blake J. Rust
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Deborah H. Fuller
- Washington National Primate Research Center (WaNPRC), Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Hans-Peter Kiem
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Afam A. Okoye
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center (ONPRC), Oregon Health & Science University, Beaverton, Oregon, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
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
|
8
|
Duggan NN, Dragic T, Chanda SK, Pache L. Breaking the Silence: Regulation of HIV Transcription and Latency on the Road to a Cure. Viruses 2023; 15:2435. [PMID: 38140676 PMCID: PMC10747579 DOI: 10.3390/v15122435] [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: 11/21/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Antiretroviral therapy (ART) has brought the HIV/AIDS epidemic under control, but a curative strategy for viral eradication is still needed. The cessation of ART results in rapid viral rebound from latently infected CD4+ T cells, showing that control of viral replication alone does not fully restore immune function, nor does it eradicate viral reservoirs. With a better understanding of factors and mechanisms that promote viral latency, current approaches are primarily focused on the permanent silencing of latently infected cells ("block and lock") or reactivating HIV-1 gene expression in latently infected cells, in combination with immune restoration strategies to eliminate HIV infected cells from the host ("shock and kill"). In this review, we provide a summary of the current, most promising approaches for HIV-1 cure strategies, including an analysis of both latency-promoting agents (LPA) and latency-reversing agents (LRA) that have shown promise in vitro, ex vivo, and in human clinical trials to reduce the HIV-1 reservoir.
Collapse
Affiliation(s)
- Natasha N. Duggan
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tatjana Dragic
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Sumit K. Chanda
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Lars Pache
- NCI Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
9
|
Sato A, Tsukiyama T, Komeno M, Iwatani C, Tsuchiya H, Kawamoto I, Murase M, Nakagawa T, Itagaki I, Seita Y, Matsumoto S, Nakaya M, Shimizu A, Yamada A, Ema M, Ogita H. Generation of a familial hypercholesterolemia model in non-human primate. Sci Rep 2023; 13:15649. [PMID: 37730951 PMCID: PMC10511719 DOI: 10.1038/s41598-023-42763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an inherited autosomal dominant disorder that is associated with a high plasma level of low-density lipoprotein (LDL) cholesterol, leading to an increased risk of cardiovascular diseases. To develop basic and translational research on FH, we here generated an FH model in a non-human primate (cynomolgus monkeys) by deleting the LDL receptor (LDLR) gene using the genome editing technique. Six LDLR knockout (KO) monkeys were produced, all of which were confirmed to have mutations in the LDLR gene by sequence analysis. The levels of plasma cholesterol and triglyceride were quite high in the monkeys, and were similar to those in FH patients with homozygous mutations in the LDLR gene. In addition, periocular xanthoma was observed only 1 year after birth. Lipoprotein profile analysis showed that the plasma very low-density lipoprotein and LDL were elevated, while the plasma high density lipoprotein was decreased in LDLR KO monkeys. The LDLR KO monkeys were also strongly resistant to medications for hypercholesterolemia. Taken together, we successfully generated a non-human primate model of hypercholesterolemia in which the phenotype is similar to that of homozygous FH patients.
Collapse
Affiliation(s)
- Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Tomoyuki Tsukiyama
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Masahiro Komeno
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Hideaki Tsuchiya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Kawamoto
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Mitsuru Murase
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Takahiro Nakagawa
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Iori Itagaki
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Yasunari Seita
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Shoma Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Masataka Nakaya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Atsushi Yamada
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Masatsugu Ema
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
10
|
Murray J, Einhaus T, Venkataraman R, Radtke S, Zhen A, Carrillo MA, Kitchen SG, Peterson CW, Kiem HP. Efficient manufacturing and engraftment of CCR5 gene-edited HSPCs following busulfan conditioning in nonhuman primates. Mol Ther Methods Clin Dev 2023; 30:276-287. [PMID: 37575091 PMCID: PMC10415663 DOI: 10.1016/j.omtm.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023]
Abstract
Hematopoietic stem cell gene therapy has been successfully used for a number of genetic diseases and is also being explored for HIV. However, toxicity of the conditioning regimens has been a major concern. Here we compared current conditioning approaches in a clinically relevant nonhuman primate model. We first customized various aspects of the therapeutic approach, including mobilization and cell collection protocols, conditioning regimens that support engraftment with minimal collateral damage, and cell manufacturing and infusing schema that reflect and build on current clinical approaches. Through a series of iterative in vivo experiments in two macaque species, we show that busulfan conditioning significantly spares lymphocytes and maintains a superior immune response to mucosal challenge with simian/human immunodeficiency virus, compared to total body irradiation and melphalan regimens. Comparative mobilization experiments demonstrate higher cell yield relative to our historical standard, primed bone marrow and engraftment of CRISPR-edited hematopoietic stem and progenitor cells (HSPCs) after busulfan conditioning. Our findings establish a detailed workflow for preclinical HSPC gene therapy studies in the nonhuman primate model, which in turn will support testing of novel conditioning regimens and more advanced HSPC gene editing techniques tailored to any disease of interest.
Collapse
Affiliation(s)
- Jason Murray
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Teresa Einhaus
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rasika Venkataraman
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Stefan Radtke
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Anjie Zhen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA
| | - Mayra A. Carrillo
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA
| | - Scott G. Kitchen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, 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
| |
Collapse
|
11
|
Galy A, Berkhout B, Breckpot K, Pichon C, Bloom K, Kiem HP, Mühlebach MD, McCune JM. Recent Advances Using Genetic Therapies Against Infectious Diseases and for Vaccination. Hum Gene Ther 2023; 34:896-904. [PMID: 37639360 DOI: 10.1089/hum.2023.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023] Open
Abstract
The development of prophylatic or therapeutic medicines for infectious diseases is one of the priorities for health organizations worldwide. Innovative solutions are required to achieve effective, safe, and accessible treatments for most if not all infectious diseases, particularly those that are chronic in nature or that emerge unexpectedly over time. Genetic technologies offer versatile possibilities to design therapies against pathogens. Recent developments such as mRNA vaccines, CRISPR gene editing, and immunotherapies provide unprecedented hope to achieve significant results in the field of infectious diseases. This review will focus on advances in this domain, showcasing the cross-fertilization with other fields (e.g., oncology), and addressing some of the logistical and economic concerns important to consider when making these advances accessible to diverse populations around the world.
Collapse
Affiliation(s)
- Anne Galy
- ART-TG, Inserm, Corbeil-Essonnes, France
| | - Ben Berkhout
- Department of Medical Microbiology Laboratory of Experimental Virology Amsterdam UMC, AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Karine Breckpot
- Department of Biomedical Sciences, Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS, ART-ARNm, Inserm, Orléans
- Institut Universitaire de France, Paris, France
| | - Kristie Bloom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Infectious Diseases and Oncology Research Institute (IDORI), Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Hans-Peter Kiem
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Joseph M McCune
- HIV Frontiers, Global Health Accelerator, Bill and Melinda Gates Foundation, Seattle, Washington, USA
| |
Collapse
|
12
|
Rothemejer FH, Lauritsen NP, Søgaard OS, Tolstrup M. Strategies for enhancing CAR T cell expansion and persistence in HIV infection. Front Immunol 2023; 14:1253395. [PMID: 37671164 PMCID: PMC10475529 DOI: 10.3389/fimmu.2023.1253395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapies are tremendously successful in hematological malignancies and show great promise as treatment and curative strategy for HIV. A major determinant for effective CAR T cell therapy is the persistence of CAR T cells. Particularly, antigen density and target cell abundance are crucial for the engagement, engraftment, and persistence of CAR T cells. The success of HIV-specific CAR T cells is challenged by limited antigen due to low cell surface expression of viral proteins and the scarcity of chronically infected cells during antiretroviral therapy. Several strategies have been explored to increase the efficacy of CAR T cells by enhancing expansion and persistence of the engineered cells. This review highlights the challenges of designing CAR T cells against HIV and other chronic viral infections. We also discuss potential strategies to enhance CAR T cell expansion and persistence in the setting of low antigen exposure.
