201
|
Abudayyeh A, Lin H, Abdelrahim M, Rondon G, Andersson BS, Martinez CS, Page VD, Tarrand JJ, Kontoyiannis DP, Marin D, Oran B, Olson A, Jones R, Popat U, Champlin RE, Chemaly RF, Shpall EJ, Rezvani K. Development and validation of a risk assessment tool for BKPyV Replication in allogeneic stem cell transplant recipients. Transpl Infect Dis 2020; 22:e13395. [PMID: 32602954 DOI: 10.1111/tid.13395] [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: 04/16/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND BK polymavirus (BKPyV), a member of the family Polyomaviridae, is associated with increased morbidity and mortality in allogeneic stem cell transplant recipients. METHODS In our previous retrospective study of 2477 stem cell transplant patients, BKPyV replication independently predicted chronic kidney disease and poor survival. In this study, using the same cohort, we derived and validated a risk grading system to identify patients at risk of BKPyV replication after transplantation in a user-friendly modality. We used 3 baseline variables (conditioning regimen, HLA match status, and underlying cancer diagnosis) that significantly predicted BKPyV replication in our initial study in a subdistribution hazard model with death as a competing risk. We also developed a nomogram of the hazard model as a visual aid. The AUC of the ROC of the risk-score-only model was 0.65. We further stratified the patients on the basis of risk score into low-, moderate-, and high-risk groups. RESULTS The total risk score was significantly associated with BKPyV replication (P < .0001). At 30 days after transplantation, the low-risk (score ≤ 0) patients had a 9% chance of developing symptomatic BKPyV replication, while the high-risk (score ≥ 8) of the population had 56% of developing BKPyV replication. We validated the risk score using a separate cohort of 1478 patients. The AUC of the ROC of the risk-score-only model was 0.59. Both the total risk score and 3-level risk variable were significantly associated with BKPyV replication in this cohort (P < .0001). CONCLUSIONS This grading system for the risk of symptomatic BKPyV replication may help in early monitoring and intervention to prevent BKPyV-associated morbidity, mortality, and kidney function decline.
Collapse
Affiliation(s)
- Ala Abudayyeh
- Division of Internal Medicine, Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maen Abdelrahim
- Institute of Academic Medicine and Weill Cornell Medical College, Houston Methodist Cancer Center, Houston, TX, USA
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Borje S Andersson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles S Martinez
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Valda D Page
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey J Tarrand
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dimitrios P Kontoyiannis
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betul Oran
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amanda Olson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roy Jones
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roy F Chemaly
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
202
|
van Balen P, Jedema I, van Loenen MM, de Boer R, van Egmond HM, Hagedoorn RS, Hoogstaten C, Veld SAJ, Hageman L, van Liempt PAG, Zwaginga JJ, Meij P, Veelken H, Falkenburg JHF, Heemskerk MHM. HA-1H T-Cell Receptor Gene Transfer to Redirect Virus-Specific T Cells for Treatment of Hematological Malignancies After Allogeneic Stem Cell Transplantation: A Phase 1 Clinical Study. Front Immunol 2020; 11:1804. [PMID: 32973756 PMCID: PMC7468382 DOI: 10.3389/fimmu.2020.01804] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
Graft-vs.-leukemia (GVL) reactivity after HLA-matched allogeneic stem cell transplantation (alloSCT) is mainly mediated by donor T cells recognizing minor histocompatibility antigens (MiHA). If MiHA are targeted that are exclusively expressed on hematopoietic cells of recipient origin, selective GVL reactivity without severe graft-vs.-host-disease (GVHD) may occur. In this phase I study we explored HA-1H TCR gene transfer into T cells harvested from the HA-1H negative stem-cell donor to treat HA-1H positive HLA-A*02:01 positive patients with high-risk leukemia after alloSCT. HA-1H is a hematopoiesis-restricted MiHA presented in HLA-A*02:01. Since we previously demonstrated that donor-derived virus-specific T-cell infusions did not result in GVHD, we used donor-derived EBV and/or CMV-specific T-cells to be redirected by HA-1H TCR. EBV and/or CMV-specific T-cells were purified, retrovirally transduced with HA-1H TCR, and expanded. Validation experiments illustrated dual recognition of viral antigens and HA-1H by HA-1H TCR-engineered virus-specific T-cells. Release criteria included products containing more than 60% antigen-specific T-cells. Patients with high risk leukemia following T-cell depleted alloSCT in complete or partial remission were eligible. HA-1H TCR T-cells were infused 8 and 14 weeks after alloSCT without additional pre-conditioning chemotherapy. For 4/9 included patients no appropriate products could be made. Their donors were all CMV-negative, thereby restricting the production process to EBV-specific T-cells. For 5 patients a total of 10 products could be made meeting the release criteria containing 3–280 × 106 virus and/or HA-1H TCR T-cells. No infusion-related toxicity, delayed toxicity or GVHD occurred. One patient with relapsed AML at time of infusions died due to rapidly progressing disease. Four patients were in remission at time of infusion. Two patients died of infections during follow-up, not likely related to the infusion. Two patients are alive and well without GVHD. In 2 patients persistence of HA-1H TCR T-cells could be illustrated correlating with viral reactivation, but no overt in-vivo expansion of infused T-cells was observed. In conclusion, HA-1H TCR-redirected virus-specific T-cells could be made and safely infused in 5 patients with high-risk AML, but overall feasibility and efficacy was too low to warrant further clinical development using this strategy. New strategies will be explored using patient-derived donor T-cells isolated after transplantation transduced with HA-1H-specific TCR to be infused following immune conditioning.
Collapse
Affiliation(s)
- Peter van Balen
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Inge Jedema
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Renate de Boer
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - H M van Egmond
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Renate S Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Conny Hoogstaten
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Sabrina A J Veld
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Lois Hageman
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - P A G van Liempt
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Jaap-Jan Zwaginga
- Center for Clinical Transfusion Research, Sanquin Research, Leiden, Netherlands.,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Pauline Meij
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - H Veelken
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - J H F Falkenburg
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | |
Collapse
|
203
|
Compagno F, Basso S, Panigari A, Bagnarino J, Stoppini L, Maiello A, Mina T, Zelini P, Perotti C, Baldanti F, Zecca M, Comoli P. Management of PTLD After Hematopoietic Stem Cell Transplantation: Immunological Perspectives. Front Immunol 2020; 11:567020. [PMID: 33042147 PMCID: PMC7526064 DOI: 10.3389/fimmu.2020.567020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/18/2020] [Indexed: 01/07/2023] Open
Abstract
Post-transplant lymphoproliferative disorders (PTLDs) are life-threatening complications of iatrogenic immune impairment after allogeneic hematopoietic stem cell transplantation (HSCT). In the pediatric setting, the majority of PTLDs are related to the Epstein-Barr virus (EBV) infection, and present as B-cell lymphoproliferations. Although considered rare events, PTLDs have been increasingly observed with the widening application of HSCT from alternative sources, including cord blood and HLA-haploidentical stem cell grafts, and the use of novel agents for the prevention and treatment of rejection and graft-vs.-host disease. The higher frequency initially paralleled a poor outcome, due to limited therapeutic options, and scarcity of controlled trials in a rare disease context. In the last 2 decades, insight into the relationship between EBV and the immune system, and advances in early diagnosis, monitoring and treatment have changed the approach to the management of PTLDs after HSCT, and significantly ameliorated the prognosis. In this review, we summarize literature on the impact of combined viro-immunologic assessment on PTLD management, describe the various strategies for PTLD prevention and preemptive/curative treatment, and discuss the potential of novel immune-based therapies in the containment of this malignant complication.
Collapse
Affiliation(s)
- Francesca Compagno
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Sabrina Basso
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Cell Factory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Arianna Panigari
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Jessica Bagnarino
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Cell Factory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Luca Stoppini
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Cell Factory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Alessandra Maiello
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Cell Factory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Tommaso Mina
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Paola Zelini
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Cell Factory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Cesare Perotti
- Immunohematology and Transfusion Service, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Fausto Baldanti
- Virology Service, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Marco Zecca
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Cell Factory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
204
|
Zinter MS, Dvorak CC, Auletta JJ. How We Treat Fever and Hypotension in Pediatric Hematopoietic Cell Transplant Patients. Front Oncol 2020; 10:581447. [PMID: 33042850 PMCID: PMC7526343 DOI: 10.3389/fonc.2020.581447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/24/2020] [Indexed: 11/13/2022] Open
Abstract
Pediatric allogeneic hematopoietic cell transplant (HCT) survival is limited by the development of post-transplant infections. In this overview, we discuss a clinical approach to the prompt recognition and treatment of fever and hypotension in pediatric HCT patients. Special attention is paid to individualized hemodynamic resuscitation, thorough diagnostic testing, novel anti-pathogen therapies, and the multimodal support required for recovery. We present three case vignettes that illustrate the complexities of post-HCT sepsis and highlight best practices that contribute to optimal transplant survival in children.
