1
|
Haney M, Devine KJ, Xavier AC, Ford JB, Audino AN, Villagomez L, Kebede A, Rubinstein JD. Posttransplant EBV-Positive Smooth Muscle Tumors in Children, Adolescents, and Young Adults: A Multi-Institution Experience. Pediatr Blood Cancer 2025; 72:e31678. [PMID: 40119578 DOI: 10.1002/pbc.31678] [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: 01/29/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/24/2025]
Abstract
Epstein-Barr virus (EBV)-positive smooth muscle tumors (SMTs) are rare tumors seen in immunocompromised patients. There is no clear standard of care for the management and treatment of EBV-SMTs. Patients are often treated with chemotherapy, surgery, and/or radiation. Additional options include antiretroviral treatment, reduction in immunosuppression, and EBV-directed virus-specific T cells (VSTs). This report describes the treatment regimens and outcomes of eight patients with EBV-associated SMTs. Although no consensus treatment for EBV-SMTs has been identified, VSTs show promise in providing a period of stable disease or partial response and surgical removal may offer long-term benefits in cases of localized disease.
Collapse
Affiliation(s)
- Meghan Haney
- University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, Ohio, USA
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, Cincinnati, Ohio, USA
| | - Kaitlin J Devine
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ana C Xavier
- Department of Pediatrics, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James B Ford
- University of Utah, Salt Lake City, Utah, USA
- Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Anthony N Audino
- Division of Pediatric Hematology/Oncology/BMT, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Lynda Villagomez
- Division of Pediatric Hematology/Oncology/BMT, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Ann Kebede
- Division of Pediatric Hematology/Oncology/BMT, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jeremy D Rubinstein
- University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, Ohio, USA
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Shahid S, Prockop SE, Flynn GC, Mauguen A, White CO, Bieler J, McAvoy D, Hosszu K, Cancio MI, Jakubowski AA, Scaradavou A, Boelens JJ, Sauter CS, Perales MA, Giralt SA, Taylor C, Chaudhari J, Wang X, Rivière I, Sadelain M, Brentjens RJ, Kernan NA, O'Reilly RJ, Curran KJ. Allogeneic off-the-shelf CAR T-cell therapy for relapsed or refractory B-cell malignancies. Blood Adv 2025; 9:1644-1657. [PMID: 39908482 PMCID: PMC11995077 DOI: 10.1182/bloodadvances.2024015157] [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: 10/23/2024] [Revised: 12/10/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
ABSTRACT Despite clinical benefit with the use of chimeric antigen receptor (CAR) T cells, the need to manufacture patient-specific products limits its clinical utility. To overcome this barrier, we developed an allogeneic "off-the-shelf" CAR T-cell product using Epstein-Barr virus (EBV)-specific T cells (EBV-VSTs) genetically modified with a CD19-specific CAR (19-28z). Patients with relapsed/refractory (R/R) B-cell malignancies were stratified into 3 treatment cohorts: cohort 1 (n = 8; disease recurrence after allogeneic or autologous hematopoietic cell transplantation [HCT]), cohort 2 (n = 6; consolidative therapy after autologous HCT), or cohort 3 (n = 2; consolidative therapy after allogeneic HCT). The primary objective of this trial was to determine the safety of multiple CAR EBV-VST infusions. Most patients (n = 12/16) received multiple doses (overall median, 2.5 [range, 1-3]) with 3 × 106 T cells per kg determined to be the optimal dose enabling multiple treatments per manufactured cell line. Severe cytokine release syndrome or neurotoxicity did not occur after infusion, and no dose-limiting toxicity was observed in the trial. Median follow-up was 48 months (range, 4-135) with 4 deaths due to disease progression. Overall survival of all patients was 81% at 12 months and 75% at 36 months. Postinfusion expansion and persistence were limited, and CAR EBV-VSTs demonstrated a unique T-cell phenotype compared with autologous 19-28z CAR T cells. Our study demonstrates the feasibility and safety of an allogeneic "off-the-shelf" CAR EBV-VST product with favorable outcomes for patients with CD19+ R/R B-cell malignancies. This trial was registered at www.ClinicalTrials.gov as #NCT01430390.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Middle Aged
- Adult
- Male
- Female
- Receptors, Chimeric Antigen/therapeutic use
- Aged
- Antigens, CD19/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transplantation, Homologous
- Hematopoietic Stem Cell Transplantation/methods
- Hematopoietic Stem Cell Transplantation/adverse effects
- Herpesvirus 4, Human
- Leukemia, B-Cell/therapy
- Treatment Outcome
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell/immunology
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/genetics
- Young Adult
Collapse
Affiliation(s)
- Sanam Shahid
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan E. Prockop
- Department of Hematopoietic Stem Cell Transplant, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Georgia C. Flynn
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Audrey Mauguen
- Department of Epidemiology-Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Charlie O. White
- Department of Epidemiology-Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jennifer Bieler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Devin McAvoy
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kinga Hosszu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria I. Cancio
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ann A. Jakubowski
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Craig S. Sauter
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | | - Sergio A. Giralt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Clare Taylor
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jagrutiben Chaudhari
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xiuyan Wang
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Isabelle Rivière
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michel Sadelain
- Department of Pharmacology, Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J. Brentjens
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Nancy A. Kernan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard J. O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kevin J. Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
3
|
Hu J, Yang C, Tan B, Xiong Q, Le Y, Hu J, Wang H, Dai X, Zhang M. Clinical and molecular genetic characteristics of pediatric PFIC3 patients: three novel variants and prognosis for parental liver transplantation. Orphanet J Rare Dis 2025; 20:164. [PMID: 40200381 PMCID: PMC11977866 DOI: 10.1186/s13023-025-03670-y] [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: 01/20/2025] [Accepted: 03/16/2025] [Indexed: 04/10/2025] Open
Abstract
Progressive Familial Intrahepatic Cholestasis Type 3 (PFIC3) is a rare inherited liver disease caused by a mutation in the ABCB4 gene, leading to dysfunction of multidrug resistance protein 3 (MDR3). The earlier the onset of PFIC3 in children is, the more severe the prognosis. The diagnosis of PFIC3 is typically based on clinical symptoms, laboratory tests, and imaging assessments, with final confirmation requiring genetic testing. The aim of this study was to investigate the associations between genetic mutations in PFIC3 and clinical features, molecular genetics, and liver histopathology to improve early recognition and understanding of this disease. By analysing the data of three children with PFIC3 who underwent parental liver transplantation, we were able to gain a deeper understanding of the complexity and diversity of the disease. With respect to molecular genetics, we identified five mutation sites in the ABCB4 gene, including three newly discovered mutations. Immunohistochemical analysis revealed reduced expression of the MDR3 protein in child 1 and no expression in child 2 or child 3, revealing an intrinsic link between the ABCB4 gene and the MDR3 protein. Histopathologically, all three patients presented with significant portal vein fibrosis or cholestatic liver cirrhosis. In conclusion, this study emphasizes the importance of molecular genetic and pathological evaluation of patients with PFIC3 mutations and elucidates the impact of these three mutations on the course of the disease in children, for whom early symptomatic treatment and early preparation for liver transplantation are options worth considering.
Collapse
Affiliation(s)
- Jiqiang Hu
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China
| | - Chenyu Yang
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China
| | - Bingqian Tan
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China
| | - Qiang Xiong
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China
| | - Ying Le
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China
| | - Jianyang Hu
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China
| | - Haoming Wang
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China
| | - Xiaoke Dai
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China.
| | - Mingman Zhang
- Department of Pediatric Hepatobiliary Surgery National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children'S Hospital of Chongqing Medical University, Chongqin, China.
| |
Collapse
|
4
|
McKenna A, Lin GA, Lee W, Raymond F, Richardson M, Agboola F. The effectiveness and value of tabelecleucel for the treatment of Epstein-Barr virus-positive posttransplant lymphoproliferative disease. J Manag Care Spec Pharm 2025; 31:429-434. [PMID: 40152797 PMCID: PMC11953852 DOI: 10.18553/jmcp.2025.31.4.429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Affiliation(s)
- Avery McKenna
- Institute for Clinical and Economic Review, Boston, MA
| | - Grace A. Lin
- Institute for Clinical and Economic Review, Boston, MA
| | - Woojung Lee
- Institute for Clinical and Economic Review, Boston, MA
| | - Finn Raymond
- Institute for Clinical and Economic Review, Boston, MA
| | | | | |
Collapse
|
5
|
Wang J, Wang R, Wang M, Ge J, Wang Y, Li Y, Chen C, He J, Zheng B, Xu M, Jiang X, Liu Y, Chen M, Long J. Cutting-Edge Therapy and Immune Escape Mechanisms in EBV-Associated Tumors. Med Res Rev 2025. [PMID: 40077924 DOI: 10.1002/med.22104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025]
Abstract
Epstein-Barr virus (EBV), the first identified human tumor virus, significantly influences the immune microenvironment of associated cancers. EBV-induced expression of viral antigens by tumor cells triggers immune recognition and elicits a pro-inflammatory response. While mild inflammation may help eliminate malignant cells, intense inflammation can accelerate tumor progression. Moreover, EBV can establish lifelong latency in human hosts, characterized by low immunogenicity of its proteins and noncoding RNAs. This enables tumor cells to evade immune detection and impair immune cell function, disrupting immune homeostasis. Consequently, EBV-associated malignancies pose a considerable public health challenge globally, often complicating the prognosis of cancer patients under conventional treatment. With deeper research into the oncogenic expressions and mechanisms of EBV, novel targeted therapies against EBV are gaining prominence. This review discusses recent advancements in understanding how EBV helps tumor cells evade immune surveillance and induce immune dysfunction. It also examines the clinical potential of targeting EBV-associated tumors, providing fresh perspectives on the mechanisms and therapeutic strategies for these cancers.
Collapse
Affiliation(s)
- Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Rong Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Junshang Ge
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Yian Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University; The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, China
| | - Yanhan Li
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Changan Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Jiale He
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Boshu Zheng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Meifang Xu
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuhang Liu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Mingfen Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Fujian Medical University, Quanzhou, China
| | - Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| |
Collapse
|
6
|
Wang V, Savoldo B, Guimaraes JA, Dotti G, Reppel L, Bensoussan D. Alloreactive-free CAR-VST therapy: a step forward in long-term tumor control in viral context. Front Immunol 2025; 15:1527648. [PMID: 39882248 PMCID: PMC11774747 DOI: 10.3389/fimmu.2024.1527648] [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: 11/13/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
CAR-T cell therapy has revolutionized immunotherapy but its allogeneic application, using various strategies, faces significant challenges including graft-versus-host disease and graft rejection. Recent advances using Virus Specific T cells to generate CAR-VST have demonstrated potential for enhanced persistence and antitumor efficacy, positioning CAR-VSTs as a promising alternative to conventional CAR-T cells in an allogeneic setting. This review provides a comprehensive overview of CAR-VST development, emphasizing strategies to mitigate immunogenicity, such as using a specialized TCR, and approaches to improve therapeutic persistence against host immune responses. In this review, we discuss the production methods of CAR-VSTs and explore optimization strategies to enhance their functionality, activation profiles, memory persistence, and exhaustion resistance. Emphasis is placed on their unique dual specificity for both antitumor and antiviral responses, along with an in-depth examination of preclinical and clinical outcomes. We highlight how these advances contribute to the efficacy and durability of CAR-VSTs in therapeutic settings, offering new perspectives for broad clinical applications. By focusing on the key mechanisms that enable CAR-VSTs to address autologous CAR-T cell challenges, this review highlights their potential as a promising strategy for developing effective allogeneic CAR-T therapies.
Collapse
Affiliation(s)
- Valentine Wang
- Unité Mixte de Recherche (UMR) 7365 Centre National de la Recherche Scientifique (CNRS), Ingénierie Moléculaire, Cellulaire et Physiopathologie (IMoPA), Université de Lorraine, Nancy, France
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - José-Arthur Guimaraes
- Unité Mixte de Recherche (UMR) 7365 Centre National de la Recherche Scientifique (CNRS), Ingénierie Moléculaire, Cellulaire et Physiopathologie (IMoPA), Université de Lorraine, Nancy, France
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Loïc Reppel
- Unité Mixte de Recherche (UMR) 7365 Centre National de la Recherche Scientifique (CNRS), Ingénierie Moléculaire, Cellulaire et Physiopathologie (IMoPA), Université de Lorraine, Nancy, France
- Centre Hospitalier Régional Universitaire (CHRU) Nancy, Cell Therapy and Tissue Bank Unit, MTInov Bioproduction and Biotherapy Integrator, Nancy, France
| | - Danièle Bensoussan
- Unité Mixte de Recherche (UMR) 7365 Centre National de la Recherche Scientifique (CNRS), Ingénierie Moléculaire, Cellulaire et Physiopathologie (IMoPA), Université de Lorraine, Nancy, France
- Centre Hospitalier Régional Universitaire (CHRU) Nancy, Cell Therapy and Tissue Bank Unit, MTInov Bioproduction and Biotherapy Integrator, Nancy, France
| |
Collapse
|
7
|
Cioni M, Muscianisi S, De Cicco M, Basso S, Hirsch HH, Fontana I, Catenacci L, Bagnarino J, Siciliano M, Montana Lampo O, Acquafredda G, Boti LTD, Rotella J, Bozza E, Zumelli J, Mebelli K, Baldanti F, Cardillo M, Zecca M, Nocera A, Luppi M, Verrina E, Ginevri F, Comoli P. Control of BKPyV-DNAemia by a Tailored Viro-Immunologic Approach Does Not Lead to BKPyV-Nephropathy Progression and Development of Donor-Specific Antibodies in Pediatric Kidney Transplantation. Microorganisms 2024; 13:48. [PMID: 39858816 PMCID: PMC11767705 DOI: 10.3390/microorganisms13010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Polyomavirus BK (BKPyV)-associated nephropathy (BKPyV-nephropathy) remains a significant cause of premature kidney allograft failure. In the absence of effective antiviral treatments, current therapeutic approaches rely on immunosuppression (IS) reduction, possibly at the risk of inducing alloimmunity. Therefore, we sought to explore the long-term effects of a tailored viro-immunologic surveillance and treatment program for BKPyV on the development of alloimmunity and kidney graft outcome. Forty-five pediatric kidney transplant recipients were longitudinally monitored for BKPyV replication, virus-specific immunity, and donor-specific HLA antibodies (DSAs). DNAemia developed in 15 patients who were treated with stepwise IS reduction. Among the other 30 patients, 17 developed DNAuria without DNAemia and 13 always resulted as BKPyV-negative. All patients with DNAemia cleared BKPyV after having mounted a virus-specific cellular immune response, and no biopsy-proven BKPyV-nephropathy was observed. The presence of cytotoxic populations directed to the BKPyV Large-T (LT) antigen early after transplantation protected kidney recipients from developing BKPyV replication, and the appearance of LT-specific T cells in viruric patients prevented the development of BKPyV-DNAemia. In our cohort, no significant correlation was observed between BKPyV-DNAemia and the development of DSA and antibody-mediated rejection. However, patients who experienced and cleared BKPyV-DNAemia had a worse allograft survival at a median follow-up of 18.9 years (p = 0.048). These data need to be confirmed in larger cohorts.
Collapse
Affiliation(s)
- Michela Cioni
- Fondazione Malattie Renali del Bambino, IRCCS G. Gaslini Institute, 16147 Genova, Italy; (M.C.); (A.N.); (E.V.); (F.G.)