Collapse
Affiliation(s)
- Frederik Holm Rothemejer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Nanna Pi Lauritsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Ole Schmeltz Søgaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
13
|
Dross S, Venkataraman R, Patel S, Huang ML, Bollard CM, Rosati M, Pavlakis GN, Felber BK, Bar KJ, Shaw GM, Jerome KR, Mullins JI, Kiem HP, Fuller DH, Peterson CW. Efficient ex vivo expansion of conserved element vaccine-specific CD8+ T-cells from SHIV-infected, ART-suppressed nonhuman primates. Front Immunol 2023; 14:1188018. [PMID: 37207227 PMCID: PMC10189133 DOI: 10.3389/fimmu.2023.1188018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023] Open
Abstract
HIV-specific T cells are necessary for control of HIV-1 replication but are largely insufficient for viral clearance. This is due in part to these cells' recognition of immunodominant but variable regions of the virus, which facilitates viral escape via mutations that do not incur viral fitness costs. HIV-specific T cells targeting conserved viral elements are associated with viral control but are relatively infrequent in people living with HIV (PLWH). The goal of this study was to increase the number of these cells via an ex vivo cell manufacturing approach derived from our clinically-validated HIV-specific expanded T-cell (HXTC) process. Using a nonhuman primate (NHP) model of HIV infection, we sought to determine i) the feasibility of manufacturing ex vivo-expanded virus-specific T cells targeting viral conserved elements (CE, CE-XTCs), ii) the in vivo safety of these products, and iii) the impact of simian/human immunodeficiency virus (SHIV) challenge on their expansion, activity, and function. NHP CE-XTCs expanded up to 10-fold following co-culture with the combination of primary dendritic cells (DCs), PHA blasts pulsed with CE peptides, irradiated GM-K562 feeder cells, and autologous T cells from CE-vaccinated NHP. The resulting CE-XTC products contained high frequencies of CE-specific, polyfunctional T cells. However, consistent with prior studies with human HXTC and these cells' predominant CD8+ effector phenotype, we did not observe significant differences in CE-XTC persistence or SHIV acquisition in two CE-XTC-infused NHP compared to two control NHP. These data support the safety and feasibility of our approach and underscore the need for continued development of CE-XTC and similar cell-based strategies to redirect and increase the potency of cellular virus-specific adaptive immune responses.
Collapse
Affiliation(s)
- Sandra Dross
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, Seattle, WA, United States
| | - Rasika Venkataraman
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Shabnum Patel
- Center for Cancer and Immunology Research, Children’s National Hospital and Department of Pediatrics, The George Washington University, Washington, DC, United States
| | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Catherine M. Bollard
- Center for Cancer and Immunology Research, Children’s National Hospital and Department of Pediatrics, The George Washington University, Washington, DC, United States
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, National Cancer Institute at Frederick, Frederick, MD, United States
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Katharine J. Bar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - George M. Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Keith R. Jerome
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Division of Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - James I. Mullins
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Hans-Peter Kiem
- Washington National Primate Research Center, Seattle, WA, United States
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Deborah Heydenburg Fuller
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, Seattle, WA, United States
| | - Christopher W. Peterson
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
14
|
Li C, Anderson AK, Wang H, Gil S, Kim J, Huang L, Germond A, Baldessari A, Nelson V, Bar KJ, Peterson CW, Bui J, Kiem HP, Lieber A. Stable HIV decoy receptor expression after in vivo HSC transduction in mice and NHPs: Safety and efficacy in protection from SHIV. Mol Ther 2023; 31:1059-1073. [PMID: 36760126 PMCID: PMC10124088 DOI: 10.1016/j.ymthe.2023.02.002] [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: 11/26/2022] [Revised: 01/15/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
We aim to develop an in vivo hematopoietic stem cell (HSC) gene therapy approach for persistent control/protection of HIV-1 infection based on the stable expression of a secreted decoy protein for HIV receptors CD4 and CCR5 (eCD4-Ig) from blood cells. HSCs in mice and a rhesus macaque were mobilized from the bone marrow and transduced by an intravenous injection of HSC-tropic, integrating HDAd5/35++ vectors expressing rhesus eCD4-Ig. In vivo HSC transduction/selection resulted in stable serum eCD4-Ig levels of ∼100 μg/mL (mice) and >20 μg/mL (rhesus) with half maximal inhibitory concentrations (IC50s) of 1 μg/mL measured by an HIV neutralization assay. After simian-human-immunodeficiency virus D (SHIV.D) challenge of rhesus macaques injected with HDAd-eCD4-Ig or a control HDAd5/35++ vector, peak plasma viral load levels were ∼50-fold lower in the eCD4-Ig-expressing animal. Furthermore, the viral load was lower in tissues with the highest eCD4-Ig expression, specifically the spleen and lymph nodes. SHIV.D challenge triggered a selective expansion of transduced CD4+CCR5+ cells, thereby increasing serum eCD4-Ig levels. The latter, however, broke immune tolerance and triggered anti-eCD4-Ig antibody responses, which could have contributed to the inability to eliminate SHIV.D. Our data will guide us in the improvement of the in vivo approach. Clearly, our conclusions need to be validated in larger animal cohorts.
Collapse
Affiliation(s)
- Chang Li
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA.
| | - Anna Kate Anderson
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Hongjie Wang
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Sucheol Gil
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Jiho Kim
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Lishan Huang
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Audrey Germond
- Washington National Primate Research Center, Division of Regenerative Medicine and Gene Therapy, Seattle, WA 98195, USA
| | - Audrey Baldessari
- Washington National Primate Research Center, Division of Regenerative Medicine and Gene Therapy, Seattle, WA 98195, USA
| | - Veronica Nelson
- Stem and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Katharine J Bar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher W Peterson
- Stem and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Washington National Primate Research Center, Division of Regenerative Medicine and Gene Therapy, Seattle, WA 98195, USA
| | - John Bui
- Stem and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Medicine, Division of Allergy and Infection Diseases, University of Washington, Seattle, WA 98195, USA
| | - Hans-Peter Kiem
- Stem and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Washington National Primate Research Center, Division of Regenerative Medicine and Gene Therapy, Seattle, WA 98195, USA; Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA 98195, USA
| | - André Lieber
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA 98195, USA; Washington National Primate Research Center, Division of Regenerative Medicine and Gene Therapy, Seattle, WA 98195, USA.
