1
|
Sato Y. Immune Aging and Its Implication for Age-Related Disease Progression. Physiology (Bethesda) 2025; 40:0. [PMID: 39887318 DOI: 10.1152/physiol.00051.2024] [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: 11/11/2024] [Revised: 12/18/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025] Open
Abstract
As life expectancy increases globally, the prevalence and severity of age-related diseases have risen, significantly impacting patients' quality of life and increasing dependency on the healthcare system. Age-related diseases share several pathological commonalities, and emerging evidence suggests that targeting these biological processes ameliorates multiple age-related diseases. Immune aging plays a critical role in the pathogenesis of age-related diseases, given its involvement not only in controlling infection and cancer but also in facilitating tissue homeostasis and repair. Aging causes compositional and functional changes in both innate and adaptive immune cells, thereby significantly contributing to the pathogenesis of age-related disease and systemic low-grade inflammation, termed "inflammaging." This review article aims to describe the current understanding of immune aging and its impact on age-related diseases with particular emphasis on kidney and autoimmune diseases. In addition, this review highlights tertiary lymphoid structures (TLS) as a hallmark of immune aging, exploring their roles in inflammation, tissue damage, and potential therapeutic targeting.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Yue YL, Liu JJ, Ma H, Pan ZD, Wang L, Zhang JW, Wang SS, Xie YQ, Jiang H, Bian YL, Wu MY, Yuan YS, Zhang BH, Xiao XD, Zhu JW. Generating potent and persistent antitumor immunity via affinity-tuned CAR-T cells targeting mesothelin. Acta Pharmacol Sin 2025:10.1038/s41401-025-01572-0. [PMID: 40341217 DOI: 10.1038/s41401-025-01572-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/20/2025] [Indexed: 05/10/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy for solid tumors faces challenges of insufficient efficacy and a high recurrence rate. Mesothelin (MSLN) is a membrane glycoprotein highly expressed in various solid tumors that has restricted low expression in normal tissues such as the pleura, peritoneum, and pericardium. We previously performed affinity maturation based on the parental antibody M912, and constructed the phage display library. In this study we identified four novel human anti-MSLN antibodies (LP12, HP4-11, HP4-41/LP6, and HP4-44/LP2) with varying degrees of enhanced affinity. These third-generation CARs targeting MSLN were packaged into lentiviral vectors to generate stable CAR-T cells. The CAR-T variants induced robust cytolytic activity, significant cytokine production, and activation-induced clonal proliferation against various MSLN-positive tumors in vitro, and effectively cleared disseminated tumors in mice. A single administration of the CAR-T variant LP12 potently eradicated various types of MSLN-positive solid tumors, achieved long-term persistence in vivo, effectively prevented tumor recurrence, and exhibited no non-specific toxicity. Therefore, optimizing the affinity of antigen-binding domain in CAR represents a promising strategy for advancing the development of safe and effective CAR-T cell therapies. The LP12 CAR-T cells developed in this study have potential applications in patients with MSLN-positive solid tumors. Schematic illustration of the generation and antitumor mechanism of affinity-tuned MSLN-targeted CAR-T cells. The expression plasmids carrying different anti-MSLN CAR genes were packaged into lentiviral vectors. Lentiviral transduction of human CD3+ T cells was performed to generate CAR-T cells, which were then expanded. After injection of moderately affinity-tuned MSLN-targeted CAR-T cells into mice, they enter the bloodstream, recognize, and infiltrate the solid tumors. They specifically recognize and bind to MSLN on the surface of tumor cells, and upon activation, release IFN-γ, IL-2, and TNF-α to exert cytolytic activity. Subsequently, they undergo clonal proliferation and primarily differentiate into effector memory CAR-T cells, maintaining long-term antitumor immunity and effectively preventing recurrence.
Collapse
Affiliation(s)
- Ya-Li Yue
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | - Jun-Jun Liu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | - Hang Ma
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | - Zhi-di Pan
- Jecho Institute Co., Ltd, Shanghai, 200240, China
| | - Lei Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | - Jia-Wei Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | | | - Yue-Qing Xie
- Jecho Laboratories, Inc, Frederick, MD, 21704, USA
| | - Hua Jiang
- Jecho Laboratories, Inc, Frederick, MD, 21704, USA
| | - Yan-Lin Bian
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | - Ming-Yuan Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | - Yun-Sheng Yuan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China
| | - Bao-Hong Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China.
| | | | - Jian-Wei Zhu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, 200240, China.
- Jecho Institute Co., Ltd, Shanghai, 200240, China.
- Jecho Laboratories, Inc, Frederick, MD, 21704, USA.
- Jecho Biopharmaceuticals Co., Ltd, Tianjin, 300467, China.
| |
Collapse
|
3
|
Levine JE. Prediction and Prognostication of Acute Graft-Versus-Host Disease by MAGIC Biomarkers. Am J Hematol 2025; 100 Suppl 3:5-13. [PMID: 40123552 PMCID: PMC11936457 DOI: 10.1002/ajh.27594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 03/25/2025]
Abstract
Recent advancements in prophylaxis for acute graft-versus-host disease (GVHD) have successfully reduced the incidence of severe cases; however, overall survival rates have not significantly improved, and GVHD continues to be a major cause of mortality. The severity of gastrointestinal (GI) damage is especially critical, as it strongly correlates with treatment failure and non-relapse mortality, but clinical symptoms do not reliably predict peak severity in its early stages. Biomarker-based algorithms, such as the Mount Sinai Acute GVHD International Consortium (MAGIC) algorithm, leverage serum levels of GI GVHD biomarkers (ST2 and REG3α) to quantify intestinal crypt damage, providing more accurate predictions of GVHD outcomes compared to clinical assessments. Clinical trials have investigated the use of biomarkers as entry criteria for treatment, with notable success in guiding treatment de-escalation, which is increasingly important as the presentation of GVHD shifts towards milder forms. The recently developed MAGIC composite scores further enhance prediction accuracy by integrating clinical symptom severity with biomarker assessments. Future clinical trials that employ these composite scores or similar algorithms are anticipated to be more efficient by identifying patients who are most likely to benefit from specific therapies and ultimately improving the management of GVHD.
