1
|
Koftori D, Kaur C, Mora Bitria L, Zhang Y, Hadcocks L, Yan AWC, Burzyński PF, Ladell K, Speiser DE, Pollock KM, Macallan D, Asquith B. Two distinct subpopulations of human stem-like memory T cells exhibit complementary roles in self-renewal and clonal longevity. PLoS Biol 2025; 23:e3003179. [PMID: 40540506 DOI: 10.1371/journal.pbio.3003179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 04/23/2025] [Indexed: 06/22/2025] Open
Abstract
T stem cell-like memory cells (TSCM cells) are considered to be essential for the maintenance of immune memory. The TSCM population has been shown to have the key properties of a stem cell population: multipotency, self-renewal and clonal longevity. Here we show that no single population has all these stem cell properties, instead the properties are distributed. We show that the human TSCM population consists of two distinct cell subpopulations which can be distinguished by the level of their CD95 expression (CD95int and CD95hi). Crucially, using long-term in vivo labelling of human volunteers, we establish that these are distinct populations rather than transient states of the same population. These two subpopulations have different functional profiles ex vivo, different transcriptional patterns, and different tissue distributions. They also have significantly different TREC content indicating different division histories and we find that the frequency of CD95hi TSCM increases with age. Most importantly, CD95hi and CD95int TSCM cells also have very different dynamics in vivo with CD95hi cells showing considerably higher proliferation but significantly reduced clonal longevity compared with CD95int TSCM. While both TSCM subpopulations exhibit considerable multipotency, no single population of TSCM cells has both the properties of self-renewal and clonal longevity. Instead, the "stemness" of the TSCM population is generated by the complementary dynamic properties of the two subpopulations: CD95int TSCM which have the property of clonal longevity and CD95hi TSCM which have the properties of expansion and self-renewal. We suggest that together, these two populations function as a stem cell population.
Collapse
Affiliation(s)
- Danai Koftori
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Charandeep Kaur
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Laura Mora Bitria
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Yan Zhang
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Linda Hadcocks
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Ada W C Yan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Piotr F Burzyński
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Katrina M Pollock
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Derek Macallan
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
2
|
Abdo L, Batista-Silva LR, Bonamino MH. Cost-effective strategies for CAR-T cell therapy manufacturing. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200980. [PMID: 40291594 PMCID: PMC12022644 DOI: 10.1016/j.omton.2025.200980] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
CAR-T cell therapy has revolutionized cancer treatment, with approvals for conditions like acute B-leukemia, large B cell lymphoma (LBCL), follicular lymphoma (FL), mantle cell lymphoma (MCL), and multiple myeloma. However, its high costs limit accessibility. Key factors driving these costs include the need for personalized, autologous treatments, transportation to specialized facilities, reliance on viral vectors requiring advanced laboratories, and lengthy cell expansion processes. To address these challenges, alternative strategies aim to simplify and reduce production complexity. Non-viral vectors, such as Sleeping Beauty, piggyBac, and CRISPR, delivered via nanoparticles or electroporation, present promising solutions. These methods could streamline manufacturing, eliminate the need for viral vectors, and reduce associated costs. Furthermore, shortening cell expansion periods and optimizing protocols could significantly accelerate production. An emerging approach involves using genetically edited T cells from healthy donors to create universal CAR-T products capable of treating multiple patients. Finally, decentralized point-of-care (POC) manufacturing of CAR-T cells minimize logistical expenses, eliminating the need for complex infrastructure, and enabling localized production closer to patients. This innovative strategy holds potential for broadening access and reducing costs, representing a step toward democratizing CAR-T therapy. Combined, these advances could make this groundbreaking treatment more feasible for healthcare systems worldwide.
Collapse
Affiliation(s)
- Luiza Abdo
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
| | - Leonardo Ribeiro Batista-Silva
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
| | - Martín Hernán Bonamino
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
- Vice-Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
3
|
Yong SB, Ha M, Cho S. Microbiome Metabolite-Incorporated Lipid Nanoparticles Augment CD8 + T Cell Memory Potential and Immunity for mRNA Cancer Vaccines. ACS Biomater Sci Eng 2025. [PMID: 40490465 DOI: 10.1021/acsbiomaterials.5c00738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
Recently, mRNA/lipid nanoparticle (LNP)-based vaccines have been successfully applied to prevent infectious diseases, and several types of neoantigen-encoding mRNA cancer vaccines are currently under clinical trials. While mRNA vaccines effectively induce adaptive immune responses to antigens, mRNA vaccine-induced immunity is shortly maintained, and the longevity of the immune memory, especially improving the CD8+ T cell memory potential, could be even more important. Previously, microbiome metabolites have shown T cell memory potential-augmenting effects via regulating the immunometabolism. Herein, we develop microbiome metabolite-incorporated LNPs (mmi-LNPs) and evaluate their potential to enhance T cell memory responses following mRNA vaccination. In various ionizable LNP formulations, mmi-LNPs elicited more stem cell-like memory T cells (T-SCMs) and augmented central and effector memory T cell responses, which indicates the general applicability of mmi-LNPs. Notably, butyrate-incorporated mmi-LNP exhibited the strongest effects. In conclusion, we suggest microbiome metabolite-incorporated LNP as a next-generation delivery vehicle for mRNA vaccines.
Collapse
Affiliation(s)
- Seok-Beom Yong
- Center for Gene & Cell Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do 28116, Republic of Korea
| | - Minki Ha
- Center for Gene & Cell Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do 28116, Republic of Korea
- College of Pharmacy, Chungbuk National Univeristy, Cheongju 28160, Republic of Korea
| | - Sungchan Cho
- Center for Gene & Cell Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do 28116, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (KUST), Daejeon-si 34113, Republic of Korea
| |
Collapse
|
4
|
Gambella M, Carlomagno S, Raiola AM, Sivori S, Angelucci E. (CAR-)T cell dynamics following chimeric antigen receptor T cells for large B cell lymphoma: a translational tale. Leuk Lymphoma 2025; 66:1036-1044. [PMID: 39945648 DOI: 10.1080/10428194.2025.2456096] [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: 10/25/2024] [Revised: 12/17/2024] [Accepted: 01/15/2025] [Indexed: 05/27/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a breakthrough in the treatment of B-cell malignancies. CAR-T cells infusion generally follows a chemotherapy regimen whose lymphodepleting properties create a favorable environment for the expansion of engineered T cells. While this process appears straightforward, emerging evidence reveals that complex mechanisms, collectively representing immune dynamics following CAR-T cell infusion, influence CAR-T cells behavior. In advance of infusion, a final-product enriched with less stressed CAR-T cells can improve their expansion and persistence, providing a biological rationale for early apheresis and administration. Following infusion, the emergence of dysfunctional CAR-T subpopulations, like regulatory or NK-like CAR-T cells, can impair efficacy. The recovery of non-CAR transduced T cells adds further complexity, as these cells could either impact outcomes or exacerbate complications, such as infections or prolonged cytopenia. In this review, we summarize the latest advances in understanding the immune dynamics following CAR-T cell infusion for large B-cell lymphomas, with a focus on both CAR-engineered and native T cell populations, and their impact on treatment efficacy and patient outcomes.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Animals
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- M Gambella
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genova, Italy
| | - S Carlomagno
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - A M Raiola
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - S Sivori
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genova, Italy
| | - E Angelucci
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
5
|
Webb L, Lofgren M, Patterson T, Watt A, Lajoie J, Zieba A, Fleury M, Liu E, Ding J, Tighe R. Membrane-bound IL-15 co-expression powers a potent and persistent CD70-targeted TRuC T-cell therapy. Front Immunol 2025; 16:1609658. [PMID: 40519930 PMCID: PMC12162932 DOI: 10.3389/fimmu.2025.1609658] [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: 04/10/2025] [Accepted: 05/12/2025] [Indexed: 06/18/2025] Open
Abstract
Introduction Although T-cell immunotherapies have been effective in the treatment of hematological malignancies, solid tumors have proven challenging due to the immunosuppressive microenvironment and lack of viable target antigens. The immune checkpoint ligand CD70, overexpressed in several solid tumors, yet with limited expression in healthy tissue, has emerged as a promising immunotherapeutic target. Method This study describes the generation and preclinical characterization of ADP-520, a high-affinity, fratricide-resistant, CD70-targeted T-cell receptor fusion construct (TRuC) T-cell therapy enhanced with constitutively expressed mbIL-15, a membrane-bound fusion protein comprising interleukin-15 (IL-15) linked to full-length IL-15 receptor-alpha. The phenotypic distribution, expansion and persistence of ADP-520 TRuC T cells were measured in vitro under autonomous and antigen-dependent conditions, with the contributions of TCR and IL-15 signaling pathways ascertained using inhibition assays. Chronic antigen stimulation was used to evaluate exhaustion-resistance, while anti-tumor potency was explored both in vitro and in vivo. Results ADP-520 was found to have potent and antigen-specific activity against hematological and solid CD70-expressing tumors, without apparent fratricide or killing of bystander T cells despite CD70 expression by activated lymphocytes. Engineered co-expression of mbIL-15 augmented antigen-dependent expansion through pro-survival effects and enrichment of an early memory T-cell phenotype, thus enhancing tumor-autonomous, exogenous cytokine-free persistence and bolstering exhaustion resistance during chronic stimulation. mbIL-15 co-expression also enhanced intratumoral T-cell infiltration in vivo for potent and persistent antitumor efficacy. Discussion These findings characterize ADP-520 as a first-in-class, CD70-targeted, fratricide-resistant autologous TRuC T-cell therapy leveraging native TCR signaling combined with constitutive IL-15 signaling to impart T cells with enhanced persistence, tumor penetration, and antitumor efficacy. This makes ADP-520 a promising cell immunotherapy candidate for clinical development, with the potential to overcome hurdles intrinsic to the treatment of solid tumors.
Collapse
Affiliation(s)
- Lindsay Webb
- TCR Therapeutics, Inc., Cambridge, MA, United States
| | | | | | - Amy Watt
- TCR Therapeutics, Inc., Cambridge, MA, United States
- Adaptimmune, Cambridge, MA, United States
| | - Jason Lajoie
- TCR Therapeutics, Inc., Cambridge, MA, United States
| | - Adam Zieba
- TCR Therapeutics, Inc., Cambridge, MA, United States
| | | | - Erica Liu
- TCR Therapeutics, Inc., Cambridge, MA, United States
| | - Jian Ding
- TCR Therapeutics, Inc., Cambridge, MA, United States
| | - Robert Tighe
- TCR Therapeutics, Inc., Cambridge, MA, United States
| |
Collapse
|
6
|
Fuentealba M, Kiprov D, Schneider K, Mu WC, Kumaar PA, Kasler H, Burton JB, Watson M, Halaweh H, King CD, Yüksel ZS, Roska-Pamaong C, Schilling B, Verdin E, Furman D. Multi-Omics Analysis Reveals Biomarkers That Contribute to Biological Age Rejuvenation in Response to Single-Blinded Randomized Placebo-Controlled Therapeutic Plasma Exchange. Aging Cell 2025:e70103. [PMID: 40424097 DOI: 10.1111/acel.70103] [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: 10/24/2024] [Revised: 04/21/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
We conducted a randomized, placebo-controlled trial to assess the safety and biological age (BA) effects of various therapeutic plasma exchange (TPE) regimens in healthy adults over 50. Participants received bi-weekly TPE with or without intravenous immunoglobulin (IVIG), monthly TPE, or placebo. Randomization was based on entry date, and treatments were blinded to maintain objectivity. Primary objectives were to assess long-term TPE safety and changes in biological clocks. Secondary goals included identifying optimal regimens. Exploratory analyses profiled baseline clinical features and longitudinal changes across the epigenome, proteome, metabolome, glycome, immune cytokines, iAge, and immune cell composition. We demonstrate in 42 individuals randomized to various treatment arms or placebo that long-term TPE was found to be safe, with only two adverse events requiring discontinuation and one related to IVIG. TPE significantly improved biological age markers, with 15 epigenetic clocks showing rejuvenation compared to placebo (FDR < 0.05). Biweekly TPE combined with intravenous immunoglobulin (TPE-IVIG) proved most effective, inducing coordinated cellular and molecular responses, reversing age-related immune decline, and modulating proteins linked to chronic inflammation. Integrative analysis identified baseline biomarkers predictive of positive outcomes, suggesting TPE-IVIG is particularly beneficial for individuals with poorer initial health status. This is the first multi-omics study to examine various TPE modalities to slow epigenetic biologic clocks, which demonstrate biological age rejuvenation and the molecular features associated with this rejuvenation. Trial Registration: Registered trial NCT06534450 on clinicaltrials.gov under the purview of the Diagnostic Investigational Review Board.
Collapse
Affiliation(s)
| | - Dobri Kiprov
- Buck Institute for Research on Aging, Novato, California, USA
- Global Apheresis Inc., Mill Valley, California, USA
- Circulate, Seattle, Washington, USA
| | - Kevin Schneider
- Buck Institute for Research on Aging, Novato, California, USA
| | - Wei-Chieh Mu
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Herbert Kasler
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jordan B Burton
- Buck Institute for Research on Aging, Novato, California, USA
| | - Mark Watson
- Buck Institute for Research on Aging, Novato, California, USA
| | - Heather Halaweh
- Buck Institute for Research on Aging, Novato, California, USA
| | | | | | | | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, California, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, California, USA
| |
Collapse
|
7
|
Szopa IM, Majchrzak-Kuligowska K, Pingwara R, Kulka M, Taşdemir M, Gajewska M. A New Method of Canine CD4 + T Lymphocyte Differentiation Towards the Th17 Phenotype with Analysis of Properties and Mitochondrial Activity. Int J Mol Sci 2025; 26:4946. [PMID: 40430086 PMCID: PMC12112516 DOI: 10.3390/ijms26104946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Th17 lymphocytes are a distinct subpopulation of T cells that are characterized by the production of interleukins IL-17, IL-21, IL-22, and IL-26, and high expression of RORγt. These cells play an important role in inflammation and autoimmune diseases. Recent studies using rodent and human models have also highlighted their promising properties as agents in cellular immunotherapy for cancer. However, much less is known about the properties of canine Th17 lymphocytes, despite the domestic dog being an important model used in comparative medicine. In this study, we developed methods of activation and differentiation of canine CD4+ T lymphocytes towards the Th17 phenotype. Additionally, we targeted the Wnt/β-catenin signaling pathway to modulate the efficiency of Th17 cells differentiation. CD4+ T cells were successfully activated with magnetic EpoxyBeads, and in combination with the appropriate programming medium, they acquired the Th17 phenotype. Furthermore, indomethacin, an inhibitor of the Wnt/β-catenin pathway, significantly increased the efficiency of differentiation, causing elevated production of IL-17 and changed T cell metabolism by promoting oxidative phosphorylation. The protocol elaborated in our study provides an efficient method of canine Th17 lymphocyte differentiation. Our findings also suggested that the modification of the Wnt/β-catenin signaling pathway could be a valuable strategy for optimizing canine Th17 cell differentiation and advancing cell-based immunotherapy.
