1
|
Wu Y, Liang X, Sun Y, Ning J, Dai Y, Jin S, Xu Y, Chen S, Pan L. A general pHLA-CD80 scaffold fusion protein to promote efficient antigen-specific T cell-based immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200827. [PMID: 39027379 PMCID: PMC11255371 DOI: 10.1016/j.omton.2024.200827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/23/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024]
Abstract
Inadequate antigen-specific T cells activation hampers immunotherapy due to complex antigen presentation. In addition, therapeutic in vivo T cell expansion is constrained by slow expansion rates and limited functionality. Herein, we introduce a model fusion protein termed antigen-presenting cell-mimic fusion protein (APC-mimic), designed to greatly mimicking the natural antigen presentation pattern of antigen-presenting cells and directly expand T cells both in vitro and in vivo. The APC-mimic comprises the cognate peptide-human leukocyte antigen (pHLA) complex and the co-stimulatory marker CD80, which are natural ligands on APCs. Following a single stimulation, APC-mimic leads to an approximately 400-fold increase in the polyclonal expansion of antigen-specific T cells compared with the untreated group in vitro without the requirement for specialized antigen-presenting cells. Through the combination of single-cell TCR sequencing (scTCR-seq) and single-cell RNA sequencing (scRNA-seq), we identify an approximately 600-fold monoclonal expansion clonotype among these polyclonal clonotypes. It also exhibits suitability for in vivo applications confirmed in the OT-1 mouse model. Furthermore, T cells expanded by APC-mimic effectively inhibits tumor growth in adoptive cell transfer (ACT) murine models. These findings pave the way for the versatile APC-mimic platform for personalized therapeutics, enabling direct expansion of polyfunctional antigen-specific T cell subsets in vitro and in vivo.
Collapse
Affiliation(s)
- Yue Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao Liang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanping Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiangtao Ning
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yukun Dai
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shijie Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingchun Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shuqing Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Precision Medicine on Tumor Therapeutics, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311200, China
| | - Liqiang Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
2
|
Sharma R, Amdare NP, Ghosh A, Schloss J, Sidney J, Garforth SJ, Lopez Y, Celikgil A, Sette A, Almo SC, DiLorenzo TP. Structural and biochemical analysis of highly similar HLA-B allotypes differentially associated with type 1 diabetes. J Biol Chem 2024; 300:107702. [PMID: 39173948 PMCID: PMC11422593 DOI: 10.1016/j.jbc.2024.107702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease involving T cell-mediated destruction of the insulin-producing beta cells in the pancreatic islets of Langerhans. CD8+ T cells, responding to beta cell peptides presented by class I major histocompatibility complex (MHC) molecules, are important effectors leading to beta cell elimination. Human leukocyte antigen (HLA) B∗39:06, B∗39:01, and B∗38:01 are closely related class I MHC allotypes that nonetheless show differential association with T1D. HLA-B∗39:06 is the most predisposing of all HLA class I molecules and is associated with early age at disease onset. B∗39:01 is also associated with susceptibility to T1D, but to a lesser extent, though differing from B∗39:06 by only two amino acids. HLA-B∗38:01, in contrast, is associated with protection from the disease. Upon identifying a peptide that binds to both HLA-B∗39:06 and B∗39:01, we determined the respective X-ray structures of the two allotypes presenting this peptide to 1.7 Å resolution. The peptide residues available for T cell receptor contact and those serving as anchors were identified. Analysis of the F pocket of HLA-B∗39:06 and B∗39:01 provided an explanation for the distinct peptide C terminus preferences of the two allotypes. Structure-based modeling of the protective HLA-B∗38:01 suggested a potential reason for its peptide preferences and its reduced propensity to present 8-mer peptides compared to B∗39:06. Notably, the three allotypes showed differential binding to peptides derived from beta cell autoantigens. Taken together, our findings should facilitate identification of disease-relevant candidate T cell epitopes and structure-guided therapeutics to interfere with peptide binding.