Collapse
Affiliation(s)
- John E Levine
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
4
|
Siegel SJ, DeWolf S, Schmalz J, Saber W, Dong J, Martens MJ, Logan B, Albanese A, Iovino L, Chen E, Kaminski J, Neuberg D, Hebert K, Keskula P, Zavistaski J, Steinberg L, Schichter I, Cagnin L, Hernandez V, Warren M, Applegate K, Bar M, Chhabra S, Choi SW, Clark W, Das S, Jenq R, Jones RJ, Levine JE, Murthy H, Rashidi A, Riches M, Sandhu K, Sung AD, Larkin K, Al Malki MM, Gooptu M, Elmariah H, Alousi A, Runaas L, Shaffer B, Rezvani A, El Jurdi N, Loren AW, Scheffey D, Sanders C, Hamadani M, Dudakov J, Bien S, Robins H, Horowitz M, Bolaños-Meade J, Holtan S, Bhatt AS, Perales MA, Kean LS. Graft-versus-host disease prophylaxis shapes T cell biology and immune reconstitution after hematopoietic cell transplant. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.25.25322901. [PMID: 40061351 PMCID: PMC11888538 DOI: 10.1101/2025.02.25.25322901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Successful hematopoietic cell transplant requires immunosuppression to prevent graft-versus-host disease (GVHD), a lethal, T-cell-mediated post-transplant complication. The phase 3 BMT CTN 1703 trial demonstrated superior GVHD-free/relapse-free survival for post-transplant cyclophosphamide (PT-Cy)-based GVHD prophylaxis versus tacrolimus/methotrexate (Tac/MTX), but did not improve overall survival. To compare T-cell biology between GVHD prophylaxis regimens, 324 patients were co-enrolled onto BMT CTN 1801 (NCT03959241). We quantified T-cell immune reconstitution using multi-modal analysis, including T-cell receptor (TCR) sequencing of 2,359 longitudinal samples (180,432,350 T-cells). Compared to Tac/MTX, PT-Cy was associated with an early, substantial reduction in TCR diversity that was sustained for 2 years. PT-Cy led to a T-cell reconstitution bottleneck, including reduced thymic output and virus-associated TCRs. Decreased D+14 TCR diversity predicted prevention of chronic GVHD, but also correlated with increased moderate-to-severe infections. This study reveals how distinct immunosuppression strategies have significant effects on the global immune repertoire, underpinning post-transplant clinical outcomes.
Collapse
Affiliation(s)
- Steven J Siegel
- Department of Pediatrics, Harvard Medical School and Divisions of Infectious Diseases and Hematology/Oncology, Boston Children's Hospital, Boston, MA
| | - Susan DeWolf
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Wael Saber
- Department of Medicine, Medical College of Wisconsin and Center for International Blood and Marrow Transplant Research (CIBMTR), Milwaukee, WI
| | - Jiayi Dong
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA
| | - Michael J Martens
- CIBMTR and Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | - Brent Logan
- CIBMTR and Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI
| | | | - Lorenzo Iovino
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Edward Chen
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA
| | - James Kaminski
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA and the Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Kyle Hebert
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Paula Keskula
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA
| | | | - Lea Steinberg
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA
| | | | - Lorenzo Cagnin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA
| | - Vanessa Hernandez
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Makya Warren
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Merav Bar
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Sung Won Choi
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - William Clark
- Division of Hematology-Oncology and Palliative Care, Virginia Commonwealth University, Richmond, VA
| | - Suman Das
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Robert Jenq
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | - Richard J Jones
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University and the Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - John E Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hemant Murthy
- Division of Hematology-Oncology and Blood and Marrow Transplant and Cellular Therapy Programs, Mayo Clinic, Jacksonville, FL
| | - Armin Rashidi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Karamjeet Sandhu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | - Anthony D Sung
- Division of Hematologic Malignancies and Cellular Therapy, Kansas University Medical Center, Kansas City, KS
| | - Karilyn Larkin
- Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Monzr M Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | - Mahasweta Gooptu
- Department of Hematology and Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Hany Elmariah
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer and Research Institute, Tampa, FL
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, the University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Lyndsey Runaas
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Brian Shaffer
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Andrew Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
| | - Najla El Jurdi
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Alison W Loren
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | | - Mehdi Hamadani
- Blood and Marrow Transplant Program and Cellular Therapy Program, Medical College of Wisconsin and CIBMTR, Milwaukee, WI
| | - Jarrod Dudakov
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center and Department of Immunology, University of Washington, Seattle, WA
| | | | | | - Mary Horowitz
- Blood and Marrow Transplant Program and Cellular Therapy Program, Medical College of Wisconsin and CIBMTR, Milwaukee, WI
| | - Javier Bolaños-Meade
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University and the Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shernan Holtan
- Blood and Marrow Transplantation Section, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Ami S Bhatt
- Division of Hematology, Departments of Medicine and Genetics, Stanford University, Palo Alto, CA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, and the Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Leslie S Kean
- Department of Pediatrics, Harvard Medical School, Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
5
|
Omdahl KI, Bermea RS, Fleming R, Kimler K, Kaminski J, Hariri LP, Ly A, Rui X, Cagnin L, Lane J, Gerdemann U, Blazar BR, Tkachev V, Kean LS. Organ-specific microenvironments drive divergent T cell evolution in acute graft-versus-host disease. Sci Transl Med 2025; 17:eads1298. [PMID: 39879321 DOI: 10.1126/scitranslmed.ads1298] [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: 07/30/2024] [Accepted: 10/28/2024] [Indexed: 01/31/2025]
Abstract
Tissue-specific T cell immune responses play a critical role in maintaining organ health but can also drive immune pathology during both autoimmunity and alloimmunity. The mechanisms controlling intratissue T cell programming remain unclear. Here, we leveraged a nonhuman primate model of acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation to probe the biological underpinnings of tissue-specific alloimmune disease using a comprehensive systems immunology approach including multiparameter flow cytometry, population-based transcriptional profiling, and multiplexed single-cell RNA sequencing and TCR sequencing. Transcriptional profiling revealed substantial biological differences between T cells infiltrating the lung and liver during aGVHD. These included enrichment for transcriptional pathways controlling extracellular matrix remodeling and chemotaxis in the lung and enrichment for transcriptional pathways linked to nucleic acid metabolism and proliferation in the liver. Single-cell RNA sequencing and TCR sequencing substantiated divergent organ-specific transcriptional programing of tissue-infiltrating T cells, which was linked to clonal expansion, with expanded clones progressively enriched for C-X3-C motif chemokine receptor 1 (CX3CR1)-expressing CD8 effector T cells in the lung and eomesodermin (EOMES)-expressing CD8 effector-memory T cells in the liver. This divergent evolution of T cells was maintained even for T cells sharing the same TCRs, indicating its independence from antigen specificity. Together, these results provide insights into the role that tissue microenvironment-derived signals play in local T cell transcriptional programming during alloimmune-mediated clonal expansion and suggest potential opportunities to develop tissue-specific therapeutics to curtail pathogenic immunity after transplant.