Collapse
Affiliation(s)
- Matt S Zinter
- Division of Critical Care Medicine, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Christopher C Dvorak
- Division of Allergy, Immunology, and Blood and Marrow Transplantation, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Jeffery J Auletta
- Division of Hematology, Oncology, Blood and Marrow Transplantation, Nationwide Children's Hospital, Columbus, OH, United States.,Division of Infectious Diseases, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
205
|
Basar R, Uprety N, Ensley E, Daher M, Klein K, Martinez F, Aung F, Shanley M, Hu B, Gokdemir E, Mendt M, Silva FR, Acharya S, Laskowski T, Muniz-Feliciano L, Banerjee P, Li Y, Li S, Garcia LM, Lin P, Shaim H, Yates SG, Marin D, Kaur I, Rao S, Mak D, Lin A, Miao Q, Dou J, Chen K, Champlin R, Shpall EJ, Rezvani K. Generation of glucocorticoid resistant SARS-CoV-2 T-cells for adoptive cell therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32995792 DOI: 10.1101/2020.09.15.298547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Adoptive cell therapy with viral-specific T cells has been successfully used to treat life-threatening viral infections, supporting the application of this approach against COVID-19. We expanded SARS-CoV-2 T-cells from the peripheral blood of COVID-19-recovered donors and non-exposed controls using different culture conditions. We observed that the choice of cytokines modulates the expansion, phenotype and hierarchy of antigenic recognition by SARS-CoV-2 T-cells. Culture with IL-2/4/7 but not other cytokine-driven conditions resulted in >1000 fold expansion in SARS-CoV-2 T-cells with a retained phenotype, function and hierarchy of antigenic recognition when compared to baseline (pre-expansion) samples. Expanded CTLs were directed against structural SARS-CoV-2 proteins, including the receptor-binding domain of Spike. SARS-CoV-2 T-cells could not be efficiently expanded from the peripheral blood of non-exposed controls. Since corticosteroids are used for the management of severe COVID-19, we developed an efficient strategy to inactivate the glucocorticoid receptor gene ( NR3C1 ) in SARS-CoV-2 CTLs using CRISPR-Cas9 gene editing.
Collapse
|
206
|
Prockop S, Doubrovina E, Suser S, Heller G, Barker J, Dahi P, Perales MA, Papadopoulos E, Sauter C, Castro-Malaspina H, Boulad F, Curran KJ, Giralt S, Gyurkocza B, Hsu KC, Jakubowski A, Hanash AM, Kernan NA, Kobos R, Koehne G, Landau H, Ponce D, Spitzer B, Young JW, Behr G, Dunphy M, Haque S, Teruya-Feldstein J, Arcila M, Moung C, Hsu S, Hasan A, O'Reilly RJ. Off-the-shelf EBV-specific T cell immunotherapy for rituximab-refractory EBV-associated lymphoma following transplantation. J Clin Invest 2020; 130:733-747. [PMID: 31689242 DOI: 10.1172/jci121127] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/22/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUNDAdoptive transfer of donor-derived EBV-specific cytotoxic T-lymphocytes (EBV-CTLs) can eradicate EBV-associated lymphomas (EBV-PTLD) after transplantation of hematopoietic cell (HCT) or solid organ (SOT) but is unavailable for most patients.METHODSWe developed a third-party, allogeneic, off-the-shelf bank of 330 GMP-grade EBV-CTL lines from specifically consented healthy HCT donors. We treated 46 recipients of HCT (n = 33) or SOT (n = 13) with established EBV-PTLD, who had failed rituximab therapy, with third-party EBV-CTLs. Treatment cycles consisted of 3 weekly infusions of EBV-CTLs and 3 weeks of observation.RESULTSEBV-CTLs did not induce significant toxicities. One patient developed grade I skin graft-versus-host disease. Complete remission (CR) or sustained partial remission (PR) was achieved in 68% of HCT recipients and 54% of SOT recipients. For patients who achieved CR/PR or stable disease after cycle 1, one year overall survival was 88.9% and 81.8%, respectively. In addition, 3 of 5 recipients with POD after a first cycle who received EBV-CTLs from a different donor achieved CR or durable PR (60%) and survived longer than 1 year. Maximal responses were achieved after a median of 2 cycles.CONCLUSIONThird-party EBV-CTLs of defined HLA restriction provide safe, immediately accessible treatment for EBV-PTLD. Secondary treatment with EBV-CTLs restricted by a different HLA allele (switch therapy) can also induce remissions if initial EBV-CTLs are ineffective. These results suggest a promising potential therapy for patients with rituximab-refractory EBV-associated lymphoma after transplantation.TRIAL REGISTRATIONPhase II protocols (NCT01498484 and NCT00002663) were approved by the Institutional Review Board at Memorial Sloan Kettering Cancer Center, the FDA, and the National Marrow Donor Program.FUNDINGThis work was supported by NIH grants CA23766 and R21CA162002, the Aubrey Fund, the Claire Tow Foundation, the Major Family Foundation, the Max Cure Foundation, the Richard "Rick" J. Eisemann Pediatric Research Fund, the Banbury Foundation, the Edith Robertson Foundation, and the Larry Smead Foundation. Atara Biotherapeutics licensed the bank of third-party EBV-CTLs from Memorial Sloan Kettering Cancer Center in June 2015.
Collapse
Affiliation(s)
- Susan Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Ekaterina Doubrovina
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Center for Immune Cellular Therapy
| | - Stephanie Suser
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Juliet Barker
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Parastoo Dahi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Miguel A Perales
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Esperanza Papadopoulos
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Craig Sauter
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Hugo Castro-Malaspina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Farid Boulad
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Kevin J Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Sergio Giralt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Boglarka Gyurkocza
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Katharine C Hsu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Ann Jakubowski
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Alan M Hanash
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Nancy A Kernan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - Rachel Kobos
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Jansen Pharmaceuticals, Raritan, New Jersey, USA
| | - Guenther Koehne
- Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Heather Landau
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Doris Ponce
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| | - James W Young
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Gerald Behr
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark Dunphy
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Julie Teruya-Feldstein
- Department of Pathology, Icahn School of Medicine, Mount Sinai Health System, New York, New York, USA
| | - Maria Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Christine Moung
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Susan Hsu
- American Red Cross, Philadelphia, Pennsylvania, USA
| | - Aisha Hasan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,GlaxoSmithKline, Oncology, Collegeville, Pennsylvania, USA
| | - Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pediatric Hematology/Oncology, New York Presbyterian Hospital, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
207
|
O'Reilly RJ, Prockop S, Hasan A, Doubrovina E. Therapeutic advantages provided by banked virus-specific T-cells of defined HLA-restriction. Bone Marrow Transplant 2020; 54:759-764. [PMID: 31431697 DOI: 10.1038/s41409-019-0614-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We have developed banks of EBV and CMV-specific T-cell lines generated from healthy seropositive third party donors and characterized them as to their HLA type, virus specificity, lack of alloreactivity, and HLA restriction. We here summarize results of studies employing these immediately accessible, broadly-applicable third party virus-specific T-cells for adoptive therapy of EBV lymphomas and CMV infections in allo-HCT recipients. We describe the characteristics contributing to their safety. We also discuss several distinctive advantages of banked third party virus-specific T-cells selected on the basis of their HLA restriction, particularly in the treatment of Rituximab-non-responsive EBV+ lymphomas and drug refractory CMV infections complicating HLA non-identical transplants.
Collapse
Affiliation(s)
- Richard J O'Reilly
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Susan Prockop
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aisha Hasan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ekaterina Doubrovina
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
208
|
"Mini" bank of only 8 donors supplies CMV-directed T cells to diverse recipients. Blood Adv 2020; 3:2571-2580. [PMID: 31481503 DOI: 10.1182/bloodadvances.2019000371] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022] Open
Abstract
Cytomegalovirus (CMV) infections remain a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT), and standard antiviral therapies are associated with significant side effects and development of drug-resistant mutants. Adoptively transferred donor-derived CMV-specific T cells (CMVSTs) can provide an alternative treatment modality with few side effects but are not widely available due to their patient-specific nature. Here we report the establishment and use of a bank of CMVSTs derived from just 8 CMV-seropositive donors, with HLA types representing the diverse US population, as an "off-the-shelf" therapy to treat drug-refractory infections. To date, we have screened 29 patients for study participation and identified a suitable line, with ≥2 of 8 shared HLA antigens, for 28 (96.6%) patients with a median of 4 shared HLA antigens. Of these, 10 patients with persistent/refractory CMV infections or disease were eligible for treatment; a single infusion of cells produced 3 partial responses and 7 complete responses, for a cumulative response rate of 100% (95% confidence interval, 69.2-100) with no graft-versus-host disease, graft failure, or cytokine release syndrome. Potential wider use of the tested CMVSTs across transplant centers is made more feasible by our ability to produce sufficient material to generate cells for >2000 infusions from a single donor collection. Our data indicate that a "mini" bank of CMVSTs prepared from just 8 well-chosen third-party donors can supply the majority of patients with an appropriately matched line that produces safe and effective anti-CMV activity post-HSCT.
Collapse
|
209
|
Fifty Years of JC Polyomavirus: A Brief Overview and Remaining Questions. Viruses 2020; 12:v12090969. [PMID: 32882975 PMCID: PMC7552028 DOI: 10.3390/v12090969] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
In the fifty years since the discovery of JC polyomavirus (JCPyV), the body of research representing our collective knowledge on this virus has grown substantially. As the causative agent of progressive multifocal leukoencephalopathy (PML), an often fatal central nervous system disease, JCPyV remains enigmatic in its ability to live a dual lifestyle. In most individuals, JCPyV reproduces benignly in renal tissues, but in a subset of immunocompromised individuals, JCPyV undergoes rearrangement and begins lytic infection of the central nervous system, subsequently becoming highly debilitating-and in many cases, deadly. Understanding the mechanisms allowing this process to occur is vital to the development of new and more effective diagnosis and treatment options for those at risk of developing PML. Here, we discuss the current state of affairs with regards to JCPyV and PML; first summarizing the history of PML as a disease and then discussing current treatment options and the viral biology of JCPyV as we understand it. We highlight the foundational research published in recent years on PML and JCPyV and attempt to outline which next steps are most necessary to reduce the disease burden of PML in populations at risk.