Collapse
Affiliation(s)
- Ruby Sharma
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Nitin P Amdare
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Agnidipta Ghosh
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jennifer Schloss
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Scott J Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Yessenia Lopez
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Alev Celikgil
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA; Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, La Jolla, California, USA
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA.
| | - Teresa P DiLorenzo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA; Division of Endocrinology and Diabetes, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA; Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, USA; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
3
|
Mi T, Soerens AG, Alli S, Kang TG, Vasandan AB, Wang Z, Vezys V, Kimura S, Iacobucci I, Baylin SB, Jones PA, Hiner C, Mueller A, Goldstein H, Mullighan CG, Zebley CC, Masopust D, Youngblood B. Conserved epigenetic hallmarks of T cell aging during immunity and malignancy. NATURE AGING 2024; 4:1053-1063. [PMID: 38867059 PMCID: PMC11333289 DOI: 10.1038/s43587-024-00649-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
Chronological aging correlates with epigenetic modifications at specific loci, calibrated to species lifespan. Such 'epigenetic clocks' appear conserved among mammals, but whether they are cell autonomous and restricted by maximal organismal lifespan remains unknown. We used a multilifetime murine model of repeat vaccination and memory T cell transplantation to test whether epigenetic aging tracks with cellular replication and if such clocks continue 'counting' beyond species lifespan. Here we found that memory T cell epigenetic clocks tick independently of host age and continue through four lifetimes. Instead of recording chronological time, T cells recorded proliferative experience through modification of cell cycle regulatory genes. Applying this epigenetic profile across a range of human T cell contexts, we found that naive T cells appeared 'young' regardless of organism age, while in pediatric patients, T cell acute lymphoblastic leukemia appeared to have epigenetically aged for up to 200 years. Thus, T cell epigenetic clocks measure replicative history and can continue to accumulate well-beyond organismal lifespan.
Collapse
Affiliation(s)
- Tian Mi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Andrew G Soerens
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Shanta Alli
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tae Gun Kang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Anoop Babu Vasandan
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhaoming Wang
- Department of Computational Biology and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Vaiva Vezys
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Shunsuke Kimura
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephen B Baylin
- The Sidney Kimmel Comprehensive Cancer Institute, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Peter A Jones
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Christopher Hiner
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - April Mueller
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Harris Goldstein
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Caitlin C Zebley
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Ben Youngblood
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
4
|
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
|
5
|
Uslu S, Lee UJ, Tavakolpour S, Abousaway O, Nili A, Bass L, Purwar P, Lacson E, Berland L, Kuhnast A, Clark LM, Picard D, Rakhshandehroo T, Mantri SR, Moravej H, Rashidian M. Development of a Stable Peptide-Major Histocompatibility Complex (MHC) via Sortase and Click Chemistry. ACS Pharmacol Transl Sci 2024; 7:1746-1757. [PMID: 38898944 PMCID: PMC11184609 DOI: 10.1021/acsptsci.3c00268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 06/21/2024]
Abstract
T cells play a crucial role in antitumor immune responses and the clearance of infected cells. They identify their targets through the binding of T-cell receptors (TCRs) to peptide-major histocompatibility complex (pMHC) molecules present in cancer cells, infected cells, and antigen-presenting cells. This interaction is often weak, requiring multimeric pMHC molecules to enhance the avidity for identifying antigen-specific T cells. Current exchangeable pMHC-I tetramerization methods may overlook TCRs recognizing less stable yet immunogenic peptides. In vivo applications targeting antigen-specific T cells demand the genetic synthesis of a pMHC fusion for each unique peptide antigen, which poses a significant challenge. To address these challenges, we developed a sortase and click chemistry-mediated approach for generating stable pMHC molecules. Leveraging sortase technology, we introduced an azide click-handle near the N-terminus of β2m, proximal to the MHC-peptide-binding groove. Simultaneously, the peptide was engineered with a multi glycine linker and a C-terminal alkyne click-handle. Azide-alkyne click reactions efficiently immobilized the peptide onto the MHC molecule, providing a versatile and efficient method for pMHC generation. The resulting peptide-clicked-MHC specifically binds to its cognate TCR and remains stable for over 3 months at 4 °C in the absence of any additional free peptide. The stability of the pMHC and its affinity to cognate TCRs are influenced by the linker's nature and length. Multi glycine linkers outperform poly(ethylene glycol) (PEG) linkers in this regard. This technology expands the toolkit for identifying and targeting antigen-specific T cells, enhancing our understanding of cancer-specific immune responses, and has the potential to streamline the development of personalized immunotherapies.