| |
Collapse
|
15
|
Campos-Gonzalez G, Martinez-Picado J, Velasco-Hernandez T, Salgado M. Opportunities for CAR-T Cell Immunotherapy in HIV Cure. Viruses 2023; 15:v15030789. [PMID: 36992496 PMCID: PMC10057306 DOI: 10.3390/v15030789] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Chimeric antigen receptor (CAR) technology is having a huge impact in the blood malignancy field and is becoming a well-established therapy for many types of leukaemia. In recent decades, efforts have been made to demonstrate that CAR-T cells have potential as a therapy to achieve a sterilizing cure for human immunodeficiency virus (HIV) infection. However, translation of this technology to the HIV scenario has not been easy, as many challenges have appeared along the way that hinder the consolidation of CAR-T cells as a putative therapy. Here, we review the origin and development of CAR-T cells, describe the advantages of CAR-T cell therapy in comparison with other therapies, and describe the major obstacles currently faced regarding application of this technology in the HIV field, specifically, viral escape, CAR-T cell infectivity, and accessibility to hidden reservoirs. Nonetheless, promising results in successfully tackling some of these issues that have been obtained in clinical trials suggest a bright future for CAR-T cells as a consolidated therapy.
Collapse
Affiliation(s)
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), 08500 Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Talia Velasco-Hernandez
- Josep Carreras Leukaemia Research Institute, 08036 Barcelona, Spain
- RICORS-TERAV, ISCIII, 28029 Madrid, Spain
| | - Maria Salgado
- IrsiCaixa AIDS Research Institute, 08916 Badalona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| |
Collapse
|
16
|
Corey D, Haeseleer F, Hou J, Corey L. Novel engineered chimeric engulfment receptors trigger T cell effector functions against SIV-infected CD4+ T cells. Mol Ther Methods Clin Dev 2023; 28:1-10. [PMID: 36514789 PMCID: PMC9720250 DOI: 10.1016/j.omtm.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
Adoptive therapy with genetically engineered T cells offers potential for infectious disease treatment in immunocompromised persons. HIV/simian immunodeficiency virus (SIV)-infected cells express phosphatidylserine (PS) early post infection. We tested whether chimeric engulfment receptor (CER) T cells designed to recognize PS-expressing cells could eliminate SIV-infected cells. Lentiviral CER constructs composed of the extracellular domain of T cell immunoglobulin and mucin domain containing 4 (TIM-4), the PS receptor, and engulfment signaling domains were transduced into primary rhesus macaque (RM) T cells. We measured PS binding and T cell engulfment of RM CD4+ T cells infected with SIV expressing GFP and in vitro, TIM-4 CER CD4+ T cells effectively killed SIV-infected cells, which was dependent on TIM-4 binding to PS. Enhanced killing of SIV-infected CD4+ T cells by CER and chimeric antigen receptor T cell combinations was also observed. This installation of innate immune functions into T cells presents an opportunity to enhance elimination of SIV-infected cells, and studies to evaluate their effect in vivo are warranted.
Collapse
Affiliation(s)
- Daniel Corey
- CERo Therapeutics, 201 Haskins Way, Suite 230, San Francisco, CA 94080, USA
| | - Francoise Haeseleer
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Joe Hou
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Lawrence Corey
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
17
|
Pampusch MS, Sevcik EN, Quinn ZE, Davey BC, Berg JM, Gorrell-Brown I, Abdelaal HM, Rakasz EG, Rendahl A, Skinner PJ. Assessment of anti-CD20 antibody pre-treatment for augmentation of CAR-T cell therapy in SIV-infected rhesus macaques. Front Immunol 2023; 14:1101446. [PMID: 36825014 PMCID: PMC9941136 DOI: 10.3389/fimmu.2023.1101446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
During chronic HIV and SIV infections, the majority of viral replication occurs within lymphoid follicles. In a pilot study, infusion of SIV-specific CD4-MBL-CAR-T cells expressing the follicular homing receptor, CXCR5, led to follicular localization of the cells and a reduction in SIV viral loads in rhesus macaques. However, the CAR-T cells failed to persist. We hypothesized that temporary disruption of follicles would create space for CAR-T cell engraftment and lead to increased abundance and persistence of CAR-T cells. In this study we treated SIV-infected rhesus macaques with CAR-T cells and preconditioned one set with anti-CD20 antibody to disrupt the follicles. We evaluated CAR-T cell abundance and persistence in four groups of SIVmac239-infected and ART-suppressed animals: untreated, CAR-T cell treated, CD20 depleted, and CD20 depleted/CAR-T cell treated. In the depletion study, anti-CD20 was infused one week prior to CAR-T infusion and cessation of ART. Anti-CD20 antibody treatment led to temporary depletion of CD20+ cells in blood and partial depletion in lymph nodes. In this dose escalation study, there was no impact of CAR-T cell infusion on SIV viral load. However, in both the depleted and non-depleted animals, CAR-T cells accumulated in and around lymphoid follicles and were Ki67+. CAR-T cells increased in number in follicles from 2 to 6 days post-treatment, with a median 15.2-fold increase in follicular CAR-T cell numbers in depleted/CAR-T treated animals compared to an 8.1-fold increase in non-depleted CAR-T treated animals. The increase in CAR T cells in depleted animals was associated with a prolonged elevation of serum IL-6 levels and a rapid loss of detectable CAR-T cells. Taken together, these data suggest that CAR-T cells likely expanded to a greater extent in depleted/CAR-T cell treated animals. Further studies are needed to elucidate mechanisms mediating the rapid loss of CAR-T cells and to evaluate strategies to improve engraftment and persistence of HIV-specific CAR-T cells. The potential for an inflammatory cytokine response appears to be enhanced with anti-CD20 antibody treatment and future studies may require CRS control strategies. These studies provide important insights into cellular immunotherapy and suggest future studies for improved outcomes.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Emily N. Sevcik
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Zoe E. Quinn
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - James M. Berg
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Ian Gorrell-Brown
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison WI, United States
| | - Aaron Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
18
|
Zhang Y, Ge T, Huang M, Qin Y, Liu T, Mu W, Wang G, Jiang L, Li T, Zhao L, Wang J. Extracellular Vesicles Expressing CD19 Antigen Improve Expansion and Efficacy of CD19-Targeted CAR-T Cells. Int J Nanomedicine 2023; 18:49-63. [PMID: 36636644 PMCID: PMC9830716 DOI: 10.2147/ijn.s390720] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
Background CAR-T cell therapy is effective in the treatment of certain hematological malignancies, and the expansion and functional persistence of CAR-T cells in vivo are crucial to clinical efficacy. The aim of this study was to investigate the potential of extracellular vesicles (EVs) modified with the CAR antigen to promote the efficacy of CAR-T cells in vivo. Methods We generated HEK293T-derived EVs to present the CD19 antigen as the CAR target. In vitro, EVs expressing CD19 antigen (CD19 EVs) were co-incubated with anti-CD19 CAR-T cells. Then, proliferation, cytokine secretion, CD107a expression, tumor killing, subsets, and immune checkpoint expression were measured to assess CAR-T cell function. After infusion of CD19 EVs pretreated CAR-T cells into a lymphoma xenograft mouse model, flow cytometry and digital PCR were used to measure the expansion of CAR-T cells, and tumor volumes were continuously monitored to assess the anti-tumor efficacy of CAR-T cells in vivo. Another mouse model was created to investigate the effect of in vivo injection of CD19 EVs on the functional persistence of CAR-T cells, and safety was determined by histopathology of the main organs. Results CD19 EVs activated CAR-T cells in an antigen-specific and dose-dependent manner and promoted the selective expansion and cytokine secretion of co-cultured CAR-T cells. Specifically, CD19 EVs preferably increased the expansion of the CAR-T subpopulation with a high surface CD19-CAR density and consequently enhanced the anti-tumor activity of CAR-T cells. Futhermore, CD19-EVs-primed CAR-T cells achieved superior proliferation and anti-tumor effects in a mouse model with lymphoma xenograft. In vivo administration of CD19 EVs promoted the functional persistence of CAR-T cells in the xenograft mouse model. Conclusion Our findings indicate that antigen-expressing EVs can be utilized as a boost to improve CAR-T cell efficacy in vitro and in vivo.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Tong Ge