Collapse
|
210
|
Bush R, Johns F, Betty Z, Goldstein S, Horn B, Shoemaker L, Upadhyay K. BK virus encephalitis and end-stage renal disease in a child with hematopoietic stem cell transplantation. Pediatr Transplant 2020; 24:e13739. [PMID: 32412694 DOI: 10.1111/petr.13739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/09/2020] [Accepted: 04/20/2020] [Indexed: 01/07/2023]
Abstract
BK virus encephalitis after HSCT is uncommon. Several reports of native kidney BKVN in patients with HSCT, hematologic malignancies, human immunodeficiency virus infection, and non-renal solid organ transplantation have been described. However, an uncommon combination of BK encephalitis and ESRD of native kidneys secondary to BK virus in a child with HSCT has not been described. We report a 10-year-old boy who presented with a gradually rising serum creatinine during treatment for severe autoimmune hemolytic anemia, which he developed 9 months after receiving an allogeneic HSCT for aplastic anemia. There was no proteinuria or hematuria present. Serum BK virus load was 5 × 106 copies/mL. A renal biopsy showed evidence of BKVN. He developed fever, seizures, and confusion, and the (CSF) showed significant presence of the BK virus (1 × 106 copies/mL) along with biochemical evidence of viral encephalitis. Cerebrospinal fluid cultures were negative. Despite significant clinical symptoms and presence of BK virus in CSF, the magnetic resonance brain imaging findings were minimal. With reduction of immunosuppression, there was resolution of BK encephalitis but BKVN remained resistant to multiple anti-BK virus agents, including leflunomide and cidofovir. He eventually became dialysis-dependent and, 6 years later, received a renal transplant from his mother. This case illustrates that BK virus in severely immunocompromised HSCT recipient may lead to BK encephalitis and BKVN of native kidneys, even without hemorrhagic cystitis, leading to ESRD. Knowledge of such is important for appropriate timely evaluation and management.
Collapse
Affiliation(s)
- Rachel Bush
- Division of Pediatric Nephrology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Felicia Johns
- Division of Pediatric Nephrology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Zachary Betty
- Division of Pediatric Nephrology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Steven Goldstein
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Biljana Horn
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Lawrence Shoemaker
- Division of Pediatric Nephrology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Kiran Upadhyay
- Division of Pediatric Nephrology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
211
|
Atilla E, Ateş C, Uslu A, Ataca Atilla P, Dolapçı I, Tekeli A, Topçuoğlu P. Prospective Analysis of Hemorrhagic Cystitis and BK Viremia in Allogeneic Hematopoietic Stem Cell Transplantation. Turk J Haematol 2020; 37:186-192. [PMID: 31852035 PMCID: PMC7463211 DOI: 10.4274/tjh.galenos.2019.2019.0296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023] Open
Abstract
Objective BK virus (BKV) infection has been shown to be related to hemorrhagic cystitis (HC) in allogeneic hematopoietic stem cell transplantation (allo-HSCT). There are conflicting data regarding the association between BKV titers in plasma and clinical disease as well as the risk factors for BKV-related HC. Our aim is to study the risk factors and relationship with plasma BK viral load for development of HC in a prospective analysis. Materials and Methods We prospectively evaluated 59 patients who received allo-HSCT between 2014 and 2016 by quantitative BK virus polymerase chain reaction (PCR) (Altona Diagnostics, Germany) from blood samples at days 0, 30, 60, and 90 after allo-HSCT. The patients were monitored for signs and symptoms of HC. Results HC was diagnosed in 22 patients (37%) at a mean of 100 days (range: 0-367 days). In multivariate analysis, the usage of cyclophosphamide (sub-distribution hazard ratio [sdHR]: 7.82, confidence interval [CI]: 1.375-39.645, p=0.02), reactivated CMV (sdHR: 6.105, CI: 1.614-23.094, p=0.008), and positive BKV viremia (sdHR: 2.15, CI: 1.456-22.065, p=0.01) significantly increased the risk of developing HC. Patients with higher viral loads at day 30 and day 60 were diagnosed with more severe HC (p<0.001). Median BK viral loads of >101.5 copies/mL at day 0 (sensitivity 0.727, specificity 0.875), >98.5 copies/mL at day 30 (sensitivity 0.909, specificity 0.875), and >90.0 copies/mL at day 60 (sensitivity 0.909, specificity 0.875) were indicative of HC. Conclusion Our study showed that administration of cyclophosphamide, CMV reactivation, and BK virus positivity were associated with HC. Plasma BK virus PCR titers at days 0, 30, and 60 after transplant were sensitive tools for predicting clinically proven HC.
Collapse
Affiliation(s)
- Erden Atilla
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Can Ateş
- Van Yüzüncü Yıl University Faculty of Medicine, Department of Biostatistics, Van, Turkey
| | - Atilla Uslu
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Pınar Ataca Atilla
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| | - Istar Dolapçı
- Ankara University Faculty of Medicine, Department of Microbiology, Ankara, Turkey
| | - Alper Tekeli
- Ankara University Faculty of Medicine, Department of Microbiology, Ankara, Turkey
| | - Pervin Topçuoğlu
- Ankara University Faculty of Medicine, Department of Hematology, Ankara, Turkey
| |
Collapse
|
212
|
Crowhurst T, Nolan J, Faull R, Holmes M, Holmes-Liew CL. BK virus-associated nephropathy in a lung transplant patient: case report and literature review. BMC Infect Dis 2020; 20:600. [PMID: 32795251 PMCID: PMC7427921 DOI: 10.1186/s12879-020-05292-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/26/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND BK virus-associated nephropathy (BKVAN) is a relatively common cause of renal dysfunction in the first six months after renal transplantation. It arises from reactivation of the latent and usually harmless BK virus (BK virus) due to immunosuppression and other factors including some that are unique to renal transplantation such as allograft injury. BKVAN is much rarer in non-renal solid organ transplantation, where data regarding diagnosis and management are extremely limited. CASE PRESENTATION We report a case of a 58-year-old man found to have worsening renal dysfunction nine months after bilateral sequential lung transplantation for chronic obstructive pulmonary disease (COPD). He had required methylprednisolone for acute allograft rejection but achieved good graft function. Urine microscopy and culture and renal ultrasound were normal. BK virus PCR was positive at high levels in urine and blood. Renal biopsy subsequently confirmed BKVAN. The patient progressed to end-stage renal failure requiring haemodialysis despite reduction in immunosuppression, including switching mycophenolate for everolimus, and the administration of intravenous immunoglobulin (IVIG). CONCLUSIONS This very rare case highlights the challenges presented by BK virus in the non-renal solid organ transplant population. Diagnosis can be difficult, especially given the heterogeneity with which BKV disease has been reported to present in such patients, and the optimal approach to management is unknown. Balancing reduction in immunosuppression against prevention of allograft rejection is delicate. Improved therapeutic options are clearly required.
Collapse
Affiliation(s)
- Thomas Crowhurst
- Discipline of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia.
- SA Lung Transplant Service, Royal Adelaide Hospital, Central Adelaide Local Health Network, 1 Port Road, Adelaide, SA, 5000, Australia.
| | - James Nolan
- SA Pathology, Royal Adelaide Hospital, Central Adelaide Local Health Network, 1 Port Road, Adelaide, SA, 5000, Australia
| | - Randall Faull
- Discipline of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Central Adelaide Local Health Network, 1 Port Road, Adelaide, SA, 5000, Australia
| | - Mark Holmes
- Discipline of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
- SA Lung Transplant Service, Royal Adelaide Hospital, Central Adelaide Local Health Network, 1 Port Road, Adelaide, SA, 5000, Australia
| | - Chien-Li Holmes-Liew
- Discipline of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
- SA Lung Transplant Service, Royal Adelaide Hospital, Central Adelaide Local Health Network, 1 Port Road, Adelaide, SA, 5000, Australia
| |
Collapse
|
213
|
Human herpesvirus 6 in transplant recipients: an update on diagnostic and treatment strategies. Curr Opin Infect Dis 2020; 32:584-590. [PMID: 31567413 DOI: 10.1097/qco.0000000000000592] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW The current review article focuses on recent advances in the approach to the diagnosis and treatment of human herpesvirus 6B (HHV-6B) in hematopoietic cell and solid organ transplant recipients. RECENT FINDINGS Over the past few years, key studies have broadened our understanding of best practices for the prevention and treatment of HHV-6B encephalitis after transplantation. Moreover, important data have been reported that support a potential role of HHV-6B reactivation in the development of acute graft-versus-host disease and lower respiratory tract disease in transplant recipients. Finally, increasing recognition of inherited chromosomally integrated HHV-6 (iciHHV-6) and an expanding array of diagnostic tools have increased our understanding of the potential for complications related to viral reactivation originating from iciHHV-6 in donors or recipients. SUMMARY Recent advances in diagnostic tools, disease associations, and potential treatments for HHV-6B present abundant opportunities for improving our understanding and management of this complex virus in transplant recipients.
Collapse
|
214
|
Adenovirus infection and disease in recipients of hematopoietic cell transplantation. Curr Opin Infect Dis 2020; 32:591-600. [PMID: 31567568 DOI: 10.1097/qco.0000000000000605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW To provide an update on risk factors associated with adenovirus (ADV) infection in patients after hematopoietic cell transplant (HCT) and on options for ADV monitoring and treatment in the setting of HCT. RECENT FINDINGS Among patients undergoing HCT, ADV infection continues to be more common amongst those receiving a T-cell-depleted or graft other than from a matched-related donor. Among children undergoing HCT, reactivation in the gastrointestinal tract appears to be the most common source, and the virus is detectable by quantitative PCR in the stool before it is detectable in the blood. Thus, screening for the virus in the stool of these children may allow for preemptive therapy to reduce mortality. Brincidofovir, although still not approved by any regulatory agency, remains a potential agent for preemptive therapy and for salvage in cases not responding to cidofovir. Rapidly generated off-the-shelf virus-specific T cells may facilitate adoptive cell therapy in populations with a special need and previously not eligible for adoptive cell therapy, such as cord blood recipients. SUMMARY ADV infection continues to adversely affect survival in HCT recipients. Screening stool in children and preemptive therapy may reduce mortality. Brincidofovir and adoptive T-cell therapy remain potential options for treatment.