Collapse
Affiliation(s)
- Kayleigh Ingersoll Omdahl
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Rene S Bermea
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Ryan Fleming
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kyle Kimler
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - James Kaminski
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lida P Hariri
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amy Ly
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xianliang Rui
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Lorenzo Cagnin
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jennifer Lane
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ulrike Gerdemann
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Bruce R Blazar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Victor Tkachev
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
6
|
Koyama M. Control of antigen-independent T-cell migration after HSCT. Blood Adv 2025; 9:207-208. [PMID: 39808447 PMCID: PMC11788125 DOI: 10.1182/bloodadvances.2024014584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Affiliation(s)
- Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
7
|
Fuchs KJ, Göransson M, Kester MGD, Ettienne NW, van de Meent M, de Jong RCM, Koster EAS, Halkes CJM, Scheeren F, Heemskerk MHM, van Balen P, Falkenburg JHF, Hadrup SR, Griffioen M. DNA barcoded peptide-MHC multimers to measure and monitor minor histocompatibility antigen-specific T cells after allogeneic stem cell transplantation. J Immunother Cancer 2024; 12:e009564. [PMID: 39653555 PMCID: PMC11629015 DOI: 10.1136/jitc-2024-009564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/26/2024] [Indexed: 12/12/2024] Open
Abstract
Allogeneic stem cell transplantation (alloSCT) provides a curative treatment option for hematological malignancies. After HLA-matched alloSCT, donor-derived T cells recognize minor histocompatibility antigens (MiHAs), which are polymorphic peptides presented by HLA on patient cells. MiHAs are absent on donor cells due to genetic differences between patient and donor. T cells targeting broadly expressed MiHAs induce graft-versus-leukemia (GvL) reactivity as well as graft-versus-host disease (GvHD), while T cells for MiHAs with restricted or preferential expression on hematopoietic or non-hematopoietic cells may skew responses toward GvL or GvHD, respectively. Besides tissue expression, overall strength of GvL and GvHD is also determined by T-cell frequencies against MiHAs.Here, we explored the use of DNA barcode-labeled peptide-MHC multimers to detect and monitor antigen-specific T cells for the recently expanded repertoire of HLA-I-restricted MiHAs. In 16 patients who experienced an immune response after donor lymphocyte infusion, variable T-cell frequencies up to 30.5% of CD8+ T cells were measured for 49 MiHAs. High T-cell frequencies above 1% were measured in 12 patients for 19 MiHAs, with the majority directed against mismatched MiHAs, typically 6-8 weeks after donor lymphocyte infusion and at the onset of GvHD. The 12 patients included 9 of 10 patients with severe GvHD, 2 of 3 patients with limited GvHD and 1 of 3 patients without GvHD.In conclusion, we demonstrated that barcoded peptide-MHC multimers reliably detect and allow monitoring for MiHA-specific T cells during treatment to investigate the kinetics of immune responses and their impact on development of GvL and GvHD after HLA-matched alloSCT.
Collapse
Affiliation(s)
- Kyra J Fuchs
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcus Göransson
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Michel G D Kester
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Natasja W Ettienne
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Marian van de Meent
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob C M de Jong
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eva A S Koster
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ferenc Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sine R Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
8
|
Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, Song G. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024; 9:340. [PMID: 39627201 PMCID: PMC11615378 DOI: 10.1038/s41392-024-02030-9] [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: 03/11/2024] [Revised: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
The dynamic regulation of chromatin accessibility is one of the prominent characteristics of eukaryotic genome. The inaccessible regions are mainly located in heterochromatin, which is multilevel compressed and access restricted. The remaining accessible loci are generally located in the euchromatin, which have less nucleosome occupancy and higher regulatory activity. The opening of chromatin is the most important prerequisite for DNA transcription, replication, and damage repair, which is regulated by genetic, epigenetic, environmental, and other factors, playing a vital role in multiple biological progresses. Currently, based on the susceptibility difference of occupied or free DNA to enzymatic cleavage, solubility, methylation, and transposition, there are many methods to detect chromatin accessibility both in bulk and single-cell level. Through combining with high-throughput sequencing, the genome-wide chromatin accessibility landscape of many tissues and cells types also have been constructed. The chromatin accessibility feature is distinct in different tissues and biological states. Research on the regulation network of chromatin accessibility is crucial for uncovering the secret of various biological processes. In this review, we comprehensively introduced the major functions and mechanisms of chromatin accessibility variation in different physiological and pathological processes, meanwhile, the targeted therapies based on chromatin dynamics regulation are also summarized.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Li
- Hepatobiliary Pancreatic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lingli Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China.
| |
Collapse
|
9
|
Sortebech D, Schoenfeldt T, Duvetorp A, Agerholm-Nielsen R, Eidsmo L. Skin-resident T Cells Contribute to the Dynamic Disease Manifestations of Psoriasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1267-1277. [PMID: 39432869 DOI: 10.4049/jimmunol.2400020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/23/2024] [Indexed: 10/23/2024]
Abstract
The human skin forms a dynamic barrier to physical injuries and microbial invasion. Constant interactions between stroma and tissue-confined immune cells maintain skin homeostasis. However, the cellular interactions that maintain skin health also contribute to focal immunopathology. Psoriasis is a common disease that manifests with focal pathology induced by environmental triggers in genetically susceptible individuals. Within psoriasis plaques, cross-talk between skin-resident T cells and stroma cells leads to chronic inflammation. Inflammatory cytokines such as TNF-α, IL-17, IL-22, and IL-23 amplify the local chronic inflammation and sustain the well-demarcated thick and scaly plaques that characterize the disease. In resolved lesions, T cells remain poised for IL-17 and IL-22 production, and postinflammatory epigenetic modifications lower the threshold for initiation of local relapse. This review focuses on how tissue-resident memory T cells contribute to the onset, maintenance, resolution, and relapse of psoriasis.