- Transfusion Service, IRCCS G. Gaslini Institute, 16147 Genova, Italy
| | - Stella Muscianisi
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Marica De Cicco
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Sabrina Basso
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Hans H. Hirsch
- Transplantation and Clinical Virology, Department of Biomedicine, University of Basel, 4009 Basel, Switzerland;
| | - Iris Fontana
- Vascular and Endovascular Department, Kidney Transplant Surgery Unit, Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Laura Catenacci
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Jessica Bagnarino
- Microbiology and Virology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (J.B.); (F.B.)
| | - Mariangela Siciliano
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Oriana Montana Lampo
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Gloria Acquafredda
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Lou Tina Diana Boti
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Jessica Rotella
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Eleonora Bozza
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
| | - Jennifer Zumelli
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Kristiana Mebelli
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
| | - Fausto Baldanti
- Microbiology and Virology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (J.B.); (F.B.)
| | - Massimo Cardillo
- Transplantation Immunology, Fondazione Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milano, Italy;
| | - Marco Zecca
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Arcangelo Nocera
- Fondazione Malattie Renali del Bambino, IRCCS G. Gaslini Institute, 16147 Genova, Italy; (M.C.); (A.N.); (E.V.); (F.G.)
- Nephrology, Dialysis and Transplantation Unit, IRCCS G. Gaslini Institute, 16147 Genova, Italy
| | - Mario Luppi
- Section of Hematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, AOU Modena, 41124 Modena, Italy;
| | - Enrico Verrina
- Fondazione Malattie Renali del Bambino, IRCCS G. Gaslini Institute, 16147 Genova, Italy; (M.C.); (A.N.); (E.V.); (F.G.)
- Nephrology, Dialysis and Transplantation Unit, IRCCS G. Gaslini Institute, 16147 Genova, Italy
| | - Fabrizio Ginevri
- Fondazione Malattie Renali del Bambino, IRCCS G. Gaslini Institute, 16147 Genova, Italy; (M.C.); (A.N.); (E.V.); (F.G.)
- Nephrology, Dialysis and Transplantation Unit, IRCCS G. Gaslini Institute, 16147 Genova, Italy
| | - Patrizia Comoli
- Cell Factory, Department of Mother and Child Health, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (S.M.); (M.D.C.); (S.B.); (L.C.); (M.S.); (O.M.L.); (G.A.); (L.T.D.B.); (J.R.); (E.B.); (J.Z.); (K.M.)
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| |
Collapse
|
8
|
Toner K, McCann CD, Bollard CM. Applications of cell therapy in the treatment of virus-associated cancers. Nat Rev Clin Oncol 2024; 21:709-724. [PMID: 39160243 DOI: 10.1038/s41571-024-00930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 08/21/2024]
Abstract
A diverse range of viruses have well-established roles as the primary driver of oncogenesis in various haematological malignancies and solid tumours. Indeed, estimates suggest that approximately 1.5 million patients annually are diagnosed with virus-related cancers. The predominant human oncoviruses include Epstein-Barr virus (EBV), Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis B and C viruses (HBV and HCV), human papillomavirus (HPV), human T-lymphotropic virus type 1 (HTLV1), and Merkel cell polyomavirus (MCPyV). In addition, although not inherently oncogenic, human immunodeficiency virus (HIV) is associated with immunosuppression that contributes to the development of AIDS-defining cancers (specifically, Kaposi sarcoma, aggressive B cell non-Hodgkin lymphoma and cervical cancer). Given that an adaptive T cell-mediated immune response is crucial for the control of viral infections, increasing research is being focused on evaluating virus-specific T cell therapies for the treatment of virus-associated cancers. In this Review, we briefly outline the roles of viruses in the pathogenesis of these malignancies before describing progress to date in the field of virus-specific T cell therapy and evaluating the potential utility of these therapies to treat or possibly even prevent virus-related malignancies.
Collapse
Affiliation(s)
- Keri Toner
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chase D McCann
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
9
|
Malpica L, Marques-Piubelli ML, Beltran BE, Chavez JC, Miranda RN, Castillo JJ. EBV-positive diffuse large B-cell lymphoma, not otherwise specified: 2024 update on the diagnosis, risk-stratification, and management. Am J Hematol 2024; 99:2002-2015. [PMID: 38957951 DOI: 10.1002/ajh.27430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
DISEASE OVERVIEW Epstein Barr virus-positive (EBV+) diffuse large B-cell lymphoma (DLBCL), not otherwise specified (NOS) is an aggressive B-cell lymphoma associated with EBV infection included in the WHO classification of lymphoid neoplasms since 2016. Although historically associated to poor prognosis, outcomes seem to have improved in the era of chemoimmunotherapy. DIAGNOSIS The diagnosis is established through meticulous pathological evaluation. Detection of EBV-encoded RNA (EBER) is the standard diagnostic method. The ICC 2022 specifies EBV+ DLBCL, NOS as occurring when >80% of malignant cells express EBER, whereas the WHO-HAEM5 emphasizes that the majority of tumor cells should be EBER positive without setting a defined threshold. The differential diagnosis includes plasmablastic lymphoma, DLBCL associated with chronic inflammation, primary effusion lymphoma, among others. RISK-STRATIFICATION The International Prognostic Index (IPI) and the Oyama score can be used for risk-stratification. The Oyama score includes age >70 years and presence of B symptoms. The expression of CD30 and PD-1/PD-L1 are emerging as potential adverse but targetable biomarkers. MANAGEMENT Patients with EBV+ DLBCL, NOS, should be staged and managed following similar guidelines than patients with EBV-negative DLBCL. EBV+ DLBCL, NOS, however, might have a worse prognosis than EBV-negative DLBCL in the era of chemoimmunotherapy. Therefore, inclusion of patients in clinical trials when available is recommended. There is an opportunity to study and develop targeted therapy in the management of patients with EBV+ DLBCL, NOS.
Collapse
Affiliation(s)
- Luis Malpica
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mario L Marques-Piubelli
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brady E Beltran
- Department of Oncology and Radiotherapy, Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru, Instituto de Ciencias Biomédicas, Universidad Ricardo Palma, Lima, Peru
| | - Julio C Chavez
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jorge J Castillo
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Schweitzer L, Muranski P. Virus-specific T cell therapy to treat refractory viral infections in solid organ transplant recipients. Am J Transplant 2024; 24:1558-1566. [PMID: 38857784 DOI: 10.1016/j.ajt.2024.05.019] [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: 02/09/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Solid organ transplant recipients require ongoing immunosuppression to prevent acute rejection, which puts them at risk of opportunistic infections. Viral infections are particularly challenging to prevent and treat as many establish latency and thus cannot be eliminated, whereas targets for small molecule antiviral medications are limited. Resistance to antivirals and unacceptable toxicity also complicate treatment. Virus-specific T cell therapies aim to restore host-specific immunity to opportunistic viruses that is lacking due to ongoing immunosuppressive therapy. This minireview will provide a state-of-the-art update of the current virus-specific T cell pipeline and translational research that is likely to lead to further treatment options for viral infections in solid organ transplant recipients.
Collapse
Affiliation(s)
- Lorne Schweitzer
- Department of Medicine, Division of Infectious Diseases, Columbia University Irving Medical Center, New York, New York, USA; Columbia Center for Translational Immunology, New York, New York, USA
| | - Pawel Muranski
- Department of Medicine, Division of Hematology, Columbia University Irving Medical Center, New York, New York, USA; Columbia Center for Translational Immunology, New York, New York, USA.
| |
Collapse
|
11
|
Khoury R, Grimley MS, Nelson AS, Leemhuis T, Cancelas JA, Cook E, Wang Y, Heyenbruch D, Bollard CM, Keller MD, Hanley PJ, Lutzko C, Pham G, Davies SM, Rubinstein JD. Third-party virus-specific T cells for the treatment of double-stranded DNA viral reactivation and posttransplant lymphoproliferative disease after solid organ transplant. Am J Transplant 2024; 24:1634-1643. [PMID: 38643944 DOI: 10.1016/j.ajt.2024.04.009] [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: 12/08/2023] [Revised: 03/29/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024]
Abstract
Reactivation or primary infection with double-stranded DNA viruses is common in recipients of solid organ transplants (SOTs) and is associated with significant morbidity and mortality. Treatment with conventional antiviral medications is limited by toxicities, resistance, and a lack of effective options for adenovirus (ADV) and BK polyomavirus (BKPyV). Virus-specific T cells (VSTs) have been shown to be an effective treatment for infections with ADV, BKPyV, cytomegalovirus (CMV), and Epstein-Barr virus (EBV). Most of these studies have been conducted in stem cell recipients, and no large studies have been published in the SOT population to date. In this study, we report on the outcome of quadrivalent third-party VST infusions in 98 recipients of SOTs in the context of an open-label phase 2 trial. The 98 patients received a total of 181 infusions, with a median of 2 infusions per patient. The overall response rate was 45% for BKPyV, 65% for cytomegalovirus, 68% for ADV, and 61% for Epstein-Barr virus. Twenty percent of patients with posttransplant lymphoproliferative disorder had a complete response and 40% of patients had a partial response. All the VST infusions were well tolerated. We conclude that VSTs are safe and effective in the treatment of viral infections in SOT recipients.
Collapse
Affiliation(s)
- Ruby Khoury
- Division of Bone Marrow Transplant and Immune Deficiencies, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA.
| | - Michael S Grimley
- Division of Bone Marrow Transplant and Immune Deficiencies, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Adam S Nelson
- Division of Bone Marrow Transplant and Immune Deficiencies, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Tom Leemhuis
- Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jose A Cancelas
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA; Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio, USA; Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Eleanor Cook
- Division of Bone Marrow Transplant and Immune Deficiencies, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - YunZu Wang
- Division of Bone Marrow Transplant and Immune Deficiencies, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Daria Heyenbruch
- Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Catherine M Bollard
- Department of Pediatrics, Center for Cancer and Immunology Research, Children's National Hospital, the George Washington University, Washington, District of Columbia, USA
| | - Michael D Keller
- Department of Pediatrics, Center for Cancer and Immunology Research, Children's National Hospital, the George Washington University, Washington, District of Columbia, USA
| | - Patrick J Hanley
- Department of Pediatrics, Center for Cancer and Immunology Research, Children's National Hospital, the George Washington University, Washington, District of Columbia, USA
| | - Carolyn Lutzko
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA; Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Giang Pham
- Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stella M Davies
- Division of Bone Marrow Transplant and Immune Deficiencies, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA; Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
12
|
Cheng J, Wistinghausen B. Clinicopathologic Spectrum of Pediatric Posttransplant Lymphoproliferative Diseases Following Solid Organ Transplant. Arch Pathol Lab Med 2024; 148:1052-1062. [PMID: 38051286 DOI: 10.5858/arpa.2023-0323-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2023] [Indexed: 12/07/2023]
Abstract
CONTEXT.— Posttransplant lymphoproliferative disorder (PTLD) remains a significant complication in pediatric patients undergoing solid organ transplant (SOT). The majority involve Epstein-Barr virus (EBV)-driven CD20+ B-cell proliferations, which respond to reduction of immunosuppression and anti-CD20-directed immunotherapy. Owing to the low overall incidence, prospective studies of pediatric PTLD are scarce, leading to a lack of comprehensive understanding of this disorder in pediatric populations. This review aims to bridge this knowledge gap by providing a comprehensive analysis of the clinical, morphologic, and molecular genetic features of PTLD in children, adolescents, and young adults after SOT. OBJECTIVE.— To examine the clinical features, pathogenesis, and classification of pediatric PTLDs after SOT. DATA SOURCES.— Personal experiences and published works in PubMed. CONCLUSIONS.— PTLD includes a broad and heterogeneous spectrum of disorders, ranging from nonmalignant lymphoproliferations to lymphomas. While most pediatric PTLDs are EBV+, an increasing number of EBV- PTLDs have been recognized. The pathologic classification of PTLDs has evolved in recent decades, reflecting advancements in understanding the underlying pathobiology. Nevertheless, there remains a great need for further research to elucidate the biology, identify patients at higher risk for aggressive disease, and establish optimal treatment strategies for relapsed/refractory disease.
Collapse
Affiliation(s)
- Jinjun Cheng
- From the Department of Pathology and Laboratory Medicine (Cheng), Center for Cancer and Blood Disorders (Wistinghausen), and Center for Cancer and Immunology Research (Cheng, Wistinghausen), Children's National Hospital, Washington, District of Columbia
| | - Birte Wistinghausen
- From the Department of Pathology and Laboratory Medicine (Cheng), Center for Cancer and Blood Disorders (Wistinghausen), and Center for Cancer and Immunology Research (Cheng, Wistinghausen), Children's National Hospital, Washington, District of Columbia
| |
Collapse
|
13
|
Juan X, Fan Z, Cao X, Ding YY, Liu H, Shang QN, Zhao X, Chang Y, Wang Y, Xu L, Zhang X, Huang X, Zhao X. CD56 bright NK cell expansion correlated with EBV reactivation control post allogeneic hematopoietic stem cell transplantation. Ann Hematol 2024; 103:3723-3735. [PMID: 38862793 DOI: 10.1007/s00277-024-05827-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Natural killer (NK) cells are equipped with anti-Epstein-Barr virus (EBV) function, however, whether EBV infection will affect NK cells reconstitution after allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains unclear. To identify the characteristics of NK cells, we prospectively enrolled 11 patients who occurred EBV reactivation post allo-HSCT and 11 patients without EBV infection as control. We found that that EBV infection induced the expansion of CD56bright and NKG2A+KIR- NK subsets,and decreased the cytotoxicity function of NK cells. The frequency of NKG2A+KIR- NK cells were higher in patients who progressed into post-transplant lymphoproliferative disorder (PTLD) than EBV viremia patients, which also correlated with decreased proliferation and cytotoxic function. By screening the activation receptors of NK cells, we found the DNAM-1+CD56bright NK cells is significantly increased after EBV stimulation, further we demonstrated that DNAM-1 is essential for EBV induced NK cells activation as the cytokine release against EBV-transformed lymphoblastoid cell lines(EBV-LCLs) of CD56bright NK cells were significantly decreased after DNAM-1 blockade. NK cells infusion suppressed the progression of EBV-related tumor mice model. A prospective cohort indicated that old donor age was an independent risk factor for EBV infection. Rapid CD56bri expansion and high expression of DNAM-1 on CD56bri NK cells in response to EBV reactivation correlated with rapid EBV clearance post allo-HSCT in patients with younger donors. In summary, our data showed that high expression of DNAM-1 receptors on NK cell may participate protective CD56bri NK cells response to EBV infection after allo-HSCT.
Collapse
Affiliation(s)
- Xie Juan
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Zeying Fan
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xunhong Cao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yi-Yang Ding
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Huixin Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Qian-Nan Shang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaosu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lanping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiangyu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.