Collapse
|
215
|
BK polyomavirus-specific antibody and T-cell responses in kidney transplantation: update. Curr Opin Infect Dis 2020; 32:575-583. [PMID: 31567736 DOI: 10.1097/qco.0000000000000602] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW BK polyomavirus (BKPyV) has emerged as a significant cause of premature graft failure after kidney transplantation. Without effective antiviral drugs, treatment is based on reducing immunosuppression to regain immune control over BKPyV replication. The paradigm of high-level viruria/decoy cells, BKPyV-DNAemia, and proven nephropathy permits early interventions. Here, we review recent findings about BKPyV-specific antibody and T-cell responses and their potential role in risk stratification, immune monitoring, and therapy. RECENT FINDING Kidney transplant recipients having low or undetectable BKPyV-specific IgG immunoglobulin G (IgG) are higher risk for developing BKPyV-DNAemia if the donor has high BKPyV-specific IgG. This observation has been extended to neutralizing antibodies. Immunosuppression, impaired activation, proliferation, and exhaustion of BKPyV-specific T cells may increase the risk of developing BKPyV-DNAemia and nephropathy. Clearance of BKPyV-DNAemia was correlated with high CD8 T cell responses to human leukocyte antigen (HLA)-types presenting BKPyV-encoded immunodominant 9mers. For clinical translation, these data need to be assessed in appropriately designed clinical studies, as outlined in recent guidelines on BKPyV in kidney transplantation. SUMMARY Evaluation of BKPyV-specific immune responses in recipient and donor may help to stratify the risk of BKPyV-DNAemia, nephropathy, and graft loss. Future efforts need to evaluate clinical translation, vaccines, and immunotherapy to control BKPyV replication.
Collapse
|
216
|
Shafat MS, Mehra V, Peggs KS, Roddie C. Cellular Therapeutic Approaches to Cytomegalovirus Infection Following Allogeneic Stem Cell Transplantation. Front Immunol 2020; 11:1694. [PMID: 32849591 PMCID: PMC7411136 DOI: 10.3389/fimmu.2020.01694] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/25/2020] [Indexed: 12/31/2022] Open
Abstract
Cytomegalovirus (CMV) infection is common following allogeneic hematopoietic stem cell transplant (HSCT) and is a major cause of morbidity and increased mortality. Whilst pharmacotherapy can be effective in the prevention and treatment of CMV, these agents are often expensive, toxic and in some cases ineffective due to viral resistance mechanisms. Immunotherapeutic approaches are compelling and early clinical trials of adoptively transferred donor-derived virus-specific T (VST) cells against CMV have demonstrated efficacy. However, significant logistical challenges limit their broad application. Strategies to optimize VST manufacture and cell banking alongside scientific developments to enhance efficacy whilst minimizing toxicity are ongoing. This review will discuss the development of CMV-specific T-cell therapies, the challenges of widespread delivery of VSTs for CMV and explore how VST therapy can change outcomes in CMV infection following HSCT.
Collapse
Affiliation(s)
- Manar S Shafat
- Research Department of Haematology, UCL Cancer Institute, University College London, Cancer Institute, London, United Kingdom
| | - Vedika Mehra
- Research Department of Haematology, UCL Cancer Institute, University College London, Cancer Institute, London, United Kingdom
| | - Karl S Peggs
- Research Department of Haematology, UCL Cancer Institute, University College London, Cancer Institute, London, United Kingdom.,Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Claire Roddie
- Research Department of Haematology, UCL Cancer Institute, University College London, Cancer Institute, London, United Kingdom.,Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
217
|
Jiang W, Withers B, Sutrave G, Clancy LE, Yong MI, Blyth E. Pathogen-Specific T Cells Beyond CMV, EBV and Adenovirus. Curr Hematol Malig Rep 2020; 14:247-260. [PMID: 31228095 DOI: 10.1007/s11899-019-00521-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Infectious diseases contribute significantly to morbidity and mortality in recipients of allogeneic haematopoietic stem cell transplantation (aHSCT), particularly in the era of highly immunosuppressive transplant regimens and alternate donor transplants. Delayed cellular immune recovery is a major mechanism for the increased risk in these patients. Adoptive cell therapy with ex vivo manipulated pathogen-specific T cells (PSTs) is increasingly taking its place as a treatment strategy using donor-derived or third party-banked cells. RECENT FINDINGS The majority of clinical trial data in the form of early-phase studies has been in the prophylaxis or treatment of cytomegalovirus (CMV), Epstein-Barr virus (EBV) and adenovirus (AdV). Advancements in methods to select and enrich PSTs offer the opportunity to target the less common viral pathogens as well as fungi with this technology. Early clinical studies of PSTs targeting polyomaviruses (BK virus and JC virus), human herpesvirus 6 (HHV6), varicella zoster virus (VZV) and Aspergillus spp. have shown promising results in small numbers of patients. Other potential targets include herpes simplex virus (HSV), respiratory viruses and other invasive fungal species. In this review, we describe the burden of disease of this wider spectrum of pathogens, the progress in the development of manufacturing capability, early clinical results and the opportunities and challenges for implementation in the clinic.
Collapse
Affiliation(s)
- Wei Jiang
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.,Westmead Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Barbara Withers
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.,Westmead Institute of Medical Research, University of Sydney, Sydney, Australia.,St Vincent's Hospital, Darlinghurst, Australia
| | - Gaurav Sutrave
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.,Westmead Institute of Medical Research, University of Sydney, Sydney, Australia.,BMT and Cell Therapies Program, Westmead Hospital, Sydney, Australia
| | - Leighton E Clancy
- Westmead Institute of Medical Research, University of Sydney, Sydney, Australia.,Sydney Cellular Therapies Laboratory, Westmead, Australia
| | - Michelle I Yong
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.,The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, Australia
| | - Emily Blyth
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia. .,Westmead Institute of Medical Research, University of Sydney, Sydney, Australia. .,St Vincent's Hospital, Darlinghurst, Australia. .,BMT and Cell Therapies Program, Westmead Hospital, Sydney, Australia.
| |
Collapse
|
218
|
Mo F, Watanabe N, McKenna MK, Hicks MJ, Srinivasan M, Gomes-Silva D, Atilla E, Smith T, Ataca Atilla P, Ma R, Quach D, Heslop HE, Brenner MK, Mamonkin M. Engineered off-the-shelf therapeutic T cells resist host immune rejection. Nat Biotechnol 2020; 39:56-63. [PMID: 32661440 PMCID: PMC7854790 DOI: 10.1038/s41587-020-0601-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Feiyan Mo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Mary K McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - M John Hicks
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Madhuwanti Srinivasan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Diogo Gomes-Silva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Erden Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Tyler Smith
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Pinar Ataca Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Royce Ma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA.,Graduate Program in Immunology, Baylor College of Medicine, Houston, TX, USA
| | - David Quach
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA. .,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA. .,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA. .,Graduate Program in Immunology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
219
|
Safety and feasibility of virus-specific T cells derived from umbilical cord blood in cord blood transplant recipients. Blood Adv 2020; 3:2057-2068. [PMID: 31292125 DOI: 10.1182/bloodadvances.2019000201] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/03/2019] [Indexed: 01/09/2023] Open
Abstract
Adoptive transfer of virus-specific T cells (VSTs) has been shown to be safe and effective in stem cell transplant recipients. However, the lack of virus-experienced T cells in donor cord blood (CB) has prevented the development of ex vivo expanded donor-derived VSTs for recipients of this stem cell source. Here we evaluated the feasibility and safety of ex vivo expansion of CB T cells from the 20% fraction of the CB unit in pediatric patients receiving a single CB transplant (CBT). In 2 clinical trials conducted at 2 separate sites, we manufactured CB-derived multivirus-specific T cells (CB-VSTs) targeting Epstein-Barr virus (EBV), adenovirus, and cytomegalovirus (CMV) for 18 (86%) of 21 patients demonstrating feasibility. Manufacturing for 2 CB-VSTs failed to meet lot release because of insufficient cell recovery, and there was 1 sterility breach during separation of the frozen 20% fraction. Delayed engraftment was not observed in patients who received the remaining 80% fraction for the primary CBT. There was no grade 3 to 4 acute graft-versus-host disease (GVHD) associated with the infusion of CB-VSTs. None of the 7 patients who received CB-VSTs as prophylaxis developed end-organ disease from CMV, EBV, or adenovirus. In 7 patients receiving CB-VSTs for viral reactivation or infection, only 1 patient developed end-organ viral disease, which was in an immune privileged site (CMV retinitis) and occurred after steroid therapy for GVHD. Finally, we demonstrated the long-term persistence of adoptively transferred CB-VSTs using T-cell receptor-Vβ clonotype tracking, suggesting that CB-VSTs are a feasible addition to antiviral pharmacotherapy.