Collapse
Affiliation(s)
- Daniel Sortebech
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Trine Schoenfeldt
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Albert Duvetorp
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Skåne University Hospital, Malmö, Sweden
| | - Rasmus Agerholm-Nielsen
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Liv Eidsmo
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Herr MM, Balderman SR, Wallace PK, Zhang Y, Tario JD, Buxbaum NP, Holtan S, Ross M, McCarthy PL, Betts B, Maslak P, Hahn TE. Outcomes of Human Leukocyte Antigen-Matched Related Donor and Haploidentical Allogeneic Hematopoietic Cell Transplantation Recipients by Immune Profiles of Recipients and Donors. Transplant Cell Ther 2024; 30:808.e1-808.e13. [PMID: 38801976 PMCID: PMC11296899 DOI: 10.1016/j.jtct.2024.05.018] [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: 02/21/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Haploidentical (Haplo) allogeneic HCTs (alloHCT) have been used more frequently over the last decade as survival is similar to HLA-matched related donor (MRD) alloHCTs. We aimed to identify donor and recipient immune signatures before alloHCT that are associated with clinically meaningful outcomes in MRD vs Haplo alloHCT recipients. This retrospective cohort study of 165 MRD (n = 132) and Haplo (n = 33) alloHCT recipients and their related donors between 2007-2019 with paired peripheral blood samples immunophenotyped for T-cell, B-cell, NK cell and dendritic cell (DC) subsets. Immune cells were quantified before alloHCT in donors and recipients; calculations of immune cell ratios were classified as high, intermediate, and low and analyzed with alloHCT outcomes. Haplo donors were younger than MRD donors (median: 35 vs 51 years), whereas Haplo recipients were older than MRD recipients (median: 68 vs 54 years), were more likely to have a Karnofsky Performance Score ≤ 70 (76% vs 57%), 3+ comorbidities (54% vs 47%), and were in complete remission prior to alloHCT (58% vs 42%). In MRD alloHCT, a lower ratio of CD4+ to CD8+ effector memory cells in the donor was associated with lower 4-yr overall survival (OS; 25% vs 61%; P = .009), lower 4-yr progression free survival (PFS; 25% vs 58%; P = .014) and higher incidence of 1-yr transplant-related mortality (TRM; 39% vs 7%; P = .009) in recipients. A higher ratio of CD8+ effector memory to total NK cells measured in MRD recipients was associated with a higher incidence of grade II-IV aGvHD (63% vs 37%; P = .004) but was not statistically significant for III-IV aGvHD (23% vs 12%). In Haplo alloHCT, a lower ratio of total T-regulatory to CD4+ central memory cells in the donor was associated with lower 4-yr PFS (22% vs 60%; P = .0091). A higher ratio of CD4+ effector memory to CD8+ effector memory cells measured in Haplo recipients pre-alloHCT was associated with lower 4-yr OS (25% vs 88%; P = .0039). In both MRD and Haplo recipients, a higher ratio of CD4+ naïve to CD4+ central memory cells was associated with a higher incidence of grade II-IV aGvHD (64% vs 38%; P = .04). Evaluation of pre-alloHCT immune signatures of the donor and recipient may influence clinically meaningful patient outcomes in both MRD and Haplo transplants.
Collapse
Affiliation(s)
- Megan M Herr
- Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| | - Sophia R Balderman
- Roswell Park Comprehensive Cancer Center, Buffalo, New York; Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Paul K Wallace
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Yali Zhang
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Joseph D Tario
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Shernan Holtan
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Maureen Ross
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Brian Betts
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Peter Maslak
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Theresa E Hahn
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
11
|
Yeh AC, Koyama M, Waltner OG, Minnie SA, Boiko JR, Shabaneh TB, Takahashi S, Zhang P, Ensbey KS, Schmidt CR, Legg SRW, Sekiguchi T, Nelson E, Bhise SS, Stevens AR, Goodpaster T, Chakka S, Furlan SN, Markey KA, Bleakley ME, Elson CO, Bradley PH, Hill GR. Microbiota dictate T cell clonal selection to augment graft-versus-host disease after stem cell transplantation. Immunity 2024; 57:1648-1664.e9. [PMID: 38876098 PMCID: PMC11236519 DOI: 10.1016/j.immuni.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 02/09/2024] [Accepted: 05/20/2024] [Indexed: 06/16/2024]
Abstract
Allogeneic T cell expansion is the primary determinant of graft-versus-host disease (GVHD), and current dogma dictates that this is driven by histocompatibility antigen disparities between donor and recipient. This paradigm represents a closed genetic system within which donor T cells interact with peptide-major histocompatibility complexes (MHCs), though clonal interrogation remains challenging due to the sparseness of the T cell repertoire. We developed a Bayesian model using donor and recipient T cell receptor (TCR) frequencies in murine stem cell transplant systems to define limited common expansion of T cell clones across genetically identical donor-recipient pairs. A subset of donor CD4+ T cell clonotypes differentially expanded in identical recipients and were microbiota dependent. Microbiota-specific T cells augmented GVHD lethality and could target microbial antigens presented by gastrointestinal epithelium during an alloreactive response. The microbiota serves as a source of cognate antigens that contribute to clonotypic T cell expansion and the induction of GVHD independent of donor-recipient genetics.
Collapse
MESH Headings
- Graft vs Host Disease/immunology
- Graft vs Host Disease/microbiology
- Animals
- Mice
- Mice, Inbred C57BL
- CD4-Positive T-Lymphocytes/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Microbiota/immunology
- Clonal Selection, Antigen-Mediated
- Transplantation, Homologous
- Bayes Theorem
- Stem Cell Transplantation/adverse effects
- Mice, Inbred BALB C
- Gastrointestinal Microbiome/immunology
- Hematopoietic Stem Cell Transplantation/adverse effects
Collapse
Affiliation(s)
- Albert C Yeh
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Olivia G Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Simone A Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Julie R Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tamer B Shabaneh
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kathleen S Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Christine R Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Samuel R W Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tomoko Sekiguchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ethan Nelson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shruti S Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Andrew R Stevens
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tracy Goodpaster
- Experimental Histopathology Core, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Saranya Chakka
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Scott N Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kate A Markey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marie E Bleakley
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Hematology, Oncology, and Bone Marrow Transplantation, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Charles O Elson
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Philip H Bradley
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
12
|
Wang Y, Ullah MA, Waltner OG, Bhise SS, Ensbey KS, Schmidt CR, Legg SR, Sekiguchi T, Nelson EL, Kuns RD, Nemychenkov NS, Atilla E, Yeh AC, Takahashi S, Boiko JR, Varelias A, Blazar BR, Koyama M, Minnie SA, Clouston AD, Furlan SN, Zhang P, Hill GR. Calcineurin inhibition rescues alloantigen-specific central memory T cell subsets that promote chronic GVHD. J Clin Invest 2024; 134:e170125. [PMID: 38828727 PMCID: PMC11142741 DOI: 10.1172/jci170125] [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: 03/02/2023] [Accepted: 04/09/2024] [Indexed: 06/05/2024] Open
Abstract
Calcineurin inhibitors (CNIs) constitute the backbone of modern acute graft-versus-host disease (aGVHD) prophylaxis regimens but have limited efficacy in the prevention and treatment of chronic GVHD (cGVHD). We investigated the effect of CNIs on immune tolerance after stem cell transplantation with discovery-based single-cell gene expression and T cell receptor (TCR) assays of clonal immunity in tandem with traditional protein-based approaches and preclinical modeling. While cyclosporin and tacrolimus suppressed the clonal expansion of CD8+ T cells during GVHD, alloreactive CD4+ T cell clusters were preferentially expanded. Moreover, CNIs mediated reversible dose-dependent suppression of T cell activation and all stages of donor T cell exhaustion. Critically, CNIs promoted the expansion of both polyclonal and TCR-specific alloreactive central memory CD4+ T cells (TCM) with high self-renewal capacity that mediated cGVHD following drug withdrawal. In contrast to posttransplant cyclophosphamide (PT-Cy), CSA was ineffective in eliminating IL-17A-secreting alloreactive T cell clones that play an important role in the pathogenesis of cGVHD. Collectively, we have shown that, although CNIs attenuate aGVHD, they paradoxically rescue alloantigen-specific TCM, especially within the CD4+ compartment in lymphoid and GVHD target tissues, thus predisposing patients to cGVHD. These data provide further evidence to caution against CNI-based immune suppression without concurrent approaches that eliminate alloreactive T cell clones.