Collapse
Affiliation(s)
- Safak
C. Uslu
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
- Medical
Scientist Training Program, Hacettepe University
Faculty of Medicine, Ankara 06230, Turkey
| | - Uk-Jae Lee
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Soheil Tavakolpour
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Omar Abousaway
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Ali Nili
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Lily Bass
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Pragallabh Purwar
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Edward Lacson
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Lea Berland
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
- CNRS,
INSERM, IRCAN, Université Côte d’Azur, 06100 Nice, France
| | - Adrien Kuhnast
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Louise M. Clark
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Delia Picard
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Taha Rakhshandehroo
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Shreya R. Mantri
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Heydar Moravej
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Mohammad Rashidian
- Department
of Cancer Immunology and Virology, Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
- Department
of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02215, United States
- Parker
Institute for Cancer Immunotherapy, San Francisco, California 94129, United States
| |
Collapse
|
6
|
Li WH, Su JY, Zhang BD, Zhao L, Zhuo SH, Wang TY, Hu HG, Li YM. Myeloid Cell-Triggered In Situ Cell Engineering for Robust Vaccine-Based Cancer Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308155. [PMID: 38295870 DOI: 10.1002/adma.202308155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/27/2023] [Indexed: 02/08/2024]
Abstract
Following the success of the dendritic cell (DC) vaccine, the cell-based tumor vaccine shows its promise as a vaccination strategy. Except for DC cells, targeting other immune cells, especially myeloid cells, is expected to address currently unmet clinical needs (e.g., tumor types, safety issues such as cytokine storms, and therapeutic benefits). Here, it is shown that an in situ injected macroporous myeloid cell adoptive scaffold (MAS) not only actively delivers antigens (Ags) that are triggered by scaffold-infiltrating cell surface thiol groups but also releases granulocyte-macrophage colony-stimulating factor and other adjuvant combos. Consequently, this promotes cell differentiation, activation, and migration from the produced monocyte and DC vaccines (MASVax) to stimulate antitumor T-cell immunity. Neoantigen-based MASVax combined with immune checkpoint blockade induces rejection of established tumors and long-term immune protection. The combined depletion of immunosuppressive myeloid cells further enhances the efficacy of MASVax, indicating the potential of myeloid cell-based therapies for immune enhancement and normalization treatment of cancer.
Collapse
Affiliation(s)
- Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Jing-Yun Su
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Bo-Dou Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Lang Zhao
- Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Shao-Hua Zhuo
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Tian-Yang Wang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Hong-Guo Hu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
- Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
7
|
Su H, Anthony-Gonda K, Orentas RJ, Dropulić B, Goldstein H. Generation of Anti-HIV CAR-T Cells for Preclinical Research. Methods Mol Biol 2024; 2807:287-298. [PMID: 38743236 DOI: 10.1007/978-1-0716-3862-0_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The inability of people living with HIV (PLWH) to eradicate human immunodeficiency virus (HIV) infection is due in part to the inadequate HIV-specific cellular immune response. The antiviral function of cytotoxic CD8+ T cells, which are crucial for HIV control, is impaired during chronic viral infection because of viral escape mutations, immune exhaustion, HIV antigen downregulation, inflammation, and apoptosis. In addition, some HIV-infected cells either localize to tissue sanctuaries inaccessible to CD8+ T cells or are intrinsically resistant to CD8+ T cell killing. The novel design of synthetic chimeric antigen receptors (CARs) that enable T cells to target specific antigens has led to the development of potent and effective CAR-T cell therapies. While initial clinical trials using anti-HIV CAR-T cells performed over 20 years ago showed limited anti-HIV effects, the improved CAR-T cell design, which enabled its success in treating cancer, has reinstated CAR-T cell therapy as a strategy for HIV cure with notable progress being made in the recent decade.Effective CAR-T cell therapy against HIV infection requires the generation of anti-HIV CAR-T cells with potent in vivo activity against HIV-infected cells. Preclinical evaluation of anti-HIV efficacy of CAR-T cells and their safety is fundamental for supporting the initiation of subsequent clinical trials in PLWH. For these preclinical studies, we developed a novel humanized mouse model supporting in vivo HIV infection, the development of viremia, and the evaluation of novel HIV therapeutics. Preclinical assessment of anti-HIV CAR-T cells using this mouse model involves a multistep process including peripheral blood mononuclear cells (PBMCs) harvested from human donors, T cell purification, ex vivo T cell activation, transduction with lentiviral vectors encoding an anti-HIV CAR, CAR-T cell expansion and infusion in mice intrasplenically injected with autologous PBMCs followed by the determination of CAR-T cell capacity for HIV suppression. Each of the steps described in the following protocol were optimized in the lab to maximize the quantity and quality of the final anti-HIV CAR-T cell products.