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Meijuan Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yun Qin
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Tianjiao Liu
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Gaoxiang Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Lijun Jiang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Tongjuan Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Lei Zhao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China,Correspondence: Lei Zhao; Jue Wang, Email ;
| | - Jue Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
19
|
Ellis GI, Deng MZ, Winn DW, Coker KE, Shukla D, Bhoj V, Milone MC, Duran-Struuck R, Riley JL. Generation of non-human primate CAR Tregs using artificial antigen-presenting cells, simian tropic lentiviral vectors, and antigen-specific restimulation. STAR Protoc 2022; 3:101784. [PMID: 36386869 PMCID: PMC9641266 DOI: 10.1016/j.xpro.2022.101784] [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] [Indexed: 11/08/2022] Open
Abstract
It is technically challenging to generate large doses of regulatory T cells (Tregs) engineered to express a chimeric antigen receptor (CAR) in non-human primates (NHP). Here, we have optimized the manufacturing of CAR Tregs by stringent sorting of Tregs, stimulation by artificial antigen-presenting cells, transduction by simian tropic lentiviral vectors, and antigen-specific expansion. The result of this method is highly suppressive CAR Tregs for use in a pre-clinical, large animal model of transplant tolerance. For complete details on the use and execution of this protocol, please refer to Ellis et al. (2022).
Collapse
Affiliation(s)
- Gavin I Ellis
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mosha Z Deng
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Delaine W Winn
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kimberly E Coker
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Divanshu Shukla
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vijay Bhoj
- Deparment of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael C Milone
- Deparment of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raimon Duran-Struuck
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Davey BC, Pampusch MS, Cartwright EK, Abdelaal HM, Rakasz EG, Rendahl A, Berger EA, Skinner PJ. Development of an anti-CAR antibody response in SIV-infected rhesus macaques treated with CD4-MBL CAR/CXCR5 T cells. Front Immunol 2022; 13:1032537. [PMID: 36582226 PMCID: PMC9793449 DOI: 10.3389/fimmu.2022.1032537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/14/2022] [Indexed: 12/15/2022] Open
Abstract
T cells expressing a simian immunodeficiency (SIV)-specific chimeric antigen receptor (CAR) and the follicular homing molecule, CXCR5, were infused into antiretroviral therapy (ART) suppressed, SIV-infected rhesus macaques to assess their ability to localize to the lymphoid follicle and control the virus upon ART interruption. While the cells showed evidence of functionality, they failed to persist in the animals beyond 28 days. Development of anti-CAR antibodies could be responsible for the lack of persistence. Potential antigenic sites on the anti-SIV CAR used in these studies included domains 1 and 2 of CD4, the carbohydrate recognition domain (CRD) of mannose-binding lectin (MBL), and an extracellular domain of the costimulatory molecule, CD28, along with short linker sequences. Using a flow cytometry based assay and target cells expressing the CAR/CXCR5 construct, we examined the serum of the CD4-MBL CAR/CXCR5-T cell treated animals to determine that the animals had developed an anti-CAR antibody response after infusion. Binding sites for the anti-CAR antibodies were identified by using alternative CARs transduced into target cells and by preincubation of the target cells with a CD4 blocking antibody. All of the treated animals developed antibodies in their serum that bound to CD4-MBL CAR/CXCR5 T cells and the majority were capable of inducing an ADCC response. The CD4 antibody-blocking assay suggests that the dominant immunogenic components of this CAR are the CD4 domains with a possible additional site of the CD28 domain with its linker. This study shows that an anti-drug antibody (ADA) response can occur even when using self-proteins, likely due to novel epitopes created by abridged self-proteins and/or the self-domain of the CAR connection to a small non-self linker. While in our study, there was no statistically significant correlation between the ADA response and the persistence of the CD4-MBL CAR/CXCR5-T cells in rhesus macaques, these findings suggest that the development of an ADA response could impact the long-term persistence of self-based CAR immunotherapies.
Collapse
Affiliation(s)
- Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Emily K. Cartwright
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Madison, WI, United States
| | - Aaron Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Edward A. Berger
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States,*Correspondence: Pamela J. Skinner,
| |
Collapse
|
21
|
Anthony-Gonda K, Ray A, Su H, Wang Y, Xiong Y, Lee D, Block A, Chilunda V, Weiselberg J, Zemelko L, Wang YY, Kleinsorge-Block S, Reese JS, de Lima M, Ochsenbauer C, Kappes JC, Dimitrov DS, Orentas R, Deeks SG, Rutishauser RL, Berman JW, Goldstein H, Dropulić B. In vivo killing of primary HIV-infected cells by peripheral-injected early memory-enriched anti-HIV duoCAR T cells. JCI Insight 2022; 7:e161698. [PMID: 36345941 PMCID: PMC9675454 DOI: 10.1172/jci.insight.161698] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/14/2022] [Indexed: 11/09/2022] Open
Abstract
HIV-specific chimeric antigen receptor-T cell (CAR T cell) therapies are candidates to functionally cure HIV infection in people with HIV (PWH) by eliminating reactivated HIV-infected cells derived from latently infected cells within the HIV reservoir. Paramount to translating such therapeutic candidates successfully into the clinic will require anti-HIV CAR T cells to localize to lymphoid tissues in the body and eliminate reactivated HIV-infected cells such as CD4+ T cells and monocytes/macrophages. Here we show that i.v. injected anti-HIV duoCAR T cells, generated using a clinical-grade anti-HIV duoCAR lentiviral vector, localized to the site of active HIV infection in the spleen of humanized mice and eliminated HIV-infected PBMCs. CyTOF analysis of preinfusion duoCAR T cells revealed an early memory phenotype composed predominantly of CCR7+ stem cell-like/central memory T cells (TSCM/TCM) with expression of some effector-like molecules. In addition, we show that anti-HIV duoCAR T cells effectively sense and kill HIV-infected CD4+ T cells and monocytes/macrophages. Furthermore, we demonstrate efficient genetic modification of T cells from PWH on suppressive ART into anti-HIV duoCAR T cells that subsequently kill autologous PBMCs superinfected with HIV. These studies support the safety and efficacy of anti-HIV duoCAR T cell therapy in our presently open phase I/IIa clinical trial (NCT04648046).