Collapse
Affiliation(s)
- Iwona Monika Szopa
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (K.M.-K.); (R.P.); (M.G.)
| | - Kinga Majchrzak-Kuligowska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (K.M.-K.); (R.P.); (M.G.)
| | - Rafał Pingwara
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (K.M.-K.); (R.P.); (M.G.)
| | - Marek Kulka
- Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Monika Taşdemir
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Małgorzata Gajewska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (K.M.-K.); (R.P.); (M.G.)
| |
Collapse
|
8
|
Chaudhry MZ, Chen E, Man HO, Jones A, Denman R, Yu H, Huang Q, Ilich A, Schreuder J, Navarro S, Tuong ZK, Belz GT. GFI1-driven transcriptional and epigenetic programs maintain CD8 + T cell stemness and persistence. Nat Immunol 2025:10.1038/s41590-025-02151-5. [PMID: 40374731 DOI: 10.1038/s41590-025-02151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 04/03/2025] [Indexed: 05/18/2025]
Abstract
Long-lived memory CD8+ T cells are essential for the control of persistent viral infections. The mechanisms that preserve memory cells are poorly understood. Fate mapping of the transcriptional repressor GFI1 identified that GFI1 was differentially regulated in virus-specific CD8+ T cells and was selectively expressed in stem cell memory and central memory cells. Deletion of GFI1 led to reduced proliferation and progressive loss of memory T cells, which in turn resulted in failure to maintain antigen-specific CD8+ T cell populations following infection with chronic lymphocytic choriomeningitis virus or murine cytomegalovirus. Ablation of GFI1 resulted in downregulation of the transcription factors EOMES and BCL-2 in memory CD8+ T cells. Ectopic expression of EOMES rescued the expression of BCL-2, but the persistence of memory CD8+ T cells was only partially rescued. These findings highlight the critical role of GFI1 in the long-term maintenance of memory CD8+ T cells in persistent infections by sustaining their proliferative potential.
Collapse
Affiliation(s)
- M Zeeshan Chaudhry
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| | - Evelyn Chen
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Hiu On Man
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Aneesha Jones
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Renae Denman
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Huiyang Yu
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Qiutong Huang
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Adrian Ilich
- QIMR Berghofer Medical Research, Herston, Brisbane, Queensland, Australia
| | - Jaring Schreuder
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Severine Navarro
- QIMR Berghofer Medical Research, Herston, Brisbane, Queensland, Australia
| | - Zewen K Tuong
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Gabrielle T Belz
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
9
|
Tichadou A, Lebrault E, Samri A, Baron M, Nakid-Cordero C, Lavergne D, Morin V, Mze O, Balegroune N, Liang X, Choquet S, Guihot A, Legembre P, Roussel M, K-VIROGREF Study Group. Soluble CD95L is a prognostic marker in central nervous system posttransplant lymphoproliferative disorders. Am J Transplant 2025:S1600-6135(25)00264-3. [PMID: 40373880 DOI: 10.1016/j.ajt.2025.05.010] [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: 12/29/2024] [Revised: 04/22/2025] [Accepted: 05/07/2025] [Indexed: 05/17/2025]
Abstract
CD95L is a transmembrane cytokine mainly expressed by activated T and natural killer cells to contract the immune response through cell-cell contact. Conversely, after cleavage by metalloproteases, this ligand releases a soluble CD95L (sCD95L) that stimulates the immune response and its antitumor activity. In posttransplant lymphoproliferative disorders (PTLDs), we hypothesized that the concentration of sCD95L could exert a biological function and affect clinical outcomes by modulating the immune response. Using the K-VIROGREF biobank, we quantified sCD95L in 163 patients with PTLD, 16 transplant controls, and 28 healthy donors. Transplant recipients had higher plasma levels of sCD95L than healthy donors. More interestingly, patients with PTLD and high concentration of sCD95L had better clinical outcomes than patients with lower concentration, particularly those with central nervous system (CNS) involvement, who are known to have poor survival. At the cellular level, only natural killer and natural killer T-like cell fractions were reduced in the blood of patients with CNS-PTLD and high concentration of sCD95L, suggesting that sCD95L may either promote the trafficking of these cells within tumors or modulate their differentiation/survival. In conclusion, we showed in this exploratory analysis that plasma concentration of sCD95L might be a prognostic marker in patients with PTLD, particularly in those with CNS involvement.
Collapse
Affiliation(s)
- Antoine Tichadou
- Hématologie Clinique et Thérapie Cellulaire, Hôpitaux Universitaires de Marseille Conception, Marseille, France
| | - Eden Lebrault
- UMR CNRS 7276, INSERM U1262, CRIBL, Université de Limoges, Limoges, France
| | - Assia Samri
- Sorbonne Université, Inserm, U1135, CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Marine Baron
- Service d'Hématologie Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Cécilia Nakid-Cordero
- Sorbonne Université, Inserm, U1135, CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - David Lavergne
- Hématologie Clinique et Thérapie Cellulaire, CHU Dupuytren, Limoges, France
| | - Véronique Morin
- Sorbonne Université, Inserm, U1135, CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Oulfata Mze
- Sorbonne Université, Inserm, U1135, CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Noureddine Balegroune
- Service d'Hématologie Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Xiaozhen Liang
- Shanghai Institute of Immunity and Infection, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Sylvain Choquet
- Service d'Hématologie Clinique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Amélie Guihot
- Sorbonne Université, Inserm, U1135, CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France; Département d'Immunologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Patrick Legembre
- UMR CNRS 7276, INSERM U1262, CRIBL, Université de Limoges, Limoges, France.
| | - Murielle Roussel
- UMR CNRS 7276, INSERM U1262, CRIBL, Université de Limoges, Limoges, France; Hématologie Clinique et Thérapie Cellulaire, CHU Dupuytren, Limoges, France.
| | | |
Collapse
|
10
|
Broomfield BJ, Tan CW, Qin RZ, Abberger H, Duckworth BC, Alvarado C, Dalit L, Lee CL, Shandre Mugan R, Mazrad ZA, Muramatsu H, Mackiewicz L, Williams BE, Chen J, Takanashi A, Fabb S, Pellegrini M, Rogers KL, Moon WJ, Pouton CW, Davis MJ, Nutt SL, Pardi N, Wimmer VC, Groom JR. Transient inhibition of type I interferon enhances CD8+ T cell stemness and vaccine protection. J Exp Med 2025; 222:e20241148. [PMID: 40062995 PMCID: PMC11893171 DOI: 10.1084/jem.20241148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/25/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Developing vaccines that promote CD8+ T cell memory is a challenge for infectious disease and cancer immunotherapy. TCF-1+ stem cell-like memory CD8+ T (TSCM) cells are important determinants of long-lived memory. Yet, the developmental requirements for TSCM cell formation are unclear. Here, we identify the temporal window for type I interferon receptor (IFNAR) blockade to drive TSCM cell generation following viral infection and mRNA-lipid nanoparticle vaccination. We reveal a reversible developmental trajectory where transcriptionally distinct TSCM cells emerged from a transitional precursor of exhausted T cellular state concomitant with viral clearance. TSCM cell differentiation correlated with T cell retention within the lymph node paracortex due to disrupted CXCR3 chemokine gradient formation. These effects were linked to increased antigen load and a counterintuitive increase in IFNγ, which controlled cell location. Vaccination with the IFNAR blockade promoted TSCM cell differentiation and enhanced protection against chronic infection. These findings propose an approach to vaccine design whereby modulation of inflammation promotes memory formation and function.
Collapse
Affiliation(s)
- Benjamin J. Broomfield
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chin Wee Tan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Raymond Z. Qin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Hanna Abberger
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Brigette C. Duckworth
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Carolina Alvarado
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Lennard Dalit
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Chee Leng Lee
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Rekha Shandre Mugan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Zihnil A.I. Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Bailey E. Williams
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Jinjin Chen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Asuka Takanashi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Stewart Fabb
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Marc Pellegrini
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, Australia
| | - Kelly L. Rogers
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | | | - Colin W. Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Melissa J. Davis
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Stephen L. Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, Philadelphia, PA, USA
| | - Verena C. Wimmer
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Joanna R. Groom
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
11
|
Peng L, Liang Q, Rong PF, Zhang S, Chen H, Liu H, Ma X, Wang W. Peripheral T lymphocyte immune characteristics dictate response to transarterial chemoembolization in unresectable hepatocellular carcinoma. Therap Adv Gastroenterol 2025; 18:17562848251333295. [PMID: 40342832 PMCID: PMC12059425 DOI: 10.1177/17562848251333295] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/17/2025] [Indexed: 05/11/2025] Open
Abstract
Background Although transcatheter arterial chemoembolization (TACE) is one of the first-line treatments for unresectable HCC (uHCC) patients, its overall efficacy varies significantly. Therefore, the identification of reliable biomarkers capable of effectively distinguishing TACE-responsive populations is clinically critical. Objectives Our research aims to investigate T-lymphocyte subpopulations and associated pathways in peripheral blood that contribute to TACE refractoriness, as well as to develop effective methods for predicting TACE efficacy. Design This is an observational study. Methods A total of 50 patients who underwent standard TACE-based therapy between January 2020 and December 2022 were included in this study. TACE response was evaluated within 1-3 months following two consecutive TACE sessions. Patients with TACE failure were assigned to the Non-Response group, whereas the remaining were categorized into the Response group. Blood samples were collected prior to treatment and subsequently analyzed using flow cytometry and RNA sequencing. Predictors were analyzed using univariate and multivariate analyses within the bivariate logistic regression models. Pathway enrichment analysis was performed using gene set enrichment analysis (GSEA). Results A total of 24 of 50 (48%) exhibited TACE failure (Non-Response). Baseline peripheral T-lymphocyte analysis revealed that the Non-Response group had a higher abundance of senescent phenotype (TSenescence, CD27-CD28-) in both CD4/CD8+ T cells (p < 0.0001), but a lower proportion of memory stem cell (TSCM) subpopulation (CD4+ TSCM: p = 0.0411; CD8+ TSCM: p < 0.0001). Furthermore, in CD8+ T cells, they exhibited higher expression of exhaustion marks (PD-1: p = 0.0005; LAG-3: p = 0.0026; TIGIT: p = 0.0014) and significantly lower production of effector molecules (TNF-α: p < 0.0001; IFN-γ: p = 0.0018; GZMB: p < 0.0001). Transcriptomics revealed that the Response group was enriched in pathways associated with energy and drug metabolism. Univariate and multivariate analyses demonstrated that the baseline CD8+ TSCM and CD8+ TSenescence subpopulations were significant predictive factors for TACE efficacy. Conclusion Our study demonstrated significant differences in the immune characteristics of peripheral T lymphocytes between the Non-Response and Response groups. The CD8+ TSCM and CD8+ TSenescence subsets are potential predictors of TACE efficacy and long-term survival. These insights into peripheral blood T lymphocytes offer valuable evidence to help clinicians more effectively identify potential TACE-responsive populations, predict survival, and develop personalized treatment regimens for patients with uHCC.
Collapse
Affiliation(s)
- Lei Peng
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qi Liang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Peng Fei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| | - Shengwang Zhang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huan Chen
- Xiangya School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Huaping Liu
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoqian Ma
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Shapiro JR, Simard N, Bolotin S, Watts TH. Fluorescent Cell Barcoding of Peripheral Blood Mononuclear Cells for High-Throughput Assessment of Vaccine-Induced T Cell Responses in Low-Volume Research Samples. Cytometry A 2025; 107:321-332. [PMID: 40202117 DOI: 10.1002/cyto.a.24933] [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: 10/24/2024] [Revised: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
T cell responses are rarely measured in large-scale human vaccine studies due to the sample volumes required, as well as the logistical, technical, and financial challenges associated with available assays. Fluorescent cell barcoding has been proposed in other contexts to allow for more high-throughput flow cytometry-based assays. Here, we aimed to expand on existing barcoding approaches to develop a reagent and sample-sparing assay for in-depth assessment of T cell responses to vaccine antigens. By using various concentrations of two fixable viability dyes in a matrix format, up to 25 samples that were pooled and acquired together could be successfully deconvoluted based on their unique fluorescent signature. This fluorescent cell barcoding approach was then combined with extracellular and intracellular staining to identify functional (i.e., producing at least one cytokine) and polyfunctional (i.e., producing multiple cytokines) T cells in response to vaccine antigen stimulation. As a proof-of-concept, we plated just 200,000 peripheral blood mononuclear cells (PBMC) per condition, and by staining and acquiring only two pooled samples, we were able to detect rare antigen-specific T cell responses in eight donors to four stimulants each. The frequencies of antigen-induced cytokine-positive cells detected in barcoded samples with 200,000 input PBMC were strongly correlated with those detected in non-barcoded samples from the same donors with 1 million input PBMC, demonstrating the validity of this approach. In conclusion, by reducing the number of PBMC needed by five-fold, and the volume of staining reagents needed by 25-fold, this assay has widespread potential applications to human vaccine studies.
Collapse
Affiliation(s)
- Janna R Shapiro
- Department of Immunology, Temerty Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
- The Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, The University of Toronto, Toronto, Ontario, Canada
| | - Nathalie Simard
- Department of Immunology, Temerty Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
| | - Shelly Bolotin
- The Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, The University of Toronto, Toronto, Ontario, Canada
- Public Health Ontario, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
| | - Tania H Watts
- Department of Immunology, Temerty Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
- The Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, The University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Song A, Zhang Y, Busch R, Asquith B, Macallan D. Deuterated water ( 2H 2O, heavy water) labelling to investigate human cell dynamics in vivo - lessons in protocol design and toxicity from the current literature. Front Immunol 2025; 16:1544193. [PMID: 40356903 PMCID: PMC12066605 DOI: 10.3389/fimmu.2025.1544193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
The use of deuterated water (also known as 'heavy water') as a tracer to measure human in vivo cell proliferation rates for specific cell subsets has expanded significantly in recent years. Although there have been several published methods papers, investigators developing new applications may be confused by differences in study design and deuterated water dose/duration. Furthermore, this approach may be met with regulatory difficulties and participant concerns about toxicity. This scoping review explores lessons that can be learnt from the current literature on the use of deuterated water in human in vivo studies measuring cell proliferation. We identified 29 such studies involving 535 study participants, both healthy volunteers and those with specific clinical conditions. Wide variations in protocols were noted with doses ranging from 40-100 ml/day of pure deuterated water (or equivalent) and durations from 4-12 weeks. Study design usually reflected the kinetics of the cell of interest. No clinical toxicity signals were noted in any studies although four studies did report transient dizziness, a recognized symptom of changing water density. These published studies provide a strong safety signal for potential participants and regulatory authorities and can act as templates for the development of new research applications.