Collapse
MESH Headings
- Humans
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Mice
- HIV Infections/immunology
- HIV Infections/therapy
- HIV Infections/virology
- Immunotherapy, Adoptive/methods
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- CD8-Positive T-Lymphocytes/immunology
- HIV-1/immunology
- T-Lymphocytes/immunology
- Transduction, Genetic
Collapse
Affiliation(s)
- Hang Su
- Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | | - Harris Goldstein
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
8
|
Schardt JS, Walseng E, Le K, Yang C, Shah P, Fu Y, Alam K, Kelton CR, Gu Y, Huang F, Lin J, Liu W, Dippel A, Zhang H, Mulgrew K, Pryts S, Chennupati V, Chen HC, Denham J, Chen X, Pradhan P, Wu Y, Hardman C, Zhao C, Kierny M, Song Y, Dovedi SJ, Cemerski S, Mazor Y. IL-2-armored peptide-major histocompatibility class I bispecific antibodies redirect antiviral effector memory CD8+ T cells to induce potent anti-cancer cytotoxic activity with limited cytokine release. MAbs 2024; 16:2395499. [PMID: 39205483 PMCID: PMC11364066 DOI: 10.1080/19420862.2024.2395499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
T cell engagers (TCEs) are becoming an integral class of biological therapeutic owing to their highly potent ability to eradicate cancer cells. Nevertheless, the widespread utility of classical CD3-targeted TCEs has been limited by narrow therapeutic index (TI) linked to systemic CD4+ T cell activation and aberrant cytokine release. One attractive approach to circumvent the systemic activation of pan CD3+ T cells and reduce the risk of cytokine release syndrome is to redirect specific subsets of T cells. A promising strategy is the use of peptide-major histocompatibility class I bispecific antibodies (pMHC-IgGs), which have emerged as an intriguing modality of TCE, based on their ability to selectively redirect highly reactive viral-specific effector memory cytotoxic CD8+ T cells to eliminate cancer cells. However, the relatively low frequency of these effector memory cells in human peripheral blood mononuclear cells (PBMCs) may hamper their redirection as effector cells for clinical applications. To mitigate this potential limitation, we report here the generation of a pMHC-IgG derivative known as guided-pMHC-staging (GPS) carrying a covalent fusion of a monovalent interleukin-2 (IL-2) mutein (H16A, F42A). Using an anti-epidermal growth factor receptor (EGFR) arm as a proof-of-concept, tumor-associated antigen paired with a single-chain HLA-A *02:01/CMVpp65 pMHC fusion moiety, we demonstrate in vitro that the IL-2-armored GPS modality robustly expands CMVpp65-specific CD8+ effector memory T cells and induces potent cytotoxic activity against target cancer cells. Similar to GPS, IL-2-armored GPS molecules induce modulated T cell activation and reduced cytokine release profile compared to an analogous CD3-targeted TCE. In vivo we show that IL-2-armored GPS, but not the corresponding GPS, effectively expands grafted CMVpp65 CD8+ T cells from unstimulated human PBMCs in an NSG mouse model. Lastly, we demonstrate that the IL-2-armored GPS modality exhibits a favorable developability profile and monoclonal antibody-like pharmacokinetic properties in human neonatal Fc receptor transgenic mice. Overall, IL-2-armored GPS represents an attractive approach for treating cancer with the potential for inducing vaccine-like antiviral T cell expansion, immune cell redirection as a TCE, and significantly widened TI due to reduced cytokine release.