Collapse
Affiliation(s)
- Yewei Wang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Olivia G. Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Shruti S. Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kathleen S. Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Christine R. Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Samuel R.W. Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Tomoko Sekiguchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Ethan L. Nelson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rachel D. Kuns
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Nicole S. Nemychenkov
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Erden Atilla
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Albert C. Yeh
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Julie R. Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Simone A. Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Scott N. Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pediatrics and
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Geoffrey R. Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Medical Oncology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
13
|
Fuchs KJ, Falkenburg JHF, Griffioen M. Minor histocompatibility antigens to predict, monitor or manipulate GvL and GvHD after allogeneic hematopoietic cell transplantation. Best Pract Res Clin Haematol 2024; 37:101555. [PMID: 39098803 DOI: 10.1016/j.beha.2024.101555] [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: 02/29/2024] [Revised: 04/22/2024] [Accepted: 05/08/2024] [Indexed: 08/06/2024]
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) provides a potential curative treatment for haematological malignancies. The therapeutic Graft-versus-Leukaemia (GvL) effect is induced by donor T cells attacking patient hematopoietic (malignant) cells. However, if healthy non-hematopoietic tissues are targeted, Graft-versus-Disease (GvHD) may develop. After HLA-matched alloHCT, GvL and GvHD are induced by donor T cells recognizing polymorphic peptides presented by HLA on patient cells, so-called minor histocompatibility antigens (MiHAs). The balance between GvL and GvHD depends on the tissue distribution of MiHAs and T-cell frequencies targeting these MiHAs. T cells against broadly expressed MiHAs induce GvL and GvHD, whereas those targeting MiHAs with hematopoietic-restricted expression induce GvL without GvHD. Recently, the MiHA repertoire identified in natural immune responses after alloHCT was expanded to 159 total HLA-I-restricted MiHAs, including 14 hematopoietic-restricted MiHAs. This review explores their potential relevance to predict, monitor, and manipulate GvL and GvHD for improving clinical outcome after HLA-matched alloHCT.
Collapse
Affiliation(s)
- Kyra J Fuchs
- Department of Hematology, Leiden University Medical Center, 2300, RC, Leiden, the Netherlands
| | - J H Frederik Falkenburg
- Department of Hematology, Leiden University Medical Center, 2300, RC, Leiden, the Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, 2300, RC, Leiden, the Netherlands.
| |
Collapse
|
14
|
Fuchs KJ, van de Meent M, Honders MW, Khatri I, Kester MGD, Koster EAS, Koutsoumpli G, de Ru AH, van Bergen CAM, van Veelen PA, ’t Hoen PAC, van Balen P, van den Akker EB, Veelken JH, Halkes CJM, Falkenburg JHF, Griffioen M. Expanding the repertoire reveals recurrent, cryptic, and hematopoietic HLA class I minor histocompatibility antigens. Blood 2024; 143:1856-1872. [PMID: 38427583 PMCID: PMC11076866 DOI: 10.1182/blood.2023022343] [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: 09/13/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 03/03/2024] Open
Abstract
ABSTRACT Allogeneic stem cell transplantation (alloSCT) is a curative treatment for hematological malignancies. After HLA-matched alloSCT, antitumor immunity is caused by donor T cells recognizing polymorphic peptides, designated minor histocompatibility antigens (MiHAs), that are presented by HLA on malignant patient cells. However, T cells often target MiHAs on healthy nonhematopoietic tissues of patients, thereby inducing side effects known as graft-versus-host disease. Here, we aimed to identify the dominant repertoire of HLA-I-restricted MiHAs to enable strategies to predict, monitor or modulate immune responses after alloSCT. To systematically identify novel MiHAs by genome-wide association screening, T-cell clones were isolated from 39 transplanted patients and tested for reactivity against 191 Epstein-Barr virus transformed B cell lines of the 1000 Genomes Project. By discovering 81 new MiHAs, we more than doubled the antigen repertoire to 159 MiHAs and demonstrated that, despite many genetic differences between patients and donors, often the same MiHAs are targeted in multiple patients. Furthermore, we showed that one quarter of the antigens are cryptic, that is translated from unconventional open reading frames, for example long noncoding RNAs, showing that these antigen types are relevant targets in natural immune responses. Finally, using single cell RNA-seq data, we analyzed tissue expression of MiHA-encoding genes to explore their potential role in clinical outcome, and characterized 11 new hematopoietic-restricted MiHAs as potential targets for immunotherapy. In conclusion, we expanded the repertoire of HLA-I-restricted MiHAs and identified recurrent, cryptic and hematopoietic-restricted antigens, which are fundamental to predict, follow or manipulate immune responses to improve clinical outcome after alloSCT.
Collapse
Affiliation(s)
- Kyra J. Fuchs
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marian van de Meent
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - M. Willy Honders
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Indu Khatri
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michel G. D. Kester
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eva A. S. Koster
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Georgia Koutsoumpli
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Peter A. van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A. C. ’t Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik B. van den Akker
- Center for Computational Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - J. Hendrik Veelken
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
15
|
Maurer K, Antin JH. The graft versus leukemia effect: donor lymphocyte infusions and cellular therapy. Front Immunol 2024; 15:1328858. [PMID: 38558819 PMCID: PMC10978651 DOI: 10.3389/fimmu.2024.1328858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many hematologic malignancies as well as non-malignant conditions. Part of the curative basis underlying HSCT for hematologic malignancies relies upon induction of the graft versus leukemia (GVL) effect in which donor immune cells recognize and eliminate residual malignant cells within the recipient, thereby maintaining remission. GVL is a clinically evident phenomenon; however, specific cell types responsible for inducing this effect and molecular mechanisms involved remain largely undefined. One of the best examples of GVL is observed after donor lymphocyte infusions (DLI), an established therapy for relapsed disease or incipient/anticipated relapse. DLI involves infusion of peripheral blood lymphocytes from the original HSCT donor into the recipient. Sustained remission can be observed in 20-80% of patients treated with DLI depending upon the underlying disease and the intrinsic burden of targeted cells. In this review, we will discuss current knowledge about mechanisms of GVL after DLI, experimental strategies for augmenting GVL by manipulation of DLI (e.g. neoantigen vaccination, specific cell type selection/depletion) and research outlook for improving DLI and cellular immunotherapies for hematologic malignancies through better molecular definition of the GVL effect.