Collapse
Affiliation(s)
- Ami Song
- Institute for Infection and Immunity, City St George's, University of London, London, United Kingdom
| | - Yan Zhang
- Institute for Infection and Immunity, City St George's, University of London, London, United Kingdom
| | - Robert Busch
- School of Life and Health Sciences, Whitelands College, University of Roehampton, London, United Kingdom
| | - Becca Asquith
- Dept of Infectious Disease, Imperial College London, London, United Kingdom
| | - Derek Macallan
- Institute for Infection and Immunity, City St George's, University of London, London, United Kingdom
- Infection Clinical Academic Group, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
14
|
Caballero AC, Ujaldón-Miró C, Pujol-Fernández P, Montserrat-Torres R, Guardiola-Perello M, Escudero-López E, Garcia-Cadenas I, Esquirol A, Martino R, Jara-Bustamante P, Ezquerra P, Soria JM, Iranzo E, Moreno-Martinez ME, Riba M, Sierra J, Alvarez-Fernández C, Escribà-Garcia L, Briones J. HSP-CAR30 with a high proportion of less-differentiated T cells promotes durable responses in refractory CD30+ lymphoma. Blood 2025; 145:1788-1801. [PMID: 39841453 DOI: 10.1182/blood.2024026758] [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: 09/03/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
ABSTRACT CD30-directed chimeric antigen receptor T-cell therapy (CART30) has limited efficacy in relapsed or refractory patients with CD30+ lymphoma, with a low proportion of durable responses. We have developed an academic CART30 cell product (HSP-CAR30) by combining strategies to improve performance. HSP-CAR30 targets a proximal epitope within the nonsoluble part of CD30, and the manufacturing process includes a modulation of ex vivo T-cell activation, as well as the addition of interleukin-21 (IL-21) to IL-7 and IL-15 to promote stemness of T cells. We translated HSP-CAR30 to a phase 1 clinical trial of 10 patients with relapsed/refractory classic Hodgkin lymphoma (HL) or CD30+ T-cell non-Hodgkin lymphoma. HSP-CAR30 was mainly composed of memory stem-like (TSCM-like) and central memory (TCM) CAR30+ T cells (87.5% ± 5%). No dose-limiting toxicities were detected. Six patients had grade 1 cytokine release syndrome, and no patient developed neurotoxicity. The overall response rate was 100%, and 5 of 8 patients with HL achieved complete remission (CR). An additional patient with HL achieved CR after a second HSP-CAR30 infusion. Remarkably, 60% of patients have ongoing CR after a mean follow-up of 34 months. CAR30+ T cells at expansion peak had a predominance of TSCM and TCM cells, and CAR30+ T cells remained detectable in 3 of 5 evaluable patients at least 12 months after infusion. Our study shows that selection of the epitope targeting CD30 and ex vivo preservation of less-differentiated memory T cells may enhance the efficacy of CART30 in patients with refractory HL. This trial is registered at www.clinicaltrials.gov (NCT04653649).
Collapse
Affiliation(s)
- Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Cristina Ujaldón-Miró
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Paula Pujol-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Rosanna Montserrat-Torres
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Maria Guardiola-Perello
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Eva Escudero-López
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | | | - Albert Esquirol
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rodrigo Martino
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Paola Jara-Bustamante
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Pol Ezquerra
- Unit of Genomics of Complex Disease, Research Institute of Sant Pau Hospital, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - José Manuel Soria
- Unit of Genomics of Complex Disease, Research Institute of Sant Pau Hospital, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - Eva Iranzo
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Maria-Estela Moreno-Martinez
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Pharmacy Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Mireia Riba
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Pharmacy Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jorge Sierra
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Laboratory of Experimental Hematology, Institut d'Investigació Biomèdica Sant Pau, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| |
Collapse
|
15
|
Plummer R, Sodergren MH, Hodgson R, Ryan BM, Raulf N, Nicholls JP, Reebye V, Voutila J, Sinigaglia L, Meyer T, Pinato DJ, Sarker D, Basu B, Blagden S, Cook N, Jeffrey Evans TR, Yachnin J, Chee CE, Li D, El-Khoueiry A, Diab M, Huang KW, Pai M, Spalding D, Talbot T, Noel MS, Keenan B, Mahalingam D, Song MS, Grosso M, Arnaud D, Auguste A, Zacharoulis D, Storkholm J, McNeish I, Habib R, Rossi JJ, Habib NA. TIMEPOINT, a phase 1 study combining MTL-CEBPA with pembrolizumab, supports the immunomodulatory effect of MTL-CEBPA in solid tumors. Cell Rep Med 2025; 6:102041. [PMID: 40168999 PMCID: PMC12047497 DOI: 10.1016/j.xcrm.2025.102041] [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: 11/16/2023] [Revised: 03/05/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025]
Abstract
Many patients with cancer do not benefit from currently approved immune checkpoint inhibitors (ICIs), suggesting that additional immunomodulation of the immunosuppressive tumor microenvironment (TME) is required. MTL-CCAAT enhancer-binding protein alpha (CEBPA) specifically upregulates the expression of the master myeloid transcription factor, CEBPA, relieving myeloid-driven immunosuppression. Here, we report the safety, tolerability, pharmacokinetics, and efficacy of MTL-CEBPA in combination with pembrolizumab in patients with advanced solid tumors that typically show ICI resistance. Multimodal exploratory analyses of paired patient biopsies demonstrate biological changes associated with the combination treatment of MTL-CEBPA and pembrolizumab, including increased infiltration of T cell and antigen-presenting cells supporting conversion from an immune-desert toward a more immune-inflamed TME. Patients with disease stabilization demonstrate reductions in immunosuppressive myeloid cells post treatment. Collectively, these data support a role for MTL-CEBPA in reducing immunosuppression in the TME. This study was registered at ClinicalTrials.gov (NCT04105335).
Collapse
Affiliation(s)
- Ruth Plummer
- The Northern Centre for Cancer Care, Freeman Hospital, NE7 7DN Newcastle, UK
| | - Mikael H Sodergren
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK
| | | | | | - Nina Raulf
- MiNA Therapeutics Ltd, W12 0BZ London, UK
| | - Joanna P Nicholls
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK; MiNA Therapeutics Ltd, W12 0BZ London, UK
| | - Vikash Reebye
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK; MiNA Therapeutics Ltd, W12 0BZ London, UK
| | | | | | - Tim Meyer
- Research Department of Oncology, UCL Cancer Institute, University College London, WC1E 6DD London, UK
| | - David J Pinato
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK; Department of Translational Medicine (DIMET), Università Del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Debashis Sarker
- Department of Research Oncology, Guys Hospital, Kings College London, SE1 9RT London, UK
| | - Bristi Basu
- University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, CB2 0QQ Cambridge, UK
| | - Sarah Blagden
- Department of Oncology, Oxford University, Churchill Hospital, OX3 7LE Oxford, UK
| | - Natalie Cook
- University of Manchester and The Christie NHS Foundation Trust, M20 4BX Manchester, UK
| | | | - Jeffrey Yachnin
- Centrum Kliniska Cancerstudier, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Cheng E Chee
- National University Hospital, National University Cancer Institute Singapore, Singapore 11928, Singapore
| | - Daneng Li
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Anthony El-Khoueiry
- Norris Comprehensive Cancer Centre, Keck Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Maria Diab
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | - Madhava Pai
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK
| | - Duncan Spalding
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK
| | - Thomas Talbot
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK
| | - Marcus S Noel
- Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - Bridget Keenan
- University of California San Francisco, San Francisco, CA 94143, USA
| | - Devalingam Mahalingam
- Robert H Lurie Comprehensive Cancer Centre, Northwestern University, Chicago, IL 60611, USA
| | - Min-Sun Song
- Beckman Research Institute, City of Hope, CA, USA
| | | | | | | | | | - Jan Storkholm
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK
| | - Iain McNeish
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK
| | | | - John J Rossi
- Beckman Research Institute, City of Hope, CA, USA
| | - Nagy A Habib
- Department of Surgery & Cancer, Imperial College London, W12 0NN London, UK; MiNA Therapeutics Ltd, W12 0BZ London, UK.
| |
Collapse
|
16
|
Vatzia E, Zhang Y, Sedaghat-Rostami E, Martini V, Paudyal B, Carr BV, McNee A, Chiu C, Moffat K, Asquith B, Beverley P, Macallan D, Tchilian E. Proliferation makes a substantive contribution to the maintenance of airway resident memory T-cell subsets in young pigs. DISCOVERY IMMUNOLOGY 2025; 4:kyaf007. [PMID: 40370578 PMCID: PMC12076203 DOI: 10.1093/discim/kyaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/03/2025] [Accepted: 04/03/2025] [Indexed: 05/16/2025]
Abstract
Tissue-resident memory (TRM) T cells play an important role in protection against respiratory infection but whether this memory is maintained by long-lived or dividing cells remains controversial. To address the rate of division of lung TRM T cells, deuterium-enriched water was administered orally to young pigs to label dividing lymphocytes. T-cell subsets were separated from blood, lymph nodes, and airways [bronchoalveolar lavage (BAL)], the latter comprising almost exclusively TRM. We show that, as in other species, circulating memory T-cell subsets divide more rapidly than naïve T cells. Rates of labelling of memory subsets were similar in blood and lymph nodes, consistent with the rapid and free exchange. Strikingly, the fraction of label in BAL was similar to those in blood/lymph nodes after 5-21 days of labelling, suggesting replacement with recently divided cells, but this was preceded at Day 2 by a phase when labelling was lower in BAL than blood/lymph node in some memory subsets. Our data exclude long-lived TRM as the source of BAL memory cells leaving three possible hypotheses: blood/airway exchange, in situ proliferation, or proliferation in the lung interstitium followed by migration to BAL. When considered in the context of other information, we favour the latter interpretation. These results indicate the dynamic nature of memory in the lung and have implications for harnessing immune responses against respiratory pathogens.
Collapse
Affiliation(s)
| | - Yan Zhang
- Institute for Infection and Immunity, City St George’s, University of London, London, UK
| | - Ehsan Sedaghat-Rostami
- The Pirbright Institute, Pirbright, UK
- Section of Immunology, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guilford, UK
| | | | | | | | | | | | | | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, UK
| | - Peter Beverley
- Department of Infectious Disease, Imperial College London, London, UK
| | - Derek Macallan
- Institute for Infection and Immunity, City St George’s, University of London, London, UK
| | | |
Collapse
|
17
|
Noorsaeed S, AlBurtamani N, Rokan A, Fassati A. Heat shock protein 90 is a chaperone regulator of HIV-1 latency. PLoS Pathog 2025; 21:e1012524. [PMID: 40168429 PMCID: PMC11981193 DOI: 10.1371/journal.ppat.1012524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 04/09/2025] [Accepted: 03/10/2025] [Indexed: 04/03/2025] Open
Abstract
An estimated 32 million people live with HIV-1 globally. Combined antiretroviral therapy suppresses viral replication but therapy interruption results in viral rebound from a latent reservoir mainly found in memory CD4+ T cells. Treatment is therefore lifelong and not curative. Eradication of this viral reservoir requires hematopoietic stem cell transplantation from hemizygous or homozygous ΔCCR5 donors, which is not broadly applicable. Alternative cure strategies include the pharmacological reactivation of latently infected cells to promote their immune-mediated clearance, or the induction of deep latency. HIV-1 latency is multifactorial and linked to the activation status of the infected CD4+ T cell. Hence to perturb latency, multiple pathways need to be simultaneously targeted without affecting CD4+ T cell function. Hsp90 has been shown to regulate HIV-1 latency, although knowledge on the pathways is limited. Because Hsp90 promotes the proper folding of numerous cellular proteins required for HIV-1 gene expression, we hypothesized that Hsp90 might be a master regulator of latency. We tested this hypothesis using a polyclonal Jurkat cell model of latency and ex-vivo latently infected primary CD4+ T cells. We found that, in the Jurkat model, Hsp90 is required for HIV-1 reactivation mediated by the T-cell receptor, phorbol esters, TNF-α, inhibition of FOXO-1, and agonists of TLR-7 and TLR-8. In primary cells, Hsp90 regulates HIV-1 gene expression induced by stimulation of the T-cell receptor or in the presence of IL-7/IL-15 or a FOXO-1 inhibitor. Chemical inhibition of Hsp90 abrogated activation of the NF-kB, NFAT and AP-1 signal transduction pathways. Within the CD4+ T cell population, CDRA45+ CCR7+ "naïve" and CD45RA- CCR7- "effector memory" cells were most sensitive to Hsp90 inhibition, which did not perturb their phenotype or activation state. Our results indicate that Hsp90 is a master regulator of HIV-1 latency that can potentially be targeted in cure strategies.