Collapse
Affiliation(s)
- John S. Schardt
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Even Walseng
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Kim Le
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Chunning Yang
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Pooja Shah
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Ying Fu
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Kausar Alam
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | | | - Yu Gu
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Fengying Huang
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Jia Lin
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Wenhai Liu
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Andrew Dippel
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Hanzhi Zhang
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | | | - Stacy Pryts
- Oncology ICC, AstraZeneca, Gaithersburg, MD, USA
| | | | - Hung-Chang Chen
- Data Science and Advanced Analytics, AstraZeneca, Gaithersburg, MD, USA
| | | | | | | | - Yuling Wu
- Oncology ICC, AstraZeneca, Cambridge, UK
| | - Colin Hardman
- Discovery Bioanalysis, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Chihao Zhao
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Michael Kierny
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Yang Song
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| | - Simon J. Dovedi
- Data Science and Advanced Analytics, AstraZeneca, Gaithersburg, MD, USA
| | | | - Yariv Mazor
- R&D Biologics Engineering, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
9
|
Mahata D, Mukherjee D, Biswas D, Basak S, Basak AJ, Jamir I, Pandey N, Khatoon H, Samanta D, Basak A, Mukherjee G. Activation and differentiation of cognate T cells by a dextran-based antigen-presenting system for cancer immunotherapy. Eur J Immunol 2023; 53:e2350528. [PMID: 37698527 DOI: 10.1002/eji.202350528] [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: 04/14/2023] [Revised: 08/14/2023] [Accepted: 09/11/2023] [Indexed: 09/13/2023]
Abstract
Immunotherapeutic modulation of antigen-specific T-cell responses instead of the whole repertoire helps avoid immune-related adverse events. We have developed an artificial antigen-presenting system (aAPS) where multiple copies of a multimeric peptide-MHC class I complex presenting a murine class I MHC restricted ovalbumin-derived peptide (signal 1), along with a costimulatory ligand (signal 2) are chemically conjugated to a dextran backbone. Cognate naive CD8+ T cells, when treated with this aAPS underwent significant expansion and showed an activated phenotype. Furthermore, elevated expression of effector cytokines led to the differentiation of these cells to cytotoxic T lymphocytes which resulted in target cell lysis, indicative of the functional efficacy of the aAPS. CD8+ T cells with decreased proliferative potential due to repeated antigenic stimulation could also be re-expanded by the developed aAPS. Thus, the developed aAPS warrants further engineering for future application as a rapidly customizable personalized immunotherapeutic agent, incorporating patient-specific MHC-restricted tumor antigens and different costimulatory signals to modulate both naive and antigen-experienced but exhausted tumor-specific T cells in cancer.
Collapse
Affiliation(s)
- Dhrubajyoti Mahata
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Debangshu Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Debarati Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Shyam Basak
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Aditya Jyoti Basak
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Imlilong Jamir
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Nidhi Pandey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Huma Khatoon
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Dibyendu Samanta
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Amit Basak
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Gayatri Mukherjee
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| |
Collapse
|
10
|
McMahon‑Cole H, Johnson A, Sadat Aghamiri S, Helikar T, Crawford LB. Modeling and Remodeling the Cell: How Digital Twins and HCMV Can Elucidate the Complex Interactions of Viral Latency, Epigenetic Regulation, and Immune Responses. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023; 10:141-151. [PMID: 37901689 PMCID: PMC10601359 DOI: 10.1007/s40588-023-00201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 10/31/2023]
Abstract
Purpose of Review Human cytomegalovirus (HCMV), while asymptomatic in most, causes significant complications during fetal development, following transplant or in immunosuppressed individuals. The host-virus interactions regulating viral latency and reactivation and viral control of the cellular environment (immune regulation, differentiation, epigenetics) are highly complex. Understanding these processes is essential to controlling infection and can be leveraged as a novel approach for understanding basic cell biology. Recent Findings Immune digital twins (IDTs) are digital simulations integrating knowledge of human immunology, physiology, and patient-specific clinical data to predict individualized immune responses and targeted treatments. Recent studies used IDTs to elucidate mechanisms of T cells, dendritic cells, and epigenetic control-all key to HCMV biology. Summary Here, we discuss how leveraging the unique biology of HCMV and IDTs will clarify immune response dynamics, host-virus interactions, and viral latency and reactivation and serve as a powerful IDT-validation platform for individualized and holistic health management.