Collapse
Affiliation(s)
- Kim Anthony-Gonda
- Caring Cross, Gaithersburg, Maryland, USA
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Alex Ray
- Department of Microbiology & Immunology and
| | - Hang Su
- Department of Microbiology & Immunology and
| | - Yuge Wang
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Ying Xiong
- Caring Cross, Gaithersburg, Maryland, USA
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Danica Lee
- Department of Microbiology & Immunology and
| | | | - Vanessa Chilunda
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jessica Weiselberg
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Lily Zemelko
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Yen Y. Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Sarah Kleinsorge-Block
- Stem Cell Transplant Program and Center for Regenerative Medicine, University Hospitals Seidman Cancer Center and Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jane S. Reese
- Stem Cell Transplant Program and Center for Regenerative Medicine, University Hospitals Seidman Cancer Center and Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Marcos de Lima
- Stem Cell Transplant Program and Center for Regenerative Medicine, University Hospitals Seidman Cancer Center and Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Christina Ochsenbauer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John C. Kappes
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham Veterans Affairs Medical Center, Research Service, Birmingham, Alabama, USA
| | - Dimiter S. Dimitrov
- Center for Antibody Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rimas Orentas
- Caring Cross, Gaithersburg, Maryland, USA
- Department of Pediatrics, University of Washington School of Medicine, and Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research lnstitute, Seattle, Washington, USA
| | - Steven G. Deeks
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Joan W. Berman
- Department of Microbiology & Immunology and
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Harris Goldstein
- Department of Microbiology & Immunology and
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Boro Dropulić
- Caring Cross, Gaithersburg, Maryland, USA
- Lentigen, a Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| |
Collapse
|
22
|
Duncan BB, Dunbar CE, Ishii K. Applying a Clinical Lens to Animal Models of CAR-T Cell Therapies. Mol Ther Methods Clin Dev 2022; 27:17-31. [PMID: 36156878 PMCID: PMC9478925 DOI: 10.1016/j.omtm.2022.08.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Chimeric antigen receptor (CAR)-T cells have emerged as a promising treatment modality for various hematologic and solid malignancies over the past decade. Animal models remain the cornerstone of pre-clinical evaluation of human CAR-T cell products and are generally required by regulatory agencies prior to clinical translation. However, pharmacokinetics and pharmacodynamics of adoptively transferred T cells are dependent on various recipient factors, posing challenges for accurately predicting human engineered T cell behavior in non-human animal models. For example, murine xenograft models did not forecast now well-established cytokine-driven systemic toxicities of CAR-T cells seen in humans, highlighting the limitations of animal models that do not perfectly recapitulate complex human immune systems. Understanding the concordance as well as discrepancies between existing pre-clinical animal data and human clinical experiences, along with established advantages and limitations of each model, will facilitate investigators’ ability to appropriately select and design animal models for optimal evaluation of future CAR-T cell products. We summarize the current state of animal models in this field, and the advantages and disadvantages of each approach depending on the pre-clinical questions being asked.
Collapse
|
23
|
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: 0] [Impact Index Per Article: 0] [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
|
24
|
Guan M, Lim L, Holguin L, Han T, Vyas V, Urak R, Miller A, Browning DL, Echavarria L, Li S, Li S, Chang WC, Scott T, Yazaki P, Morris KV, Cardoso AA, Blanchard MS, Le Verche V, Forman SJ, Zaia JA, Burnett JC, Wang X. Pre-clinical data supporting immunotherapy for HIV using CMV-HIV-specific CAR T cells with CMV vaccine. Mol Ther Methods Clin Dev 2022; 25:344-359. [PMID: 35573050 PMCID: PMC9062763 DOI: 10.1016/j.omtm.2022.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/10/2022] [Indexed: 01/22/2023]
Abstract
T cells engineered to express HIV-specific chimeric antigen receptors (CARs) represent a promising strategy to clear HIV-infected cells, but to date have not achieved clinical benefits. A likely hurdle is the limited T cell activation and persistence when HIV antigenemia is low, particularly during antiretroviral therapy (ART). To overcome this issue, we propose to use a cytomegalovirus (CMV) vaccine to stimulate CMV-specific T cells that express CARs directed against the HIV-1 envelope protein gp120. In this study, we use a GMP-compliant platform to engineer CMV-specific T cells to express a second-generation CAR derived from the N6 broadly neutralizing antibody, one of the broadest anti-gp120 neutralizing antibodies. These CMV-HIV CAR T cells exhibit dual effector functions upon in vitro stimulation through their endogenous CMV-specific T cell receptors or the introduced CARs. Using a humanized HIV mouse model, we show that CMV vaccination during ART accelerates CMV-HIV CAR T cell expansion in the peripheral blood and that higher numbers of CMV-HIV CAR T cells were associated with a better control of HIV viral load and fewer HIV antigen p24+ cells in the bone marrow upon ART interruption. Collectively, these data support the clinical development of CMV-HIV CAR T cells in combination with a CMV vaccine in HIV-infected individuals.
Collapse
Affiliation(s)
- Min Guan
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Laura Lim
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Leo Holguin
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Tianxu Han
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Vibhuti Vyas
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Ryan Urak
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Aaron Miller
- Department of Molecular Imaging and Therapy, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Diana L. Browning
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Liliana Echavarria
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Shasha Li
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Shirley Li
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Wen-Chung Chang
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - Tristan Scott
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Paul Yazaki
- Department of Molecular Imaging and Therapy, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Kevin V. Morris
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Angelo A. Cardoso
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - M. Suzette Blanchard
- Division of Biostatistics, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Virginia Le Verche
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Stephen J. Forman
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| | - John A. Zaia
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - John C. Burnett
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Xiuli Wang
- T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
- Corresponding author Xiuli Wang, T Cell Therapeutics Research Laboratory, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010-3000, USA.
| |
Collapse
|
25
|
Kreider EF, Bar KJ. HIV-1 Reservoir Persistence and Decay: Implications for Cure Strategies. Curr HIV/AIDS Rep 2022; 19:194-206. [PMID: 35404007 PMCID: PMC10443186 DOI: 10.1007/s11904-022-00604-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Despite suppressive antiretroviral therapy (ART), a viral reservoir persists in individuals living with HIV that can reignite systemic replication should treatment be interrupted. Understanding how HIV-1 persists through effective ART is essential to develop cure strategies to induce ART-free virus remission. RECENT FINDINGS The HIV-1 reservoir resides in a pool of CD4-expressing cells as a range of viral species, a subset of which is genetically intact. Recent studies suggest that the reservoir on ART is highly dynamic, with expansion and contraction of virus-infected cells over time. Overall, the intact proviral reservoir declines faster than defective viruses, suggesting enhanced immune clearance or cellular turnover. Upon treatment interruption, rebound viruses demonstrate escape from adaptive and innate immune responses, implicating these selective pressures in restriction of virus reactivation. Cure strategies employing immunotherapy are poised to test whether host immune pressure can be augmented to enhance reservoir suppression or clearance. Alternatively, genomic engineering approaches are being applied to directly eliminate intact viruses and shrink the replication-competent virus pool. New evidence suggests host immunity exerts selective pressure on reservoir viruses and clears HIV-1 infected cells over years on ART. Efforts to build on the detectable, but insufficient, reservoir clearance via empiric testing in clinical trials will inform our understanding of mechanisms of viral persistence and the direction of future cure strategies.
