1
|
Shneider NA, Nesta AV, Rifai OM, Yasek J, Elyaman W, Aziz-Zaman S, Lyu MA, Levy SHS, Hoover BN, Vlad G, Huang M, Zeng K, Sadeghi T, Reddy A, Flowers CR, Parmar S. Clinical Safety and Preliminary Efficacy of Regulatory T Cells for ALS. NEJM EVIDENCE 2025; 4:EVIDoa2400249. [PMID: 40261116 DOI: 10.1056/evidoa2400249] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BACKGROUND Peripheral and neuroinflammation have been previously associated with progression in amyotrophic lateral sclerosis (ALS), a neurodegenerative disease involving progressive loss of motor neurons. We hypothesize that regulatory T cell (Treg) therapy can resolve inflammation and preserve function in those patients with ALS. METHODS Participants with ALS received infusions of a fixed dose (100×106 cells) of umbilical cord blood-derived, allogeneic, nonhuman leukocyte antigen-matched, cryopreserved Treg product (TREG), administered as four weekly infusions followed by six monthly infusions. No lymphodepletion, immunosuppression, or interleukin 2 was administered. The primary outcome was dose-limiting toxicity, including infusion reaction within 24 hours (as graded by National Cancer Institute - Common Terminology Criteria for Adverse Events, Version 4.0) and/or regimen-related death, or grade 3 or 4 cytokine release syndrome within 14 days postinfusion. We measured clinical response using the Revised ALS Functional Rating Scale (ALSFRS-R; range 0 to 48, with lower numbers indicating lower functional ability). Exploratory analyses measured serum and plasma neurofilament light (NfL) and inflammatory biomarkers. RESULTS Six participants with a median age of 48.5 years (range 27 to 66 years) and baseline ALSFRS-R score of 31.5 (range 23 to 43) were treated with a median of 11 (range 6 to 22) TREG infusions in an ambulatory setting. No dose-limiting toxicity was observed. In participants with sufficient data points (n=4), the mean ALSFRS-R slope of decline was -1.66±1.03 points/month before treatment, -0.41±0.45/month during treatment, and -0.60±0.59/month posttreatment. Biomarkers including NfL and inflammatory markers MIP-1δ (macrophage inflammatory protein-1 delta), CTACK (cutaneous T cell-attracting chemokine), and GROα (growth-regulated oncogene alpha) exhibited different relationships with ALSFRS-R score between participants. CONCLUSIONS This study demonstrates the preliminary safety of "off-the-shelf", allogeneic Treg-cell therapy.
Collapse
Affiliation(s)
| | | | | | - Julia Yasek
- Department of Neurology, Columbia University, New York
| | | | | | - Mi-Ae Lyu
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston
| | | | | | - George Vlad
- Department of Neurology, Columbia University, New York
| | - Meixian Huang
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston
| | - Ke Zeng
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Houston
| | | | | | | | - Simrit Parmar
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, Bryan, TX
| |
Collapse
|
2
|
Mughal TI, Mascarenhas J, Rampal RK, Bose P, Lion T, Ajufo H, Yacoub A, Meshinchi S, Masarova L, Mesa R, Jamieson C, Barbui T, Saglio G, Van Etten RA. Impact of Recent Translational and Therapeutic Developments on Clinical Course of BCR::ABL1-Positive and -Negative Myeloproliferative Neoplasms. Hematol Oncol 2025; 43:e70013. [PMID: 39825826 DOI: 10.1002/hon.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/23/2024] [Indexed: 01/20/2025]
Abstract
Despite the study of BCR::ABL1-positive and -negative myeloproliferative neoplasms (MPNs) providing seminal insights into cancer biology, tumor evolution and precision oncology over the past half century, significant challenges remain. MPNs are clonal hematopoietic stem cell-derived neoplasms with heterogenous clinical phenotypes and a clonal architecture which impacts the often-complex underlying genetics and microenvironment. The major driving molecular abnormalities have been well characterized, but debate on their role as disease-initiating molecular lesions continues. The introduction of the ABL1 tyrosine kinase inhibitors have been extremely successful in the treatment of chronic myeloid leukemia with most patients having a near-normal life expectancy. Similar success has, however, not been achieved for BCR::ABL1-negative MPNs in terms of disease course modification and most patients remain incurable. In both disease categories, genomic instability seems to increase the risk of disease progression to accelerated/blast phase, which is resistant/refractory to conventional treatment and associated with a poor prognosis. To address some of these issues, the late John Goldman and Tariq Mughal founded a scientific and clinical platform in 2006, the Post-American Society of Hematology (ASH) MPN workshop, to appraise novel cancer biology, candidate therapeutic targets, treatments and other clinical challenges and pay tribute to all the many scientists and clinicians around the world instrumental to the progress made and continuing advances being made. This paper summarizes some of the recent data discussed at the 18th edition of the workshop and includes reference to some data presented or published after the workshop, including the 26th John Goldman CML conference.