Collapse
Affiliation(s)
- Hana McMahon‑Cole
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Alicia Johnson
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Sara Sadat Aghamiri
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Lindsey B. Crawford
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Center for Virology, Lincoln, NE, USA
- Nebraska Center for Integrated Biomolecular Communication, Lincoln, NE, USA
| |
Collapse
|
11
|
Salek-Ardakani S, Zajonc DM, Croft M. Agonism of 4-1BB for immune therapy: a perspective on possibilities and complications. Front Immunol 2023; 14:1228486. [PMID: 37662949 PMCID: PMC10469789 DOI: 10.3389/fimmu.2023.1228486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Costimulatory receptors on immune cells represent attractive targets for immunotherapy given that these molecules can increase the frequency of individual protective immune cell populations and their longevity, as well as enhance various effector functions. 4-1BB, a member of the TNF receptor superfamily, also known as CD137 and TNFRSF9, is one such molecule that is inducible on several cell types, including T cells and NK cells. Preclinical studies in animal models have validated the notion that stimulating 4-1BB with agonist reagents or its natural ligand could be useful to augment conventional T cell and NK cell immunity to protect against tumor growth and against viral infection. Additionally, stimulating 4-1BB can enhance regulatory T cell function and might be useful in the right context for suppressing autoimmunity. Two human agonist antibodies to 4-1BB have been produced and tested in clinical trials for cancer, with variable results, leading to the production of a wealth of second-generation antibody constructs, including bi- and multi-specifics, with the hope of optimizing activity and selectivity. Here, we review the progress to date in agonism of 4-1BB, discuss the complications in targeting the immune system appropriately to elicit the desired activity, together with challenges in engineering agonists, and highlight the untapped potential of manipulating this molecule in infectious disease and autoimmunity.
Collapse
Affiliation(s)
| | - Dirk M. Zajonc
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California (UC) San Diego, La Jolla, CA, United States
| |
Collapse
|
12
|
Britsch I, van Wijngaarden AP, Helfrich W. Applications of Anti-Cytomegalovirus T Cells for Cancer (Immuno)Therapy. Cancers (Basel) 2023; 15:3767. [PMID: 37568582 PMCID: PMC10416821 DOI: 10.3390/cancers15153767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Infection with cytomegalovirus (CMV) is highly prevalent in the general population and largely controlled by CD8pos T cells. Intriguingly, anti-CMV T cells accumulate over time to extraordinarily high numbers, are frequently present as tumor-resident 'bystander' T cells, and remain functional in cancer patients. Consequently, various strategies for redirecting anti-CMV CD8pos T cells to eliminate cancer cells are currently being developed. Here, we provide an overview of these strategies including immunogenic CMV peptide-loading onto endogenous HLA complexes on cancer cells and the use of tumor-directed fusion proteins containing a preassembled CMV peptide/HLA-I complex. Additionally, we discuss conveying the advantageous characteristics of anti-CMV T cells in adoptive cell therapy. Utilization of anti-CMV CD8pos T cells to generate CAR T cells promotes their in vivo persistence and expansion due to appropriate co-stimulation through the endogenous (CMV-)TCR signaling complex. Designing TCR-engineered T cells is more challenging, as the artificial and endogenous TCR compete for expression. Moreover, the use of expanded/reactivated anti-CMV T cells to target CMV peptide-expressing glioblastomas is discussed. This review highlights the most important findings and compares the benefits, disadvantages, and challenges of each strategy. Finally, we discuss how anti-CMV T cell therapies can be further improved to enhance treatment efficacy.