Collapse
|
220
|
McGuire HM, Rizzetto S, Withers BP, Clancy LE, Avdic S, Stern L, Patrick E, Fazekas de St Groth B, Slobedman B, Gottlieb DJ, Luciani F, Blyth E. Mass cytometry reveals immune signatures associated with cytomegalovirus (CMV) control in recipients of allogeneic haemopoietic stem cell transplant and CMV-specific T cells. Clin Transl Immunology 2020; 9:e1149. [PMID: 32642063 PMCID: PMC7332355 DOI: 10.1002/cti2.1149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/22/2022] Open
Abstract
Objectives Cytomegalovirus (CMV) is known to have a significant impact on immune recovery post‐allogeneic haemopoietic stem cell transplant (HSCT). Adoptive therapy with donor‐derived or third‐party virus‐specific T cells (VST) can restore CMV immunity leading to clinical benefit in prevention and treatment of post‐HSCT infection. We developed a mass cytometry approach to study natural immune recovery post‐HSCT and assess the mechanisms underlying the clinical benefits observed in recipients of VST. Methods A mass cytometry panel of 38 antibodies was utilised for global immune assessment (72 canonical innate and adaptive immune subsets) in HSCT recipients undergoing natural post‐HSCT recovery (n = 13) and HSCT recipients who received third‐party donor‐derived CMV‐VST as salvage for unresponsive CMV reactivation (n = 8). Results Mass cytometry identified distinct immune signatures associated with CMV characterised by a predominance of innate cells (monocytes and NK) seen early and an adaptive signature with activated CD8+ T cells seen later. All CMV‐VST recipients had failed standard antiviral pharmacotherapy as a criterion for trial involvement; 5/8 had failed to develop the adaptive immune signature by study enrolment despite significant CMV antigen exposure. Of these, VST administration resulted in development of the adaptive signature in association with CMV control in three patients. Failure to respond to CMV‐VST in one patient was associated with persistent absence of the adaptive immune signature. Conclusion The clinical benefit of CMV‐VST may be mediated by the recovery of an adaptive immune signature characterised by activated CD8+ T cells.
Collapse
Affiliation(s)
- Helen M McGuire
- Ramaciotti Facility for Human Systems Biology The University of Sydney Sydney NSW Australia.,Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Discipline of Pathology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - Simone Rizzetto
- Kirby Institute for Infection and Immunity University of New South Wales Sydney NSW Australia.,School of Medical Sciences University of New South Wales Kensington NSW Australia
| | - Barbara P Withers
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,St Vincent's Hospital Darlinghurst NSW Australia
| | - Leighton E Clancy
- Sydney Cellular Therapies Laboratory Westmead NSW Australia.,BMT and Cell Therapies Program Westmead Hospital Sydney NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| | - Selmir Avdic
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| | - Lauren Stern
- Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Discipline of Infectious Diseases and Immunology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - Ellis Patrick
- Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia.,School of Mathematics and Statistics Faculty of Science The University of Sydney Sydney NSW Australia
| | - Barbara Fazekas de St Groth
- Ramaciotti Facility for Human Systems Biology The University of Sydney Sydney NSW Australia.,Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Discipline of Pathology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - Barry Slobedman
- Charles Perkins Centre The University of Sydney Sydney NSW Australia.,Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Discipline of Infectious Diseases and Immunology Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia
| | - David J Gottlieb
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,BMT and Cell Therapies Program Westmead Hospital Sydney NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| | - Fabio Luciani
- Kirby Institute for Infection and Immunity University of New South Wales Sydney NSW Australia.,School of Medical Sciences University of New South Wales Kensington NSW Australia
| | - Emily Blyth
- Faculty of Medicine and Health The University of Sydney Camperdown NSW Australia.,Sydney Cellular Therapies Laboratory Westmead NSW Australia.,BMT and Cell Therapies Program Westmead Hospital Sydney NSW Australia.,Westmead Institute for Medical Research The University of Sydney Sydney NSW Australia
| |
Collapse
|
221
|
Bennett KM, Fernandes PM. Novel treatments for progressive multifocal leukoencephalopathy. Br J Hosp Med (Lond) 2020; 81:1-9. [PMID: 32730143 DOI: 10.12968/hmed.2020.0093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Progressive multifocal leukoencephalopathy is a rare demyelinating disorder of the CNS, caused by John Cunningham virus, that occurs in those with impaired immune systems. Existing treatment options are ineffective or unproven. This article reviews research into novel therapies: immune checkpoint-blocking antibodies (nivolumab and pembrolizumab), allogenic BK virus-specific T cell treatment and filgrastim. Results for these therapies in small clinical trials are promising, but further research is required to assess efficacy fully.
Collapse
Affiliation(s)
- Karina M Bennett
- Department of Neurology, NHS Lothian, Western General Hospital, Edinburgh, UK
| | - Peter M Fernandes
- Department of Neurology, NHS Lothian, Western General Hospital, Edinburgh, UK.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
222
|
Wu HL, Weber WC, Shriver-Munsch C, Swanson T, Northrup M, Price H, Armantrout K, Robertson-LeVay M, Reed JS, Bateman KB, Mahyari E, Thomas A, Junell SL, Hobbs TR, Martin LD, MacAllister R, Bimber BN, Slifka MK, Legasse AW, Moats C, Axthelm MK, Smedley J, Lewis AD, Colgin L, Meyers G, Maziarz RT, Burwitz BJ, Stanton JJ, Sacha JB. Viral opportunistic infections in Mauritian cynomolgus macaques undergoing allogeneic stem cell transplantation mirror human transplant infectious disease complications. Xenotransplantation 2020; 27:e12578. [PMID: 31930750 PMCID: PMC7354885 DOI: 10.1111/xen.12578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/11/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) and xenotransplantation are accompanied by viral reactivations and virus-associated complications resulting from immune deficiency. Here, in a Mauritian cynomolgus macaque model of fully MHC-matched allogeneic HSCT, we report reactivations of cynomolgus polyomavirus, lymphocryptovirus, and cytomegalovirus, macaque viruses analogous to HSCT-associated human counterparts BK virus, Epstein-Barr virus, and human cytomegalovirus. Viral replication in recipient macaques resulted in characteristic disease manifestations observed in HSCT patients, such as polyomavirus-associated hemorrhagic cystitis and tubulointerstitial nephritis or lymphocryptovirus-associated post-transplant lymphoproliferative disorder. However, in most cases, the reconstituted immune system, alone or in combination with short-term pharmacological intervention, exerted control over viral replication, suggesting engraftment of functional donor-derived immunity. Indeed, the donor-derived reconstituted immune systems of two long-term engrafted HSCT recipient macaques responded to live attenuated yellow fever 17D vaccine (YFV 17D) indistinguishably from untransplanted controls, mounting 17D-targeted neutralizing antibody responses and clearing YFV 17D within 14 days. Together, these data demonstrate that this macaque model of allogeneic HSCT recapitulates clinical situations of opportunistic viral infections in transplant patients and provides a pre-clinical model to test novel prophylactic and therapeutic modalities.
Collapse
Affiliation(s)
- Helen L. Wu
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | | | - Tonya Swanson
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Mina Northrup
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Heidi Price
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Kimberly Armantrout
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | | | - Jason S. Reed
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Katherine B. Bateman
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Eisa Mahyari
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Archana Thomas
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Stephanie L. Junell
- Divison of Medical Physics, Department of Radiation Medicine, Oregon Health & Science University, Portland, OR Vaccine and Gene Therapy Institute, Oregon Health
| | - Theodore R. Hobbs
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Lauren D. Martin
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Rhonda MacAllister
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Benjamin N. Bimber
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Alfred W. Legasse
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Cassandra Moats
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Michael K. Axthelm
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Anne D. Lewis
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Lois Colgin
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Gabrielle Meyers
- Divison of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Richard T. Maziarz
- Divison of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Benjamin J. Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jeffrey J. Stanton
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jonah B. Sacha
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| |
Collapse
|
223
|
Saade A, Styczynski J, Cesaro S. BK virus infection in allogeneic hematopoietic cell transplantation: An update on pathogenesis, immune responses, diagnosis and treatments. J Infect 2020; 81:372-382. [PMID: 32526327 DOI: 10.1016/j.jinf.2020.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 12/15/2022]
Abstract
In hematopoietic cell transplantation (HCT) patients, BK polyomavirus (BKPyV) infection results in significant morbidity mainly due to hemorrhagic cystitis (HC). Despite increased knowledge acquired over recent decades, no treatment has shown effectiveness in the management of organ damage in HCT allografts. This review summarizes the current knowledge on BKPyV, from the virus constitution to the pathophysiology and immune-related mechanisms. We next focus on BKPyV-induced HC in HCT to discuss the benefit of monitoring BKPyV viruria and viremia in the management of patients. At last, we review currently used therapeutics, along with future promising therapies to propose clinical and practical guidelines and further interesting research areas.
Collapse
Affiliation(s)
- Anastasia Saade
- Department of Hematology, Ponchaillou, Centre Hospitalier Universitaire de Rennes, France.
| | - Jan Styczynski
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland
| | - Simone Cesaro
- Pediatric Hematology Oncology, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| | | |
Collapse
|
224
|
Yu F, Jia R, Tang Y, Liu J, Wei B. SARS-CoV-2 infection and stem cells: Interaction and intervention. Stem Cell Res 2020; 46:101859. [PMID: 32570174 PMCID: PMC7263221 DOI: 10.1016/j.scr.2020.101859] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 02/08/2023] Open
Abstract
The emergence of the novel severe acute respiratory coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread have created a global health emergency. The resemblance with SARS-CoV in spike protein suggests that SARS-CoV-2 employs spike-driven entry into angiotensin-converting enzyme 2 (ACE2)-expressing cells. From a stem cell perspective, this review focuses on the possible involvement of ACE2+ stem/progenitor cells from both the upper and lower respiratory tracts in coronavirus infection. Viral infection-associated acute respiratory distress syndrome and acute lung injury occur because of dysregulation of the immune response. Mesenchymal stem cells appear to be a promising cell therapy given that they favorably modulate the immune response to reduce lung injury. The use of exogenous stem cells may lead to lung repair. Therefore, intervention by transplantation of exogenous stem cells may be required to replace, repair, remodel, and regenerate lung tissue in survivors infected with coronavirus. Ultimately, vaccines, natural killer cells and induced-pluripotent stem cell-derived virus-specific cytotoxic T lymphocytes may offer off-the-shelf therapeutics for preventing coronavirus reemergence.