- Collaborative Innovation Center of Hematology, Beijing, China.
| |
Collapse
|
14
|
Palianina D, Mietz J, Stühler C, Arnold B, Bantug G, Münz C, Chijioke O, Khanna N. Stem cell memory EBV-specific T cells control EBV tumor growth and persist in vivo. SCIENCE ADVANCES 2024; 10:eado2048. [PMID: 39178248 PMCID: PMC11343021 DOI: 10.1126/sciadv.ado2048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/19/2024] [Indexed: 08/25/2024]
Abstract
Adoptive T cell therapy (ACT), the therapeutic transfer of defined T cell immunity to patients, offers great potential in the fight against different human diseases including difficult-to-treat viral infections, but persistence and longevity of the cells are areas of concern. Very-early-differentiated stem cell memory T cells (TSCMs) have superior self-renewal, engraftment, persistence, and anticancer efficacy, but their potential for antiviral ACT remains unknown. Here, we developed a clinically scalable protocol for expanding Epstein-Barr virus (EBV)-specific TSCM-enriched T cells with high proportions of CD4+ T cells and broad EBV antigen coverage. These cells showed tumor control in a xenograft model of EBV-induced lymphoma and were superior to previous ACT protocols in terms of tumor infiltration, in vivo proliferation, persistence, proportion of functional CD4+ T cells, and diversity of EBV antigen specificity. Thus, our protocol may pave the way for the next generation of potent unmodified antigen-specific cell therapies for EBV-associated diseases, including tumors, and other indications.
Collapse
Affiliation(s)
- Darya Palianina
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Juliane Mietz
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Claudia Stühler
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Brice Arnold
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Glenn Bantug
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Obinna Chijioke
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Nina Khanna
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
15
|
Law N, Logan C, Taplitz R. EBV Reactivation and Disease in Allogeneic Hematopoietic Stem Cell Transplant (HSCT) Recipients and Its Impact on HSCT Outcomes. Viruses 2024; 16:1294. [PMID: 39205268 PMCID: PMC11359191 DOI: 10.3390/v16081294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
The acquisition or reactivation of Epstein-Barr virus (EBV) after allogeneic Hematopoietic Stem Cell Transplant (HSCT) can be associated with complications including the development of post-transplant lymphoproliferative disorder (PTLD), which is associated with significant morbidity and mortality. A number of risk factors for PTLD have been defined, including T-cell depletion, and approaches to monitoring EBV, especially in high-risk patients, with the use of preemptive therapy upon viral activation have been described. Newer therapies for the preemption or treatment of PTLD, such as EBV-specific cytotoxic T-cells, hold promise. Further studies to help define risks, diagnosis, and treatment of EBV-related complications are needed in this at-risk population.
Collapse
Affiliation(s)
- Nancy Law
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Cathy Logan
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, La Jolla, CA 92093, USA
| | - Randy Taplitz
- Division of Infectious Diseases, Department of Medicine, City of Hope National Medical Center, Duarte, CA 91010, USA;
| |
Collapse
|
16
|
Grosso D, Wagner JL, O’Connor A, Keck K, Huang Y, Wang ZX, Mehler H, Leiby B, Flomenberg P, Gergis U, Nikbakht N, Morris M, Karp J, Peedin A, Flomenberg N. Safety and feasibility of third-party cytotoxic T lymphocytes for high-risk patients with COVID-19. Blood Adv 2024; 8:4113-4124. [PMID: 38885482 PMCID: PMC11345373 DOI: 10.1182/bloodadvances.2024013344] [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: 04/08/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
ABSTRACT Cytotoxic T lymphocytes (CTLs) destroy virally infected cells and are critical for the elimination of viral infections such as those caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Delayed and dysfunctional adaptive immune responses to SARS-CoV-2 are associated with poor outcomes. Treatment with allogeneic SARS-CoV-2-specific CTLs may enhance cellular immunity in high-risk patients providing a safe, direct mechanism of treatment. Thirty high-risk ambulatory patients with COVID-19 were enrolled in a phase 1 trial assessing the safety of third party, SARS-CoV-2-specific CTLs. Twelve interventional patients, 6 of whom were immunocompromised, matched the HLA-A∗02:01 restriction of the CTLs and received a single infusion of 1 of 4 escalating doses of a product containing 68.5% SARS-CoV-2-specific CD8+ CTLs/total cells. Symptom improvement and resolution in these patients was compared with an observational group of 18 patients lacking HLA-A∗02:01 who could receive standard of care. No dose-limiting toxicities were observed at any dosing level. Nasal swab polymerase chain reaction testing showed ≥88% and >99% viral elimination from baseline in all patients at 4 and 14 days after infusion, respectively. The CTLs did not interfere with the development of endogenous anti-SARS-CoV-2 humoral or cellular responses. T-cell receptor β analysis showed persistence of donor-derived SARS-CoV-2-specific CTLs through the end of the 6-month follow-up period. Interventional patients consistently reported symptomatic improvement 2 to 3 days after infusion, whereas improvement was more variable in observational patients. SARS-CoV-2-specific CTLs are a potentially feasible cellular therapy for COVID-19 illness. This trial was registered at www.clinicaltrials.gov as #NCT04765449.
Collapse
Affiliation(s)
- Dolores Grosso
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - John L. Wagner
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Allyson O’Connor
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Kaitlyn Keck
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Yanping Huang
- Department of Pathology and Genomic Medicine, Histocompatibility and Immunogenetics Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Zi-Xuan Wang
- Departments of Surgery and Pathology, Molecular and Genomic Pathology Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Hilary Mehler
- Department of Pathology and Genomic Medicine, Histocompatibility and Immunogenetics Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Benjamin Leiby
- Division of Biostatistics, Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Phyllis Flomenberg
- Division of Infectious Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Usama Gergis
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Neda Nikbakht
- Department of Dermatology and Cutaneous Biology, Cutaneous Lymphoma Clinic, Thomas Jefferson University, Philadelphia, PA
| | - Michael Morris
- Department of Emergency Medicine, Thomas Jefferson University Washington Township Hospital, Sewell, NJ
| | - Julie Karp
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Alexis Peedin
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Neal Flomenberg
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
17
|
Allen UD, L'Huillier AG, Bollard CM, Gross TG, Hayashi RJ, Höcker B, Maecker-Kolhoff B, Marks SD, Mazariegos GV, Smets F, Trappe RU, Visner G, Chinnock RE, Comoli P, Danziger-Isakov L, Dulek DE, Dipchand AI, Ferry JA, Martinez OM, Metes DM, Michaels MG, Preiksaitis J, Squires JE, Swerdlow SH, Wilkinson JD, Dharnidharka VR, Green M, Webber SA, Esquivel CO. The IPTA Nashville consensus conference on post-transplant lymphoproliferative disorders after solid organ transplantation in children: IV-consensus guidelines for the management of post-transplant lymphoproliferative disorders in children and adolescents. Pediatr Transplant 2024; 28:e14781. [PMID: 38808744 DOI: 10.1111/petr.14781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
The International Pediatric Transplant Association convened an expert consensus conference to assess current evidence and develop recommendations for various aspects of care relating to post-transplant lymphoproliferative disorders (PTLD) after pediatric solid organ transplantation. This report addresses the outcomes of deliberations by the PTLD Management Working Group. A strong recommendation was made for reduction in immunosuppression as the first step in management. Similarly, strong recommendations were made for the use of the anti-CD20 monoclonal antibody (rituximab) as was the case for chemotherapy in selected scenarios. In some scenarios, there is uncoupling of the strength of the recommendations from the available evidence in situations where such evidence is lacking but collective clinical experiences drive decision-making. Of note, there are no large, randomized phase III trials of any treatment for PTLD in the pediatric age group. Current gaps and future research priorities are highlighted.
Collapse
Affiliation(s)
- Upton D Allen
- Division of Infectious Diseases, Department of Paediatrics, Transplant and Regenerative Medicine Center, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Arnaud G L'Huillier
- Pediatric Infectious Diseases Unit and Laboratory of Virology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, The George Washington University, Washington, District of Columbia, USA
| | - Thomas G Gross
- Center for Cancer and Blood Diseases, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Robert J Hayashi
- Division of Pediatric Hematology/Oncology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Britta Höcker
- Department of Pediatrics I, Medical Faculty, University Children's Hospital, Heidelberg University, Heidelberg, Germany
| | | | - Stephen D Marks
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - George Vincent Mazariegos
- Hillman Center for Pediatric Transplantation, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francoise Smets
- Pediatric Gastroenterology and Hepatology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Ralf U Trappe
- Department of Hematology and Oncology, DIAKO Ev. Diakonie-Krankenhaus Bremen, Bremen, Germany
- Department of Internal Medicine II: Hematology and Oncology, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - Gary Visner
- Division of Pulmonary Medicine, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | | | - Patrizia Comoli
- Cell Factory & Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lara Danziger-Isakov
- Division of Infectious Disease, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Daniel E Dulek
- Division of Pediatric Infectious Diseases, Monroe Carell Junior Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anne I Dipchand
- Department of Paediatrics, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Judith A Ferry
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Olivia M Martinez
- Department of Surgery and Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Diana M Metes
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marian G Michaels
- Division of Pediatric Infectious Diseases, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jutta Preiksaitis
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - James E Squires
- Division of Gastroenterology, Hepatology and Nutrition, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Steven H Swerdlow
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James D Wilkinson
- Department of Pediatrics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - Vikas R Dharnidharka
- Division of Pediatric Nephrology, Hypertension & Apheresis, Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Michael Green
- Division of Pediatric Infectious Diseases, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Steven A Webber
- Department of Pediatrics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | | |
Collapse
|
18
|
Obermaier B, Braun C, Hensen L, Ahmad O, Faul C, Lang P, Bethge W, Lengerke C, Vogel W. Adenovirus- and cytomegalovirus-specific adoptive T-cell therapy in the context of hematologic cell transplant or HIV infection - A single-center experience. Transpl Infect Dis 2024; 26:e14296. [PMID: 38830809 DOI: 10.1111/tid.14296] [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: 02/14/2024] [Revised: 04/17/2024] [Accepted: 05/05/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Reactivation of viral infections, in particular cytomegalovirus (CMV) and adenovirus (ADV), cause morbidity and non-relapse-mortality in states of immune deficiency, especially after allogeneic hematopoietic cell transplantation (allo-HCT). Against the background of few available pharmacologic antiviral agents, limited by toxicities and resistance, adoptive transfer of virus-specific T-cells (VST) is a promising therapeutic approach. METHODS We conducted a single-center retrospective analysis of adult patients treated with ADV- or CMV-specific T-cells in 2012-2022. Information was retrieved by review of electronic health records. Primary outcome was a response to VST by decreasing viral load or clinical improvement. Secondary outcomes included overall survival and safety of VST infusion, in particular association with graft-versus-host disease (GVHD). RESULTS Ten patients were included, of whom four were treated for ADV, five for CMV, and one for ADV-CMV-coinfection. Cells were derived from stem cell donors (6/10) or third-party donors (4/10). Response criteria were met by six of 10 patients (4/4 ADV, 2/5 CMV, and 0/1 ADV-CMV). Overall survival was 40%. No infusion related adverse events were documented. Aggravation of GVHD after adoptive immunotherapy was observed in two cases, however in temporal association with a conventional donor lymphocyte infusion and a stem cell boost, respectively. CONCLUSION In this cohort, CMV- and ADV-specific T-cell therapy appear to be safe and effective. We describe the first reported case of virus-specific T-cell therapy for CMV reactivation not associated with transplantation but with advanced HIV infection. This encourages further evaluation of adoptive immunotherapy beyond the context of allo-HCT.
Collapse
Affiliation(s)
- Benedikt Obermaier
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Christiane Braun
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Luca Hensen
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Osama Ahmad
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Christoph Faul
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Peter Lang
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Wolfgang Bethge
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Claudia Lengerke
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Wichard Vogel
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
19
|
Pociupany M, Snoeck R, Dierickx D, Andrei G. Treatment of Epstein-Barr Virus infection in immunocompromised patients. Biochem Pharmacol 2024; 225:116270. [PMID: 38734316 DOI: 10.1016/j.bcp.2024.116270] [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: 03/05/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Epstein-Barr Virus (EBV), is a ubiquitous γ-Herpesvirus that infects over 95% of the human population and can establish a life-long infection without causing any clinical symptoms in healthy individuals by residing in memory B-cells. Primary infection occurs in childhood and is mostly asymptomatic, however in some young adults it can result in infectious mononucleosis (IM). In immunocompromised individuals however, EBV infection has been associated with many different malignancies. Since EBV can infect both epithelial and B-cells and very rarely NK cells and T-cells, it is associated with both epithelial cancers like nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC), with lymphomas including Burkitt Lymphoma (BL) or Post-transplant Lymphoproliferative Disorder (PTLD) and rarely with NK/T-cell lymphomas. Currently there are no approved antivirals active in PTLD nor in any other malignancy. Moreover, lytic phase disease almost never requires antiviral treatment. Although many novel therapies against EBV have been described, the management and/or prevention of EBV primary infections or reactivations remains difficult. In this review, we discuss EBV infection, therapies targeting EBV in both lytic and latent state with novel therapeutics developed that show anti-EBV activity as well as EBV-associated malignancies both, epithelial and lymphoproliferative malignancies and emerging therapies targeting the EBV-infected cells.
Collapse
Affiliation(s)
- Martyna Pociupany
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Robert Snoeck
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Daan Dierickx
- Laboratory of Experimental Hematology, Department of Oncology, KU Leuven, Leuven, Belgium; Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Graciela Andrei
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
Nikiforow S, Whangbo JS, Reshef R, Tsai DE, Bunin N, Abu-Arja R, Mahadeo KM, Weng WK, Van Besien K, Loeb D, Nasta SD, Nemecek ER, Zhao W, Sun Y, Galderisi F, Wahlstrom J, Mehta A, Gamelin L, Dinavahi R, Prockop S. Tabelecleucel for EBV+ PTLD after allogeneic HCT or SOT in a multicenter expanded access protocol. Blood Adv 2024; 8:3001-3012. [PMID: 38625984 PMCID: PMC11215195 DOI: 10.1182/bloodadvances.2023011626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/18/2024] Open
Abstract
ABSTRACT Patients with Epstein-Barr virus (EBV)-positive posttransplant lymphoproliferative disease (EBV+ PTLD) in whom initial treatment fails have few options and historically low median overall survival (OS) of 0.7 months after allogeneic hematopoietic cell transplant (HCT) and 4.1 months after solid organ transplant (SOT). Tabelecleucel is an off-the-shelf, allogeneic EBV-specific cytotoxic T-lymphocyte immunotherapy for EBV+ PTLD. Previous single-center experience showed responses in patients with EBV+ PTLD after HCT or SOT. We now report outcomes from a multicenter expanded access protocol in HCT (n = 14) and SOT (n = 12) recipients treated with tabelecleucel for EBV+ PTLD that was relapsed/refractory (R/R) to rituximab with/without chemotherapy. The investigator-assessed objective response rate was 65.4% overall (including 38.5% with a complete and 26.9% with a partial response), 50.0% in HCT, and 83.3% in SOT. The estimated 1- and 2-year OS rates were both 70.0% (95% confidence interval [CI], 46.5-84.7) overall, both 61.5% (95% CI, 30.8-81.8) in HCT, and both 81.5% (95% CI, 43.5-95.1) in SOT (median follow-up: 8.2, 2.8, and 22.5 months, respectively). Patients responding to tabelecleucel had higher 1- and 2-year OS rates (94.1%) than nonresponders (0%). Treatment was well tolerated, with no reports of tumor flare, cytokine release syndrome, or rejection of marrow and SOT. Results demonstrate clinically meaningful outcomes across a broad population treated with tabelecleucel, indicating a potentially transformative and accessible treatment advance for R/R EBV+ PTLD after HCT or SOT. This trial was registered at www.ClinicalTrials.gov as #NCT02822495.