Collapse
Affiliation(s)
- Somaya Noorsaeed
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Division of Infection & Immunity and Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Nawal AlBurtamani
- Division of Infection & Immunity and Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Ahmed Rokan
- Division of Infection & Immunity and Institute of Immunity and Transplantation, University College London, London, United Kingdom
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudia Arabia
| | - Ariberto Fassati
- Division of Infection & Immunity and Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
18
|
Zhai Y, Li G, Pan C, Yu M, Hu H, Wang D, Shi Z, Jiang T, Zhang W. The development and potent antitumor efficacy of CD44/CD133 dual-targeting IL7Rα-armored CAR-T cells against glioblastoma. Cancer Lett 2025; 614:217541. [PMID: 39952598 DOI: 10.1016/j.canlet.2025.217541] [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: 11/18/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Tumor heterogeneity and an immunosuppressive microenvironment pose significant challenges for immunotherapy against solid tumors, particularly glioblastoma multiforme (GBM). Recent studies have highlighted the crucial role of glioma stem cells (GSCs) in tumor recurrence and therapeutic resistance. In this context, we developed a tandem chimeric antigen receptor (CAR)-T cell targeting CD44 and CD133 (PROM1), containing a truncated IL-7 receptor alpha intracellular domain (Δ7R) between the CD28 costimulatory receptor and the CD3ζ signaling chain (Tanζ-T28-Δ7R). Our target identification and validation were carried out using GSCs, samples from GBM patients, and the corresponding sequencing data. The antitumor efficacy of CAR-T cells was evaluated in patient-derived GSCs, intracranial xenograft models, patient-derived xenograft models, and glioblastoma organoids (GBOs). Single-cell RNA sequencing and mass cytometry were used to determine the immune phenotypes of CAR-T cells. We showed that locoregionally administered Tanζ-T28-Δ7R CAR-T cells induced long-term tumor regression with the desired safety outcomes. Patient-derived autologous Tanζ-T28-Δ7R CAR-T cells showed robust antitumor activity against GBOs. Our pre-clinical data has demonstrated the translational potential of Tanζ-T28-Δ7R CAR-T cell against GBM.
Collapse
Affiliation(s)
- You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China.
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Changqing Pan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Mingchen Yu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China
| | - Zhongfang Shi
- Department of Pathophysiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China; China National Clinical Research Center for Neurological Diseases, Beijing, PR China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, PR China; Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, PR China; Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, PR China; Beijing Engineering Research Center of Targeted Drugs and Cell Therapy for CNS Tumors, Beijing, PR China.
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China; China National Clinical Research Center for Neurological Diseases, Beijing, PR China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, PR China; Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, PR China; Beijing Engineering Research Center of Targeted Drugs and Cell Therapy for CNS Tumors, Beijing, PR China.
| |
Collapse
|
19
|
Fazeli P, Abolhasani S, Karamali N, Hajivalili M, Daryabor G, Panji M, Karimian M, Hosseini M. The role of memory T cells in type 1 diabetes: Phenotypes, mechanisms, and therapeutic implications. Autoimmun Rev 2025; 24:103759. [PMID: 39880347 DOI: 10.1016/j.autrev.2025.103759] [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: 08/27/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by the loss of insulin-producing cells in the pancreatic islets. Patients with T1D have autoreactive CD4+ and CD8+ T cells that show specific features, indicating previous exposure to self-antigens. Despite that memory T cells are vital components of the adaptive immune system, providing enduring protection against pathogens; individuals with T1D have a higher proportion of memory T cells compared to healthy individuals with naїve phenotypes. Targeting memory T cells in newly diagnosed T1D patients has shown promising results, providing evidence for the significant role of memory T cells in this disease. There are various types of memory T cells, each with unique characteristics and functions. Recent advancements in understanding the complexity and heterogeneity of T cell subpopulations have shown that T1D cannot be fully understood through simple categorization. This review aims to discuss various types of memory T cells in the immunopathogenesis of T1D, focusing on their phenotypes and frequencies, as well as epigenetic and metabolic alterations. Additionally, it will address novel immunotherapeutic approaches targeting memory T cell subsets in T1D.
Collapse
Affiliation(s)
- Pooria Fazeli
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shiva Abolhasani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Karamali
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Hajivalili
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Daryabor
- Autoimmune Disease Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Panji
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Karimian
- Brigham and Women's Hospital, Harvard Medical School Brigham and Women's Hospital, Boston, USA
| | - Maryam Hosseini
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
20
|
Liu C, Zhang H, Zhai YY, Dong J, Zhou Y, Li H, Zhang M, Yang CL, Zhang P, Li XL, Duan RS, Du T. Phenotypic and functional dysregulations of CD8 + T Cells in myasthenia gravis. Clin Exp Med 2025; 25:96. [PMID: 40131529 PMCID: PMC11937161 DOI: 10.1007/s10238-025-01603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
Myasthenia Gravis (MG) is a heterogeneous autoimmune disorder characterized by fluctuating muscle weakness caused by autoantibodies targeting neuromuscular junction components. While the role of CD4 + T cells in MG is well established, the contribution of CD8 + T cells remains poorly understood. In this study, we analyze CD8 + T cells in 36 MG patients and 38 age- and gender-matched controls using flow cytometry to evaluate subset distribution, granzyme expression, and cytokine production. MG patients exhibit an altered CD4 + /CD8 + T cell ratio and significant changes in CD8 + T cell subsets, including increased central memory CD8 + T cell (Tcm) proportions and decreased effector memory CD8 + T cell (Tem) proportions. Granzyme B expression in Tcm cells is significantly elevated in MG patients, whereas no significant changes are observed in other subsets or GZMK expression. Cytokine analysis reveals increased IL-21, GM-CSF, and IL-17A production by CD8 + T cells in MG patients. These phenotypic and functional alterations of CD8 + T cells persist during the acute phase of the disease, regardless of immunotherapy usage, and vary between ocular and generalized MG. Subgroup and correlation analyses further identify age-dependent and age-independent dysregulations of CD8 + T cells, indicating complex and subtype-specific roles of CD8 + T cells in the immunopathological processes underlying MG. Our findings provide novel insights into the involvement of CD8 + T cells in MG pathogenesis, laying a foundation for future research and potential therapeutic strategies targeting CD8 + T cells.
Collapse
Affiliation(s)
- Chang Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Hao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Yu-Yao Zhai
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Jing Dong
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Yang Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Heng Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Min Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Chun-Lin Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Xiao-Li Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Rui-Sheng Duan
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China.
| | - Tong Du
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China.
| |
Collapse
|
21
|
Rassek K, Misiak J, Ołdak T, Rozwadowska N, Basak G, Kolanowski T. New player in CAR-T manufacture field: comparison of umbilical cord to peripheral blood strategies. Front Immunol 2025; 16:1561174. [PMID: 40191201 PMCID: PMC11968755 DOI: 10.3389/fimmu.2025.1561174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
One of the most successful treatments in hematologic cancer is chimeric antigen receptor (CAR)-T cell-based immunotherapy. However, CAR-T therapy is not without challenges like the costly manufacturing process required to personalize each treatment for individual patients or graft-versus-host disease. Umbilical cord blood (UCB) has been most commonly used for hematopoietic cell transplant as it offers several advantages, including its rich source of hematopoietic stem cells, lower risk of graft-versus-host disease, and easier matching for recipients due to less stringent HLA requirements compared to bone marrow or peripheral blood stem cells. In this review, we have discussed the advantages and disadvantages of different CAR-T cell manufacturing strategies with the use of allogeneic and autologous peripheral blood cells. We compare them to the UCB approach and discuss ongoing pre-clinical and clinical trials in the field. Finally, we propose a cord blood bank as a readily available source of CAR-T cells.
Collapse
Affiliation(s)
- Karolina Rassek
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Tomasz Ołdak
- FamicordTx, Warsaw, Poland
- Polish Stem Cell Bank (PBKM), Warsaw, Poland
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- FamicordTx, Warsaw, Poland
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Kolanowski
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- FamicordTx, Warsaw, Poland
| |
Collapse
|
22
|
Yin H, Chen J, Li C. Immune Memory: A New Frontier in Treating Recurrent Inflammatory Skin Diseases. Clin Rev Allergy Immunol 2025; 68:31. [PMID: 40100550 DOI: 10.1007/s12016-025-09039-0] [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] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
The recurrence of inflammatory skin diseases represents a significant challenge in clinical practice, primarily mediated by immune memory. In inflammatory skin diseases, immune memory encompasses adaptive immune memory, trained immunity, and inflammatory memory, which are conducted by adaptive immune cells, innate immune cells, and structural cells, respectively. Adaptive immune memory is established through gene rearrangement, leading to antigen-specific immune memory. In contrast, trained immunity and inflammatory memory are formed through epigenetic and metabolic reprogramming, resulting in non-specific immune memory. Different types of immune memory work synergistically to aggravate localized inflammation in recurrent inflammatory skin diseases. However, immune memory in specific cells, such as macrophages, may also play an immunoregulatory role under certain conditions. We reviewed the immune memory mechanisms in different inflammatory skin diseases and discussed future strategies for targeted regulation of the molecular mechanisms underlying immune memory, such as targeted biological agents and epigenetic modifications. Additionally, we explored the potential for precise regulation of immune memory and its application in personalized treatment for recurrent inflammatory skin diseases.
Collapse
Affiliation(s)
- Hang Yin
- Department of Dermatology, Xijing Hospital, Forth Military Medical University, Xi'an, 710032, China
| | - Jianru Chen
- Department of Dermatology, Xijing Hospital, Forth Military Medical University, Xi'an, 710032, China.
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, 200433, China.
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Forth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
23
|
Yin X, Chen W, Ao X, Xu L, Cao J, Huang T, Liang J, Hu J, Liu J, Wang X, Li W, Zhou M, He L, Guo Z. Sodium citrate pretreatment enhances CAR-T cell persistence and anti-tumor efficacy through inhibition of calcium signaling. Front Immunol 2025; 16:1540754. [PMID: 40165944 PMCID: PMC11955688 DOI: 10.3389/fimmu.2025.1540754] [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: 12/06/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Chimeric antigen receptor T cell (CAR-T) therapy has shown success in treating hematological malignancies, but its effectiveness against solid tumors is hindered by T cell exhaustion. During in vitro expansion, tonic signaling induced by CAR expression contributes to CAR-T cell exhaustion, which can be mitigated by inhibiting calcium signaling. Given that sodium citrate can chelate calcium ions and inhibit calcium signaling, in this study, we investigated whether sodium citrate could reduce exhaustion and enhance CAR-T cell function. Methods We constructed anti-CD70 CAR-T cells and cultured them in the presence of sodium citrate. The characteristics and functionality of sodium citrate-pretreated CAR-T cells were assessed through in vitro and in vivo experiments. To further validate our observation, we also treated anti-mesothelin (MSLN) CAR-T cells with sodium citrate and detected the phenotypes and anti-tumor function of CAR-T cells. Results We found that sodium citrate-pretreated anti-CD70 CAR-T cells exhibited reduced exhaustion, increased memory T cell proportions, and enhanced anti-tumor efficacy both in vitro and in vivo. Notably, sodium citrate treatment improved the in vivo persistence of CAR-T cells and prevented tumor recurrence. These beneficial effects were also observed in anti-MSLN CAR-T cells. Transcriptomic and metabolite analyses revealed that sodium citrate inhibited calcium signaling, mTORC1 activity, and glycolysis pathways, thus modulating T cell exhaustion and differentiation. Discussion Our findings suggest that sodium citrate supplementation during CAR-T cell expansion could be a promising strategy to improve CAR-T therapy for solid tumors by preventing exhaustion and promoting memory T cell formation.
Collapse
Affiliation(s)
- Xuechen Yin
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wenwen Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xudong Ao
- Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Luxia Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jiujiu Cao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tinghui Huang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Junqing Liang
- Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Jianhua Hu
- Center of Biotherapy, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Jiaqi Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xinping Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wenying Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Muya Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
24
|
Chang E, Cavallo K, Behar SM. CD4 T cell dysfunction is associated with bacterial recrudescence during chronic tuberculosis. Nat Commun 2025; 16:2636. [PMID: 40097414 PMCID: PMC11914476 DOI: 10.1038/s41467-025-57819-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
While most people contain Mycobacterium tuberculosis infection, some individuals develop active disease, usually within two years of infection. Why immunity fails after initially controlling infection is unknown. C57BL/6 mice control Mycobacterium tuberculosis for up to a year but ultimately succumb to disease. We hypothesize that the development of CD4 T cell dysfunction permits bacterial recrudescence. We developed a reductionist model to assess antigen-specific T cells during chronic infection and found evidence of CD4 T cell senescence and exhaustion. In C57BL/6 mice, CD4 T cells upregulate coinhibitory receptors and lose effector cytokine production. Single cell RNAseq shows that only a small number of CD4 T cells in the lungs of chronically infected mice are polyfunctional. While the origin and causal relationship between T-cell dysfunction and recrudescence remains uncertain, we propose T cell dysfunction leads to a feed-forward loop that causes increased bacillary numbers, greater T cell dysfunction, and progressive disease.
Collapse
Affiliation(s)
- Evelyn Chang
- Immunology and Microbiology Program, Morningside Graduate School of Biomedical Sciences, Worcester, MA, USA
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelly Cavallo
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Samuel M Behar
- Immunology and Microbiology Program, Morningside Graduate School of Biomedical Sciences, Worcester, MA, USA.
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
25
|
O’Sullivan A, Case S, McCrudden A, Hackett E, Gallagher L, Martin D, Johnson GP, Mahadik K, Kienzle T, Lim JK, Nashat A, Srinivasan K, Lowdell MW, O’Flynn L, Frankish J. Increased functional potency of multi-edited CAR-T cells manufactured by a non-viral transfection system. Mol Ther Methods Clin Dev 2025; 33:101389. [PMID: 39811687 PMCID: PMC11730244 DOI: 10.1016/j.omtm.2024.101389] [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: 05/26/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy represents a breakthrough for the treatment of hematological malignancies. However, to treat solid tumors and certain hematologic cancers, next-generation CAR-T cells require further genetic modifications to overcome some of the current limitations. Improving manufacturing processes to preserve cell health and function of edited T cells is equally critical. Here, we report that Solupore, a Good Manufacturing Practice-aligned, non-viral physicochemical transfection system, can be used to manufacture multi-edited CAR-T cells using CRISPR-Cas9 ribonucleoproteins while maintaining robust cell functionality. When compared to electroporation, an industry standard, T cells that were triple edited using Solupore had reduced levels of apoptosis and maintained similar proportions of stem cell memory T cells with higher oxidative phosphorylation levels. Following lentiviral transduction with a CD19 CAR, and subsequent cryopreservation, triple-edited CAR-T cells manufactured using Solupore demonstrated improved immunological synapse strength to target CD19+ Raji cells and enhanced cellular cytotoxicity compared with electroporated CAR-T cells. In an in vivo mouse model (NSG), Solupore triple-edited CAR-T cells enhanced tumor growth inhibition by more than 30-fold compared to electroporated cells.