Collapse
Affiliation(s)
| | - Joseph H. Antin
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Wang Y, He S, Calendo G, Bui T, Tian Y, Lee CY, Zhou Y, Zhao X, Abraham C, Mo W, Chen M, Sanders-Braggs R, Madzo J, Issa JP, Hexner EO, Wiest DL, Reshef R, Xue HH, Zhang Y. Tissue-infiltrating alloreactive T cells require Id3 to deflect PD-1-mediated immune suppression during GVHD. Blood 2024; 143:166-177. [PMID: 37871574 PMCID: PMC10797551 DOI: 10.1182/blood.2023021126] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/22/2023] [Accepted: 09/22/2023] [Indexed: 10/25/2023] Open
Abstract
ABSTRACT Persisting alloreactive donor T cells in target tissues are a determinant of graft-versus-host disease (GVHD), but the transcriptional regulators that control the persistence and function of tissue-infiltrating T cells remain elusive. We demonstrate here that Id3, a DNA-binding inhibitor, is critical for sustaining T-cell responses in GVHD target tissues in mice, including the liver and intestine. Id3 loss results in aberrantly expressed PD-1 in polyfunctional T helper 1 (Th1) cells, decreased tissue-infiltrating PD-1+ polyfunctional Th1 cell numbers, impaired maintenance of liver TCF-1+ progenitor-like T cells, and inhibition of GVHD. PD-1 blockade restores the capacity of Id3-ablated donor T cells to mediate GVHD. Single-cell RNA-sequencing analysis revealed that Id3 loss leads to significantly decreased CD28- and PI3K/AKT-signaling activity in tissue-infiltrating polyfunctional Th1 cells, an indicator of active PD-1/PD-L1 effects. Id3 is also required for protecting CD8+ T cells from the PD-1 pathway-mediated suppression during GVHD. Genome-wide RNA-sequencing analysis reveals that Id3 represses transcription factors (e.g., Nfatc2, Fos, Jun, Ets1, and Prdm1) that are critical for PD-1 transcription, exuberant effector differentiation, and interferon responses and dysfunction of activated T cells. Id3 achieves these effects by restraining the chromatin accessibility for these transcription factors. Id3 ablation in donor T cells preserved their graft vs tumor effects in mice undergoing allogeneic hematopoietic stem cell transplantation. Furthermore, CRISPR/Cas9 knockout of ID3 in human CD19-directed chimeric antigen receptor T cells retained their antitumor activity in NOD/SCID/IL2Rg-/- mice early after administration. These findings identify that ID3 is an important target to reduce GVHD, and the gene-editing program of ID3 may have broad implications in T-cell-based immunotherapy.
Collapse
Affiliation(s)
- Ying Wang
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | - Shan He
- Fels Institute and Department of Cancer Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | | | - Tien Bui
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | - Yuanyuan Tian
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | - Che Young Lee
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | - Yan Zhou
- Fox Chase Cancer Center, Temple University, Philadelphia, PA
| | - Xin Zhao
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | - Ciril Abraham
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
- Fels Institute and Department of Cancer Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Wenbin Mo
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | - Mimi Chen
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | | | - Jozef Madzo
- Coriell Institute for Medical Research, Camden, NJ
| | | | - Elizabeth O. Hexner
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David L. Wiest
- Fox Chase Cancer Center, Temple University, Philadelphia, PA
| | - Ran Reshef
- Blood and Marrow Transplantation and Cell Therapy Program, Columbia University Irving Medical Center, New York, NY
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
| | - Yi Zhang
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ
- Fels Institute and Department of Cancer Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
17
|
Bernardi C, Simonetta F. Id3 keeps the PD-1 checkpoint in check in GVHD. Blood 2024; 143:103-104. [PMID: 38206642 DOI: 10.1182/blood.2023022712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024] Open
|
18
|
Teshima T, Hashimoto D. Separation of GVL from GVHD -location, location, location. Front Immunol 2023; 14:1296663. [PMID: 38116007 PMCID: PMC10728488 DOI: 10.3389/fimmu.2023.1296663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for various hematologic malignancies. However, alloimmune response is a double-edged sword that mediates both beneficial graft-versus-leukemia (GVL) effects and harmful graft-versus-host disease (GVHD). Separation of GVL effects from GVHD has been a topic of intense research to improve transplant outcomes, but reliable clinical strategies have not yet been established. Target tissues of acute GVHD are the skin, liver, and intestine, while leukemic stem cells reside in the bone marrow. Tissue specific effector T-cell migration is determined by a combination of inflammatory and chemotactic signals that interact with specific receptors on T cells. Specific inhibition of donor T cell migration to GVHD target tissues while preserving migration to the bone marrow may represent a novel strategy to separate GVL from GVHD. Furthermore, tissue specific GVHD therapy, promoting tissue tolerance, and targeting of the tumor immune microenvironment may also help to separate GVHD and GVL.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | | |
Collapse
|
19
|
Quann K, Sacirbegovic F, Shlomchik WD. Natalizumab for GVHD: too little or too late? Blood Adv 2023; 7:5187-5188. [PMID: 37698891 PMCID: PMC10505719 DOI: 10.1182/bloodadvances.2023010486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023] Open
Affiliation(s)
- Kevin Quann
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Faruk Sacirbegovic
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Warren D Shlomchik
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
20
|
Tang L, Huang Z, Mei H, Hu Y. Immunotherapy in hematologic malignancies: achievements, challenges and future prospects. Signal Transduct Target Ther 2023; 8:306. [PMID: 37591844 PMCID: PMC10435569 DOI: 10.1038/s41392-023-01521-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/19/2023] Open
Abstract
The immune-cell origin of hematologic malignancies provides a unique avenue for the understanding of both the mechanisms of immune responsiveness and immune escape, which has accelerated the progress of immunotherapy. Several categories of immunotherapies have been developed and are being further evaluated in clinical trials for the treatment of blood cancers, including stem cell transplantation, immune checkpoint inhibitors, antigen-targeted antibodies, antibody-drug conjugates, tumor vaccines, and adoptive cell therapies. These immunotherapies have shown the potential to induce long-term remission in refractory or relapsed patients and have led to a paradigm shift in cancer treatment with great clinical success. Different immunotherapeutic approaches have their advantages but also shortcomings that need to be addressed. To provide clinicians with timely information on these revolutionary therapeutic approaches, the comprehensive review provides historical perspectives on the applications and clinical considerations of the immunotherapy. Here, we first outline the recent advances that have been made in the understanding of the various categories of immunotherapies in the treatment of hematologic malignancies. We further discuss the specific mechanisms of action, summarize the clinical trials and outcomes of immunotherapies in hematologic malignancies, as well as the adverse effects and toxicity management and then provide novel insights into challenges and future directions.