Collapse
Affiliation(s)
- Tariq I Mughal
- Tufts University Medical Center, Boston, Massachusetts, USA
- Beckmann Research Institute of City of Hope, Duarte, California, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Raajit K Rampal
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Thomas Lion
- St.Anna Children's Cancer Research Institute, Vienna, Austria
| | - Helen Ajufo
- John T. Milliken Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Abdulraheem Yacoub
- Division of Hematologic Malignancies, University of Kansas, Kansas City, Kansas, USA
| | | | | | - Ruben Mesa
- Wake Forest University School of Medicine, Atrium Health Levine Cancer Institute, Charlotte, North Carolina, USA
| | - Catriona Jamieson
- University of San Diego, Moores Cancer Center, La Jolla, California, USA
| | | | | | | |
Collapse
|
3
|
Zeng K, Ma H, Huang M, Lyu MA, Sadeghi T, Flowers CR, Parmar S. Cord blood T regulatory cells synergize with ruxolitinib to improve GVHD outcomes. FRONTIERS IN TRANSPLANTATION 2024; 3:1448650. [PMID: 39722683 PMCID: PMC11668690 DOI: 10.3389/frtra.2024.1448650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024]
Abstract
Background Adoptive therapy with umbilical cord blood (UCB) T-regulatory (Treg) cells can prevent graft vs. host disease (GVHD). We hypothesize that UCB Tregs can treat GVHD and synergize with ruxolitinib, Jak2 inhibitor, to improve outcomes. Methods UCB Treg potency and efficacy was examined using cell suppression assay and xenogeneic GVHD model, respectively. Ruxolitinib was fed continuously in presence or absence of CellTraceViolet tagged UCB Tregs on days +4, +7, +11, +18. Mice were followed for survival, GVHD score, hematology parameters and inflammation. Results Addition of ruxolitinib to UCB Tregs exerted synergistic suppressor function in vitro and improved persistence of UCB Tregs in vivo. Lower GVHD score, improved survival, increased hemoglobin level and platelet count, decreased inflammatory cytokines and decrease in CD3+ T cell lung infiltrate was observed in UCB Tregs+ruxolitinib recipients. Conclusion UCB Treg+Ruxolitinib combination improves outcomes in xenogeneic GVHD and should be explored in a clinical setting.
Collapse
Affiliation(s)
- Ke Zeng
- Department of Lymphoma/Myeloma, The University of Texas at MD Anderson Cancer Center, Houston, TX, United States
| | - Hongbing Ma
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Meixian Huang
- Department of Lymphoma/Myeloma, The University of Texas at MD Anderson Cancer Center, Houston, TX, United States
| | - Mi-Ae Lyu
- Department of Lymphoma/Myeloma, The University of Texas at MD Anderson Cancer Center, Houston, TX, United States
| | | | - Christopher R. Flowers
- Department of Lymphoma/Myeloma, The University of Texas at MD Anderson Cancer Center, Houston, TX, United States
| | - Simrit Parmar
- Department of Microbial Pathogenesis & Immunology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
4
|
Bulliard Y, Freeborn R, Uyeda MJ, Humes D, Bjordahl R, de Vries D, Roncarolo MG. From promise to practice: CAR T and Treg cell therapies in autoimmunity and other immune-mediated diseases. Front Immunol 2024; 15:1509956. [PMID: 39697333 PMCID: PMC11653210 DOI: 10.3389/fimmu.2024.1509956] [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: 10/11/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
Autoimmune diseases, characterized by the immune system's attack on the body's own tissues, affect millions of people worldwide. Current treatments, which primarily rely on broad immunosuppression and symptom management, are often associated with significant adverse effects and necessitate lifelong therapy. This review explores the next generation of therapies for immune-mediated diseases, including chimeric antigen receptor (CAR) T cell and regulatory T cell (Treg)-based approaches, which offer the prospect of targeted, durable disease remission. Notably, we highlight the emergence of CD19-targeted CAR T cell therapies, and their ability to drive sustained remission in B cell-mediated autoimmune diseases, suggesting a possible paradigm shift. Further, we discuss the therapeutic potential of Type 1 and FOXP3+ Treg and CAR-Treg cells, which aim to achieve localized immune modulation by targeting their activity to specific tissues or cell types, thereby minimizing the risk of generalized immunosuppression. By examining the latest advances in this rapidly evolving field, we underscore the potential of these innovative cell therapies to address the unmet need for long-term remission and potential tolerance induction in individuals with autoimmune and immune-mediated diseases.