Collapse
Affiliation(s)
- Fenggang Yu
- Institute of Life Science, Yingfeng Bilogical Group, Jinan, Shandong Province, China; Institute for Advanced Interdisciplinary Researc (iAIR), University of Jinan, Jinan 250022, China.
| | - Rufu Jia
- Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Yongyong Tang
- Yinfeng Dingcheng Bioengineering and Technology Ltd, Beijing, China
| | - Jin Liu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Benjie Wei
- Institute of Life Science, Yingfeng Bilogical Group, Jinan, Shandong Province, China
| |
Collapse
|
225
|
Hanley B, Roufosse CA, Osborn M, Naresh KN. Convalescent donor SARS-COV-2-specific cytotoxic T lymphocyte infusion as a possible treatment option for COVID-19 patients with severe disease has not received enough attention till date. Br J Haematol 2020; 189:1062-1063. [PMID: 32369628 DOI: 10.1111/bjh.16780] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Brian Hanley
- Department of Cellular & Molecular Pathology, Northwest London Pathology, Imperial College Healthcare NHS Trust & Department of Immunology & Inflammation, Imperial College London, London, UK
| | - Candice A Roufosse
- Department of Cellular & Molecular Pathology, Northwest London Pathology, Imperial College Healthcare NHS Trust & Department of Immunology & Inflammation, Imperial College London, London, UK
| | - Michael Osborn
- Department of Cellular & Molecular Pathology, Northwest London Pathology, Imperial College Healthcare NHS Trust & Department of Immunology & Inflammation, Imperial College London, London, UK
| | - Kikkeri N Naresh
- Department of Cellular & Molecular Pathology, Northwest London Pathology, Imperial College Healthcare NHS Trust & Department of Immunology & Inflammation, Imperial College London, London, UK
| |
Collapse
|
226
|
Aftab BT, Sasu B, Krishnamurthy J, Gschweng E, Alcazer V, Depil S. Toward “off‐the‐shelf” allogeneic CAR T cells. ACTA ACUST UNITED AC 2020. [DOI: 10.1002/acg2.86] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Barbra Sasu
- Allogene Therapeutics South San Francisco CA USA
| | | | | | | | - Stéphane Depil
- Centre de Recherche en Cancérologie de Lyon Lyon France
- Centre Léon Bérard Lyon France
- Université Claude Bernard Lyon 1 Lyon France
| |
Collapse
|
227
|
Naik S, Vasileiou S, Aguayo-Hiraldo P, Mukhi S, Sasa G, Martinez C, Krance RA, Gottschalk S, Leen A. Toward Functional Immune Monitoring in Allogeneic Stem Cell Transplant Recipients. Biol Blood Marrow Transplant 2020; 26:911-919. [PMID: 31927102 DOI: 10.1016/j.bbmt.2020.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/06/2020] [Accepted: 01/06/2020] [Indexed: 12/28/2022]
Abstract
Serious viral infections, due to delayed immune reconstitution, are a leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). Thus, many transplant centers prospectively track cellular immune recovery by evaluating absolute cell numbers and the phenotypic profile of reconstituting T cell subsets to identify individuals who are at highest risk of infection. Conventional assessments, however, fail to measure either the antigen specificity or functional capacity of reconstituting cells-both factors that correlate with endogenous antiviral protection. In this pilot study, we sought to address this limitation by prospectively investigating the tempo of endogenous immune reconstitution in a cohort of 23 pediatric HSCT patients using both quantitative (flow cytometry) and qualitative (IFNγ ELISpot) measures, which we correlated with either the presence or absence of infections associated with cytomegalovirus, adenovirus, Epstein-Barr virus, BK virus, human herpes virus 6, respiratory syncytial virus, parainfluenza, influenza, and human metapneumovirus. We present data spanning 12 months post-transplant demonstrating the influence of conditioning on immune recovery and highlighting the differential impact of active viral replication on the quantity and quality of reconstituting cells. Judicious use of standard (phenotypic) and novel (functional) monitoring strategies can help guide the clinical care and personalized management of allogenic HSCT recipients with infections.
Collapse
Affiliation(s)
- Swati Naik
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, and Medicine Baylor College of Medicine, Houston, Texas.
| | - Spyridoula Vasileiou
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Paibel Aguayo-Hiraldo
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, and Medicine Baylor College of Medicine, Houston, Texas
| | - Shivani Mukhi
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Ghadir Sasa
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, and Medicine Baylor College of Medicine, Houston, Texas
| | - Caridad Martinez
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, and Medicine Baylor College of Medicine, Houston, Texas
| | - Robert A Krance
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, and Medicine Baylor College of Medicine, Houston, Texas
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, and Medicine Baylor College of Medicine, Houston, Texas
| | - Ann Leen
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston Methodist Hospital, Houston, Texas; Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, and Medicine Baylor College of Medicine, Houston, Texas
| |
Collapse
|
228
|
Maryamchik E, Gallagher KME, Preffer FI, Kadauke S, Maus MV. New directions in chimeric antigen receptor T cell [CAR-T] therapy and related flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:299-327. [PMID: 32352629 DOI: 10.1002/cyto.b.21880] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor (CAR) T cells provide a promising approach to the treatment of hematologic malignancies and solid tumors. Flow cytometry is a powerful analytical modality, which plays an expanding role in all stages of CAR T therapy, from lymphocyte collection, to CAR T cell manufacturing, to in vivo monitoring of the infused cells and evaluation of their function in the tumor environment. Therefore, a thorough understanding of the new directions is important for designing and implementing CAR T-related flow cytometry assays in the clinical and investigational settings. However, the speed of new discoveries and the multitude of clinical and preclinical trials make it challenging to keep up to date in this complex field. In this review, we summarize the current state of CAR T therapy, highlight the areas of emergent research, discuss applications of flow cytometry in modern cell therapy, and touch upon several considerations particular to CAR detection and assessing the effectiveness of CAR T therapy.
Collapse
Affiliation(s)
- Elena Maryamchik
- Department of Pathology and Laboratory Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Frederic I Preffer
- Clinical Cytometry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephan Kadauke
- Department of Pathology and Laboratory Medicine, Cell and Gene Therapy Laboratory, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marcela V Maus
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Cellular Immunotherapy Program, Department of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
229
|
Weber EW, Maus MV, Mackall CL. The Emerging Landscape of Immune Cell Therapies. Cell 2020; 181:46-62. [PMID: 32243795 PMCID: PMC8900215 DOI: 10.1016/j.cell.2020.03.001] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022]
Abstract
Cell therapies present an entirely new paradigm in drug development. Within this class, immune cell therapies are among the most advanced, having already demonstrated definitive evidence of clinical benefits in cancer and infectious disease. Numerous features distinguish these "living therapies" from traditional medicines, including their ability to expand and contract in proportion to need and to mediate therapeutic benefits for months or years following a single application. Continued advances in fundamental immunology, genetic engineering, gene editing, and synthetic biology exponentially expand opportunities to enhance the sophistication of immune cell therapies, increasing potency and safety and broadening their potential for treatment of disease. This perspective will summarize the current status of immune cell therapies for cancer, infectious disease, and autoimmunity, and discuss advances in cellular engineering to overcome barriers to progress.
Collapse
Affiliation(s)
- Evan W Weber
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
230
|
Arroyo J, Pello O. Adoptive immunotherapy with antiviral T cells: Materials and methods. Rev Clin Esp 2020. [DOI: 10.1016/j.rceng.2019.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
231
|
Abstract
PURPOSE OF REVIEW CMV DNA polymerase inhibitors such as ganciclovir and foscarnet have dramatically reduced the burden of CMV infection in the HCT recipient. However, their use is often limited by toxicities and resistance. Agents with novel mechanisms and favorable toxicity profiles are critically needed. We review recent developments in CMV antivirals and immune-based approaches to mitigating CMV infection. RECENT FINDINGS Letermovir, an inhibitor of the CMV terminase complex, was approved in 2017 for primary CMV prophylaxis in adult seropositive allogeneic HCT recipients. Maribavir, an inhibitor of the CMV UL97 kinase, is currently in two phase 3 treatment studies. Adoptive immunotherapy using third-party T cells has proven safe and effective in preliminary studies. Vaccine development continues, with several promising candidates currently under study. No longer limited to DNA polymerase inhibitors, the prevention and treatment of CMV infections in the HCT recipient is a rapidly evolving field which should translate into improvements in CMV-related outcomes.
Collapse
Affiliation(s)
- Morgan Hakki
- Division of Infectious Diseases, Department of Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Mail code L457, Portland, OR, 97239, USA.
| |
Collapse
|
232
|
HHV-6-Associated Neurological Disease in Children: Epidemiologic, Clinical, Diagnostic, and Treatment Considerations. Pediatr Neurol 2020; 105:10-20. [PMID: 31932119 DOI: 10.1016/j.pediatrneurol.2019.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/12/2019] [Accepted: 10/17/2019] [Indexed: 02/04/2023]
Abstract
Human herpesviruses 6A and 6B, often referred to collectively as human herpesvirus 6, are a pair of beta-herpesviruses known to cause a variety of clinical syndromes in both immunocompetent and immunocompromised individuals. Most humans are infected with human herpesvirus 6B, and many with human herpesvirus 6A. Primary infection typically occurs in early childhood, although large-scale reviews on the topic are limited. Herein, the authors explore the clinical manifestations of human herpesvirus 6-associated disease in both immunocompetent and immunocompromised pediatric patients, the risk factors for development of human herpesvirus 6-associated neurological disease, the risk of autoimmunity associated with development of active or latent infection, the relevance of human herpesvirus 6-specific diagnostic tests, and the medications used to treat human herpesvirus 6. The goal of this review is to improve the current understanding of human herpesvirus 6 in pediatric populations and to examine the most effective diagnostic and therapeutic interventions in this disease state.