Collapse
Affiliation(s)
| | - Sarah Case
- Avectas, Cherrywood Business Park, Dublin, Ireland
| | | | - Emer Hackett
- Avectas, Cherrywood Business Park, Dublin, Ireland
| | | | | | | | | | | | | | - Aya Nashat
- Avectas, Cherrywood Business Park, Dublin, Ireland
| | | | | | - Lisa O’Flynn
- Avectas, Cherrywood Business Park, Dublin, Ireland
| | | |
Collapse
|
26
|
Luostarinen A, Vuorela A, Kerkelä E, Patrikoski M, Kotovuori A, Koski J, Ahoniemi J, Lähteenmäki K, Lehtisalo J, Oja T, Paavilainen H, Autio A, Nyman M, Nikoskelainen V, Kergourlay V, Elbasani E, van Veen B, Thotakura A, Monzo H, Ojala PM, Korhonen M, Hongisto H, Laitinen A. Establishing a GMP-compliant manufacturing process and phase-appropriate analytics for early development of a FiCAR T-cell product with a novel CAR spacer. Sci Rep 2025; 15:8093. [PMID: 40057567 PMCID: PMC11890757 DOI: 10.1038/s41598-025-92736-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/03/2025] [Indexed: 05/13/2025] Open
Abstract
There is a growing demand for chimeric antigen receptor (CAR) -T cells for clinical trials. Consequently, new centers capable of manufacturing advanced therapy medicinal products (ATMPs) are needed. In this study, we established a good manufacturing practice -compliant manufacturing process and phase-appropriate analytics for a novel autologous CD19-targeted CAR T-cell product, 19-FiCART. We evaluated the stability of fresh, healthy donor-derived leukapheresis products (LPs), produced 19-FiCART using a 12-day semi-automated process with CD4/CD8-positive cell enrichment and lentiviral transduction, and evaluated the in vivo efficacy of 19-FiCART in a xenograft mouse lymphoma model. The optimal hold time and temperature to maintain LP stability were up to 73 h at 2-8 °C. The 19-FiCART manufacturing process consistently yielded more than 2 × 109 highly viable CAR+ T cells, which is considered sufficient for a clinical product. The 19-FiCART products also demonstrated potent anti-tumor activity both in vitro and in vivo. This paper provides a detailed description of the manufacturing process and analytics for 19-FiCART and provides insights into the development of a release strategy for novel CAR T-cell products intended for early clinical studies. Additionally, we present data on LP stability, which has broader implications for the development of various immune cell-based ATMPs.
Collapse
Affiliation(s)
- Annu Luostarinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland.
| | - Arja Vuorela
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland
| | - Erja Kerkelä
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland
| | - Mimmi Patrikoski
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland
| | - Annika Kotovuori
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland
| | - Jan Koski
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Jonna Ahoniemi
- Quality Management, Finnish Red Cross Blood Service, Vantaa, Finland
| | | | - Jenni Lehtisalo
- Pharmaceutical Sciences, Orion Corporation Orion Pharma, Turku, Finland
| | - Terhi Oja
- Pharmaceutical Sciences, Orion Corporation Orion Pharma, Turku, Finland
| | | | - Anu Autio
- Immuno-Oncology, Oncology Research, Orion Corporation, Turku, Finland
| | - Marie Nyman
- Immuno-Oncology, Oncology Research, Orion Corporation, Turku, Finland
| | - Veera Nikoskelainen
- Protein and Antibody Engineering, Medicine Design, Orion Corporation, Turku, Finland
| | | | - Endrit Elbasani
- Immuno-Oncology, Oncology Research, Orion Corporation, Turku, Finland
| | - Bert van Veen
- Pharmaceutical Sciences, Orion Corporation Orion Pharma, Turku, Finland
| | - Anil Thotakura
- Immuno-Oncology, Oncology Research, Orion Corporation, Turku, Finland
| | - Hector Monzo
- Translational Cancer Medicine Research Program, University of Helsinki, Helsinki, Finland
| | - Päivi M Ojala
- Translational Cancer Medicine Research Program, University of Helsinki, Helsinki, Finland
| | - Matti Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland
- Research and Development, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Heidi Hongisto
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland
| | - Anita Laitinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Härkälenkki 13, 01,730, Vantaa, Finland
| |
Collapse
|
27
|
Chéret A. Acute HIV-1 Infection: Paradigm and Singularity. Viruses 2025; 17:366. [PMID: 40143294 PMCID: PMC11945883 DOI: 10.3390/v17030366] [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: 02/02/2025] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Acute HIV-1 infection (AHI) is a transient period where the virus causes evident damage to the immune system, including an extensive apoptosis of CD4+ T cells associated with a high level of activation and a major cytokine storm to fight the invading virus. HIV infection establishes persistence by integrating the viral genome into host cell DNA in both replicating and non-replicating forms, effectively hiding from immune surveillance within infected lymphocytes as cellular reservoirs. The measurement of total HIV-1 DNA in peripheral blood mononuclear cells (PBMCs) is a reliable reflection of this reservoir. Initiating treatments during AHI with nucleoside reverse transcriptase inhibitors (NRTIs) and/or integrase strand transfer inhibitors (INSTIs) is essential to alter the dynamics of the global reservoir expansion, and to reduce the establishment of long-lived cellular and tissue reservoirs, while preserving and enhancing specific and non-specific immune responses. Furthermore, some of the patients treated at the AHI stage may become post-treatment controllers and should be informative regarding the mechanism of viral control, so patients treated during AHI are undoubtedly the best candidates to test innovative remission strategies toward a functional cure that could play a pivotal role in long-term HIV control. AHI is characterized by high levels of viral replication, with a significant increase in the risk of HIV transmission. Detecting AHI and initiating early treatment following diagnosis provides a window of opportunity to control the epidemic, particularly in high-risk populations.
Collapse
Affiliation(s)
- Antoine Chéret
- Inserm U1016, CNRS UMR 8104, Institut Cochin, Université Paris Descartes, 75014 Paris, France;
- Service Plateforme de Diagnostic et Thérapeutique Pluridisciplinaire, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
- INSERM-CIC-1424, Centre Hospitalier Universitaire, 97159 Pointe à Pitre, Guadeloupe, France
| |
Collapse
|
28
|
Li R, Grosskopf AK, Joslyn LR, Stefanich EG, Shivva V. Cellular Kinetics and Biodistribution of Adoptive T Cell Therapies: from Biological Principles to Effects on Patient Outcomes. AAPS J 2025; 27:55. [PMID: 40032717 DOI: 10.1208/s12248-025-01017-w] [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: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 03/05/2025] Open
Abstract
Cell-based immunotherapy has revolutionized cancer treatment in recent years and is rapidly expanding as one of the major therapeutic options in immuno-oncology. So far ten adoptive T cell therapies (TCTs) have been approved by the health authorities for cancer treatment, and they have shown remarkable anti-tumor efficacy with potent and durable responses. While adoptive T cell therapies have shown success in treating hematological malignancies, they are lagging behind in establishing promising efficacy in treating solid tumors, partially due to our incomplete understanding of the cellular kinetics (CK) and biodistribution (including tumoral penetration) of cell therapy products. Indeed, recent clinical studies have provided ample evidence that CK of TCTs can influence clinical outcomes in both hematological malignancies and solid tumors. In this review, we will discuss the current knowledge on the CK and biodistribution of anti-tumor TCTs. We will first describe the typical CK and biodistribution characteristics of these "living" drugs, and the biological factors that influence these characteristics. We will then review the relationships between CK and pharmacological responses of TCT, and potential strategies in enhancing the persistence and tumoral penetration of TCTs in the clinic. Finally, we will also summarize bioanalytical methods, preclinical in vitro and in vivo tools, and in silico modeling approaches used to assess the CK and biodistribution of TCTs.
Collapse
Affiliation(s)
- Ran Li
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Abigail K Grosskopf
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Louis R Joslyn
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Eric Gary Stefanich
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Vittal Shivva
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
29
|
Kumar D, Gaikwad K, Gunnale R, Vishwakarma S, Shukla S, Srivastava S, Gopal J, Vaidya B, Saraf A, Gurjar R, Kaviraj S, Singh A, Raghuwanshi A, Agarwal P, Savergave L, Singh S. Cellular immune breadth of an Omicron-specific, self-amplifying monovalent mRNA vaccine booster for COVID-19. NPJ Vaccines 2025; 10:42. [PMID: 40025095 PMCID: PMC11873296 DOI: 10.1038/s41541-025-01076-2] [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: 08/17/2024] [Accepted: 01/16/2025] [Indexed: 03/04/2025] Open
Abstract
Selecting a booster vaccine strategy that generates cellular immune breadth is crucial for effectively recalling cellular reservoirs upon infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants. This post hoc analysis from a multicentre, randomized phase 3 study (CTRI/2022/10/046475) compared the cellular immune breadth induced by self-replicating mRNA (samRNA) vaccine GEMCOVAC-OM, encoding Omicron B.1.1.529 Spike protein, with the adenovector vaccine ChAdOx1 nCoV-19, encoding Wuhan variant Spike protein, when administered as a booster. GEMCOVAC-OM elicited significant expansion of memory B-cells (MBCs) specific to Omicron B.1.1.529, compared to ChAdOx1 nCoV-19. GEMCOVAC-OM also induced more B-cells reactive to Omicron XBB.1.5 and BA.2.86 Spike proteins. Additionally, GEMCOVAC-OM triggered higher frequencies of Omicron-Spike-specific T-cells, including stem cell, central, and effector memory subsets. In summary, while ChAdOx1 nCoV-19 showed some cross-reactivity, GEMCOVAC-OM induced a more targeted immune response. GEMCOVAC-OM offers a broader, longer-lasting immunity, making it a promising candidate for future vaccine development and global distribution.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Amit Saraf
- Gennova Biopharmaceutical Limited, Pune, India
| | | | | | - Ajay Singh
- Gennova Biopharmaceutical Limited, Pune, India
| | | | | | | | | |
Collapse
|
30
|
Traynor R, Vignola I, Sarkar S, Prochazkova M, Cai Y, Shi R, Underwood S, Ramanujam S, Yates B, Silbert S, Jin P, Dreyzin A, Shah NN, Somerville RP, Stroncek DF, Song HW, Highfill SL. Efficient manufacturing of CAR-T cells from whole blood: a scalable approach to reduce costs and enhance accessibility in cancer therapy. Cytotherapy 2025; 27:400-409. [PMID: 39652017 PMCID: PMC11810577 DOI: 10.1016/j.jcyt.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 02/12/2025]
Abstract
BACKGROUND Chimeric antigen receptor T (CAR-T) cells have significantly advanced the treatment of cancers such as leukemia and lymphoma. Traditionally, T cells are collected from patients through leukapheresis, an expensive and potentially invasive process that requires specialized equipment and trained personnel. Although whole blood collections are much more technically straightforward, whole blood starting material has not been widely utilized for clinical CAR-T cell manufacturing, in part due to lack of manufacturing processes designed for use in a good manufacturing practice (GMP) environment. Collecting cellular starting material from whole blood without leukapheresis could reduce manufacturing complexity and cost, thereby improving accessibility to CAR-T cell therapy. METHODS Whole blood samples were collected from eight healthy donors and one pediatric B-cell acute lymphoblastic leukemia (B-ALL) patient. These samples were processed using the Sepax C-Pro (Cytiva) instrument to isolate mononuclear cells (MNCs) via density gradient separation. CAR-T cells were then manufactured from the isolated MNCs using a GMP-compliant 7-day protocol, whereby T cells were activated with anti-CD3 and IL-2, transduced with GMP lentiviral vector encoding a CD19/CD22 bispecific CAR, and expanded in gas permeable cell culture bags. The resulting CAR-T cells were then evaluated for their phenotypic and functional properties using flow cytometry, cytokine release and cytotoxicity assays. RESULTS From an average 77.7 mL of whole blood from healthy donors (range = 29-96 mL), we isolated an average of 42.2 × 106 CD3⁺ T cells (range 7.3-63.0) postprocessing. CAR-T cell cultures were initiated from thaw using 1-10 × 106 starting CD3+ T cells, yielding a median T cell number of 105 × 106 cells on day 7 (range 61-188 × 106). We observed 66 ± 11% mean transduction efficiency and produced a mean of 77.4 × 106 transduced CAR-T cells (range 30.8-143.5 × 106). Similar results were obtained when using a blood sample (28mL) obtained from a patient with relapsed B-ALL who had received recent chemotherapy. CONCLUSIONS Therapeutically relevant doses of CD19/CD22 CAR-T cells can be successfully manufactured from whole blood. On average, 80 mL of whole blood yields enough CAR-T cells to create a single dose for a pediatric patient (50 kg) at a dosage of 1 × 106 CAR-T cells/kg. For larger patients, scaling up is straightforward by collecting a larger blood volume. This method also demonstrates a cost-effective approach to T cell activation and expansion which, alongside a more straightforward collection of whole blood, makes it more widely accessible especially for middle- and low-income countries. By reducing costs and labor, this strategy has the potential to significantly expand global access to CAR-T cell therapy.
Collapse
Affiliation(s)
- Roshini Traynor
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Isabella Vignola
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarmila Sarkar
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Michaela Prochazkova
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Yihua Cai
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Rongye Shi
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarah Underwood
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Supriya Ramanujam
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Bonnie Yates
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara Silbert
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Jin
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexandra Dreyzin
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert P Somerville
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Hannah W Song
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven L Highfill
- Department of Transfusion Medicine, Center for Cellular Engineering, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
31
|
Yue T, Sun Y, Dai Y, Jin F. Mechanisms for resistance to BCMA-targeted immunotherapies in multiple myeloma. Blood Rev 2025; 70:101256. [PMID: 39818472 DOI: 10.1016/j.blre.2025.101256] [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: 10/16/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Multiple myeloma (MM) remains incurable and patients eventually face the relapse/refractory dilemma. B cell maturation antigen (BCMA)-targeted immunotherapeutic approaches have shown great effectiveness in patients with relapsed/refractory MM, mainly including chimeric antigen receptor T cells (CAR-T), bispecific T cell engagers (TCEs), and antibody-drug conjugates (ADCs). However, their impact on long-term survival remains to be determined. Nonetheless, resistance to these novel therapies is still inevitable, raising a challenge that we have never met in both laboratory research and clinical practice. In this scenario, the investigation aiming to enhance and prolong the anti-MM activity of BCMA-targeted therapies has been expanding rapidly. Despite considerable uncertainty in our understanding of the mechanisms for their resistance, they have mainly been attributed to antigen-dependency, T cell-driven factors, and (immune) tumor microenvironment. In this review, we summarize the current understanding of the mechanisms for resistance to BCMA-targeted immunotherapies and discuss potential strategies for overcoming it.