Collapse
Affiliation(s)
- Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jennifer S. Whangbo
- VOR Bio, Cambridge, MA
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Ran Reshef
- Blood and Marrow Transplantation and Cell Therapy Program, Columbia University Irving Medical Center, New York, NY
| | - Donald E. Tsai
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Nancy Bunin
- Division of Pediatric Hematology/Oncology and Blood and Marrow Transplant, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Rolla Abu-Arja
- Division of Pediatric Hematology/Oncology and Blood and Marrow Transplant, Nationwide Children’s Hospital, Columbus, OH
| | - Kris Michael Mahadeo
- Division of Pediatric Transplant and Cellular Therapy, Duke University Medical Center, Durham, NC
| | - Wen-Kai Weng
- BMT-Cellular Therapy, Department of Medicine, Stanford University, School of Medicine, Stanford, CA
| | - Koen Van Besien
- Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, OH
| | - David Loeb
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Sunita Dwivedy Nasta
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Eneida R. Nemecek
- Pediatric Transplant & Cellular Therapy, Oregon Health and Science University, Portland, OR
| | | | - Yan Sun
- Atara Biotherapeutics, Thousand Oaks, CA
| | | | | | | | | | | | - Susan Prockop
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- VOR Bio, Cambridge, MA
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
- Dana-Farber Cancer Institute/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA
| |
Collapse
|
21
|
Quach DH, Ganesh HR, Briones YD, Nouraee N, Ma A, Hadidi YF, Sharma S, Rooney CM. Rejection resistant CD30.CAR-modified Epstein-Barr virus-specific T cells as an off-the-shelf platform for CD30 + lymphoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200814. [PMID: 38966037 PMCID: PMC11223124 DOI: 10.1016/j.omton.2024.200814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/14/2024] [Accepted: 05/10/2024] [Indexed: 07/06/2024]
Abstract
Off-the-shelf (OTS) adoptive T cell therapies have many benefits such as immediate availability, improved access and reduced cost, but face the major challenges of graft-vs-host disease (GVHD) and graft rejection, mediated by alloreactive T cells present in the graft and host, respectively. We have developed a platform for OTS T cell therapies by using Epstein-Bar virus (EBV)-specific T cells (EBVSTs) expressing a chimeric antigen receptor (CAR) targeting CD30. Allogeneic EBVSTs have not caused GVHD in several clinical trials, while the CD30.CAR, that is effective for the treatment of lymphoma, can also target alloreactive T cells that upregulate CD30 on activation. Although EBVSTs express high levels of CD30, they were protected from fratricide in cis, by the CD30.CAR. Hence, they could proliferate extensively and maintained function both through their native EBV-specific T cell receptor and the CD30.CAR. The CD30.CAR enabled EBVSTs to persist in co-cultures with naive and primed alloreactive T cells and eliminate activated natural killer cells that can also be alloreactive. In conclusion, we show that CD30.CAR EBVSTs have the potential to be an effective OTS therapy against CD30+ tumors and, if successful, could then be used as a platform to target other tumor antigens.
Collapse
Affiliation(s)
- David H. Quach
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haran R. Ganesh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Yolanda D. Briones
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Nazila Nouraee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Audrey Ma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Yezan F. Hadidi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Sandhya Sharma
- Graduate program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cliona M. Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston TX 77030, USA
- Department of Molecular Virology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
22
|
Yeo SP, Kua L, Tan JW, Lim JK, Wong FHS, Santos MD, Poh CM, Goh AXH, Koh XY, Zhou X, Rajarethinam R, Chen Q, Her Z, Horak ID, Low L, Tan KW. B7-H3-Targeting Chimeric Antigen Receptors Epstein-Barr Virus-specific T Cells Provides a Tumor Agnostic Off-The-Shelf Therapy Against B7-H3-positive Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2024; 4:1410-1429. [PMID: 38717140 PMCID: PMC11149603 DOI: 10.1158/2767-9764.crc-23-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/14/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024]
Abstract
Encouraged by the observations of significant B7-H3 protein overexpression in many human solid tumors compared to healthy tissues, we directed our focus towards targeting B7-H3 using chimeric antigen receptor (CAR) T cells. We utilized a nanobody as the B7-H3-targeting domain in our CAR construct to circumvent the stability issues associated with single-chain variable fragment-based domains. In efforts to expand patient access to CAR T-cell therapy, we engineered our nanobody-based CAR into human Epstein-Barr virus-specific T cells (EBVST), offering a readily available off-the-shelf treatment. B7H3.CAR-armored EBVSTs demonstrated potent in vitro and in vivo activities against multiple B7-H3-positive human tumor cell lines and patient-derived xenograft models. Murine T cells expressing a murine equivalent of our B7H3.CAR exhibited no life-threatening toxicities in immunocompetent mice bearing syngeneic tumors. Further in vitro evaluation revealed that while human T, B, and natural killer cells were unaffected by B7H3.CAR EBVSTs, monocytes were targeted because of upregulation of B7-H3. Such targeting of myeloid cells, which are key mediators of cytokine release syndrome (CRS), contributed to a low incidence of CRS in humanized mice after B7H3.CAR EBVST treatment. Notably, we showed that B7H3.CAR EBVSTs can target B7-H3-expressing myeloid-derived suppressor cells (MDSC), thereby mitigating MDSC-driven immune suppression. In summary, our data demonstrate that our nanobody-based B7H3.CAR EBVSTs are effective as an off-the-shelf therapy for B7-H3-positive solid tumors. These cells also offer an avenue to modulate the immunosuppressive tumor microenvironment, highlighting their promising clinical potential in targeting solid tumors. SIGNIFICANCE Clinical application of EBVSTs armored with B7-H3-targeting CARs offer an attractive solution to translate off-the-shelf CAR T cells as therapy for solid tumors.
Collapse
Affiliation(s)
| | - Lindsay Kua
- Tessa Therapeutics Ltd, Singapore
- Tikva Allocell Pte Ltd, Singapore
| | - Jin Wei Tan
- Tessa Therapeutics Ltd, Singapore
- Tikva Allocell Pte Ltd, Singapore
| | | | - Fiona HS Wong
- Tessa Therapeutics Ltd, Singapore
- Tikva Allocell Pte Ltd, Singapore
| | | | | | - Angeline XH Goh
- Tessa Therapeutics Ltd, Singapore
- Tikva Allocell Pte Ltd, Singapore
| | | | | | - Ravisankar Rajarethinam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ivan D. Horak
- Tessa Therapeutics Ltd, Singapore
- Tikva Allocell Pte Ltd, Singapore
| | - Lionel Low
- Tessa Therapeutics Ltd, Singapore
- Tikva Allocell Pte Ltd, Singapore
| | - Kar Wai Tan
- Tessa Therapeutics Ltd, Singapore
- Tikva Allocell Pte Ltd, Singapore
| |
Collapse
|
23
|
Rischall A, Olson A. SOHO State of the Art Updates and Next Questions | CTLs for Infections Following Stem Cell Transplantation. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:340-347. [PMID: 38267354 DOI: 10.1016/j.clml.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/26/2024]
Abstract
Allogeneic hematopoietic stem cell transplantation (AHSCT) is an important modality in the treatment of acute leukemia and other hematologic disorders. The post-transplant period is associated with prolonged periods of impaired immune function. Delayed T-cell immune reconstitution is correlated with increased risk of viral, bacterial, and fungal infections. This risk increases with high intensity inductions regimens often required for alternative donor sources. Current therapies for prophylaxis and treatment of these infections are limited by poor efficacy and significant toxicity. Adoptive cell therapy with cytotoxic T lymphocytes (CTL) has proven to be both efficacious and safe in the management of post-transplant viral infections. Recent advances have led to faster production of CTLs and broadened applications for their use. In particular, the generation of third party CTLs has helped ameliorate the problems related to donor availability and product generation time. In this review we aim to describe both the history of CTL use and current advances in the field.
Collapse
Affiliation(s)
- Ariel Rischall
- Department of Medical Oncology, The University of Texas Medical Branch, Galveston, TX
| | - Amanda Olson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
24
|
Ali FEM, Ibrahim IM, Althagafy HS, Hassanein EHM. Role of immunotherapies and stem cell therapy in the management of liver cancer: A comprehensive review. Int Immunopharmacol 2024; 132:112011. [PMID: 38581991 DOI: 10.1016/j.intimp.2024.112011] [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: 02/02/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
Liver cancer (LC) is the sixth most common disease and the third most common cause of cancer-related mortality. The WHO predicts that more than 1 million deaths will occur from LC by 2030. Hepatocellular carcinoma (HCC) is a common form of primary LC. Today, the management of LC involves multiple disciplines, and multimodal therapy is typically selected on an individual basis, considering the intricate interactions between the patient's overall health, the stage of the tumor, and the degree of underlying liver disease. Currently, the treatment of cancers, including LC, has undergone a paradigm shift in the last ten years because of immuno-oncology. To treat HCC, immune therapy approaches have been developed to enhance or cause the body's natural immune response to specifically target tumor cells. In this context, immune checkpoint pathway inhibitors, engineered cytokines, adoptive cell therapy, immune cells modified with chimeric antigen receptors, and therapeutic cancer vaccines have advanced to clinical trials and offered new hope to cancer patients. The outcomes of these treatments are encouraging. Additionally, treatment using stem cells is a new approach for restoring deteriorated tissues because of their strong differentiation potential and capacity to release cytokines that encourage cell division and the formation of blood vessels. Although there is no proof that stem cell therapy works for many types of cancer, preclinical research on stem cells has shown promise in treating HCC. This review provides a recent update regarding the impact of immunotherapy and stem cells in HCC and promising outcomes.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt; Michael Sayegh, Faculty of Pharmacy, Aqaba University of Technology, Aqaba 77110, Jordan.
| | - Islam M Ibrahim
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| |
Collapse
|
25
|
Ergisi M, Ooi B, Salim O, Papalois V. Post-transplant lymphoproliferative disorders following kidney transplantation: A literature review with updates on risk factors, prognostic indices, screening strategies, treatment and analysis of donor type. Transplant Rev (Orlando) 2024; 38:100837. [PMID: 38430887 DOI: 10.1016/j.trre.2024.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Post-transplant lymphoproliferative disorders (PTLD) is a devastating complication of kidney transplantation with an insidious presentation and potential to disseminate aggressively. This review delineates the risk factors, prognostic indexes, screening, current management algorithm and promising treatment strategies for PTLD. Kidneys from both extended criteria donors (ECD) and living donors (LD) are being increasingly used to expand the donor pool. This review also delineates whether PTLD outcomes vary based on these donor sources. While Epstein-Barr virus (EBV) is a well-known risk factor for PTLD development, the use of T-cell depleting induction agents has been increasingly implicated in aggressive, monomorphic forms of PTLD. Research regarding maintenance therapy is sparse. The international prognostic index seems to be the most validate prognostic tool. Screening for PTLD is controversial, as annual PET-CT is most sensitive but costly, while targeted monitoring of EBV-seronegative patients was more economically feasible, is recommended by the American Society of Transplantation, but is limited to a subset of the population. Other screening strategies such as using Immunoglobulin/T-cell receptor require further validation. A risk-stratified approach is taken in the treatment of PTLD. The first step is the reduction of immunosuppressants, after which rituximab and chemotherapy may be introduced if unsuccessful. Some novel treatments have also shown potential benefit in studies: brentuximab vedotin, chimeric antigen receptor T-cell therapy and EBV-specific cytotoxic T lymphocytes. Analysis of LD v DD recipients show no significant difference in incidence and mortality of PTLD but did reveal a shortened time to development of PTLD from transplant. Analysis of SCD vs ECD recipients show a higher incidence of PTLD in the ECD group, which might be attributed to longer time on dialysis for these patients, age, and the pro-inflammatory nature of these organs. However, incidence of PTLD overall is still extremely low. Efforts should be focused on optimising recipients instead. Minimising the use of T-cell depleting therapy while encouraging research on the effect of new immunosuppressants on PTLD, screening for EBV status are essential, while enabling shared decision-making during counselling when choosing kidney donor types and individualised risk tailoring are strongly advocated.
Collapse
Affiliation(s)
- Mehmet Ergisi
- Norfolk and Norwich University Hospital, Norfolk and Norwich University Hospitals NHS Foundation Trust, Department of Medicine, Norwich, United Kingdom.
| | - Bryan Ooi
- Department of Medicine, Imperial College London, London, United Kingdom.
| | - Omar Salim
- Isle of Wight NHS Trust, Parkhurst Road, Newport, United Kingdom
| | - Vassilios Papalois
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, Department of Transplant and General Surgery, London, United Kingdom.
| |
Collapse
|
26
|
Keller MD, Schattgen SA, Chandrakasan S, Allen EK, Jensen-Wachspress MA, Lazarski CA, Qayed M, Lang H, Hanley PJ, Tanna J, Pai SY, Parikh S, Berger SI, Gottschalk S, Pulsipher MA, Thomas PG, Bollard CM. Secondary bone marrow graft loss after third-party virus-specific T cell infusion: Case report of a rare complication. Nat Commun 2024; 15:2749. [PMID: 38553461 PMCID: PMC10980733 DOI: 10.1038/s41467-024-47056-3] [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: 09/19/2022] [Accepted: 12/20/2023] [Indexed: 04/02/2024] Open
Abstract
Virus-specific T cells (VST) from partially-HLA matched donors have been effective for treatment of refractory viral infections in immunocompromised patients in prior studies with a good safety profile, but rare adverse events have been described. Here we describe a unique and severe adverse event of VST therapy in an infant with severe combined immunodeficiency, who receives, as part of a clinical trial (NCT03475212), third party VSTs for treating cytomegalovirus viremia following bone marrow transplantation. At one-month post-VST infusion, rejection of graft and reversal of chimerism is observed, as is an expansion of T cells exclusively from the VST donor. Single-cell gene expression and T cell receptor profiling demonstrate a narrow repertoire of predominantly activated CD4+ T cells in the recipient at the time of rejection, with the repertoire overlapping more with that of peripheral blood from VST donor than the infused VST product. This case thus demonstrates a rare but serious side effect of VST therapy.
Collapse
Affiliation(s)
- Michael D Keller
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Division of Allergy and Immunology, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University, Washington, DC, USA
| | - Stefan A Schattgen
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - E Kaitlynn Allen
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Christopher A Lazarski
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Hospital of Atlanta, Atlanta, GA, USA
| | - Haili Lang
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Jay Tanna
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Sung-Yun Pai
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Suhag Parikh
- Aflac Cancer and Blood Disorders Center, Children's Hospital of Atlanta, Atlanta, GA, USA
| | - Seth I Berger
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Stephen Gottschalk
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael A Pulsipher
- Division of Pediatric Hematology/Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Paul G Thomas
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA.
- GW Cancer Center, George Washington University, Washington, DC, USA.