Collapse
Affiliation(s)
- Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Zhongpei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
21
|
Senjo H, Harada S, Kubota SI, Tanaka Y, Tateno T, Zhang Z, Okada S, Chen X, Kikuchi R, Miyashita N, Onozawa M, Goto H, Endo T, Hasegawa Y, Ohigashi H, Ara T, Hasegawa Y, Murakami M, Teshima T, Hashimoto D. Calcineurin inhibitor inhibits tolerance induction by suppressing terminal exhaustion of donor T cells after allo-HCT. Blood 2023; 142:477-492. [PMID: 37216687 DOI: 10.1182/blood.2023019875] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/11/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Calcineurin inhibitor-based graft-versus-host disease (GVHD) prophylaxis is standard in allogeneic hematopoietic stem cell transplantation (HCT) but fails to induce long-term tolerance without chronic GVHD (cGVHD) in a considerable number of patients. In this study, we addressed this long-standing question in mouse models of HCT. After HCT, alloreactive donor T cells rapidly differentiated into PD-1+ TIGIT+ terminally exhausted T cells (terminal Tex). GVHD prophylaxis with cyclosporine (CSP) suppressed donor T-cell expression of TOX, a master regulator to promote differentiation of transitory exhausted T cells (transitory Tex), expressing both inhibitory receptors and effector molecules, into terminal Tex, and inhibited tolerance induction. Adoptive transfer of transitory Tex, but not terminal Tex, into secondary recipients developed cGVHD. Transitory Tex maintained alloreactivity and thus PD-1 blockade restored graft-versus-leukemia (GVL) activity of transitory Tex and not terminal Tex. In conclusion, CSP inhibits tolerance induction by suppressing the terminal exhaustion of donor T cells, while maintaining GVL effects to suppress leukemia relapse.
Collapse
Affiliation(s)
- Hajime Senjo
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinpei Harada
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shimpei I Kubota
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Tanaka
- Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takahiro Tateno
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Zixuan Zhang
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Satomi Okada
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Xuanzhong Chen
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Kikuchi
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoki Miyashita
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiro Onozawa
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideki Goto
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tomoyuki Endo
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuta Hasegawa
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Ohigashi
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahide Ara
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshinori Hasegawa
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Masaaki Murakami
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Group of Quantum Immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
- Division of Molecular Neuroimmunology, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
- Division of Biological Response Analysis, Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daigo Hashimoto
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
22
|
Xin Y, Xiong Y, Liu F, Qu L, Li W, Yang L, Liu Y, Zhu J. Quantitative analysis of contrast-enhanced ultrasonography in rat models of hepatic acute graft-versus-host disease. Quant Imaging Med Surg 2023; 13:4908-4918. [PMID: 37581062 PMCID: PMC10423398 DOI: 10.21037/qims-22-1145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/09/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Hepatic acute graft-versus-host disease (aGVHD) is a major life-threatening complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). We hypothesized that contrast-enhanced ultrasound (CEUS) could serve as a new imaging biomarker in early diagnosis of hepatic aGVHD by detecting liver microcirculation. METHODS Thirty Wistar rats received allo-HSCT were finally included after excluding 9 rats, and they were randomly divided into 5 groups (1- to 5-week groups, 6 per group). Six rats were used for the control group without any intervention. We observed the clinical scores, serum liver enzyme levels and liver CEUS parameters of rats in each group. Hepatic aGVHD was finally confirmed by histopathologic analysis. The diagnostic performance of CEUS parameters in detecting GVHD was evaluated by comparing the area under the receiver operating curve (AUC) values. RESULTS After HSCT, the rats developed ruffling of fur, maculopapular rash, weight loss, accompanied by increased clinical scores. Serum liver enzymes were significantly higher than those in the control group from the third week, especially alkaline phosphatase, while CEUS parameters, peak intensity (PI) and mean transit time (MTT), changed in the second week (P<0.001). Compared with non-aGVHD group, the PI was significantly decreased while time to peak and MTT were prolonged in aGVHD group. CEUS parameters were more strongly correlated with pathological grade than serology. PI was an independent predictor for hepatic aGVHD. The AUC of CEUS parameters for diagnosing hepatic aGVHD was 0.933 (95% CI: 0.779-0.992), which was higher than that of clinical scores (AUC =0.748, 95% CI: 0.557-0.888, P=0.032) and serological markers (AUC =0.902, 95% CI: 0.737-0.980, P=0.694). CONCLUSIONS CEUS exhibits promising applications as a quantitative method to detect hepatic aGVHD and early liver damage.
Collapse
Affiliation(s)
- Yuwei Xin
- Department of Ultrasound, Peking University People’s Hospital, Beijing, China
| | - Yu Xiong
- Department of Ultrasound, Peking University People’s Hospital, Beijing, China
| | - Feifei Liu
- Department of Ultrasound, Peking University People’s Hospital, Beijing, China
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, China
| | - Linlin Qu
- Department of Pathology, Peking University People’s Hospital, Beijing, China
| | - Wenxue Li
- Department of Ultrasound, Peking University People’s Hospital, Beijing, China
| | - Li Yang
- Department of Ultrasound, Peking University People’s Hospital, Beijing, China
| | - Yiqun Liu
- Department of Ultrasound, Peking University People’s Hospital, Beijing, China
| | - Jia’an Zhu
- Department of Ultrasound, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
23
|
Brandstadter JD, Outen R, Maillard I. Having it both ways: how STAT3 deficiency blocks graft-versus-host disease while preserving graft-versus-leukemia activity. J Clin Invest 2023; 133:e172251. [PMID: 37526083 PMCID: PMC10378150 DOI: 10.1172/jci172251] [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] [Indexed: 08/02/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation can cure patients with high-risk leukemia through graft-versus-leukemia (GVL) effects, the process by which malignant leukemic cells are cleared by donor-derived immune cells from the graft. The problem of harnessing GVL effects while controlling inflammation and host-organ damage linked with graft-versus-host disease (GVHD) has been the most formidable hurdle facing allogeneic hematopoietic cell transplantation. This powerful, curative-intent therapy remains among the most toxic treatments in the hematologist's armamentarium due to the combined risks of GVHD-related morbidity, infections, and leukemia relapse. In this issue of the JCI, Li, Wang, et al. report that T cell Stat3 deficiency can extricate GVL effects from GVHD through tissue-specific programmed death-ligand 1/programmed cell death protein 1-dependent (PD-L1/PD-1-dependent) bioenergetic alterations that blunt harmful T cell effects in GVHD target organs, while preserving their beneficial antitumor activity in lymphohematopoietic tissues.