Collapse
Affiliation(s)
- Tingting Yue
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin, China; Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yue Sun
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Fengyan Jin
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
32
|
Álvarez B, Revilla C, Ezquerra Á, Domínguez J. Phenotypes of porcine blood CD8β T cells and their capacity for IFN gamma production in the context of PRV vaccination. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 165:105347. [PMID: 39988100 DOI: 10.1016/j.dci.2025.105347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
CD8 T cells play a key role in elimination of intracellular pathogens. Here, we have carried out a detailed phenotypic and functional analysis of swine blood CD8β T cells, after vaccination with a live attenuated PRV vaccine. Based on the expression of six surface molecules (CD11a, CD27, CD45RA, CD95, CCR7 and SLA-DR), up to eight subsets can be identified within the circulating compartment of CD8β T cells of adult pigs; six of which correlate phenotypically with naïve, stem cell memory, central memory, transitional memory, effector memory, and terminal effector subsets described in humans. The remaining two subsets appear to correspond to intermediate stages between naïve and central memory cells, and between transitional memory and effector memory or terminal effector cells, respectively. Although CD45RA has been proposed as a marker to distinguish between porcine naïve and central memory CD8β T cells, we found that a substantial proportion of naïve T cells, which varies among animals, lack this marker and that the combination of CD95 and CD11a allows a more accurate discrimination of these subsets among CD27hi CCR7+ cells. The majority of virus-specific IFN-γ-producing CD8β T cells in the blood of PRV-vaccinated pigs, collected at 10-16-days postvaccination, showed a CD27+ CD11ahi CD45RA- phenotype, consistent with that of central, and/or transitional memory T cells.
Collapse
Affiliation(s)
- Belén Álvarez
- Departamento de Biotecnología. Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain.
| | - Concepción Revilla
- Departamento de Biotecnología. Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Ángel Ezquerra
- Departamento de Biotecnología. Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| | - Javier Domínguez
- Departamento de Biotecnología. Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, 28040, Spain
| |
Collapse
|
33
|
Yount KS, Chen CJ, Kollipara A, Liu C, Mokashi NV, Zheng X, Bagwell CB, Poston TB, Wiesenfeld HC, Hillier SL, O’Connell CM, Stanley N, Darville T. T cell signatures associated with reduced Chlamydia trachomatis reinfection in a highly exposed cohort. JCI Insight 2025; 10:e189388. [PMID: 40014387 PMCID: PMC11991011 DOI: 10.1172/jci.insight.189388] [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: 11/18/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
Chlamydia trachomatis (CT) is the most common bacterial sexually transmitted infection globally. Understanding natural immunity to CT will inform vaccine design. This study aimed to profile immune cells and associated functional features in CT-infected women and determine immune profiles associated with reduced risk of ascended endometrial CT infection and CT reinfection. PBMCs from CT-exposed women were profiled by mass cytometry, and random forest models identified key features that distinguished outcomes. CT+ participants exhibited higher frequencies of CD4+ Th2, Th17, and Th17 double-negative (Th17 DN) CD4+ T effector memory (TEM) cells than uninfected participants with decreased expression of T cell activation and differentiation markers. Minimal differences were detected between women with or without endometrial CT infection. Participants who remained follow-up negative (FU-) showed higher frequencies of CD4+ T central memory (TCM) Th1, Th17, Th1/17, and Th17 DN but reduced CD4+ TEM Th2 cells than FU+ participants. Expression of markers associated with central memory and Th17 lineage was increased on T cell subsets among FU- participants. These data indicate that peripheral T cells exhibit distinct features associated with resistance to CT reinfection. The highly plastic Th17 lineage appears to contribute to protection. Addressing these immune nuances could promote efficacy of CT vaccines.
Collapse
Affiliation(s)
| | | | | | - Chuwen Liu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Neha V. Mokashi
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Xiaojing Zheng
- Department of Pediatrics, School of Medicine
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | - Harold C. Wiesenfeld
- University of Pittsburgh School of Medicine and Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Sharon L. Hillier
- University of Pittsburgh School of Medicine and Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | | | - Natalie Stanley
- Department of Computer Science; and
- Computational Medicine Program and
| | - Toni Darville
- Department of Pediatrics, School of Medicine
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
34
|
Yoon JW, Kim KM, Cho S, Cho MJ, Park S, Hwang D, Kim HR, Park SH, Cho JH, Jeong H, Choi JM. Th1-poised naive CD4 T cell subpopulation reflects anti-tumor immunity and autoimmune disease. Nat Commun 2025; 16:1962. [PMID: 40000667 PMCID: PMC11861895 DOI: 10.1038/s41467-025-57237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Naïve CD4 T cells are traditionally viewed as a quiescent, homogeneous, resting population, but emerging evidence reveals their heterogeneity, which can be crucial for understanding disease contexts and therapeutic outcomes. In this study, we identify distinct subpopulations within both murine and human naïve CD4 T cells by single cell-RNA-sequencing (scRNA-seq), particularly focusing on a subpopulation that expresses super-high levels of interleukin-7 receptor (IL-7Rsup-hi), along with CD97, IL-18R, and Ly6C. This subpopulation, absent in the thymus and peripherally induced, exhibits type 1 helper T cell (Th1)-poised characteristics and contributes to the inhibition of cancer progression in B16F10 tumor-bearing mice. In humans, this IL-7Rsup-hi subpopulation expressing CD97 correlates with the responsiveness to anti-PD-1 therapy in cancer patients and the disease state of multiple sclerosis. By elucidating the heterogeneity of naive CD4 T cells and identifying a Th1-poised subpopulation capable of robust type 1 responses, we highlight the importance of this heterogeneity in inflammatory conditions for defining the disease states and predicting drug responsiveness.
Collapse
Affiliation(s)
- Jae-Won Yoon
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Kyung Min Kim
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sookyung Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Min-Ji Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Seonjun Park
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science & Technology (UNIST), Ulsan, Republic of Korea
| | - Jae-Ho Cho
- Medical Research Center for Combinatorial Tumor Immunotherapy, Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun, 58128, Korea
| | - Hyobin Jeong
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea.
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea.
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea.
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
35
|
Tang WW, Battistone B, Bauer KM, Weis AM, Barba C, Fadlullah MZH, Ghazaryan A, Tran VB, Lee SH, Agir ZB, Nelson MC, Victor ES, Thibeaux A, Hernandez C, Tantalla J, Tan AC, Rao D, Williams M, Drummond MJ, Beswick EJ, Round JL, Ekiz HA, Voth WP, O'Connell RM. A microRNA-regulated transcriptional state defines intratumoral CD8 + T cells that respond to immunotherapy. Cell Rep 2025; 44:115301. [PMID: 39951377 PMCID: PMC11924119 DOI: 10.1016/j.celrep.2025.115301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/24/2024] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
The rising incidence of advanced-stage colorectal cancer (CRC) and poor survival outcomes necessitate new and effective therapies. Immune checkpoint inhibitors (ICIs), specifically anti-PD-1 therapy, show promise, yet clinical determinants of a positive response are suboptimal. Here, we identify microRNA-155 (miR-155) as necessary for CD8+ T cell-infiltrated tumors through an unbiased in vivo CRISPR-Cas9 screen identifying functional tumor antigen-specific CD8+ T cell-expressed microRNAs. T cell miR-155 is required for anti-PD-1 responses and for a vital intratumor CD8+ T cell differentiation cascade by repressing Ship-1, inhibiting Tcf-1 and stemness, and subsequently enhancing Cxcr6 expression, anti-tumor immunity, and effector functions. Based on an underlying miR-155-dependent CD8+ T cell transcriptional profile, we identify a gene signature that predicts ICI responses across 12 diverse cancers. Together, our findings support a model whereby miR-155 serves as a central regulator of CD8+ T cell-dependent cancer immunity and ICI responses that may be leveraged for future therapeutics.
Collapse
Affiliation(s)
- William W Tang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ben Battistone
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Kaylyn M Bauer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Allison M Weis
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Cindy Barba
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Muhammad Zaki Hidayatullah Fadlullah
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Arevik Ghazaryan
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Van B Tran
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Soh-Hyun Lee
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Z Busra Agir
- Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey
| | - Morgan C Nelson
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Emmanuel Stephen Victor
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Amber Thibeaux
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Colton Hernandez
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Jacob Tantalla
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Aik C Tan
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Dinesh Rao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew Williams
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Micah J Drummond
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT 84108, USA
| | - Ellen J Beswick
- Division of Digestive Disease and Nutrition, Department of Internal Medicine, University of Kentucky, Lexington, KY 40508, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - H Atakan Ekiz
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Warren P Voth
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Deecke L, Ohlei O, Goldeck D, Homann J, Toepfer S, Demuth I, Bertram L, Pawelec G, Lill CM. Peripheral Immune Profiles in Individuals at Genetic Risk of Amyotrophic Lateral Sclerosis and Alzheimer's Disease. Cells 2025; 14:250. [PMID: 39996723 PMCID: PMC11852917 DOI: 10.3390/cells14040250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
The immune system plays a crucial role in the pathogenesis of neurodegenerative diseases. Here, we explored whether blood immune cell profiles are already altered in healthy individuals with a genetic predisposition to amyotrophic lateral sclerosis (ALS) or Alzheimer's disease (AD). Using multicolor flow cytometry, we analyzed 92 immune cell phenotypes in the blood of 448 healthy participants from the Berlin Aging Study II. We calculated polygenic risk scores (PGSs) using genome-wide significant SNPs from recent large genome-wide association studies on ALS and AD. Linear regression analyses were then performed of the immune cell types on the PGSs in both the overall sample and a subgroup of older participants (>60 years). While we did not find any significant associations between immune cell subtypes and ALS and AD PGSs when controlling for the false discovery rate (FDR = 0.05), we observed several nominally significant results (p < 0.05) with consistent effect directions across strata. The strongest association was observed with CD57+ CD8+ early-memory T cells and ALS risk (p = 0.006). Other immune cell subtypes associated with ALS risk included PD-1+ CD8+ and CD57+ CD4+ early-memory T cells, non-classical monocytes, and myeloid dendritic cells. For AD, naïve CD57+ CD8+ T cells and mature NKG2A+ natural killer cells showed nominally significant associations. We did not observe major immune cell changes in individuals at high genetic risk of ALS or AD, suggesting they may arise later in disease progression. Additional studies are required to validate our nominally significant findings.
Collapse
Affiliation(s)
- Laura Deecke
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
| | - Olena Ohlei
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
| | - David Goldeck
- Department of Immunology, University of Tübingen, 72076 Tübingen, Germany (G.P.)
| | - Jan Homann
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
| | - Sarah Toepfer
- Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Ilja Demuth
- Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- BCRT—Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, 23562 Lübeck, Germany
| | - Graham Pawelec
- Department of Immunology, University of Tübingen, 72076 Tübingen, Germany (G.P.)
- Health Sciences North Research Institute of Canada, Sudbury, ON P3E 2H3, Canada
| | - Christina M. Lill
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
- Ageing and Epidemiology Unit (AGE), School of Public Health, Imperial College London, London W6 8RP, UK
| |
Collapse
|
37
|
Ramaswami R, Kask AS, D'Amico L, Menon MP, Lurain K, Yarchoan R, Ekwede I, Couey P, Burnham E, Angeldekao A, Ha Lee B, Kaiser JC, Cheever M, Uldrick TS, Kwok LL, Wright A, Fling SP, Wang CCJ. Phase I study of efineptakin alfa (NT-I7) for the treatment of Kaposi sarcoma. J Immunother Cancer 2025; 13:e010291. [PMID: 39915263 PMCID: PMC11804200 DOI: 10.1136/jitc-2024-010291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/03/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND CD4+ T-cell lymphocytopenia and immune dysfunction are factors that drive the onset and persistence of Kaposi sarcoma (KS) in people with (PWH) and without HIV. Standard chemotherapy agents for KS can contribute to increasing CD4+ T cell lymphocytopenia. IL-7 is a cytokine that is essential in T-cell development, proliferation and homeostasis. In PWH, IL-7 administration leads to increased numbers of circulating central memory and naïve T-cell phenotypes. METHODS In this multicenter phase I study with a 3+3 dose escalation design, participants with KS with or without HIV received up to four intramuscular injections of IL-7 (NT-I7) every 9 weeks. The primary endpoint of the study was to evaluate safety over three escalating dose levels (DL) of NT-I7 (DL1:480 µg/kg, DL2: 960 µg/kg and DL3: 1200 µg/kg) and identify a maximum tolerated dose. Secondary endpoints included evaluation of antitumor activity per the modified AIDS Clinical Trials Group Criteria and assessment of the effect of NT-I7 on the kinetics of CD4+ and CD8+ T-cells. RESULTS Eight cisgender male participants (five with HIV infection) were enrolled. Six participants were treated at DL1, and two were treated at DL2. The study was closed to accrual after enrolment of the second participant on DL2 due to termination of study funding. Four of the eight participants (three in DL1 and one in DL2) completed all four doses of the NT-I7. With regard to treatment-emergent adverse events (AEs), all participants had CONCLUSIONS Preliminary data demonstrate safety and activity of IL-7 in patients with KS and activity specifically among individuals HIV-associated KS. TRIAL REGISTRATION NUMBER NCT04893018.