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA.
| |
Collapse
|
27
|
Wistinghausen B, Toner K, Barkauskas DA, Jerkins LP, Kinoshita H, Chansky P, Pezzella G, Saguilig L, Hayashi RJ, Abhyankar H, Scull B, Karri V, Tanna J, Hanley P, Hermiston ML, Allen CE, Bollard CM. Durable immunity to EBV after rituximab and third-party LMP-specific T cells: a Children's Oncology Group study. Blood Adv 2024; 8:1116-1127. [PMID: 38163318 PMCID: PMC10909726 DOI: 10.1182/bloodadvances.2023010832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/07/2023] [Accepted: 11/12/2023] [Indexed: 01/03/2024] Open
Abstract
ABSTRACT Posttransplant lymphoproliferative disease (PTLD) in pediatric solid organ transplant (SOT) recipients is characterized by uncontrolled proliferation of Epstein-Barr virus-infected (EBV+) B cells due to decreased immune function. This study evaluated the feasibility, safety, clinical and immunobiological outcomes in pediatric SOT recipients with PTLD treated with rituximab and third-party latent membrane protein-specific T cells (LMP-TCs). Newly diagnosed (ND) patients without complete response to rituximab and all patients with relapsed/refractory (R/R) disease received LMP-TCs. Suitable LMP-TC products were available for all eligible subjects. Thirteen of 15 patients who received LMP-TCs were treated within the prescribed 14-day time frame. LMP-TC therapy was generally well tolerated. Notable adverse events included 3 episodes of rejection in cardiac transplant recipients during LMP-TC therapy attributed to subtherapeutic immunosuppression and 1 episode of grade 3 cytokine release syndrome. Clinical outcomes were associated with disease severity. Overall response rate (ORR) after LMP-TC cycle 1 was 70% (7/10) for the ND cohort and 20% (1/5) for the R/R cohort. For all cohorts combined, the best ORR for LMP-TC cycles 1 and 2 was 53% and the 2-year overall survival was 70.7%. vβT-cell receptor sequencing showed persistence of adoptively transferred third-party LMP-TCs for up to 8 months in the ND cohort. This study establishes the feasibility of administering novel T-cell therapies in a cooperative group clinical trial and demonstrates the potential for positive outcomes without chemotherapy for ND patients with PTLD. This trial was registered at www.clinicaltrials.gov as #NCT02900976 and at the Children's Oncology Group as ANHL1522.
Collapse
Affiliation(s)
- Birte Wistinghausen
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Keri Toner
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Donald A. Barkauskas
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA
- Children’s Oncology Group Statistics and Data Center, Monrovia, CA
| | - Lauren P Jerkins
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Hannah Kinoshita
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Pamela Chansky
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Gloria Pezzella
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
| | - Lauren Saguilig
- Children’s Oncology Group Statistics and Data Center, Monrovia, CA
| | - Robert J. Hayashi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis Children’s Hospital, St. Louis, MO
| | - Harshal Abhyankar
- Baylor College of Medicine, Texas Children’s Hospital Cancer Center, Houston, TX
| | - Brooks Scull
- Baylor College of Medicine, Texas Children’s Hospital Cancer Center, Houston, TX
| | | | - Jay Tanna
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
| | - Patrick Hanley
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Michelle L. Hermiston
- Department of Pediatrics, Benioff Children’s Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Carl E. Allen
- Baylor College of Medicine, Texas Children’s Hospital Cancer Center, Houston, TX
| | - Catherine M. Bollard
- Center for Cancer and Blood Disorders, Children’s National Hospital, Washington, DC
- Center for Cancer and Immunology Research, Children’s National Research Institute, Children’s National Hospital, Washington, DC
- The George Washington University School of Medicine and Health Sciences, Washington, DC
| |
Collapse
|
28
|
Mark C, Martin G, Baadjes B, Geerlinks AV, Punnett A, Lafay-Cousin L. Treatment of Monomorphic Posttransplant Lymphoproliferative Disorder in Pediatric Solid Organ Transplant: A Multicenter Review. J Pediatr Hematol Oncol 2024; 46:e127-e130. [PMID: 38145403 DOI: 10.1097/mph.0000000000002804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023]
Abstract
Posttransplant lymphoproliferative disorder (PTLD) is the most common posttransplant malignancy in children. We reviewed data from 3 Canadian pediatric centers to determine patient characteristics, treatment approaches, and outcomes for children with monomorphic PTLD. There were 55 eligible children diagnosed between January 2001 to December 2021. Forty-eight patients (87.2%) had B-cell PTLD: Burkitt lymphoma (n = 25; 45.4%) and diffuse large B-cell lymphoma (n = 23; 41.2%), the remainder had natural killer (NK)/T-cell lymphoma (n = 5; 9.1%), Hodgkin lymphoma (n = 1;1.8%), or other (n = 1;1.8%). Thirty-nine (82.1%) patients with B-cell PTLD were treated with rituximab and chemotherapy with or without a reduction in immunosuppression (reduced immune suppression). The chemotherapy used was primarily one of 2 regimens: Mature Lymphoma B-96 protocol in 22 patients (56.4%) and low-dose cyclophosphamide with prednisone in 14 patients (35%). Most patients with T/NK-cell lymphoma were treated with reduced immune suppression + chemotherapy (n = 4; 80%). For all patients with monomorphic PTLD, the projected 3-year event-free survival/3-year overall survival was 62% and 77%, respectively. Of the patients, 100% with T/NK-cell PTLD 100% progressed or relapsed and, subsequently, died of disease. For patients with B-cell PTLD, there was no significant difference in outcome between the two main chemotherapy regimens employed.
Collapse
Affiliation(s)
- Catherine Mark
- Division of Hematology/Oncology, Toronto Hospital for Sick Children, Toronto, ON
| | - Georgina Martin
- Department of Hematology/Oncology, Jim Pattison Children's Hospital, Saskatoon, SK
| | - Bjorn Baadjes
- Department of Paediatric Hematology/Oncology, BC Children's Hospital, Vancouver, British CO
| | - Ashley V Geerlinks
- Department of Paediatric Hematology/Oncology, Children's Hospital, London Health Sciences Centre, Western University, London, ON
| | - Angela Punnett
- Division of Hematology/Oncology, Toronto Hospital for Sick Children, Toronto, ON
| | - Lucie Lafay-Cousin
- Department of Paediatric Hematology/Oncology and Bone Marrow Transplant, Alberta Children's Hospital, Calgary, AB, Canada
| |
Collapse
|
29
|
Carbone A, Chadburn A, Gloghini A, Vaccher E, Bower M. Immune deficiency/dysregulation -associated lymphoproliferative disorders. Revised classification and management. Blood Rev 2024; 64:101167. [PMID: 38195294 DOI: 10.1016/j.blre.2023.101167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/13/2023] [Accepted: 12/30/2023] [Indexed: 01/11/2024]
Abstract
Significant advances in the field of lymphoma have resulted in two recent classification proposals, the International Consensus Classification (ICC) and the 5th edition WHO. A few entities are categorized differently in the ICC compared to the WHO. Nowhere is this more apparent than the immunodeficiency lymphoproliferative disorders. The three previous versions of the WHO classification (3rd, 4th and revised 4th editions) and the ICC focused on four clinical settings in which these lesions arise for primary categorization. In contrast the 2023 WHO 5th edition includes pathologic characteristics including morphology and viral status, in addition to clinical setting, as important information for lesion classification. In addition, the 2023 WHO recognizes a broader number of clinical scenarios in which these lesions arise, including not only traditional types of immune deficiency but also immune dysregulation. With this classification it is hoped that new treatment strategies will be developed leading to better patient outcomes.
Collapse
Affiliation(s)
- Antonino Carbone
- Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, National Cancer Institute, Aviano, Italy.
| | - Amy Chadburn
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States of America.
| | - Annunziata Gloghini
- Department of Advanced Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Emanuela Vaccher
- Infectious Diseases and Tumors Unit, Department of Medical Oncology, Centro di Riferimento Oncologico (CRO), IRCCS, National Cancer Institute, Aviano, Italy.
| | - Mark Bower
- Department of Oncology and National Centre for HIV Malignancy, Chelsea & Westminster Hospital, London SW109NH, UK.
| |
Collapse
|
30
|
Mahadeo KM, Baiocchi R, Beitinjaneh A, Chaganti S, Choquet S, Dierickx D, Dinavahi R, Duan X, Gamelin L, Ghobadi A, Guzman-Becerra N, Joshi M, Mehta A, Navarro WH, Nikiforow S, O'Reilly RJ, Reshef R, Ruiz F, Spindler T, Prockop S. Tabelecleucel for allogeneic haematopoietic stem-cell or solid organ transplant recipients with Epstein-Barr virus-positive post-transplant lymphoproliferative disease after failure of rituximab or rituximab and chemotherapy (ALLELE): a phase 3, multicentre, open-label trial. Lancet Oncol 2024; 25:376-387. [PMID: 38309282 DOI: 10.1016/s1470-2045(23)00649-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/20/2023] [Accepted: 12/14/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Survival in Epstein-Barr virus (EBV)-positive post-transplant lymphoproliferative disease following haematopoietic stem-cell transplant (HSCT) or solid organ transplant (SOT) is poor after failure of initial therapy, indicating an urgent need for therapies for this ultra-rare disease. With recent EU marketing authorisation, tabelecleucel is the first off-the-shelf, allogeneic, EBV-specific T-cell immunotherapy to receive approval for treatment of relapsed or refractory EBV-positive post-transplant lymphoproliferative disease. We aimed to determine the clinical benefit of tabelecleucel in patients with relapsed or refractory EBV-positive post-transplant lymphoproliferative disease following HSCT or SOT. METHODS In this global, multicentre, open-label, phase 3 trial, eligible patients (of any age) had biopsy-proven EBV-positive post-transplant lymphoproliferative disease, disease that was relapsed or refractory to rituximab after HSCT and rituximab with or without chemotherapy after SOT, and partially HLA-matched and appropriately HLA-restricted tabelecleucel available. Patients received tabelecleucel administered intravenously at 2 × 106 cells per kg on days 1, 8, and 15 in 35-day cycles and are assessed for up to 5 years for survival post-treatment initiation. The primary endpoint was objective response rate. All patients who received at least one dose of tabelecleucel were included in safety and efficacy analyses. This trial is registered with ClinicalTrials.gov, NCT03394365, and is ongoing. FINDINGS From June 27, 2018, to Nov 5, 2021, 63 patients were enrolled, of whom 43 (24 [56%] male and 19 [44%] female) were included, 14 had prior HSCT, 29 had SOT. Seven (50%, 95% CI 23-77) of 14 participants in the HSCT group and 15 (52%, 33-71) of 29 participants in the SOT group had an objective response, with a median follow-up of 14·1 months (IQR 5·7-23·9) and 6·0 months (1·8-18·4), respectively. The most common grade 3 or 4 treatment-emergent adverse events were disease progression (in four [29%] of 14 in HSCT and eight [28%] of 29 in SOT) and decreased neutrophil count (in four [29%] of 14 in HSCT and four [14%] of 29 in SOT). Treatment-emergent serious adverse events were reported in 23 (53%) of 43 patients and fatal treatment-emergent adverse events in five (12%); no fatal treatment-emergent adverse event was treatment-related. There were no reports of tumour flare reaction, cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, transmission of infectious diseases, marrow rejection, or infusion reactions. No events of graft-versus-host disease or SOT rejection were reported as related to tabelecleucel. INTERPRETATION Tabelecleucel provides clinical benefit in patients with relapsed or refractory EBV-positive post-transplant lymphoproliferative disease, for whom there are no other approved therapies, without evidence of safety concerns seen with other adoptive T-cell therapies. These data represent a potentially transformative and accessible treatment advance for patients with relapsed or refractory disease with few treatment options. FUNDING Atara Biotherapeutics.
Collapse
Affiliation(s)
| | - Robert Baiocchi
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Amer Beitinjaneh
- Division of Transplantation and Cellular Therapy, University of Miami Hospital and Clinics, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Sridhar Chaganti
- Centre for Clinical Haematology, University Hospital Birmingham, Birmingham, UK
| | - Sylvain Choquet
- Clinical Hematology Unit, Groupe Hospitalier Pitié Salpêtrière, APHP, Sorbonne Université, Paris, France
| | | | | | | | | | - Armin Ghobadi
- Division of Oncology, Washington University, St Louis, MO, USA
| | | | | | - Aditi Mehta
- Atara Biotherapeutics, Thousand Oaks, CA, USA
| | | | - Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Richard J O'Reilly
- Transplant Service, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ran Reshef
- Blood and Marrow Transplantation and Cell Therapy Program, Columbia University Medical Center, New York, NY, USA
| | - Fiona Ruiz
- Atara Biotherapeutics, Thousand Oaks, CA, USA
| | | | - Susan Prockop
- Department of Pediatrics, Boston Children's Hospital-Dana Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
31
|
Rudilla F, Carrasco-Benso MP, Pasamar H, López-Montañés M, Andrés-Rozas M, Tomás-Marín M, Company D, Moya C, Larrea L, Guerreiro M, Barba P, Arbona C, Querol S. Development and characterization of a cell donor registry for virus-specific T cell manufacture in a blood bank. HLA 2024; 103:e15419. [PMID: 38450972 DOI: 10.1111/tan.15419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/19/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024]
Abstract
Adoptive cell therapy using virus-specific T cells (VST) is a strategy for treating common opportunistic viral infections after transplantation, particularly when these infections do not resolve through antiviral drug therapy. The availability of third-party healthy donors allows for the immediate use of cells for allogeneic therapy in cases where patients lack an appropriate donor. Here, we present the creation of a cell donor registry of human leukocyte antigen (HLA)-typed blood donors, REDOCEL, a strategic initiative to ensure the availability of compatible cells for donation when needed. Currently, the registry consists of 597 healthy donors with a median age of 29 years, 54% of whom are women. The most represented blood groups were A positive and O positive, with 36.52% and 34.51%, respectively. Also, donors were screened for cytomegalovirus (CMV) and Epstein-Barr virus (EBV). Almost 65% of donors were CMV-seropositive, while less than 5% were EBV-seronegative. Of the CMV-seropositive donors, 98% were also EBV-seropositive. High-resolution HLA-A, -B, -C, -DRB1 and -DQB1 allele and haplotype frequencies were determined in the registry. Prevalent HLA alleles and haplotypes were well represented to ensure donor-recipient HLA-matching, including alleles reported to present viral immunodominant epitopes. Since the functional establishment of REDOCEL, in May 2019, 87 effective donations have been collected, and the effective availability of donors with the first call has been greater than 75%. Thus, almost 89% of patients receiving an effective donation had available at least 5/10 HLA-matched cell donors (HLA-A, -B, -C, -DRB1, and -DQB1). To summarize, based on our experience, a cell donor registry from previously HLA-typed blood donors is a useful tool for facilitating access to VST therapy.