Collapse
Affiliation(s)
| | | | - Wijnand Helfrich
- Department of Surgery, Translational Surgical Oncology, University of Groningen, UMC Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (I.B.)
| |
Collapse
|
13
|
Fraker S, Atkinson B, Heredia A. Humanized mouse models for preclinical evaluation of HIV cure strategies. AIDS Rev 2022; 24:139-151. [PMID: 35622983 PMCID: PMC9643647 DOI: 10.24875/aidsrev.22000013] [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: 04/04/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
Abstract
Although the world is currently focused on the COVID-19 pandemic, HIV/AIDS remains a significant threat to public health. To date, the HIV/AIDS pandemic has claimed the lives of over 36 million people, while nearly 38 million people are currently living with the virus. Despite the undeniable success of antiretroviral therapy (ART) in controlling HIV, the medications are not curative. Soon after initial infection, HIV integrates into the genome of infected cells as a provirus, primarily, within CD4+ T lymphocytes and tissue macrophages. When not actively transcribed, the provirus is referred to as a latent reservoir because it is hidden to the immune system and ART. Following ART discontinuation, HIV may emerge from the replication-competent proviruses and resumes the infection of healthy cells. Thus, these latent reservoirs are a major obstacle to an HIV cure, and their removal remains a priority. A vital aspect in the development of curative therapies is the demonstration of efficacy in an animal model, such as the humanized mouse model. Therefore, optimization, standardization, and validation of the humanized mouse model are a priority. The purpose of this review article is to provide an update on existing humanized mouse models, highlighting the advantages and disadvantages of each as they pertain to HIV cure studies and to review the approaches to curative therapies that are under investigation.
Collapse
Affiliation(s)
- Sally Fraker
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Benjamin Atkinson
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Alonso Heredia
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
14
|
Favaretto F, Bettini S, Busetto L, Milan G, Vettor R. Adipogenic progenitors in different organs: Pathophysiological implications. Rev Endocr Metab Disord 2022; 23:71-85. [PMID: 34716543 PMCID: PMC8873140 DOI: 10.1007/s11154-021-09686-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/14/2022]
Abstract
In physiological conditions, the adipose organ resides in well-defined areas, where it acts providing an energy supply and as an endocrine organ involved in the control of whole-body energy metabolism. Adipose tissue adipokines connect the body's nutritional status to the regulation of energy balance. When it surrounds organs, it provides also for mechanical protection. Adipose tissue has a complex and heterogenous cellular composition that includes adipocytes, adipose tissue-derived stromal and stem cells (ASCs) which are mesenchymal stromal cells, and endothelial and immune cells, which signal to each other and to other tissues to maintain homeostasis. In obesity and in other nutrition related diseases, as well as in age-related diseases, biological and functional changes of adipose tissue give rise to several complications. Obesity triggers alterations of ASCs, impairing adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance and other metabolic disorders. Adipose tissue grows by hyperplasia recruiting new ASCs and by hypertrophy, up to its expandability limit. To overcome this limitation and to store the excess of nutrients, adipose tissue develops ectopically, involving organs such as muscle, bone marrow and the heart. The origin of ectopic adipose organ is not clearly elucidated, and a possible explanation lies in the stimulation of the adipogenic differentiation of mesenchymal precursor cells which normally differentiate toward a lineage specific for the organ in which they reside. The chronic exposition of these newly-formed adipose depots to the pathological environment, will confer to them all the phenotypic characteristics of a dysfunctional adipose tissue, perpetuating the organ alterations. Visceral fat, but also ectopic fat, either in the liver, muscle or heart, can increase the risk of developing insulin resistance, type 2 diabetes, and cardiovascular diseases. Being able to prevent and to target dysfunctional adipose tissue will avoid the progression towards the complications of obesity and other nutrition-related diseases. The aim of this review is to summarize some of the knowledge regarding the presence of adipose tissue in particular tissues (where it is not usually present), describing the composition of its adipogenic precursors, and the interactions responsible for the development of organ pathologies.
Collapse
Affiliation(s)
- Francesca Favaretto
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Silvia Bettini
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Luca Busetto
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Gabriella Milan
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Roberto Vettor
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| |
Collapse
|