Collapse
Affiliation(s)
- Edward F Kreider
- Perelman School of Medicine, University of Pennsylvania, Stemmler Hall Room 130-150, 3450 Hamilton Walk, Philadelphia, PA, 19104-6073, USA
| | - Katharine J Bar
- Perelman School of Medicine, University of Pennsylvania, 502D Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA, 19104‑0673, USA.
| |
Collapse
|
26
|
Ellis GI, Coker KE, Winn DW, Deng MZ, Shukla D, Bhoj V, Milone MC, Wang W, Liu C, Naji A, Duran-Struuck R, Riley JL. Trafficking and persistence of alloantigen-specific chimeric antigen receptor regulatory T cells in Cynomolgus macaque. Cell Rep Med 2022; 3:100614. [PMID: 35551746 PMCID: PMC9133392 DOI: 10.1016/j.xcrm.2022.100614] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/16/2022] [Accepted: 03/29/2022] [Indexed: 01/13/2023]
Abstract
Adoptive transfer of chimeric antigen receptor regulatory T cells (CAR Tregs) is a promising way to prevent allograft loss without the morbidity associated with current therapies. Non-human primates (NHPs) are a clinically relevant model to develop transplant regimens, but manufacturing and engraftment of NHP CAR Tregs have not been demonstrated yet. Here, we describe a culture system that massively expands CAR Tregs specific for the Bw6 alloantigen. In vitro, these Tregs suppress in an antigen-specific manner without pro-inflammatory cytokine secretion or cytotoxicity. In vivo, Bw6-specific CAR Tregs preferentially traffic to and persist in bone marrow for at least 1 month. Following transplant of allogeneic Bw6+ islets and autologous CAR Tregs into the bone marrow of diabetic recipients, CAR Tregs traffic to the site of islet transplantation and maintain a phenotype of suppressive Tregs. Our results establish a framework for the optimization of CAR Treg therapy in NHP disease models.
Collapse
Affiliation(s)
- Gavin I Ellis
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly E Coker
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Delaine W Winn
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Mosha Z Deng
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Divanshu Shukla
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Vijay Bhoj
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Wang
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | - James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Peterson CW, Venkataraman R, Reddy SS, Pande D, Enstrom MR, Radtke S, Humbert O, Kiem HP. Intracellular RNase activity dampens zinc finger nuclease-mediated gene editing in hematopoietic stem and progenitor cells. Mol Ther Methods Clin Dev 2022; 24:30-39. [PMID: 34977270 PMCID: PMC8671732 DOI: 10.1016/j.omtm.2021.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/19/2021] [Indexed: 12/21/2022]
Abstract
Over the past decade, numerous gene-editing platforms which alter host DNA in a highly specific and targeted fashion have been described. Two notable examples are zinc finger nucleases (ZFNs), the first gene-editing platform to be tested in clinical trials, and more recently, CRISPR/Cas9. Although CRISPR/Cas9 approaches have become arguably the most popular platform in the field, the therapeutic advantages and disadvantages of each strategy are only beginning to emerge. We have established a nonhuman primate (NHP) model that serves as a strong predictor of successful gene therapy and gene-editing approaches in humans; our recent work shows that ZFN-edited hematopoietic stem and progenitor cells (HSPCs) engraft at lower levels than CRISPR/Cas9-edited cells. Here, we investigate the mechanisms underlying this difference. We show that optimized culture conditions, including defined serum-free media, augment engraftment of gene-edited NHP HSPCs in a mouse xenograft model. Furthermore, we identify intracellular RNases as major barriers for mRNA-encoded nucleases relative to preformed enzymatically active CRISPR/Cas9 ribonucleoprotein (RNP) complexes. We conclude that CRISPR/Cas9 RNP gene editing is more stable and efficient than ZFN mRNA-based delivery and identify co-delivered RNase inhibitors as a strategy to enhance the expression of gene-editing proteins from mRNA intermediates.
Collapse
Affiliation(s)
- Christopher W. Peterson
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Rasika Venkataraman
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Sowmya S. Reddy
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Dnyanada Pande
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Mark R. Enstrom
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Stefan Radtke
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Olivier Humbert
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
28
|
Pampusch MS, Abdelaal HM, Cartwright EK, Molden JS, Davey BC, Sauve JD, Sevcik EN, Rendahl AK, Rakasz EG, Connick E, Berger EA, Skinner PJ. CAR/CXCR5-T cell immunotherapy is safe and potentially efficacious in promoting sustained remission of SIV infection. PLoS Pathog 2022; 18:e1009831. [PMID: 35130312 PMCID: PMC8853520 DOI: 10.1371/journal.ppat.1009831] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/17/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
During chronic human immunodeficiency virus (HIV) or simian immunodeficiency virus (SIV) infection prior to AIDS progression, the vast majority of viral replication is concentrated within B cell follicles of secondary lymphoid tissues. We investigated whether infusion of T cells expressing an SIV-specific chimeric antigen receptor (CAR) and the follicular homing receptor, CXCR5, could successfully kill viral-RNA+ cells in targeted lymphoid follicles in SIV-infected rhesus macaques. In this study, CD4 and CD8 T cells from rhesus macaques were genetically modified to express antiviral CAR and CXCR5 moieties (generating CAR/CXCR5-T cells) and autologously infused into a chronically infected animal. At 2 days post-treatment, the CAR/CXCR5-T cells were located primarily in spleen and lymph nodes both inside and outside of lymphoid follicles. Few CAR/CXCR5-T cells were detected in the ileum, rectum, and lung, and no cells were detected in the bone marrow, liver, or brain. Within follicles, CAR/CXCR5-T cells were found in direct contact with SIV-viral RNA+ cells. We next infused CAR/CXCR5-T cells into ART-suppressed SIV-infected rhesus macaques, in which the animals were released from ART at the time of infusion. These CAR/CXCR5-T cells replicated in vivo within both the extrafollicular and follicular regions of lymph nodes and accumulated within lymphoid follicles. CAR/CXR5-T cell concentrations in follicles peaked during the first week post-infusion but declined to undetectable levels after 2 to 4 weeks. Overall, CAR/CXCR5-T cell-treated animals maintained lower viral loads and follicular viral RNA levels than untreated control animals, and no outstanding adverse reactions were noted. These findings indicate that CAR/CXCR5-T cell treatment is safe and holds promise as a future treatment for the durable remission of HIV.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Emily K. Cartwright
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jhomary S. Molden
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jordan D. Sauve
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Emily N. Sevcik
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Aaron K. Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, Arizona, United States of America
| | - Edward A. Berger
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| |
Collapse
|
29
|
Uldrick TS, Adams SV, Fromentin R, Roche M, Fling SP, Gonçalves PH, Lurain K, Ramaswami R, Jackie Wang CC, Gorelick RJ, Welker JL, O’Donoghue L, Choudhary H, Lifson JD, Rasmussen TA, Rhodes A, Tumpach C, Yarchoan R, Maldarelli F, Cheever MA, Sékaly R, Chomont N, Deeks SG, Lewin SR. Pembrolizumab induces HIV latency reversal in people living with HIV and cancer on antiretroviral therapy. Sci Transl Med 2022; 14:eabl3836. [PMID: 35080914 PMCID: PMC9014398 DOI: 10.1126/scitranslmed.abl3836] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In people living with HIV (PLWH) on antiretroviral therapy (ART), virus persists in a latent form where there is minimal transcription or protein expression. Latently infected cells are a major barrier to curing HIV. Increasing HIV transcription and viral production in latently infected cells could facilitate immune recognition and reduce the pool of infected cells that persist on ART. Given that programmed cell death protein 1 (PD-1) expressing CD4+ T cells are preferentially infected with HIV in PLWH on ART, we aimed to determine whether administration of antibodies targeting PD-1 would reverse HIV latency in vivo. We therefore evaluated the impact of intravenous administration of pembrolizumab every 3 weeks on HIV latency in 32 PLWH and cancer on ART. After the first infusion of anti-PD-1, we observed a median 1.32-fold increase in unspliced HIV RNA and 1.61-fold increase in unspliced RNA:DNA ratio in sorted blood CD4+ T cells compared to baseline. We also observed a 1.65-fold increase in plasma HIV RNA. The frequency of CD4+ T cells with inducible virus evaluated using the tat/rev limiting dilution assay was higher after 6 cycles compared to baseline. Phylogenetic analyses of HIV env sequences in a participant who developed low concentrations of HIV viremia after 6 cycles of pembrolizumab did not demonstrate clonal expansion of HIV-infected cells. These data are consistent with anti-PD-1 being able to reverse HIV latency in vivo and support the rationale for combining anti-PD-1 with other interventions to reduce the HIV reservoir.