Collapse
Affiliation(s)
- Francesc Rudilla
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Immunogenetics and Histocompatibility Laboratory, Blood and Tissue Bank, Barcelona, Spain
| | - María Paz Carrasco-Benso
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (Fisabio), Valencia, Spain
| | - Helena Pasamar
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - María López-Montañés
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - María Andrés-Rozas
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - Maria Tomás-Marín
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| | - Desirée Company
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (Fisabio), Valencia, Spain
| | - Cristina Moya
- Blood Donors Management Department, Blood and Tissue Bank, Barcelona, Spain
| | - Luis Larrea
- Centro de Transfusión de la Comunitat Valenciana, Valencia, Spain
| | - Manuel Guerreiro
- Department of Hematology, La Fe Polytechnic and University Hospital, Valencia, Spain
| | - Pere Barba
- Hospital Vall d'Hebron, Barcelona, Spain
| | - Cristina Arbona
- Centro de Transfusión de la Comunitat Valenciana, Valencia, Spain
| | - Sergio Querol
- Transfusional Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma of Barcelona (VHIR-UAB), Barcelona, Spain
- Advanced & Cell Therapy Services, Blood and Tissue Bank, Barcelona, Spain
| |
Collapse
|
32
|
Work E, Gupta D, Slayton WB, Rees J, Coppola JA, Seifert R, Bleiweis MS, Jacobs JP, Peek G, Philip J, Brock A, Rivera JH, Sullivan K, Narasimhulu SS. Epstein Barr virus-directed T-cell therapy for refractory EBV-PTLD in a toddler post Orthotopic heart transplantation. Pediatr Transplant 2024; 28:e14707. [PMID: 38419558 DOI: 10.1111/petr.14707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 01/08/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
Epstein-Barr Virus (EBV) is a ubiquitous herpes type virus that is associated with post-transplant lymphoproliferative disorder (PTLD). Usual management includes reduction or cessation of immunosuppression and in some cases chemotherapy including rituximab. However, limited therapies are available if PTLD is refractory to rituximab. Several clinical trials have investigated the use of EBV-directed T cells in rituximab-refractory patients; however, data regarding response is scarce and inconclusive. Herein, we describe a patient with EBV-PTLD refractory to rituximab after orthotopic heart transplantation (OHT) requiring EBV-directed T-cell therapy. This article aims to highlight the unique and aggressive clinical presentation and progression of PTLD with utilization of EBV-directed T-cell therapy for management and associated pitfalls.
Collapse
Affiliation(s)
- Emily Work
- University of Florida, Gainesville, Florida, USA
| | - Dipankar Gupta
- Congenital Heart Center, University of Florida, Gainesville, Florida, USA
| | - William B Slayton
- Division of Hematology and Oncology, University of Florida, Gainesville, Florida, USA
| | - John Rees
- Department of Radiology, University of Florida, Gainesville, Florida, USA
| | | | - Robert Seifert
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | - Mark S Bleiweis
- Congenital Heart Center, University of Florida, Gainesville, Florida, USA
| | - Jeffrey P Jacobs
- Congenital Heart Center, University of Florida, Gainesville, Florida, USA
| | - Giles Peek
- Congenital Heart Center, University of Florida, Gainesville, Florida, USA
| | - Joseph Philip
- Congenital Heart Center, University of Florida, Gainesville, Florida, USA
| | - Alan Brock
- Congenital Heart Center, University of Florida, Gainesville, Florida, USA
| | | | - Kevin Sullivan
- Congenital Heart Center, University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|
33
|
Green A, Rubinstein JD, Grimley M, Pfeiffer T. Virus-Specific T Cells for the Treatment of Systemic Infections Following Allogeneic Hematopoietic Cell and Solid Organ Transplantation. J Pediatric Infect Dis Soc 2024; 13:S49-S57. [PMID: 38417086 DOI: 10.1093/jpids/piad077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/25/2023] [Indexed: 03/01/2024]
Abstract
Viral infections are a major source of morbidity and mortality in the context of immune deficiency and immunosuppression following allogeneic hematopoietic cell (allo-HCT) and solid organ transplantation (SOT). The pharmacological treatment of viral infections is challenging and often complicated by limited efficacy, the development of resistance, and intolerable side effects. A promising strategy to rapidly restore antiviral immunity is the adoptive transfer of virus-specific T cells (VST). This therapy involves the isolation and ex vivo expansion or direct selection of antigen-specific T cells from healthy seropositive donors, followed by infusion into the patient. This article provides a practical guide to VST therapy by reviewing manufacturing techniques, donor selection, and treatment indications. The safety and efficacy data of VSTs gathered in clinical trials over nearly 30 years is summarized. Current challenges and limitations are discussed, as well as opportunities for further research and development.
Collapse
Affiliation(s)
- Abby Green
- Department of Pediatrics, Division of Hematology/Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jeremy D Rubinstein
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michael Grimley
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Thomas Pfeiffer
- Department of Pediatrics, Division of Hematology/Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
34
|
Kong IY, Giulino-Roth L. Targeting latent viral infection in EBV-associated lymphomas. Front Immunol 2024; 15:1342455. [PMID: 38464537 PMCID: PMC10920267 DOI: 10.3389/fimmu.2024.1342455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Epstein-Barr virus (EBV) contributes to the development of a significant subset of human lymphomas. As a herpes virus, EBV can transition between a lytic state which is required to establish infection and a latent state where a limited number of viral antigens are expressed which allows infected cells to escape immune surveillance. Three broad latency programs have been described which are defined by the expression of viral proteins RNA, with latency I being the most restrictive expressing only EBV nuclear antigen 1 (EBNA1) and EBV-encoded small RNAs (EBERs) and latency III expressing the full panel of latent viral genes including the latent membrane proteins 1 and 2 (LMP1/2), and EBNA 2, 3, and leader protein (LP) which induce a robust T-cell response. The therapeutic use of EBV-specific T-cells has advanced the treatment of EBV-associated lymphoma, however this approach is only effective against EBV-associated lymphomas that express the latency II or III program. Latency I tumors such as Burkitt lymphoma (BL) and a subset of diffuse large B-cell lymphomas (DLBCL) evade the host immune response to EBV and are resistant to EBV-specific T-cell therapies. Thus, strategies for inducing a switch from the latency I to the latency II or III program in EBV+ tumors are being investigated as mechanisms to sensitize tumors to T-cell mediated killing. Here, we review what is known about the establishment and regulation of latency in EBV infected B-cells, the role of EBV-specific T-cells in lymphoma, and strategies to convert latency I tumors to latency II/III.
Collapse
|
35
|
Wang J, Xu Z, Lai Y, Zhang Y, Zhang P, Mu Q, Yang S, Sun Y, Sheng L, Ouyang G. Silencing of PD-1 combined with EBV-specific killer T cells for the treatment of EBV-associated B lymphoma. Transl Oncol 2024; 40:101831. [PMID: 38039947 PMCID: PMC10716022 DOI: 10.1016/j.tranon.2023.101831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Epstein-Barr Virus (EBV) infection is closely associated with the development of lymphoma, as it plays a significant role in the malignant transformation of lymphocytes. The expression of programmed death-1 (PD-1), which binds to PD-L1 in tumor cells, can lead to immune evasion by lymphoma cells and promote tumor progression. In this study, immortalized B lymphoblastoid cell lines (B-LCLs) positive for EBV-specific proteins were established from human peripheral mononuclear cells (PBMCs) using EBV induction along with CpG-ODN 2006 and cyclosporin A. EBV-specific T cells (EBVST) were generated by multiple immunizations of CD3+ T lymphocytes using irradiated B-LCLs. Flow cytometry analysis confirmed the activation of EBVST through the detection of CD3+, CD4+, and CD8+ markers. Co-incubation of EBVST with EBV-positive B lymphocyte cell lines resulted in the secretion of perforin by EBVST, leading to granzyme B-mediated cell death and an increase in LDH levels. Silencing PD-1 in EBVST cells enhanced perforin production, increased granzyme B release, and upregulated cell death in co-incubated B lymphocytes. In a nude mice tumor transplantation model, silencing PD-1 in combination with EBV-specific killer T cells exhibited the maximum inhibition of B-lymphoblastoma. This treatment upregulated the expression of proteins associated with apoptosis and immune response, while inhibiting anti-apoptotic protein expression in tumor tissues. Silencing PD-1 also increased the infiltration of EBV-specific killer T cells in the tumor tissues. Overall, PD-1 silencing enhanced the tumor targeting effect of EBV-specific killer T cells on EBV-infected B lymphocytes and attenuated the immune escape effect mediated by the PD-1 pathway.
Collapse
Affiliation(s)
- Jiaping Wang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Zhijuan Xu
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Yanli Lai
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Yanli Zhang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Ping Zhang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Qitian Mu
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Shujun Yang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Yongcheng Sun
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China
| | - Lixia Sheng
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China.
| | - Guifang Ouyang
- Ningbo Clinical Research Center for Hematological Malignancies, Department of hematology, the First Affiliated Hospital of Ningbo University, Ningbo 315000, Zhejiang, China.
| |
Collapse
|
36
|
Preiksaitis J, Allen U, Bollard CM, Dharnidharka VR, Dulek DE, Green M, Martinez OM, Metes DM, Michaels MG, Smets F, Chinnock RE, Comoli P, Danziger-Isakov L, Dipchand AI, Esquivel CO, Ferry JA, Gross TG, Hayashi RJ, Höcker B, L'Huillier AG, Marks SD, Mazariegos GV, Squires J, Swerdlow SH, Trappe RU, Visner G, Webber SA, Wilkinson JD, Maecker-Kolhoff B. The IPTA Nashville Consensus Conference on Post-Transplant lymphoproliferative disorders after solid organ transplantation in children: III - Consensus guidelines for Epstein-Barr virus load and other biomarker monitoring. Pediatr Transplant 2024; 28:e14471. [PMID: 37294621 DOI: 10.1111/petr.14471] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/10/2022] [Accepted: 01/02/2023] [Indexed: 06/11/2023]
Abstract
The International Pediatric Transplant Association convened an expert consensus conference to assess current evidence and develop recommendations for various aspects of care relating to post-transplant lymphoproliferative disorders after solid organ transplantation in children. In this report from the Viral Load and Biomarker Monitoring Working Group, we reviewed the existing literature regarding the role of Epstein-Barr viral load and other biomarkers in peripheral blood for predicting the development of PTLD, for PTLD diagnosis, and for monitoring of response to treatment. Key recommendations from the group highlighted the strong recommendation for use of the term EBV DNAemia instead of "viremia" to describe EBV DNA levels in peripheral blood as well as concerns with comparison of EBV DNAemia measurement results performed at different institutions even when tests are calibrated using the WHO international standard. The working group concluded that either whole blood or plasma could be used as matrices for EBV DNA measurement; optimal specimen type may be clinical context dependent. Whole blood testing has some advantages for surveillance to inform pre-emptive interventions while plasma testing may be preferred in the setting of clinical symptoms and treatment monitoring. However, EBV DNAemia testing alone was not recommended for PTLD diagnosis. Quantitative EBV DNAemia surveillance to identify patients at risk for PTLD and to inform pre-emptive interventions in patients who are EBV seronegative pre-transplant was recommended. In contrast, with the exception of intestinal transplant recipients or those with recent primary EBV infection prior to SOT, surveillance was not recommended in pediatric SOT recipients EBV seropositive pre-transplant. Implications of viral load kinetic parameters including peak load and viral set point on pre-emptive PTLD prevention monitoring algorithms were discussed. Use of additional markers, including measurements of EBV specific cell mediated immunity was discussed but not recommended though the importance of obtaining additional data from prospective multicenter studies was highlighted as a key research priority.
Collapse
Affiliation(s)
- Jutta Preiksaitis
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Upton Allen
- Division of Infectious Diseases and the Transplant and Regenerative Medicine Center, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, The George Washington University, Washington, District of Columbia, USA
| | - Vikas R Dharnidharka
- Department of Pediatrics, Division of Pediatric Nephrology, Hypertension & Pheresis, Washington University School of Medicine & St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Daniel E Dulek
- Division of Pediatric Infectious Diseases, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael Green
- Division of Pediatric Infectious Diseases, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Olivia M Martinez
- Department of Surgery and Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Diana M Metes
- Departments of Surgery and Immunology, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marian G Michaels
- Division of Pediatric Infectious Diseases, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Françoise Smets
- Pediatric Gastroenterology and Hepatology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | | | - Patrizia Comoli
- Cell Factory & Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico, Pavia, Italy
| | - Lara Danziger-Isakov
- Division of Infectious Disease, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Anne I Dipchand
- Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | - Judith A Ferry
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Thomas G Gross
- Center for Cancer and Blood Diseases, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Robert J Hayashi
- Division of Pediatric Hematology/Oncology, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Britta Höcker
- University Children's Hospital, Pediatrics I, Heidelberg, Germany
| | - Arnaud G L'Huillier
- Faculty of Medicine, Pediatric Infectious Diseases Unit and Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland
| | - Stephen D Marks
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health, London, UK
| | - George Vincent Mazariegos
- Department of Surgery, Hillman Center for Pediatric Transplantation, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James Squires
- Division of Gastroenterology, Hepatology and Nutrition, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Steven H Swerdlow
- Division of Hematopathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ralf U Trappe
- Department of Hematology and Oncology, DIAKO Ev. Diakonie-Krankenhaus Bremen, Bremen, Germany
- Department of Internal Medicine II: Hematology and Oncology, University Medical Centre Schleswig-Holstein, Kiel, Germany
| | - Gary Visner
- Division of Pulmonary Medicine, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Steven A Webber
- Department of Pediatrics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - James D Wilkinson
- Department of Pediatrics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | | |
Collapse
|
37
|
Green M, Squires JE, Chinnock RE, Comoli P, Danziger-Isakov L, Dulek DE, Esquivel CO, Höcker B, L'Huillier AG, Mazariegos GV, Visner GA, Bollard CM, Dipchand AI, Ferry JA, Gross TG, Hayashi R, Maecker-Kolhoff B, Marks S, Martinez OM, Metes DM, Michaels MG, Preiksaitis J, Smets F, Swerdlow SH, Trappe RU, Wilkinson JD, Allen U, Webber SA, Dharnidharka VR. The IPTA Nashville consensus conference on Post-Transplant lymphoproliferative disorders after solid organ transplantation in children: II-consensus guidelines for prevention. Pediatr Transplant 2024; 28:e14350. [PMID: 36369745 DOI: 10.1111/petr.14350] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022]
Abstract
The International Pediatric Transplant Association (IPTA) convened an expert consensus conference to assess current evidence and develop recommendations for various aspects of care relating to post-transplant lymphoproliferative disorder after solid organ transplantation in children. In this report from the Prevention Working Group, we reviewed the existing literature regarding immunoprophylaxis and chemoprophylaxis, and pre-emptive strategies. While the group made a strong recommendation for pre-emptive reduction of immunosuppression at the time of EBV DNAemia (low to moderate evidence), no recommendations for use could be made for any prophylactic strategy or alternate pre-emptive strategy, largely due to insufficient or conflicting evidence. Current gaps and future research priorities are highlighted.