Collapse
|
24
|
DeWolf S, Elhanati Y, Nichols K, Waters NR, Nguyen CL, Slingerland JB, Rodriguez N, Lyudovyk O, Giardina PA, Kousa AI, Andrlová H, Ceglia N, Fei T, Kappagantula R, Li Y, Aleynick N, Baez P, Murali R, Hayashi A, Lee N, Gipson B, Rangesa M, Katsamakis Z, Dai A, Blouin AG, Arcila M, Masilionis I, Chaligne R, Ponce DM, Landau HJ, Politikos I, Tamari R, Hanash AM, Jenq RR, Giralt SA, Markey KA, Zhang Y, Perales MA, Socci ND, Greenbaum BD, Iacobuzio-Donahue CA, Hollmann TJ, van den Brink MR, Peled JU. Tissue-specific features of the T cell repertoire after allogeneic hematopoietic cell transplantation in human and mouse. Sci Transl Med 2023; 15:eabq0476. [PMID: 37494469 PMCID: PMC10758167 DOI: 10.1126/scitranslmed.abq0476] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/06/2023] [Indexed: 07/28/2023]
Abstract
T cells are the central drivers of many inflammatory diseases, but the repertoire of tissue-resident T cells at sites of pathology in human organs remains poorly understood. We examined the site-specificity of T cell receptor (TCR) repertoires across tissues (5 to 18 tissues per patient) in prospectively collected autopsies of patients with and without graft-versus-host disease (GVHD), a potentially lethal tissue-targeting complication of allogeneic hematopoietic cell transplantation, and in mouse models of GVHD. Anatomic similarity between tissues was a key determinant of TCR repertoire composition within patients, independent of disease or transplant status. The T cells recovered from peripheral blood and spleens in patients and mice captured a limited portion of the TCR repertoire detected in tissues. Whereas few T cell clones were shared across patients, motif-based clustering revealed shared repertoire signatures across patients in a tissue-specific fashion. T cells at disease sites had a tissue-resident phenotype and were of donor origin based on single-cell chimerism analysis. These data demonstrate the complex composition of T cell populations that persist in human tissues at the end stage of an inflammatory disorder after lymphocyte-directed therapy. These findings also underscore the importance of studying T cell in tissues rather than blood for tissue-based pathologies and suggest the tissue-specific nature of both the endogenous and posttransplant T cell landscape.
Collapse
Affiliation(s)
- Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yuval Elhanati
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katherine Nichols
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas R. Waters
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chi L. Nguyen
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John B. Slingerland
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasia Rodriguez
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olga Lyudovyk
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul A. Giardina
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anastasia I. Kousa
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hana Andrlová
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nick Ceglia
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rajya Kappagantula
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yanyun Li
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nathan Aleynick
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Priscilla Baez
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rajmohan Murali
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Akimasa Hayashi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, Kyorin University, Mitaka City, Tokyo, Japan
| | - Nicole Lee
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brianna Gipson
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Madhumitha Rangesa
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zoe Katsamakis
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anqi Dai
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amanda G. Blouin
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Arcila
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ignas Masilionis
- Program for Computational and System Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronan Chaligne
- Program for Computational and System Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doris M. Ponce
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Heather J. Landau
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Ioannis Politikos
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Roni Tamari
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Alan M. Hanash
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert R. Jenq
- Departments of Genomic Medicine and Stem Cell Transplantation Cellular Therapy, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sergio A. Giralt
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Kate A. Markey
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington; Seattle, WA, USA
| | - Yanming Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Nicholas D. Socci
- Bioinformatics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin D. Greenbaum
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Travis J. Hollmann
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Bristol Myers Squibb, Lawrenceville, NJ 08540
| | - Marcel R.M. van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jonathan U. Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
25
|
Nizharadze T, Becker NB, Höfer T. Quantitating CD8 + T cell memory development. Trends Immunol 2023; 44:519-529. [PMID: 37277233 DOI: 10.1016/j.it.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/07/2023]
Abstract
In acute immune responses to infection, memory T cells develop that can spawn recall responses. This process has not been observable directly in vivo. Here we highlight the utility of mathematical inference to derive quantitatively testable models of mammalian CD8+ T cell memory development from complex experimental data. Previous inference studies suggested that precursors of memory T cells arise early during the immune response. Recent work has both validated a crucial prediction of this T cell diversification model and refined the model. While multiple developmental routes to distinct memory subsets might exist, a branch point occurs early in proliferating T cell blasts, from which separate differentiation pathways emerge for slowly dividing precursors of re-expandable memory cells and rapidly dividing effectors.
Collapse
Affiliation(s)
- Tamar Nizharadze
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Nils B Becker
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
26
|
Abstract
Cytotoxic CD8+ T cells recognize and eliminate infected or cancerous cells. A subset of CD8+ memory T cells called tissue-resident memory T cells (TRM ) resides in peripheral tissues, monitors the periphery for pathogen invasion, and offers a rapid and potent first line of defense at potential sites of re-infection. TRM cells are found in almost all tissues and are transcriptionally and epigenetically distinct from circulating memory populations, which shows their ability to acclimate to the tissue environment to allow for long-term survival. Recent work and the broader availability of single-cell profiling have highlighted TRM heterogeneity among different tissues, as well as identified specialized subsets within individual tissues, that are time and infection dependent. TRM cell phenotypic and transcriptional heterogeneity has implications for understanding TRM function and longevity. This review aims to summarize and discuss the latest findings on CD8+ TRM heterogeneity using single-cell molecular profiling and explore the potential implications for immune protection and the design of immune therapies.
Collapse
Affiliation(s)
- Maximilian Heeg
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, USA
| | - Ananda W Goldrath
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
27
|
Gail LM, Schell KJ, Łacina P, Strobl J, Bolton SJ, Steinbakk Ulriksen E, Bogunia-Kubik K, Greinix H, Crossland RE, Inngjerdingen M, Stary G. Complex interactions of cellular players in chronic Graft-versus-Host Disease. Front Immunol 2023; 14:1199422. [PMID: 37435079 PMCID: PMC10332803 DOI: 10.3389/fimmu.2023.1199422] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/07/2023] [Indexed: 07/13/2023] Open
Abstract
Chronic Graft-versus-Host Disease is a life-threatening inflammatory condition that affects many patients after allogeneic hematopoietic stem cell transplantation. Although we have made substantial progress in understanding disease pathogenesis and the role of specific immune cell subsets, treatment options are still limited. To date, we lack a global understanding of the interplay between the different cellular players involved, in the affected tissues and at different stages of disease development and progression. In this review we summarize our current knowledge on pathogenic and protective mechanisms elicited by the major involved immune subsets, being T cells, B cells, NK cells and antigen presenting cells, as well as the microbiome, with a special focus on intercellular communication of these cell types via extracellular vesicles as up-and-coming fields in chronic Graft-versus-Host Disease research. Lastly, we discuss the importance of understanding systemic and local aberrant cell communication during disease for defining better biomarkers and therapeutic targets, eventually enabling the design of personalized treatment schemes.
Collapse
Affiliation(s)
- Laura Marie Gail
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kimberly Julia Schell
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Piotr Łacina
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Steven J. Bolton
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Hildegard Greinix
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Rachel Emily Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|