Collapse
Affiliation(s)
- Ramya Ramaswami
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Angela Shaulov Kask
- Cancer Immunotherapy Trials Network (CITN), Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Leonard D'Amico
- Cancer Immunotherapy Trials Network (CITN), Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Manoj P Menon
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kathryn Lurain
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Irene Ekwede
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Paul Couey
- Division of Hematology and Medical Oncology, University of California San Francisco (UCSF) Helen Diller Comprehensive Cancer Center, San Francisco, California, USA
| | - Eli Burnham
- Harborview Medical Center, Seattle, Washington, USA
| | | | | | - Judith C Kaiser
- Cancer Immunotherapy Trials Network (CITN), Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Martin Cheever
- Cancer Immunotherapy Trials Network (CITN), Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Thomas S Uldrick
- Cancer Immunotherapy Trials Network (CITN), Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Anna Wright
- Cancer Immunotherapy Trials Network (CITN), Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Steven P Fling
- Cancer Immunotherapy Trials Network (CITN), Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Chia-Ching Jackie Wang
- Division of Hematology and Medical Oncology, University of California San Francisco (UCSF) Helen Diller Comprehensive Cancer Center, San Francisco, California, USA
| |
Collapse
|
38
|
Shafer AM, Kenna E, Golden LAF, Elhossiny AM, Perry KD, Wilkowski J, Yan W, Kaczkofsky B, McGue J, Bresler SC, Courtney AH, Dalman JM, Galban CJ, Jiang W, Espinoza CE, Chugh R, Iyer MK, Frankel TL, Pasca di Magliano M, Dlugosz AA, Angeles CV. An immunocompetent mouse model of liposarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.634916. [PMID: 40297505 PMCID: PMC12036434 DOI: 10.1101/2025.01.31.634916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Liposarcoma (LPS) is the most prevalent soft tissue sarcoma. The most common biological subtypes are well-differentiated (WDLPS), a low-grade disease that can evolve to high-grade dedifferentiated LPS (DDLPS), with increased rates of recurrence and metastasis and low response rates to chemotherapy and targeted therapies. Preclinical testing of immunotherapeutics for LPS has been held back by the lack of an immunocompetent mouse model. Here, we present a spontaneous immunocompetent LPS mouse model, ACPP, with targeted deletion of Trp53 and Pten in adipocytes to mimic signaling alterations observed in human LPS. Similar to human LPS, tumors arising in ACPP mice produce WDLPS and DDLPS, along with tumors that exhibit both WD and DD components. Murine and human DDLPS tumors possess transcriptional similarities, including increased expression of oncogenes Cdk4 and Hmga2 and reduced expression of the tumor suppressor Cebpa; further, both mouse and human DDLPS exhibit either high or low T cell infiltration. Syngeneic cell lines derived from spontaneous ACPP DDLPS reliably produce tumors following orthotopic injection, each with distinct growth patterns, aggressiveness and tumor infiltrating lymphocyte profiles. These models provide much needed tools to understand the complex immunobiology of LPS and greatly accelerate the pace of preclinical studies to uncover new therapies for patients with this aggressive malignancy.
Collapse
|
39
|
Ouyang SX, Xu YG, Ding P, Long Y, Zhang Z, Sun SJ, Zhang Y, Yin H, Zhang JB, Cao Q, Shen FM, Wang P, Liu J, Li DJ. Dynamic analysis of intrahepatic T cells reveals a unique group of restorative Cxcr3 + tissue-resident CD4 T cells in acute liver injury. Toxicology 2025; 511:154058. [PMID: 39828240 DOI: 10.1016/j.tox.2025.154058] [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: 10/01/2024] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Acetaminophen (APAP) stands as one of the most prevalent triggers of drug-induced acute liver injury (ALI). The intricate modulation of immune system activation and inflammatory cascades by hepatic immune cells is paramount in managing liver injury and subsequent restoration. In this study, we employed an integrative approach that fused our proprietary flow cytometry analyses across various time points post-APAP injury with publicly available single-cell RNA sequencing (scRNA-seq) datasets, encompassing time-series data from liver tissue of mice subjected to APAP intoxication. This allowed us to delve into the dynamics of T cell profiles during APAP-induced ALI. Our comprehensive analyses unveiled the intricate temporal shifts in intrahepatic T cell populations across different phases following APAP-induced ALI. Notably, we observed a persistent augmentation of intrahepatic CD4+ T cells post-APAP injury. Amongst these, a distinct population of restorative Cxcr3+ tissue-resident CD4+ T cells emerged. Inhibition of CXCR3 using a neutralizing antibody exacerbated APAP-induced liver function impairment and hepatocyte death. Furthermore, we identified that the Cxcr3+ tissue-resident CD4+ T cells were tightly regulated by intrahepatic ''Lgals9-Cd45'' and 'CXCL13-Cxcr3' signaling pathways. These discoveries underscore the novel protective function of CXCR3, a vital biological macromolecule, in mitigating APAP-induced ALI, and may shed lights on new therapeutic strategies targeting this condition.
Collapse
Affiliation(s)
- Shen-Xi Ouyang
- Department of Pharmacology, Shanghai Tenth People's Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China; Department of Pharmacology, Shanghai Pulmonary Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China; Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Yong-Gang Xu
- Department of Cardiology, The 921th Hospital of the PLA Joint Logistics Support Force, The Second Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Peng Ding
- Department of Anesthesiology, The 983th Hospital PLA Joint Logistics Support Force, Tianjin, China
| | - Yue Long
- Department of Anesthesiology, The 921th Hospital of the PLA Joint Logistics Support Force, The Second Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhen Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China
| | - Si-Jia Sun
- Department of Pharmacology, Shanghai Tenth People's Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China; Department of Pharmacology, Shanghai Pulmonary Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China
| | - Yan Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China
| | - Hang Yin
- Department of Pharmacology, Shanghai Tenth People's Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China
| | - Jia-Bao Zhang
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Qi Cao
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacology, Shanghai Tenth People's Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China
| | - Pei Wang
- Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Jian Liu
- Department of Hepatic Surgery, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University/Second Military Medical University, Shanghai, China.
| | - Dong-Jie Li
- Department of Pharmacology, Shanghai Tenth People's Hospital Affiliated to School of Medicine of Tongji University, Shanghai, China.
| |
Collapse
|
40
|
da Graça CG, Sheikh AA, Newman DM, Wen L, Li S, Shen J, Zhang Y, Gabriel SS, Chisanga D, Seow J, Poch A, Rausch L, Nguyen MHT, Singh J, Su CH, Cluse LA, Tsui C, Burn TN, Park SL, Von Scheidt B, Mackay LK, Vasanthakumar A, Bending D, Shi W, Cui W, Schröder J, Johnstone RW, Kallies A, Utzschneider DT. Stem-like memory and precursors of exhausted T cells share a common progenitor defined by ID3 expression. Sci Immunol 2025; 10:eadn1945. [PMID: 39888981 PMCID: PMC7617396 DOI: 10.1126/sciimmunol.adn1945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/23/2024] [Indexed: 02/02/2025]
Abstract
Stem-like T cells are attractive immunotherapeutic targets in patients with cancer given their ability to proliferate and differentiate into effector progeny. Thus, identifying T cells with enhanced stemness and understanding their developmental requirements are of broad clinical and therapeutic interest. Here, we demonstrate that during acute infection, the transcriptional regulator inhibitor of DNA binding 3 (ID3) identifies stem-like T cells that are uniquely adapted to generate precursors of exhausted T (Tpex) cells in response to chronic infection or cancer. Expression of ID3 itself enables Tpex cells to sustain T cell responses in chronic infection or cancer, whereas loss of ID3 results in impaired maintenance of CD8 T cell immunity. Furthermore, we demonstrate that interleukin-1 (IL-1) family members, including IL-36β and IL-18, promote the generation of ID3+ T cells that mediate superior tumor control. Overall, we identify ID3 as a common denominator of stem-like T cells in both acute and chronic infections that is specifically required to sustain T cell responses to chronic stimulation.
Collapse
Affiliation(s)
- Catarina Gago da Graça
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Amania A. Sheikh
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Dane M. Newman
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Lifen Wen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Sining Li
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jian Shen
- Department of Pathology, Northwestern University, Chicago, IL
| | - Yuqi Zhang
- Department of Pathology, Northwestern University, Chicago, IL
| | - Sarah S. Gabriel
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
| | - Justine Seow
- Computational Sciences Initiative (CSI), The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Annika Poch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Lisa Rausch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Minh-Hanh T. Nguyen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jayendra Singh
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
| | - Chun-Hsi Su
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Leonie A. Cluse
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Carlson Tsui
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Thomas N. Burn
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Simone L. Park
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Bianca Von Scheidt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Laura K. Mackay
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - David Bending
- Department of Immunology and Immunotherapy, College of Medicine and Health, University of Birmingham, BirminghamB15 2TT, UK
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Weiguo Cui
- Department of Pathology, Northwestern University, Chicago, IL
| | - Jan Schröder
- Computational Sciences Initiative (CSI), The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Ricky W. Johnstone
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Daniel T. Utzschneider
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| |
Collapse
|
41
|
Habl MS, Emara MM, Zayed RA, Sultan AM, Elsabagh A, Elsaid AM, Abdel-Khalek EE, El-Saadany MM, Wahab MA, Shehta A. Allograft tolerance after adult living donor liver transplantation: a case-control study. BMC Surg 2025; 25:52. [PMID: 39885500 PMCID: PMC11783700 DOI: 10.1186/s12893-025-02780-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND To investigate the incidence and potential predictors of immune tolerance among adult living donor liver transplant (LDLT) recipients. METHODS This case-control study included adult recipients who underwent LDLT between May 2004 and January 2018, with at least a 5-year follow-up after LDLT. We divided the study recipients into two groups: Group 1 (Tolerance Group) included recipients who achieved operational or prope tolerance for at least one year; Group 2 (Control Group) included recipients who did not achieve tolerance. We used logistic regression analysis to study the potential predictors of tolerance after LDLT. RESULTS We included 368 recipients, 275 (74.7%) in Group 1 and 93 (25.3%) in Group 2. Operational tolerance occurred in 13/275 (4.7%) recipients and prope tolerance in 262/275 (95.3%) recipients. Age was significantly higher in Group 1. The median time for tolerance among the study recipients was 60 months (36-168). During follow-up, Group 1 showed lower serum levels of bilirubin, liver enzymes, alkaline phosphatase, and gamma-glutamyl transferase. Group 1 had a lower incidence of acute cellular rejection (ACR), recurrent viral hepatitis, and biliary complications. Logistic regression identified preoperative MELD, indication for LDLT, ACR, recurrent viral hepatitis, and biliary complications as significant predictors for allograft tolerance after LDLT. CONCLUSION Allograft tolerance occurred in 74.7% of this cohort. We suggest that the MELD score, indication for LT, ACR, recurrent viral hepatitis, and biliary complications are predictors of allograft tolerance after LDLT.
Collapse
Affiliation(s)
- Mohamed S Habl
- Department of Hepatology and Gastroenterology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Moataz Maher Emara
- Department of Anesthesiology and Intensive Care and Pain Medicine, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Reham A Zayed
- Department of Hepatology and Gastroenterology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed M Sultan
- Department of Anesthesiology and Intensive Care and Pain Medicine, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Elsabagh
- Department of Hepatology and Gastroenterology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Marwan Elsaid
- Department of Hepatology and Gastroenterology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ehab E Abdel-Khalek
- Department of Hepatology and Gastroenterology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed M El-Saadany
- Department of Hepatology and Gastroenterology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Abdel Wahab
- Department of Anesthesiology and Intensive Care and Pain Medicine, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Shehta
- Department of Anesthesiology and Intensive Care and Pain Medicine, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
- Gastrointestinal Surgery Center, Department of Surgery, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
- Liver Transplantation Program, Gastrointestinal Surgery Center, Faculty of Medicine, Mansoura University, Mansoura, 35511, Egypt.
| |
Collapse
|
42
|
Osum KC, Becker SH, Krueger PD, Mitchell JS, Hong SW, Magill IR, Jenkins MK. A minority of Th1 and Tfh effector cells express survival genes shared by memory cell progeny that require IL-7 or TCR signaling to persist. Cell Rep 2025; 44:115111. [PMID: 39723889 PMCID: PMC12009130 DOI: 10.1016/j.celrep.2024.115111] [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: 12/08/2023] [Revised: 10/24/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
It is not clear how CD4+ memory T cells are formed from a much larger pool of earlier effector cells. We found that transient systemic bacterial infection rapidly generates several antigen-specific T helper (Th)1 and T follicular helper (Tfh) cell populations with different tissue residence behaviors. Although most cells of all varieties had transcriptomes indicative of cell stress and death at the peak of the response, some had already acquired a memory cell signature characterized by expression of genes involved in cell survival. Each Th1 and Tfh cell type was maintained long term by interleukin (IL)-7, except germinal center Tfh cells, which depended on a T cell antigen receptor (TCR) signal. The results indicate that acute infection induces rapid differentiation of Th1 and Tfh cells, a minority of which quickly adopt the gene expression profile of memory cells and survive by signals from the IL-7 receptor or TCR.
Collapse
Affiliation(s)
- Kevin C Osum
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Samuel H Becker
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Peter D Krueger
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jason S Mitchell
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sung-Wook Hong
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Ian R Magill
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
43
|
Chang E, Cavallo K, Behar SM. CD4 T cell dysfunction is associated with bacterial recrudescence during chronic tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634376. [PMID: 39896548 PMCID: PMC11785196 DOI: 10.1101/2025.01.22.634376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
While most people contain Mycobacterium tuberculosis infection, some individuals develop active disease, usually within two years of infection. Why immunity fails after initially controlling infection is unknown. C57BL/6 mice control Mycobacterium tuberculosis for up to a year but ultimately succumb to disease. We hypothesize that the development of CD4 T cell dysfunction permits bacterial recrudescence. We developed a reductionist model to assess antigen-specific T cells during chronic infection and found evidence of CD4 T cell senescence and exhaustion. In C57BL/6 mice, CD4 T cells upregulate coinhibitory receptors and lose effector cytokine production. Single cell RNAseq shows that only a small number of CD4 T cells in the lungs of chronically infected mice are polyfunctional. While the origin and causal relationship between T-cell dysfunction and recrudescence remains uncertain, we propose T cell dysfunction leads to a feed-forward loop that causes increased bacillary numbers, greater T cell dysfunction, and progressive disease.
Collapse
Affiliation(s)
- Evelyn Chang
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kelly Cavallo
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Samuel M. Behar
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
44
|
Filoni J, Ferrari A, Jofra T, Putignano AR, Da Dalt L, Cesarano S, Di Dedda C, Bonacina F, Marchesi F, Norata GD, Bonini C, Piemonti L, Monti P. Metabolic reprogramming of naïve regulatory T cells by IL-7 and IL-15 promotes their persistence and performance upon adoptive transfer. Commun Biol 2025; 8:99. [PMID: 39838096 PMCID: PMC11751088 DOI: 10.1038/s42003-024-07381-1] [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: 07/22/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Tregs for adoptive therapy are traditionally expanded ex vivo using high doses of IL-2. However, the final Treg product has limited survival once infused in patients, potentially affecting therapeutic effectiveness. Here, we tested a novel expansion protocol in which highly purified naïve Tregs were expanded with a combination of IL-7 and IL-15, in the absence of IL-2. The final Treg product was enriched with cells displaying an immature CD45RA+CD62L+CD95+ phenotype, reminiscent of conventional memory stem T cells. The combination of IL-7 and IL-15 confers Tregs a glycolytic metabolism and improved metabolic fitness, characterized by an increased capacity to adapt metabolism according to glucose and oxygen availability. Tregs expanded with IL-7 and IL-15 showed longer persistence and an improved capacity to control xeno-GvHD in NSG mice. This work suggests that metabolic reprogramming induced by IL-7 and IL-15 provides better Treg performance for adoptive therapy.