Collapse
Affiliation(s)
- Thomas S. Uldrick
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- University of Washington, Seattle, WA 98109, USA
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Scott V. Adams
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Remi Fromentin
- Department of Microbiology, Infectiology, and Immunology, Université de Montréal and Centre de Recherche du CHUM, Montréal H2X0A9, Canada
| | - Michael Roche
- RMIT University, Melbourne, VIC 3083, Australia
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Steven P. Fling
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Kathryn Lurain
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Ramya Ramaswami
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Robert J. Gorelick
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jorden L. Welker
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Liz O’Donoghue
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Thomas A. Rasmussen
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Ajantha Rhodes
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Carolin Tumpach
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Frank Maldarelli
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | - Nicolas Chomont
- Department of Microbiology, Infectiology, and Immunology, Université de Montréal and Centre de Recherche du CHUM, Montréal H2X0A9, Canada
| | - Steven G. Deeks
- University of California, San Francisco, San Francisco, CA 94110, USA
| | - Sharon R. Lewin
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
30
|
Hupperetz C, Lah S, Kim H, Kim CH. CAR T Cell Immunotherapy Beyond Haematological Malignancy. Immune Netw 2022; 22:e6. [PMID: 35291659 PMCID: PMC8901698 DOI: 10.4110/in.2022.22.e6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/03/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells, which express a synthetic receptor engineered to target specific antigens, have demonstrated remarkable potential to treat haematological malignancies. However, their transition beyond haematological malignancy has so far been unsatisfactory. Here, we discuss recent challenges and improvements for CAR T cell therapy against solid tumors: Antigen heterogeneity which provides an effective escape mechanism against conventional mono-antigen-specific CAR T cells; and the immunosuppressive tumor microenvironment which provides physical and molecular barriers that respectively prevent T cell infiltration and drive T cell dysfunction and hypoproliferation. Further, we discuss the application of CAR T cells in infectious disease and autoimmunity.
Collapse
Affiliation(s)
- Cedric Hupperetz
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Sangjoon Lah
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hyojin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Chan Hyuk Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
31
|
Deeks SG, Archin N, Cannon P, Collins S, Jones RB, de Jong MAWP, Lambotte O, Lamplough R, Ndung'u T, Sugarman J, Tiemessen CT, Vandekerckhove L, Lewin SR. Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2021. Nat Med 2021; 27:2085-2098. [PMID: 34848888 DOI: 10.1038/s41591-021-01590-5] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/27/2021] [Indexed: 12/21/2022]
Abstract
Despite the success of antiretroviral therapy (ART) for people living with HIV, lifelong treatment is required and there is no cure. HIV can integrate in the host genome and persist for the life span of the infected cell. These latently infected cells are not recognized as foreign because they are largely transcriptionally silent, but contain replication-competent virus that drives resurgence of the infection once ART is stopped. With a combination of immune activators, neutralizing antibodies, and therapeutic vaccines, some nonhuman primate models have been cured, providing optimism for these approaches now being evaluated in human clinical trials. In vivo delivery of gene-editing tools to either target the virus, boost immunity or protect cells from infection, also holds promise for future HIV cure strategies. In this Review, we discuss advances related to HIV cure in the last 5 years, highlight remaining knowledge gaps and identify priority areas for research for the next 5 years.
Collapse
Affiliation(s)
- Steven G Deeks
- University of California San Francisco, San Fransisco, CA, USA.
| | - Nancie Archin
- UNC HIV Cure Center, Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Paula Cannon
- University of Southern California, Los Angeles, CA, USA
| | | | - R Brad Jones
- Weill Cornell Medicine, Cornell University, New York, NY, USA
| | | | - Olivier Lambotte
- University Paris Saclay, AP-HP, Bicêtre Hospital, UMR1184 INSERM CEA, Le Kremlin Bicêtre, Paris, France
| | | | - Thumbi Ndung'u
- Africa Health Research Institute and University of KwaZulu-Natal, Durban, South Africa
- University College London, London, UK
- Ragon Institute of MGH, MIT and Harvard University, Cambridge, MA, USA
| | - Jeremy Sugarman
- Berman Institute of Bioethics and Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Caroline T Tiemessen
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Sharon R Lewin
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia.
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| |
Collapse
|
32
|
Genome editing in large animal models. Mol Ther 2021; 29:3140-3152. [PMID: 34601132 DOI: 10.1016/j.ymthe.2021.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/26/2021] [Accepted: 09/26/2021] [Indexed: 12/21/2022] Open
Abstract
Although genome editing technologies have the potential to revolutionize the way we treat human diseases, barriers to successful clinical implementation remain. Increasingly, preclinical large animal models are being used to overcome these barriers. In particular, the immunogenicity and long-term safety of novel gene editing therapeutics must be evaluated rigorously. However, short-lived small animal models, such as mice and rats, cannot address secondary pathologies that may arise years after a gene editing treatment. Likewise, immunodeficient mouse models by definition lack the ability to quantify the host immune response to a novel transgene or gene-edited locus. Large animal models, including dogs, pigs, and non-human primates (NHPs), bear greater resemblance to human anatomy, immunology, and lifespan and can be studied over longer timescales with clinical dosing regimens that are more relevant to humans. These models allow for larger scale and repeated blood and tissue sampling, enabling greater depth of study and focus on rare cellular subsets. Here, we review current progress in the development and evaluation of novel genome editing therapies in large animal models, focusing on applications in human immunodeficiency virus 1 (HIV-1) infection, cancer, and genetic diseases including hemoglobinopathies, Duchenne muscular dystrophy (DMD), hypercholesterolemia, and inherited retinal diseases.