Collapse
|
233
|
Nelson RC, Fellowes V, Jin P, Liu H, Highfill SL, Ren J, Szymanski J, Flegel WA, Stroncek DF. Group O plasma as a media supplement for CAR-T cells and other adoptive T-cell therapies. Transfusion 2020; 60:1004-1014. [PMID: 32167176 DOI: 10.1111/trf.15745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Most chimeric antigen receptor T (CAR-T) cells and other adoptive T-cell therapies (ACTs) are currently manufactured by ex vivo expansion of patient lymphocytes in culture media supplemented with human plasma from group AB donors. As lymphocytes do not express A or B antigens, the isoagglutinins of non-AB plasmas are unlikely to cause deleterious effects on lymphocytes in culture. STUDY DESIGN AND METHODS Seeding cultures with peripheral blood mononuclear cell (PBMNC) concentrates from group A1 donors and using a CAR-T culture protocol, parallel cultures were performed, each with unique donor plasmas as media supplements (including group O plasmas with high-titer anti-A and group AB plasmas as control). An additional variable, a 3% group A1 red blood cell (RBC) spike, was added to simulate a RBC-contaminated PBMNC collection. Cultures were monitored by cell count, viability, flow cytometric phenotype, gene expression analysis, and supernatant chemokine analysis. RESULTS There was no difference in lymphocyte expansion or phenotype when cultured with AB plasma or O plasma with high-titer anti-A. Compared to controls, the presence of contaminating RBCs in lymphocyte culture led to poor lymphocyte expansion and a less desirable phenotype-irrespective of the isoagglutinin titer of the plasma supplement used. CONCLUSIONS This study suggests that ABO incompatible plasma may be used as a media supplement when culturing cell types that do not express ABO antigens-such as lymphocytes for CAR-T or other ACT. The presence of contaminating RBCs in culture was disadvantageous independent of isoagglutinin titer.
Collapse
Affiliation(s)
- Randin C Nelson
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland.,Department of Pathology, Montefiore Medical Center & Albert Einstein College of Medicine, Bronx, New York
| | - Vicki Fellowes
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Ping Jin
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Hui Liu
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Steven L Highfill
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Jiaqiang Ren
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - James Szymanski
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland.,Department of Pathology, Montefiore Medical Center & Albert Einstein College of Medicine, Bronx, New York
| | - Willy A Flegel
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - David F Stroncek
- Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland.,Center for Cellular Engineering, National Institutes of Health Clinical Center, Bethesda, Maryland
| |
Collapse
|
234
|
Hong J, Ni J, Ruan M, Yang M, Dong Q, Li Q. LMP1-specific cytotoxic T cells for the treatment of EBV-related post-transplantation lymphoproliferative disorders. Int J Hematol 2020; 111:851-857. [PMID: 32162095 DOI: 10.1007/s12185-020-02844-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022]
Abstract
Epstein-Barr virus-specific cytotoxic T lymphocytes (EBV-CTLs) represent a promising treatment option for EBV-associated post-transplantation lymphoproliferative disorders (PTLD). However, production of EBV-CTLs is often complicated and expensive. In the present study, we sought to establish an easy-to-use and economical production protocol for EBV-CTLs. EBV-CTLs were generated using latent membrane protein 1 (LMP1) peptides based on a modified generation protocol of cytokine-induced killer (CIK) cells. After 2-week culture, cells were well expanded (median total cell number: 9.82 × 109; median expansion fold: 107.8) and the median EBV LMP1-specific CD8+ T cell number was 8.94 × 108 (median frequency: 6.7%). However, the EBV-CTL products, unlike CIK cells, did not exhibit NK-like anti-tumor activity. Furthermore, the clinical efficacy of EBV-CTLs was demonstrated with a successful treatment of PTLD on a compassionate use basis in a patient following haploidentical hematopoietic stem cell transplantation. This study indicates the safety and efficacy of EBV LMP1-specific CTLs generated based on a modified generation protocol of CIK cells. Further investigation in a well-designed clinical study is warranted.
Collapse
Affiliation(s)
- Jian Hong
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Jixi Road No. 218, Hefei, 230022, Anhui, China
| | - Jing Ni
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Jixi Road No. 218, Hefei, 230022, Anhui, China
| | - Min Ruan
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Jixi Road No. 218, Hefei, 230022, Anhui, China
| | - Mingzhen Yang
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Jixi Road No. 218, Hefei, 230022, Anhui, China
| | | | - Qingsheng Li
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Jixi Road No. 218, Hefei, 230022, Anhui, China.
| |
Collapse
|
235
|
Rubinstein JD, Burns K, Absalon M, Lutzko C, Leemhuis T, Chandra S, Hanley PJ, Keller MD, Davies SM, Nelson A, Grimley M. EBV-directed viral-specific T-lymphocyte therapy for the treatment of EBV-driven lymphoma in two patients with primary immunodeficiency and DNA repair defects. Pediatr Blood Cancer 2020; 67:e28126. [PMID: 31850668 DOI: 10.1002/pbc.28126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/11/2019] [Accepted: 11/26/2019] [Indexed: 11/12/2022]
Abstract
Children with ataxia telangiectasia (AT), a primary immunodeficiency caused by mutations in ATM, which is critical for repairing DNA defects, are at risk for the development of hematologic malignancy, frequently driven by infection with Epstein-Barr virus (EBV). Conventional chemotherapy is poorly tolerated by patients with AT, with excessive toxicity even when doses are reduced. Here, we report on two patients with AT and EBV-positive neoplasms who were treated with EBV-targeted viral-specific T cells (VST). One patient had a prolonged complete response to VSTs while the other had a partial response. Therapy was well tolerated without infusion toxicity or graft-versus-host disease.
Collapse
Affiliation(s)
- Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Karen Burns
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Oncology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael Absalon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Oncology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Carolyn Lutzko
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Experimental Hematology, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tom Leemhuis
- Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio
| | - Sharat Chandra
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Health System and Department of Pediatrics, The George Washington University, Washington, District of Columbia
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children's National Health System and Department of Pediatrics, The George Washington University, Washington, District of Columbia
| | - Stella M Davies
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam Nelson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael Grimley
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
236
|
Keller MD, Bollard CM. Virus-specific T-cell therapies for patients with primary immune deficiency. Blood 2020; 135:620-628. [PMID: 31942610 PMCID: PMC7046606 DOI: 10.1182/blood.2019000924] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023] Open
Abstract
Viral infections are common and are potentially life-threatening in patients with moderate to severe primary immunodeficiency disorders. Because T-cell immunity contributes to the control of many viral pathogens, adoptive immunotherapy with virus-specific T cells (VSTs) has been a logical and effective way of combating severe viral disease in immunocompromised patients in multiple phase 1 and 2 clinical trials. Common viral targets include cytomegalovirus, Epstein-Barr virus, and adenovirus, though recent published studies have successfully targeted additional pathogens, including HHV6, BK virus, and JC virus. Though most studies have used VSTs derived from allogenic stem cell donors, the use of banked VSTs derived from partially HLA-matched donors has shown efficacy in multicenter settings. Hence, this approach could shorten the time for patients to receive VST therapy thus improving accessibility. In this review, we discuss the usage of VSTs for patients with primary immunodeficiency disorders in clinical trials, as well as future potential targets and methods to broaden the applicability of virus-directed T-cell immunotherapy for this vulnerable patient population.
Collapse
Affiliation(s)
- Michael D Keller
- Center for Cancer and Immunology Research and
- Division of Allergy and Immunology, Children's National Health System, Washington, DC
- GW Cancer Center, George Washington University, Washington, DC; and
| | - Catherine M Bollard
- Center for Cancer and Immunology Research and
- GW Cancer Center, George Washington University, Washington, DC; and
- Division of Blood and Marrow Transplantation, Children's National Health System, Washington, DC
| |
Collapse
|
237
|
Grau-Vorster M, López-Montañés M, Cantó E, Vives J, Oliver-Vila I, Barba P, Querol S, Rudilla F. Characterization of a Cytomegalovirus-Specific T Lymphocyte Product Obtained Through a Rapid and Scalable Production Process for Use in Adoptive Immunotherapy. Front Immunol 2020; 11:271. [PMID: 32161589 PMCID: PMC7052482 DOI: 10.3389/fimmu.2020.00271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Immunosuppressed patients are susceptible to virus reactivation or de novo infection. Adoptive immunotherapy, based on virus-specific T lymphocytes (VST), can prevent or treat viral diseases. However, donor availability, HLA-compatibility restrictions, high costs, and time required for the production of personalized medicines constitute considerable limitations to this treatment. Ex vivo rapid and large-scale expansion of VST, compliant with current good manufacturing practice (cGMP) standards, with an associated cell donor registry would overcome these limitations. This study aimed to characterize a VST product obtained through an expansion protocol transferable to cGMP standards. Antigenic stimulus consisted of cytomegalovirus (CMV) pp65 peptide pool-pulsed autologous dendritic cells (DCs) derived from monocytes. G-Rex technology, cytokines IL-2, IL-7, and IL-15, and anti-CD3 and anti-CD28 antibodies were used for culture. At day 14 of cell culture, the final product was characterized regarding T cell subsets, specificity, and functionality. The final product, comprised mainly CD4+ and CD8+ T lymphocytes (49.2 ± 24.7 and 42.3 ± 25.2, respectively). The culture conditions made it possible to achieve at least a 98.89-fold increase in pp65-specific CD3+ IFN-γ+ cells. These cells were specific, as pp65-specific cytotoxicity was demonstrated. Additionally, in complete HLA mismatch and without the presence of pp65, alloreactivity resulted in <5% cell lysis. In conclusion, a cGMP scalable process for the generation of a large number of doses of CMV-specific cytotoxic T cells was successfully performed.