Collapse
Affiliation(s)
- Yannick Bulliard
- Department of Research and Development, Tr1X, Inc., San Diego, CA, United States
| | - Robert Freeborn
- Department of Research and Development, Tr1X, Inc., San Diego, CA, United States
| | - Molly Javier Uyeda
- Department of Research and Development, Tr1X, Inc., San Diego, CA, United States
| | - Daryl Humes
- Department of Research and Development, Tr1X, Inc., San Diego, CA, United States
| | - Ryan Bjordahl
- Department of Research and Development, Tr1X, Inc., San Diego, CA, United States
| | - David de Vries
- Department of Research and Development, Tr1X, Inc., San Diego, CA, United States
| | - Maria Grazia Roncarolo
- Department of Research and Development, Tr1X, Inc., San Diego, CA, United States
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
5
|
Arora S, Vachhani P, Bose P. Investigational drugs in early phase trials for myelofibrosis. Expert Opin Investig Drugs 2024; 33:1231-1244. [PMID: 39604120 PMCID: PMC11669310 DOI: 10.1080/13543784.2024.2434696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Myelofibrosis (MF) is a chronic myeloproliferative neoplasm characterized by bone marrow fibrosis, cytopenias, and organomegaly. Four JAK inhibitors are US-FDA approved for treatment of MF. While these drugs reduce symptom burden and spleen size to varying degrees, they do not affect the natural disease course or decrease the risk of leukemic transformation. Therefore, there is a strong need for newer therapies to further advance the field and improve the outcomes of MF. In this review, we cover novel therapies for MF currently in early stages of development. AREAS COVERED We present the latest data from early phase clinical trials in MF using drugs with diverse therapeutic mechanisms, including novel JAK-STAT pathway inhibitors, epigenetic therapies, antifibrotic agents, and immunotherapeutic strategies. Additionally, we cover drugs targeted toward anemia improvement in MF. EXPERT OPINION Numerous agents representing diverse drug classes are in clinical development for MF. While deeper and durable improvements in splenomegaly, symptoms, and anemia are the main clinical objectives, a number of putative biomarkers are being assessed as measures of potential 'disease modification.' Although JAK inhibitor monotherapy represents the current standard, it is hoped that JAK inhibitor-based rational combinations and driver mutation-specific therapies will soon usher in a new era.
Collapse
Affiliation(s)
- Sankalp Arora
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pankit Vachhani
- Department of Medicine, Division of Hematology and Oncology, The University of Alabama at Birmingham, Birmingham, AL
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
6
|
Huang M, Ke Z, Lyu MA, Masarova L, Sadeghi T, Flowers CR, Parmar S. CXCR4-enriched T regulatory cells preferentially home to bone marrow and resolve inflammation. iScience 2024; 27:110830. [PMID: 39314243 PMCID: PMC11418154 DOI: 10.1016/j.isci.2024.110830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/15/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
CXCR4 cell surface expression is critical for the homing of T regulatory (Treg) cells to the bone marrow (BM). We hypothesize that CXCR4 enrichment on Tregs cell surface may abbreviate their transit time to reach BM. Umbilical cord-blood CD25+ Tregs underwent CXCR4 dual enrichment and ex vivo expansion using the CRANE process to generate CXCR4-enriched Tregs (TregCXCR4) cells, which showed a faster migration across the Transwell membrane toward CXCL12/stromal cell-derived factor 1α (SDF1α) at 15, 30, and 60 min, when compared to unmanipulated Tregcontrol cells (p < 0.0001). TregCXCR4 exhibited preferential homing to BM in vivo at 12 and 24 h. Metacluster analysis of BM showed a decrease in CD8+ and an increase in CD39 and CD73 and CXCR5 when compared to Tregcontrol. TregCXCR4 decreased plasma TGF-β1/β2 and IFN-γ levels. When compared to control, TregCXCR4 cells decreased in CD8+ T cell, IFN-γ, and TNF-α expression in BM. We conclude that TregCXCR4 show enhanced migration toward CXCL12/SDF1α and a preferential homing to BM resulting in resolution of inflammation.
Collapse
Affiliation(s)
- Meixian Huang
- Department of Lymphoma/ Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Zeng Ke
- Department of Lymphoma/ Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Mi-Ae Lyu
- Department of Lymphoma/ Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Lucia Masarova
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Simrit Parmar
- Department of Lymphoma/ Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
7
|
Seguin-Devaux C, Zimmer J. Adoptive T Regulatory Cell Therapy Takes an Important Step. NEJM EVIDENCE 2024; 3:EVIDe2400109. [PMID: 38804780 DOI: 10.1056/evide2400109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Affiliation(s)
- Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| |
Collapse
|