Collapse
Affiliation(s)
- Michael Green
- Division of Pediatric Infectious Diseases, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James E Squires
- Division of Gastroenterology, Hepatology and Nutrition, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Patrizia Comoli
- Cell Factory & Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico, San Matteo, Pavia, Italy
| | - Lara Danziger-Isakov
- Division of Infectious Disease, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Daniel E Dulek
- Division of Pediatric Infectious Diseases, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Britta Höcker
- Pediatrics I, University Children's Hospital, Heidelberg, Germany
| | - Arnaud G L'Huillier
- Pediatric Infectious Diseases Unit and Laboratory of Virology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - George Vincent Mazariegos
- Hillman Center for Pediatric Transplantation, UPMC Children's Hospital of Pittsburgh and Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gary A Visner
- Division of Pulmonary Medicine, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, The George Washington University, Washington, District of Columbia, USA
| | - Anne I Dipchand
- Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Judith A Ferry
- Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Thomas G Gross
- Center for Cancer and Blood Diseases, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Robert Hayashi
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | | | - Stephen Marks
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health, London, UK
| | - Olivia M Martinez
- Department of Surgery and Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Diana M Metes
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marian G Michaels
- Division of Pediatric Infectious Diseases, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jutta Preiksaitis
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Françoise Smets
- Pediatric Gastroenterology and Hepatology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Stephen H Swerdlow
- Division of Hematopathology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ralf U Trappe
- Department of Hematology and Oncology, DIAKO Ev. Diakonie-Krankenhaus Bremen, Bremen, Germany and Department of Internal Medicine II: Hematology and Oncology, University Medical Centre Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - James D Wilkinson
- Department of Pediatrics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - Upton Allen
- Division of Infectious Diseases and the Transplant and Regenerative Medicine Center, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Steven A Webber
- Department of Pediatrics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - Vikas R Dharnidharka
- Department of Pediatrics, Division of Pediatric Nephrology, Hypertension & Pheresis, Washington University School of Medicine & St. Louis Children's Hospital, St. Louis, Missouri, USA
| |
Collapse
|
38
|
Bednarska K, Chowdhury R, Tobin JWD, Swain F, Keane C, Boyle S, Khanna R, Gandhi MK. Epstein-Barr virus-associated lymphomas decoded. Br J Haematol 2024; 204:415-433. [PMID: 38155519 DOI: 10.1111/bjh.19255] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/15/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023]
Abstract
Epstein-Barr virus (EBV)-associated lymphomas cover a range of histological B- and T-cell non-Hodgkin and Hodgkin lymphoma subtypes. The role of EBV on B-cell malignant pathogenesis and its impact on the tumour microenvironment are intriguing but incompletely understood. Both the International Consensus Classification (ICC) and 5th Edition of the World Health Organization (WHO-HAEM5) proposals give prominence to the distinct clinical, prognostic, genetic and tumour microenvironmental features of EBV in lymphoproliferative disorders. There have been major advances in our biological understanding, in how to harness features of EBV and its host immune response for targeted therapy, and in using EBV as a method to monitor disease response. In this article, we showcase the latest developments and how they may be integrated to stimulate new and innovative approaches for further lines of investigation and therapy.
Collapse
Affiliation(s)
- Karolina Bednarska
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Rakin Chowdhury
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Joshua W D Tobin
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Fiona Swain
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Colm Keane
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Stephen Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Rajiv Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Maher K Gandhi
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
O’Reilly RJ, Prockop S, Oved JH. Virus-specific T-cells from third party or transplant donors for treatment of EBV lymphoproliferative diseases arising post hematopoietic cell or solid organ transplantation. Front Immunol 2024; 14:1290059. [PMID: 38274824 PMCID: PMC10808771 DOI: 10.3389/fimmu.2023.1290059] [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: 09/06/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
EBV+ lymphomas constitute a significant cause of morbidity and mortality in recipients of allogeneic hematopoietic cell (HCT) and solid organ transplants (SOT). Phase I and II trials have shown that in HCT recipients, adoptive transfer of EBV-specific T-cells from the HCT donor can safely induce durable remissions of EBV+ lymphomas including 70->90% of patients who have failed to respond to treatment with Rituximab. More recently, EBV-specific T-cells generated from allogeneic 3rd party donors have also been shown to induce durable remission of EBV+ lymphomas in Rituximab refractory HCT and SOT recipients. In this review, we compare results of phase I and II trials of 3rd party and donor derived EBV-specific T-cells. We focus on the attributes and limitations of each product in terms of access, safety, responses achieved and durability. The limited data available regarding donor and host factors contributing to T cell persistence is also described. We examine factors contributing to treatment failures and approaches to prevent or salvage relapse. Lastly, we summarize strategies to further improve results for virus-specific immunotherapies for post-transplant EBV lymphomas.
Collapse
Affiliation(s)
- Richard J. O’Reilly
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Susan Prockop
- Pediatric Stem Cell Transplantation, Boston Children’s Hospital/Dana-Farber Cancer Institute, Boston, MA, United States
| | - Joseph H. Oved
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
40
|
Seňavová J, Rajmonová A, Heřman V, Jura F, Veľasová A, Hamová I, Tkachenko A, Kupcová K, Havránek O. Immune Checkpoints and Their Inhibition in T-Cell Lymphomas. Folia Biol (Praha) 2024; 70:123-151. [PMID: 39644109 DOI: 10.14712/fb2024070030123] [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] [Indexed: 12/09/2024]
Abstract
T-cell lymphomas (TCLs) are a rare and heterogeneous subgroup of non-Hodgkin lymphomas (NHLs), forming only 10 % of all NHL cases in Western countries. Resulting from their low incidence and heterogeneity, the current treatment outcome is generally unfavorable, with limited availability of novel therapeutic approaches. Therefore, the recent success of immune checkpoint inhibitors (ICIs) in cancer treatment motivated their clinical investigation in TCLs as well. Multiple studies showed promising results; however, cases of TCL hyperprogression following ICI treatment and secondary T-cell-derived malignancies associated with ICI treatment of other cancer types were also reported. In our review, we first briefly summarize classification of T-cell-derived malignancies, general anti-tumor immune response, immune evasion, and immune checkpoint signaling. Next, we provide an overview of immune checkpoint molecule deregulation in TCLs, summarize available studies of ICIs in TCLs, and review the above-mentioned safety concerns associa-ted with ICI treatment and T-cell-derived malignancies. Despite initial promising results, further studies are necessary to define the most suitable clinical applications and ICI therapeutic combinations with other novel treatment approaches within TCL treatment. ICIs, and their combinations, might hopefully bring the long awaited improvement for the treatment of T-cell-derived malignancies.
Collapse
Affiliation(s)
- Jana Seňavová
- 1st Department of Medicine - Department of Haematology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Anežka Rajmonová
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Václav Heřman
- 1st Department of Medicine - Department of Haematology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Filip Jura
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Adriana Veľasová
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Iva Hamová
- 1st Department of Medicine - Department of Haematology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristýna Kupcová
- 1st Department of Medicine - Department of Haematology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ondřej Havránek
- 1st Department of Medicine - Department of Haematology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
- BIOCEV, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
41
|
Schreiber B, Tripathi S, Nikiforow S, Chandraker A. Adoptive Immune Effector Cell Therapies in Cancer and Solid Organ Transplantation: A Review. Semin Nephrol 2024; 44:151498. [PMID: 38555223 DOI: 10.1016/j.semnephrol.2024.151498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Cancer is one of the most devastating complications of kidney transplantation and constitutes one of the leading causes of morbidity and mortality among solid organ transplantation (SOT) recipients. Immunosuppression, although effective in preventing allograft rejection, inherently inhibits immune surveillance against oncogenic viral infections and malignancy. Adoptive cell therapy, particularly immune effector cell therapy, has long been a modality of interest in both cancer and transplantation, though has only recently stepped into the spotlight with the development of virus-specific T-cell therapy and chimeric antigen receptor T-cell therapy. Although these modalities are best described in hematopoietic cell transplantation and hematologic malignancies, their potential application in the SOT setting may hold tremendous promise for those with limited therapeutic options. In this review, we provide a brief overview of the development of adoptive cell therapies with a focus on virus-specific T-cell therapy and chimeric antigen receptor T-cell therapy. We also describe the current experience of these therapies in the SOT setting as well as the challenges in their application and future directions in their development.
Collapse
Affiliation(s)
- Brittany Schreiber
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sudipta Tripathi
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sarah Nikiforow
- Division of Medical Oncology, Department of Medicine, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anil Chandraker
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Renal Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA.
| |
Collapse
|
42
|
Bachar-Lustig E, Lask A, Eidelstein Y, Or-Geva N, Gidron-Budovsky R, Nathansohn-Levy B, Eyrich M, Liu WH, Dang G, Miranda KC, Ramirez A, Kaur I, Rezvani K, Shpall E, Champlin RE, Nagler A, Shimoni A, Barnees-Kagan S, Reisner Y. Generation of Non-Alloreactive Antiviral Central Memory CD8 Human Veto T Cells for Cell Therapy. Transplant Cell Ther 2024; 30:71.e1-71.e13. [PMID: 37890590 DOI: 10.1016/j.jtct.2023.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023]
Abstract
Previous studies in mice demonstrated that CD8 T cells exhibit marked veto activity enhancing engraftment in several models for T cell-depleted bone marrow (TDBM) allografting. To reduce the risk of graft-versus-host disease (GVHD) associated with allogeneic CD8 veto T cells, these studies made use of naive CD8 T cells stimulated against third-party stimulators under cytokine deprivation and subsequent expansion in the presence of IL-15. More recently, it was shown that mouse CD8 veto T cells can be generated by stimulating CD8 memory T cells from ovalbumin immunized mice under cytokine deprivation, using ovalbumin as a third-party antigen. These cells also exhibited substantial enhancement of BM allografting without GVHD. In this study, we tested the hypothesis that stimulation and expansion of human CD8 memory T cells under IL-15 and IL-7 deprivation during the early phase of activation against recall viral antigens can lead to substantial loss of alloreactive T clones while retaining marked veto activity. Memory CD8 T cells were enriched by removal of CD45RA+, CD4+, and CD56+ cells from peripheral blood of cytomegalovirus (CMV)- and Epstein-Barr virus (EBV)-positive donors. In parallel, CD14+ monocytes were isolated; differentiated into mature dendritic cells (mDCs); pulsed with a library of CMV, EBV, adenovirus, and BK virus peptides; and irradiated. The CD8 T cell-enriched fraction was then cultured with the pulsed mDCs in the presence of IL-21 for 3 days, after which IL-15 and IL-7 were added. After 12 days of culture, the cells were tested by limiting dilution analysis for the frequency of alloreactive T cell clones and their veto activity. In preclinical runs using GMP reagents, we established that within 12 days of culture, a large number of highly homogenous CD8 T cells, predominantly expressing a central memory phenotype, could be harvested. These cells exhibited marked veto activity in vitro and >3-log depletion of alloreactivity. Based on these preclinical data, a phase 1-2 clinical trial was initiated to test the safety and efficacy of these antiviral CD8 central memory veto cells in the context of nonmyeloablative (NMA) T cell-depleted haploidentical hematopoietic stem cell transplantation (HSCT). In 2 validation runs and 11 clinical runs using GMP reagents, >1 × 1010 cells were generated from a single leukapheresis in 12 out of 13 experiments. At the end of 12 days of culture, there were 97 ± 2.5% CD3+CD8+ T cells, of which 84 ± 9.0% (range, 71.5% to 95.1%) exhibited the CD45RO+CD62L+ CM phenotype. Antiviral activity tested by intracellular expression of INF-γ and TNF-α and showed an average of 38.8 ± 19.6% positive cells on 6 hours of stimulation against the viral peptide mixture. Our results demonstrate a novel approach for depleting alloreactive T cell clones from preparations of antiviral CD8 veto cells. Based on these results, a phase 1-2 clinical trial is currently in progress to test the safety and efficacy of these veto cells in the context of NMA haploidentical T cell-depleted HSCT. Studies testing the hypothesis that these non-alloreactive CD8 T cells could potentially offer a platform for off-the-shelf veto chimeric antigen receptor T cell therapy in allogenic recipients, are warranted.
Collapse
Affiliation(s)
- Esther Bachar-Lustig
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Assaf Lask
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yaki Eidelstein
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Noga Or-Geva
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Matthias Eyrich
- Children's Department of Oncology, Hematology and Stem Cell Transplantation, University, Hospital Wurzburg, Wurzburg, Germany
| | - Wei-Hsin Liu
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Giang Dang
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Karla Castro Miranda
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Alejandro Ramirez
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Indreshpal Kaur
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Katayoun Rezvani
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth Shpall
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Avichai Shimoni
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | | | - Yair Reisner
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Hematopoietic Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas; Cancer Prevention and Research Institute of Texas Scholars in Cancer Research, Houston, Texas.
| |
Collapse
|
43
|
Storek J, Lindsay J. Rituximab for posttransplant lymphoproliferative disorder - therapeutic, preemptive, or prophylactic? Bone Marrow Transplant 2024; 59:6-11. [PMID: 38001229 DOI: 10.1038/s41409-023-02155-5] [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: 09/09/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023]
Abstract
To minimize mortality due to posttransplant lymphoproliferative disorder (PTLD), the following strategies have been used: (1) Therapy without EBV Monitoring, i.e., administration of rituximab after PTLD diagnosis, usually by biopsy, in the absence of routine Epstein-Barr virus (EBV) DNAemia monitoring, (2) Prompt Therapy, i.e., monitoring EBV DNAemia, searching for PTLD by imaging when the DNAemia has exceeded a pre-specified threshold, and administration of rituximab if the imaging is consistent with PTLD, (3) Preemptive Therapy, i.e., monitoring EBV DNAemia and administration of rituximab when the DNAemia has exceeded a pre-specified threshold, and (4) Prophylaxis, i.e., administration of rituximab to all transplant recipients. The superiority of one of these strategies over the other strategies has not been established. Here we review the pros and cons of each strategy. Preemptive therapy or prophylaxis may currently be preferred for patients who are at a high risk of dying due to PTLD. However, Therapy without EBV Monitoring may be used for both high- and low-risk patients in the future, if effective and relatively non-toxic therapies for rituximab-refractory PTLD (e.g., EBV-specific T cells) have become easily available.
Collapse
Affiliation(s)
- Jan Storek
- University of Calgary, Calgary, AB, Canada.
| | - Julian Lindsay
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- National Centre for Infection in Cancer and Transplantation, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
44
|
Dharnidharka VR, Ruzinova MB, Marks LJ. Post-Transplant Lymphoproliferative Disorders. Semin Nephrol 2024; 44:151503. [PMID: 38519279 PMCID: PMC11213680 DOI: 10.1016/j.semnephrol.2024.151503] [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] [Indexed: 03/24/2024]
Abstract
Post-transplant lymphoproliferative disorders (PTLDs) are a heterogenous set of unregulated lymphoid cell proliferations after organ or tissue transplant. A majority of cases are associated with the Epstein-Barr virus and higher intensity of pharmacologic immunosuppression. The clinical presentations are numerous. The diagnosis is ideally by histology, except in cases where the tumor is inaccessible to biopsy. While some pre-emptive therapies and treatment strategies are available have reasonable success are available, they do not eliminate the high morbidity and significant mortality after PTLD.