Collapse
|
33
|
Abstract
Over the past decade, chimeric antigen receptor (CAR) T cells have emerged as the prototype gene therapy for B cell leukemias. These so-called living drugs are derived from a patient's own cells, reprogrammed to recognize and destroy cancer cells, and then reintroduced into the body. The huge success of this therapy for cancer is rooted in pioneering clinical and preclinical studies, established more than three decades ago, focused on persistent HIV-1 infection. In this issue of the JCI, Bingfeng Liu et al. revisit HIV-specific CAR T cells in an important clinical study that supports broader application of this groundbreaking therapy. Although curative endpoints were not achieved, these findings lay the foundation for augmented approaches applying combinatorial technologies including antigen supplementation.
Collapse
Affiliation(s)
- Christopher W Peterson
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
34
|
Haeseleer F, Fukazawa Y, Park H, Varco-Merth B, Rust BJ, Smedley JV, Eichholz K, Peterson CW, Mason R, Kiem HP, Roederer M, Picker LJ, Okoye AA, Corey L. Immune inactivation of anti-simian immunodeficiency virus chimeric antigen receptor T cells in rhesus macaques. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:304-319. [PMID: 34485613 PMCID: PMC8403686 DOI: 10.1016/j.omtm.2021.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/15/2021] [Indexed: 12/04/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapies are being investigated as potential HIV cures and designed to target HIV reservoirs. Monoclonal antibodies (mAbs) targeting the simian immunodeficiency virus (SIV) envelope allowed us to investigate the potency of single-chain variable fragment (scFv)-based anti-SIV CAR T cells. In vitro, CAR T cells expressing the scFv to both the variable loop 1 (V1) or V3 of the SIV envelope were highly potent at eliminating SIV-infected T cells. However, in preclinical studies, in vivo infusion of these CAR T cells in rhesus macaques (RMs) resulted in lack of expansion and no detectable in vivo antiviral activity. Injection of envelope-expressing antigen-presenting cells (APCs) 1 week post-CAR T cell infusion also failed to stimulate CAR T cell expansion in vivo. To investigate this in vitro versus in vivo discrepancy, we examined host immune responses directed at CAR T cells. A humoral immune response against the CAR scFv was detected post-infusion of the anti-SIV CAR T cells; anti-SIV IgG antibodies present in plasma of SIV-infected animals were associated with inhibited CAR T cell effector functions. These data indicate that lack of in vivo expansion and efficacy of CAR T cells might be due to antibodies blocking the interaction between the CAR scFv and its epitope.
Collapse
Affiliation(s)
- Françoise Haeseleer
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yoshinori Fukazawa
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Haesun Park
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Benjamin Varco-Merth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Blake J Rust
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jeremy V Smedley
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Karsten Eichholz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Christopher W Peterson
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rosemarie Mason
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hans-Peter Kiem
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.,Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Afam A Okoye
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Lawrence Corey
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
35
|
Interests of the Non-Human Primate Models for HIV Cure Research. Vaccines (Basel) 2021; 9:vaccines9090958. [PMID: 34579195 PMCID: PMC8472852 DOI: 10.3390/vaccines9090958] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Non-human primate (NHP) models are important for vaccine development and also contribute to HIV cure research. Although none of the animal models are perfect, NHPs enable the exploration of important questions about tissue viral reservoirs and the development of intervention strategies. In this review, we describe recent advances in the use of these models for HIV cure research and highlight the progress that has been made as well as limitations using these models. The main NHP models used are (i) the macaque, in which simian immunodeficiency virus (SIVmac) infection displays similar replication profiles as to HIV in humans, and (ii) the macaque infected by a recombinant virus (SHIV) consisting of SIVmac expressing the HIV envelope gene serving for studies analyzing the impact of anti-HIV Env broadly neutralizing antibodies. Lessons for HIV cure that can be learned from studying the natural host of SIV are also presented here. An overview of the most promising and less well explored HIV cure strategies tested in NHP models will be given.
Collapse
|
36
|
Liu B, Zhang W, Xia B, Jing S, Du Y, Zou F, Li R, Lu L, Chen S, Li Y, Hu Q, Lin Y, Zhang Y, He Z, Zhang X, Chen X, Peng T, Tang X, Cai W, Pan T, Li L, Zhang H. Broadly neutralizing antibody-derived CAR-T cells reduce viral reservoir in HIV-1-infected individuals. J Clin Invest 2021; 131:e150211. [PMID: 34375315 DOI: 10.1172/jci150211] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-modified T cells have emerged as a novel approach to treat malignant tumors. This strategy has also been proposed for the treatment of HIV-1 infection. We have developed a broadly neutralizing antibody (bNAb)-derived CAR-T cell therapy which can exerted specific cytotoxic activity against HIV-1-infected cells. METHODS We conducted an open-label trial of the safety, side-effect profile, pharmacokinetic properties, and antiviral activity of bNAb-derived CAR-T cell therapy in HIV-1-infected individuals who were undergoing analytical interruption of antiretroviral therapy (ART). RESULTS A total of 14 participants completed only a single administration of bNAb-derived CAR-T cells. CAR-T administration was safe and well tolerated. Six participants discontinued ART, and viremia rebound occurred in all of them, with a 5.3-week median time. Notably, the cell-associated viral RNA and intact proviruses decreased significantly after CAR-T treatment. Analyses of HIV-1 variants before or after CAR-T administration suggested that CAR-T cells exerted pressure on rebound viruses, resulting in a selection of viruses with less diversity and mutations against CAR-T-mediated cytotoxicity. CONCLUSIONS No safety concerns were identified with adoptive transfer of bNAb-derived CAR-T cells. They reduced viral reservoir. All the rebounds were due to preexisting or emergence of viral escape mutations. TRIAL REGISTRATION ClinicalTrials.gov number, NCT03240328. FUNDING Ministry of Science and Technology of China, National Natural Science Foundation of China, and Department of Science and Technology of Guangdong Province.
Collapse
Affiliation(s)
- Bingfeng Liu
- Institute of Human Virology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wanying Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Baijin Xia
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuliang Jing
- Institute of Human Virology, Institute of Human Virology of Zhongshan School of Medicine Zhongshan Schoo, Guangzhou, China
| | - Yingying Du
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Fan Zou
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Rong Li
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Lu
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shaozhen Chen
- Infectious Diseases Center, Guangzhou Eighth People's Hospital, Guangzhou, China
| | - Yonghong Li
- Infectious Diseases Center, Guangzhou Eighth People's Hospital, Guangzhou, China
| | - Qifei Hu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yingtong Lin
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhangping He
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Xu Zhang
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Xiejie Chen
- Infectious Diseases Center, Guangzhou Eighth People's Hospital, Guangzhou, China
| | - Tao Peng
- State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Xiaoping Tang
- department of infectious disease, Institute of Infectious Disease, Guangzhou Eighth People's Hospital, Guangz, guangzhou, China
| | - Weiping Cai
- Institute of Infectious Disease, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ting Pan
- Institute of Human Virology, Zhongshan medicine shcool, Sun Yat-sen University, Guangzhou, China
| | - Linghua Li
- Institute of Infectious Disease, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hui Zhang
- Institute of Human Virology of Zhongshan School of Medicine, Guangzhou, China
| |
Collapse
|
37
|
|