Collapse
Affiliation(s)
- Jessica Filoni
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Arianna Ferrari
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Tatiana Jofra
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Anna Rita Putignano
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital Rozzano, Rozzano, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Susanna Cesarano
- Experimental Hematology Unit, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Carla Di Dedda
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Federica Marchesi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Monti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy.
| |
Collapse
|
45
|
Sweis RF, Chatta GS, Jain RK, Moon H, Delacroix SE, Fang A, D’Amico L, Kask AS, Cheever MA, Fling S, Sharon E, Lacroix A, Kaiser JC, Pachynski RK, Yu EY. A Phase II Open-Label, Randomized Clinical Trial of Atezolizumab with or without Human Recombinant IL-7 (CYT107) in Advanced Urothelial Cancer. Clin Cancer Res 2025; 31:299-307. [PMID: 39576210 PMCID: PMC11747792 DOI: 10.1158/1078-0432.ccr-24-1728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/08/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE Advanced urothelial cancer generally has high mortality despite modern anti-PD-1/L1 antibody-based combinations. Augmenting checkpoint inhibitor-mediated immune responses with lymphocyte growth factors may improve outcomes. We conducted a randomized phase II study (Cancer Immunotherapy Trials Network-14) in 47 patients to explore whether human recombinant IL-7 (CYT107) could be safely combined with PD-L1 inhibition to enhance responses. PATIENTS AND METHODS Patients with urothelial cancer after platinum chemotherapy were randomized to atezolizumab alone or with CYT107 weekly for four doses. The primary objective was clinical efficacy by the objective response rate (ORR). Secondary objectives included safety, toxicity, and other clinical outcomes. Correlative endpoints included peripheral immunophenotyping and quantification of cytokines. RESULTS CYT107 plus atezolizumab was well-tolerated, without dose-limiting toxicities and lower grade 3 to 4 treatment-related adverse events compared with atezolizumab monotherapy. The ORR was 26.3% for the combination therapy versus 23.8% for atezolizumab alone (P = 0.428). The complete response rate was 10.5% for the combination therapy versus 4.8% for monotherapy. Three patients on combination therapy had responses >21 months versus one with monotherapy. CD4+ and CD8+ T-lymphocyte expansion occurred in patients with response to combination therapy, with the greatest effect in T memory stem cells. Patients who responded to treatment exhibited elevated baseline levels of CCL4 and reduced levels of VEGFA and TNF. CONCLUSIONS Combining CYT107 with atezolizumab was safe and resulted in lymphocyte expansion, a doubling of the complete response rate, and durable responses exceeding 2 years. However, the ORR was similar to atezolizumab alone. Increased and sustained doses of CYT107 coupled with patient selection strategies should be further investigated.
Collapse
Affiliation(s)
- Randy F. Sweis
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | | | | | | | - Scott Edward Delacroix
- Louisiana State University School of Medicine and Stanley S. Scott Cancer Ctr, New Orleans, LA
| | | | | | | | | | | | | | | | | | | | - Evan Y. Yu
- Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington
| |
Collapse
|
46
|
Ferreira-Hermosillo A, Santana-Sánchez P, Vaquero-García R, García-Sáenz MR, Castro-Ríos A, Chávez-Rueda AK, Gómez-Díaz RA, Chávez-Sánchez L, Legorreta-Haquet MV. Circulating T Cell Subsets in Type 1 Diabetes. Cells 2025; 14:48. [PMID: 39791749 PMCID: PMC11719944 DOI: 10.3390/cells14010048] [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: 12/09/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Type 1 diabetes (T1D) is a complex disease driven by the immune system attacking the insulin-producing beta cells in the pancreas. Understanding the role of different T cell subpopulations in the development and progression of T1D is crucial. By employing flow cytometry to compare the characteristics of T cells, we can pinpoint potential indicators of treatment response or therapeutic inefficacy. Our study reveals elevated prolactin (PRL) levels in T1D patients, along with a decreased production of key cytokines. Additionally, PD1 appears to play a significant role in T1D. Notably, PRL levels correlate with an earlier disease onset and a specific T cell phenotype, hinting at the potential influence of PRL. These findings highlight the need for further research to identify promising cellular targets for more effective and tailored therapies.
Collapse
Affiliation(s)
- Aldo Ferreira-Hermosillo
- Unidad de Investigación en Enfermedades Endócrinas de la UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Paola Santana-Sánchez
- Unidad de Investigación Médica en Inmunología, de la UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (P.S.-S.); (R.V.-G.); (A.K.C.-R.); (L.C.-S.)
| | - Ricardo Vaquero-García
- Unidad de Investigación Médica en Inmunología, de la UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (P.S.-S.); (R.V.-G.); (A.K.C.-R.); (L.C.-S.)
| | - Manuel R. García-Sáenz
- Servicio de Endocrinología, de la UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Angélica Castro-Ríos
- Unidad de Investigación en Epidemiología Clínica de la UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Adriana K. Chávez-Rueda
- Unidad de Investigación Médica en Inmunología, de la UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (P.S.-S.); (R.V.-G.); (A.K.C.-R.); (L.C.-S.)
| | - Rita A. Gómez-Díaz
- Unidad de Investigación en Epidemiología Clínica de la UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Luis Chávez-Sánchez
- Unidad de Investigación Médica en Inmunología, de la UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (P.S.-S.); (R.V.-G.); (A.K.C.-R.); (L.C.-S.)
| | - María V. Legorreta-Haquet
- Unidad de Investigación Médica en Inmunología, de la UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (P.S.-S.); (R.V.-G.); (A.K.C.-R.); (L.C.-S.)
| |
Collapse
|
47
|
Ahmadi M, Putnam N, Dotson M, Hayoun D, Padilla J, Fatima N, Bhanap P, Nonterah G, de Mollerat du Jeu X, Ji Y. Accelerating CAR T cell manufacturing with an automated next-day process. Curr Res Transl Med 2025; 73:103489. [PMID: 39705851 DOI: 10.1016/j.retram.2024.103489] [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: 06/05/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024]
Abstract
The traditional method of CAR T cell production involves lengthy ex-vivo culture times which can result in the reduction of crucial naïve T cell subsets. Moreover, traditional CAR T cell therapy manufacturing processes can prolong time-to-patient, potentially delaying patient treatment, and contribute to disease progression. In this study, we describe an innovative and semi-automated 24-hour CAR T manufacturing process that yields a higher percentage of naïve/stem-cell like T cells which showed high cytotoxic activity and cytokine release in vitro. The data supports the feasibility of implementing this streamlined manufacturing process in clinics. This approach also has the potential to enhance CAR T therapy efficacy and improve patient access to therapy.
Collapse
Affiliation(s)
- Moloud Ahmadi
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | - Nicholas Putnam
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | - Max Dotson
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | - Danny Hayoun
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | - Jasmine Padilla
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | - Nujhat Fatima
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | - Prajakta Bhanap
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | - Gertrude Nonterah
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA
| | | | - Yongchang Ji
- Thermo Fisher Scientific, 5781 Van Allen Way, Carlsbad CA 92008, USA.
| |
Collapse
|
48
|
Biwott K, Singh P, Baráth S, Nyariki JN, Hevessy Z, Bacso Z. Dynamic P-glycoprotein expression in early and late memory states of human CD8 + T cells and the protective role of ruxolitinib. Biomed Pharmacother 2025; 182:117780. [PMID: 39740391 DOI: 10.1016/j.biopha.2024.117780] [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: 09/27/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
ABCB1/MDR-1/P-glycoprotein (Pgp) is an ABC transporter responsible for cancer cell multi-drug resistance. It is expressed in cytotoxic T lymphocytes (CTL). Eliminating sensitive cancer cells during high-dose chemotherapy can also damage immune cells. Our study aimed to assess which maturing human CD8 + CTL memory subsets may be affected based on their Pgp protein expression. In an in vitro CTL differentiation model system, we tracked the maturation of naive, effector, and memory cells and the expression of Pgp. This system involves co-culturing blood lymphocytes with proliferation-inhibited JY antigen-presenting B-lymphoblastoid cells expressing HLA-I A2. These JY-primed maturing CTLs were TCR-activated using beads, and the effect of the maturation-modifying JAK1/2 inhibitor ruxolitinib was examined. Multidimensional analysis identified six major CTL subsets: naive, young memory (Tym), stem cell memory (Tscm), central memory (Tcm), effector memory (Tem), and effectors (Te). These subsets were further divided into thirteen specific subsets: TymCD127 + , TymCD127-, Tscm, TcmCD95 + , TcmCD73 +CD95 + , TcmCD95+CD127 + , TcmPD1 + , TemCD95 + , TemraCD127 + , TemraCD127-, TeCD95 + , and TeCD73 +CD95 + . Pgp expression was detectable in naïve cells and dynamically changed across the thirteen identified subsets. Increased Pgp was detected in young memory T cells and in Tscm, TcmCD95 + , and TcmPD1 + human CTL subsets. Unlike other transiently appearing memory cells, the number of cells in these core Pgp-expressing memory subsets stabilized by the end of the contraction phase. Ruxolitinib treatment downregulated effector T-cell polarization while upregulating small memory subsets expressing Pgp. In conclusion, activation increased Pgp expression, whereas ruxolitinib treatment preserved small early and late memory subset core that primarily expressed Pgp.
Collapse
Affiliation(s)
- Kipchumba Biwott
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen 4032, Hungary; Department of Biochemistry and Biotechnology, Technical University of Kenya, Kenya.
| | - Parvind Singh
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | - Sándor Baráth
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | | | - Zsuzsanna Hevessy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen 4032, Hungary; Dean's office, Faculty of Pharmacy, University of Debrecen, Debrecen 4032, Hungary.
| |
Collapse
|
49
|
Agliardi G, Dias J, Rampotas A, Garcia J, Roddie C. Accelerating and optimising CAR T-cell manufacture to deliver better patient products. Lancet Haematol 2025; 12:e57-e67. [PMID: 39510106 DOI: 10.1016/s2352-3026(24)00273-4] [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: 05/30/2024] [Revised: 08/17/2024] [Accepted: 08/20/2024] [Indexed: 11/15/2024]
Abstract
Autologous chimeric antigen receptor (CAR) T-cell therapy has transformed the management of B-cell leukaemia and lymphoma. However, current manufacturing processes present logistical hurdles, restricting broader application. As clinical outcomes can be heavily influenced by the quality of autologous starting materials and production processes, strategies to improve product phenotype are crucial. Short manufacturing processes have the advantage of bringing products to patients more quickly and, in parallel, avoiding the highly differentiated and exhausted CAR T-cell phenotypes associated with prolonged ex vivo manufacture. This Review examines advances in our understanding of what constitutes an effective CAR T-cell product and approaches to improve product quality. Historically, strategies have relied on adjustments in medium composition and selection of less differentiated cell subtypes. Since 2020, the field has been shifting towards reduced-expansion protocols, no-activation protocols, and point-of-care manufacturing. These approaches have the advantage of a rapid turnaround while maintaining a less differentiated and exhausted phenotype. These efforts are leading to ultrarapid production methods and even elimination of ex vivo manipulation with the use of in vivo manufacturing approaches. In this Review, we focus on the advances needed to accelerate CAR T-cell manufacture (including near-patient methods), with an emphasis on improved therapeutic efficacy and rapid turnaround time, and simplified quality control procedures required to fully realise the clinical potential of CAR T-cell therapies.
Collapse
Affiliation(s)
- Giulia Agliardi
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Juliana Dias
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Alexandros Rampotas
- Cancer Institute, University College London, London, UK; Department of Haematology, University College London Hospitals, London, UK
| | - John Garcia
- Cancer Institute, University College London, London, UK; Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital London, NHS Foundation Trust, London, UK
| | - Claire Roddie
- Cancer Institute, University College London, London, UK; Department of Haematology, University College London Hospitals, London, UK.
| |
Collapse
|
50
|
Nguema L, Picard F, El Hajj M, Dupaty L, Fenwick C, Cardinaud S, Wiedemann A, Pantaleo G, Zurawski S, Centlivre M, Zurawski G, Lévy Y, Godot V. Subunit protein CD40.SARS.CoV2 vaccine induces SARS-CoV-2-specific stem cell-like memory CD8 + T cells. EBioMedicine 2025; 111:105479. [PMID: 39667270 PMCID: PMC11697708 DOI: 10.1016/j.ebiom.2024.105479] [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: 09/10/2023] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Ideally, vaccination should induce protective long-lived humoral and cellular immunity. Current licensed COVID-19 mRNA vaccines focused on the spike (S) region induce neutralizing antibodies that rapidly wane. METHODS Herein, we show that a subunit vaccine (CD40.CoV2) targeting spike and nucleocapsid antigens to CD40-expressing cells elicits broad specific human (hu)Th1 CD4+ and CD8+ T cells in humanized mice. FINDINGS CD40.CoV2 vaccination selectively enriched long-lived spike- and nucleocapsid-specific CD8+ progenitors with stem-cell-like memory (Tscm) properties, whereas mRNA BNT162b2 induced effector memory CD8+ T cells. CD8+ Tscm cells produced IFNγ and TNF upon antigenic restimulation and showed a high proliferation rate. We demonstrate that CD40 activation is specifically required for the generation of huCD8+ Tscm cells. INTERPRETATION These results support the development of a CD40-vaccine platform capable of eliciting long-lasting T-cell immunity. FUNDING This work was supported by Inserm, Université Paris-Est Créteil, and the Investissements d'Avenir program, Vaccine Research Institute (VRI), managed by the ANR.
Collapse
Affiliation(s)
- Laury Nguema
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Florence Picard
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Marwa El Hajj
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Léa Dupaty
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Craig Fenwick
- Service of Immunology and Allergy Lausanne University Hospital, Swiss Vaccine Research Institute, University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Sylvain Cardinaud
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Aurélie Wiedemann
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Giuseppe Pantaleo
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Service of Immunology and Allergy Lausanne University Hospital, Swiss Vaccine Research Institute, University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Sandra Zurawski
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Mireille Centlivre
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Gerard Zurawski
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Baylor Scott and White Research Institute, Dallas, TX, United States
| | - Yves Lévy
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service Immunologie Clinique, Créteil, France.
| | - Véronique Godot
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France.
| |
Collapse
|