Collapse
Affiliation(s)
- Vikas R Dharnidharka
- Division of Pediatric Nephrology, Hypertension and Apheresis, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO.
| | - Marianna B Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Lianna J Marks
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
45
|
Leroyer EH, Petitpain N, Morisset S, Neven B, Castelle M, Winter S, Souchet L, Morel V, Le Cann M, Fahd M, Yacouben K, Mechinaud F, Ouachée-Chardin M, Renard C, Wallet HL, Angoso M, Jubert C, Chevallier P, Léger A, Rialland F, Dhedin N, Robin C, Maury S, Beckerich F, Beauvais D, Cluzeau T, Loschi M, Fernster A, Bittencourt MDC, Cravat M, Bilger K, Clément L, Decot V, Gauthier M, Legendre A, Larghero J, Ouedrani A, Martin-Blondel G, Pochon C, Reppel L, Rouard H, Nguyen-Quoc S, Dalle JH, D'Aveni M, Bensoussan D. On behalf of the SFGM-TC: Real-life use of third-party virus-specific T-cell transfer in immunocompromised transplanted patients. Hemasphere 2024; 8:e40. [PMID: 38434523 PMCID: PMC10878191 DOI: 10.1002/hem3.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/14/2023] [Indexed: 03/05/2024] Open
Affiliation(s)
| | - Nadine Petitpain
- Regional Centre of Pharmacovigilance Nancy University Hospital Vandoeuvre-les-Nancy France
| | | | - Bénédicte Neven
- Assistance Publique des Hôpitaux de Paris, Department of Pediatric Immuno-hematology Necker Children Hospital Paris France
| | - Martin Castelle
- Assistance Publique des Hôpitaux de Paris, Department of Pediatric Immuno-hematology Necker Children Hospital Paris France
| | - Sarah Winter
- Assistance Publique des Hôpitaux de Paris, Department of Pediatric Immuno-hematology Necker Children Hospital Paris France
| | - Laetitia Souchet
- Assistance Publique des Hôpitaux de Paris, Department of Hematology La Pitié-Salpêtrière Hospital Paris France
| | - Véronique Morel
- Assistance Publique des Hôpitaux de Paris, Department of Hematology La Pitié-Salpêtrière Hospital Paris France
| | - Marie Le Cann
- Assistance Publique des Hôpitaux de Paris, Department of Hematology La Pitié-Salpêtrière Hospital Paris France
| | - Mony Fahd
- Department of Pediatric Hematology and Immunology, Robert Debré Academic Hospital GHU APHP Nord Université Paris Cité Paris France
| | - Karima Yacouben
- Department of Pediatric Hematology and Immunology, Robert Debré Academic Hospital GHU APHP Nord Université Paris Cité Paris France
| | - Françoise Mechinaud
- Department of Pediatric Hematology and Immunology, Robert Debré Academic Hospital GHU APHP Nord Université Paris Cité Paris France
| | - Marie Ouachée-Chardin
- Institute of Pediatric Hematology and Oncology (IHOPe) Hospices Civils de Lyon and Claude Bernard University Lyon France
| | - Cécile Renard
- Institute of Pediatric Hematology and Oncology (IHOPe) Hospices Civils de Lyon and Claude Bernard University Lyon France
| | - Hélène Labussière Wallet
- Institute of Pediatric Hematology and Oncology (IHOPe) Hospices Civils de Lyon and Claude Bernard University Lyon France
| | - Marie Angoso
- Department of Pediatric Hematology Oncology University Hospital of Bordeaux Bordeaux France
| | - Charlotte Jubert
- Department of Pediatric Hematology Oncology University Hospital of Bordeaux Bordeaux France
| | | | - Alexandra Léger
- Department of Pediatric Hematology, Hôpital Mère-Enfant Nantes University Hospital Nantes France
| | - Fanny Rialland
- Department of Pediatric Hematology, Hôpital Mère-Enfant Nantes University Hospital Nantes France
| | - Nathalie Dhedin
- Assistance Publique des Hôpitaux de Paris, Department of Hematology Saint-Louis Hospital Paris France
| | - Christine Robin
- Assistance Publique des Hôpitaux de Paris, Department of Hematology Henri Mondor Hospital and Université Créteil France
| | - Sébastien Maury
- Assistance Publique des Hôpitaux de Paris, Department of Hematology Henri Mondor Hospital and Université Créteil France
| | - Florence Beckerich
- Assistance Publique des Hôpitaux de Paris, Department of Hematology Henri Mondor Hospital and Université Créteil France
| | - David Beauvais
- Department of Hematology, Allogeneic Stem Cell Transplantation Unit Lille University Hospital Lille France
| | - Thomas Cluzeau
- Department of Hematology, Université Cote d'Azur Nice University Hospital Nice France
| | - Michaël Loschi
- Department of Hematology, Université Cote d'Azur Nice University Hospital Nice France
| | - Alina Fernster
- Hôpital Universitaire des Enfants de la Reine Fabiola, Department of Pediatric Hematology Brussels University Hospital Brussels Belgium
| | | | - Maxime Cravat
- Cytometry Platform Nancy University Hospital Vandoeuvre-les-Nancy France
| | - Karin Bilger
- INCANS Department of Hematology Strasbourg France
| | - Laurence Clément
- Department of Hematology Bordeaux University Hospital Bordeaux France
| | - Véronique Decot
- Cell Therapy Unit Nancy University Hospital Vandoeuvre-les-Nancy France
| | - Mélanie Gauthier
- Cell Therapy Unit Nancy University Hospital Vandoeuvre-les-Nancy France
| | | | - Jérôme Larghero
- Assistance Publique des Hôpitaux de Paris, Cell Therapy Unit, INSERM CICBT 501 Saint-Louis Hospital Paris France
| | - Amani Ouedrani
- Assistance Publique des Hôpitaux de Paris, Department of Immunology and Histocompatibily Saint-Louis Hospital Paris France
| | - Guillaume Martin-Blondel
- Department of Infectious and Tropical Diseases, and Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291-CNRS UMR5051-Université Toulouse III Toulouse University Hospital Toulouse France
| | - Cécile Pochon
- Department of Pediatric Hematology Nancy University Hospital Vandoeuvre-les-Nancy France
- CNRS Unit UMR 7365 IMoPA Lorraine University Vandoeuvre-les-Nancy France
| | - Loïc Reppel
- Cell Therapy Unit Nancy University Hospital Vandoeuvre-les-Nancy France
- CNRS Unit UMR 7365 IMoPA Lorraine University Vandoeuvre-les-Nancy France
| | | | - Stéphanie Nguyen-Quoc
- Assistance Publique des Hôpitaux de Paris, Department of Hematology La Pitié-Salpêtrière Hospital Paris France
| | - Jean-Hugues Dalle
- Department of Pediatric Hematology and Immunology, Robert Debré Academic Hospital GHU APHP Nord Université Paris Cité Paris France
| | - Maud D'Aveni
- Department of Hematology Nancy University Hospital Vandoeuvre-les-Nancy France
- CNRS Unit UMR 7365 IMoPA Lorraine University Vandoeuvre-les-Nancy France
| | - Danièle Bensoussan
- Cell Therapy Unit Nancy University Hospital Vandoeuvre-les-Nancy France
- CNRS Unit UMR 7365 IMoPA Lorraine University Vandoeuvre-les-Nancy France
| |
Collapse
|
46
|
Koukoulias K, Papayanni PG, Jones J, Kuvalekar M, Watanabe A, Velazquez Y, Gilmore S, Papadopoulou A, Leen AM, Vasileiou S. Assessment of the cytolytic potential of a multivirus-targeted T cell therapy using a vital dye-based, flow cytometric assay. Front Immunol 2023; 14:1299512. [PMID: 38187380 PMCID: PMC10766817 DOI: 10.3389/fimmu.2023.1299512] [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: 09/22/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Reliable and sensitive characterization assays are important determinants of the successful clinical translation of immunotherapies. For the assessment of cytolytic potential, the chromium 51 (51Cr) release assay has long been considered the gold standard for testing effector cells. However, attaining the approvals to access and use radioactive isotopes is becoming increasingly complex, while technical aspects [i.e. sensitivity, short (4-6 hours) assay duration] may lead to suboptimal performance. This has been the case with our ex vivo expanded, polyclonal (CD4+ and CD8+) multivirus-specific T cell (multiVST) lines, which recognize 5 difficult-to-treat viruses [Adenovirus (AdV), BK virus (BKV), cytomegalovirus (CMV), Epstein Barr virus (EBV), and human herpes virus 6 (HHV6)] and when administered to allogeneic hematopoietic stem cell (HCT) or solid organ transplant (SOT) recipients have been associated with clinical benefit. However, despite mediating potent antiviral effects in vivo, capturing in vitro cytotoxic potential has proven difficult in a traditional 51Cr release assay. Now, in addition to cytotoxicity surrogates, including CD107a and Granzyme B, we report on an alternative, vital dye -based, flow cytometric platform in which superior sensitivity and prolonged effector:target co-culture duration enabled the reliable detection of both CD4- and CD8-mediated in vitro cytolytic activity against viral targets without non-specific effects.
Collapse
Affiliation(s)
- Kiriakos Koukoulias
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Penelope G. Papayanni
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Julia Jones
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Manik Kuvalekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Ayumi Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Yovana Velazquez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | | | - Anastasia Papadopoulou
- Hematology Department- Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, “George Papanikolaou” Hospital, Thessaloniki, Greece
| | - Ann M. Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
47
|
Sausen DG, Poirier MC, Spiers LM, Smith EN. Mechanisms of T cell evasion by Epstein-Barr virus and implications for tumor survival. Front Immunol 2023; 14:1289313. [PMID: 38179040 PMCID: PMC10764432 DOI: 10.3389/fimmu.2023.1289313] [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: 09/05/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Epstein-Barr virus (EBV) is a prevalent oncogenic virus estimated to infect greater than 90% of the world's population. Following initial infection, it establishes latency in host B cells. EBV has developed a multitude of techniques to avoid detection by the host immune system and establish lifelong infection. T cells, as important contributors to cell-mediated immunity, make an attractive target for these immunoevasive strategies. Indeed, EBV has evolved numerous mechanisms to modulate T cell responses. For example, it can augment expression of programmed cell death ligand-1 (PD-L1), which inhibits T cell function, and downregulates the interferon response, which has a strong impact on T cell regulation. It also modulates interleukin secretion and can influence major histocompatibility complex (MHC) expression and presentation. In addition to facilitating persistent EBV infection, these immunoregulatory mechanisms have significant implications for evasion of the immune response by tumor cells. This review dissects the mechanisms through which EBV avoids detection by host T cells and discusses how these mechanisms play into tumor survival. It concludes with an overview of cancer treatments targeting T cells in the setting of EBV-associated malignancy.
Collapse
Affiliation(s)
- D. G. Sausen
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | | | | | | |
Collapse
|
48
|
Furukawa Y, Ishii M, Ando J, Ikeda K, Igarashi KJ, Kinoshita S, Azusawa Y, Toyota T, Honda T, Nakanishi M, Ohshima K, Masuda A, Yoshida E, Kitade M, Porteus M, Terao Y, Nakauchi H, Ando M. iPSC-derived hypoimmunogenic tissue resident memory T cells mediate robust anti-tumor activity against cervical cancer. Cell Rep Med 2023; 4:101327. [PMID: 38091985 PMCID: PMC10772465 DOI: 10.1016/j.xcrm.2023.101327] [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: 02/10/2023] [Revised: 07/20/2023] [Accepted: 11/17/2023] [Indexed: 12/22/2023]
Abstract
Functionally rejuvenated human papilloma virus-specific cytotoxic T lymphocytes (HPV-rejTs) generated from induced pluripotent stem cells robustly suppress cervical cancer. However, autologous rejT generation is time consuming, leading to difficulty in treating patients with advanced cancer. Although use of allogeneic HPV-rejTs can obviate this, the major obstacle is rejection by the patient immune system. To overcome this, we develop HLA-A24&-E dual integrated HPV-rejTs after erasing HLA class I antigens. These rejTs effectively suppress recipient immune rejection while maintaining more robust cytotoxicity than original cytotoxic T lymphocytes. Single-cell RNA sequencing performed to gain deeper insights reveal that HPV-rejTs are highly enriched with tissue resident memory T cells, which enhance cytotoxicity against cervical cancer through TGFβR signaling, with increased CD103 expression. Genes associated with the immunological synapse also are upregulated, suggesting that these features promote stronger activation of T cell receptor (TCR) and increased TCR-mediated target cell death. We believe that our work will contribute to feasible "off-the-shelf" T cell therapy with robust anti-cervical cancer effects.
Collapse
Affiliation(s)
- Yoshiki Furukawa
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Midori Ishii
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Jun Ando
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Division of Cell Therapy & Blood Transfusion Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Kazuya Ikeda
- Department of Pediatrics, School of Medicine, Stanford University, 291 Campus Drive, Stanford, CA 94305, USA
| | - Kyomi J Igarashi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Shintaro Kinoshita
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoko Azusawa
- Division of Cell Therapy & Blood Transfusion Medicine, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Tokuko Toyota
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Tadahiro Honda
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Mahito Nakanishi
- TOKIWA-Bio, Inc., Tsukuba Center Inc. (TCI), Building G, 2-1-6 Sengen, Tsukuba, Ibaraki 305-0047, Japan
| | - Koichi Ohshima
- Department of Pathology, School of Medicine, Kurume University, Fukuoka 830-0011, Japan
| | - Ayako Masuda
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Emiko Yoshida
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Mari Kitade
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Matthew Porteus
- Department of Pediatrics, School of Medicine, Stanford University, 291 Campus Drive, Stanford, CA 94305, USA
| | - Yasuhisa Terao
- Department of Obstetrics and Gynecology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| | - Miki Ando
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
49
|
He L, Chen N, Dai L, Peng X. Advances and challenges of immunotherapies in NK/T cell lymphomas. iScience 2023; 26:108192. [PMID: 38026157 PMCID: PMC10651691 DOI: 10.1016/j.isci.2023.108192] [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] [Indexed: 12/01/2023] Open
Abstract
Natural killer (NK)/T cell lymphoma (NKTCL) is a rare subtype of Epstein-Barr virus (EBV)-associated non-Hodgkin lymphoma characterized by poor clinical outcomes. It is more common in East Asian and Latin American countries. Despite the introduction of asparaginase/pegaspargase-based chemotherapy, the prognosis of patients with advanced NKTCL needs to be improved, and few salvage treatment options are available for relapsed/refractory patients who fail chemotherapy. Although many unknowns remain, novel treatment strategies to further improve outcomes are urgently needed. Immunotherapy has emerged and shown favorable antitumor activity in NKTCL, including monoclonal antibodies targeting immune checkpoint inhibitors, other receptors on the cellular membrane, and cellular immunotherapy, which could enhance immune cells attack on tumor cells. In this review, we provide an overview of recent immunotherapy in NKTCL, focusing on programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1), cytotoxic T lymphocyte-associated protein 4 (CTLA-4), chimeric antigen receptor (CAR) T cells, EBV-specific cytotoxic T lymphocytes, immunomodulatory agents, and other targeted agents, as well as the current progress and challenges in the field.
Collapse
Affiliation(s)
- Ling He
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
50
|
Zaffiri L, Chambers ET. Screening and Management of PTLD. Transplantation 2023; 107:2316-2328. [PMID: 36949032 DOI: 10.1097/tp.0000000000004577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Posttransplant lymphoproliferative disorder (PTLD) represents a heterogeneous group of lymphoproliferative diseases occurring in the setting of immunosuppression following hematopoietic stem cells transplant and solid organ transplantation. Despite its overall low incidence, PTLD is a serious complication following transplantation, with a mortality rate as high as 50% in transplant recipients. Therefore, it is important to establish for each transplant recipient a personalized risk evaluation for the development of PTLD based on the determination of Epstein-Barr virus serostatus and viral load following the initiation of immunosuppression. Due to the dynamic progression of PTLD, reflected in the diverse pathological features, different therapeutic approaches have been used to treat this disorder. Moreover, new therapeutic strategies based on the administration of virus-specific cytotoxic T cells have been developed. In this review, we summarize the available data on screening and treatment to suggest a strategy to identify transplant recipients at a higher risk for PTLD development and to review the current therapeutic options for PTLD.
Collapse
Affiliation(s)
- Lorenzo Zaffiri
- Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | | |
Collapse
|