Collapse
Affiliation(s)
- Marta Grau-Vorster
- Cell Therapy Service, Banc de Sang i Teixits, Barcelona, Spain.,Transfusion Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María López-Montañés
- Cell Therapy Service, Banc de Sang i Teixits, Barcelona, Spain.,Transfusion Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ester Cantó
- Cell Therapy Service, Banc de Sang i Teixits, Barcelona, Spain.,Transfusion Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquim Vives
- Cell Therapy Service, Banc de Sang i Teixits, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.,Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Pere Barba
- Hematology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sergi Querol
- Cell Therapy Service, Banc de Sang i Teixits, Barcelona, Spain
| | - Francesc Rudilla
- Cell Therapy Service, Banc de Sang i Teixits, Barcelona, Spain.,Transfusion Medicine Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
238
|
Baumeister SHC, Rambaldi B, Shapiro RM, Romee R. Key Aspects of the Immunobiology of Haploidentical Hematopoietic Cell Transplantation. Front Immunol 2020; 11:191. [PMID: 32117310 PMCID: PMC7033970 DOI: 10.3389/fimmu.2020.00191] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/24/2020] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cell transplantation from a haploidentical donor is increasingly used and has become a standard donor option for patients lacking an appropriately matched sibling or unrelated donor. Historically, prohibitive immunological barriers resulting from the high degree of HLA-mismatch included graft-vs.-host disease (GVHD) and graft failure. These were overcome with increasingly sophisticated strategies to manipulate the sensitive balance between donor and recipient immune cells. Three different approaches are currently in clinical use: (a) ex vivo T-cell depletion resulting in grafts with defined immune cell content (b) extensive immunosuppression with a T-cell replete graft consisting of G-CSF primed bone marrow and PBSC (GIAC) (c) T-cell replete grafts with post-transplant cyclophosphamide (PTCy). Intriguing studies have recently elucidated the immunologic mechanisms by which PTCy prevents GVHD. Each approach uniquely affects post-transplant immune reconstitution which is critical for the control of post-transplant infections and relapse. NK-cells play a key role in haplo-HCT since they do not mediate GVHD but can successfully mediate a graft-vs.-leukemia effect. This effect is in part regulated by KIR receptors that inhibit NK cell cytotoxic function when binding to the appropriate HLA-class I ligands. In the context of an HLA-class I mismatch in haplo-HCT, lack of inhibition can contribute to NK-cell alloreactivity leading to enhanced anti-leukemic effect. Emerging work reveals immune evasion phenomena such as copy-neutral loss of heterozygosity of the incompatible HLA alleles as one of the major mechanisms of relapse. Relapse and infectious complications remain the leading causes impacting overall survival and are central to scientific advances seeking to improve haplo-HCT. Given that haploidentical donors can typically be readily approached to collect additional stem- or immune cells for the recipient, haplo-HCT represents a unique platform for cell- and immune-based therapies aimed at further reducing relapse and infections. The rapid advancements in our understanding of the immunobiology of haplo-HCT are therefore poised to lead to iterative innovations resulting in further improvement of outcomes with this compelling transplant modality.
Collapse
Affiliation(s)
- Susanne H C Baumeister
- Division of Pediatric Hematology-Oncology, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Benedetta Rambaldi
- Harvard Medical School, Boston, MA, United States.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, United States.,Bone Marrow Transplant Unit, Clinical and Experimental Sciences Department, ASST Spedali Civili, University of Pavia, Brescia, Italy
| | - Roman M Shapiro
- Harvard Medical School, Boston, MA, United States.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Rizwan Romee
- Harvard Medical School, Boston, MA, United States.,Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA, United States
| |
Collapse
|
239
|
Hanajiri R, Sani GM, Saunders D, Hanley PJ, Chopra A, Mallal SA, Sosnovtsev SV, Cohen JI, Green KY, Bollard CM, Keller MD. Generation of Norovirus-Specific T Cells From Human Donors With Extensive Cross-Reactivity to Variant Sequences: Implications for Immunotherapy. J Infect Dis 2020; 221:578-588. [PMID: 31562500 PMCID: PMC7325618 DOI: 10.1093/infdis/jiz491] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic norovirus infection in immunocompromised patients can be severe, and presently there is no effective treatment. Adoptive transfer of virus-specific T cells has proven to be safe and effective for the treatment of many viral infections, and this could represent a novel treatment approach for chronic norovirus infection. Hence, we sought to generate human norovirus-specific T cells (NSTs) that can recognize different viral sequences. METHODS Norovirus-specific T cells were generated from peripheral blood of healthy donors by stimulation with overlapping peptide libraries spanning the entire coding sequence of the norovirus genome. RESULTS We successfully generated T cells targeting multiple norovirus antigens with a mean 4.2 ± 0.5-fold expansion after 10 days. Norovirus-specific T cells comprised both CD4+ and CD8+ T cells that expressed markers for central memory and effector memory phenotype with minimal expression of coinhibitory molecules, and they were polyfunctional based on cytokine production. We identified novel CD4- and CD8-restricted immunodominant epitopes within NS6 and VP1 antigens. Furthermore, NSTs showed a high degree of cross-reactivity to multiple variant epitopes from clinical isolates. CONCLUSIONS Our findings identify immunodominant human norovirus T-cell epitopes and demonstrate that it is feasible to generate potent NSTs from third-party donors for use in antiviral immunotherapy.
Collapse
Affiliation(s)
- Ryo Hanajiri
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, District of Columbia, USA
| | - Gelina M Sani
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, District of Columbia, USA
| | - Devin Saunders
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, District of Columbia, USA
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, District of Columbia, USA
- GW Cancer Center, George Washington University, Washington, District of Columbia, USA
- Division of Blood and Marrow Transplantation, Children’s National Health System, Washington, District of Columbia, USA
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia
- Division of Infectious Diseases, Department of Medicine Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Simon A Mallal
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia
- Division of Infectious Diseases, Department of Medicine Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Stanislav V Sosnovtsev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey I Cohen
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kim Y Green
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, District of Columbia, USA
- GW Cancer Center, George Washington University, Washington, District of Columbia, USA
- Division of Blood and Marrow Transplantation, Children’s National Health System, Washington, District of Columbia, USA
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, District of Columbia, USA
- GW Cancer Center, George Washington University, Washington, District of Columbia, USA
- Division of Allergy and Immunology, Children’s National Health System, Washington, District of Columbia, USA
| |
Collapse
|
240
|
Zhang DKY, Cheung AS, Mooney DJ. Activation and expansion of human T cells using artificial antigen-presenting cell scaffolds. Nat Protoc 2020; 15:773-798. [DOI: 10.1038/s41596-019-0249-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/30/2019] [Indexed: 01/03/2023]
|
241
|
Bhatt ST, Schulz G, Hente M, Slater A, Murray L, Shenoy S, Bednarski JJ. A single-center experience using alemtuzumab, fludarabine, melphalan, and thiotepa as conditioning for transplantation in pediatric patients with chronic granulomatous disease. Pediatr Blood Cancer 2020; 67:e28030. [PMID: 31599480 DOI: 10.1002/pbc.28030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/05/2019] [Accepted: 09/23/2019] [Indexed: 11/08/2022]
Abstract
Chronic granulomatous disease (CGD) is an immune deficiency characterized by defective neutrophil function and increased risk of life-threatening infections. Allogeneic hematopoietic cell transplantation is curative for CGD, and conditioning regimen impacts transplant-related outcomes. We report a single-center prospective study (NCT01821781) of four patients with CGD transplanted using a reduced-intensity conditioning regimen (RIC) containing alemtuzumab, fludarabine, melphalan, and thiotepa. Patients had early immune reconstitution with low incidence of infections. Disease-free survival was 75% at a median of five years after transplant. This RIC regimen presents an alternative approach for transplant of patients with CGD who may not tolerate busulfan-based conditioning.
Collapse
Affiliation(s)
- Sima T Bhatt
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Ginny Schulz
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Monica Hente
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Ashley Slater
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Lisa Murray
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Shalini Shenoy
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | | |
Collapse
|
242
|
What the Intensivist Needs to Know About Hematopoietic Stem Cell Transplantation? ONCOLOGIC CRITICAL CARE 2020. [PMCID: PMC7121262 DOI: 10.1007/978-3-319-74588-6_99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is a potential curative therapy for some patients with hematologic conditions. There are two main types of HSCT. This includes autologous HSCT, for which the stem cells are obtained from the patient, and allogeneic HSCT, for which the stem cells are obtained from a related or unrelated donor. The most common indications for autologous stem cell transplant are multiple myeloma and relapsed/refractory lymphoma, whereas leukemia and bone marrow failure syndromes remain the most common indications for allogeneic stem cell transplant. This chapter will review the different types, indications, processes, and main complications of HSCT. This chapter will also discuss end-of-life issues that patients and providers face when transplant patients are admitted for the intensive care unit.
Collapse
|
243
|
Genetically Modified T-Cell Therapy for Osteosarcoma: Into the Roaring 2020s. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:109-131. [PMID: 32483735 DOI: 10.1007/978-3-030-43032-0_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-cell immunotherapy may offer an approach to improve outcomes for patients with osteosarcoma who fail current therapies. In addition, it has the potential to reduce treatment-related complications for all patients. Generating tumor-specific T cells with conventional antigen-presenting cells ex vivo is time-consuming and often results in T-cell products with a low frequency of tumor-specific T cells. Furthermore, the generated T cells remain sensitive to the immunosuppressive tumor microenvironment. Genetic modification of T cells is one strategy to overcome these limitations. For example, T cells can be genetically modified to render them antigen specific, resistant to inhibitory factors, or increase their ability to home to tumor sites. Most genetic modification strategies have only been evaluated in preclinical models; however, early clinical phase trials are in progress. In this chapter, we will review the current status of gene-modified T-cell therapy with special focus on osteosarcoma, highlighting potential antigenic targets, preclinical and clinical studies, and strategies to improve current T-cell therapy approaches.
Collapse
|
244
|
Immunological and Clinical Impact of Manipulated and Unmanipulated DLI after Allogeneic Stem Cell Transplantation of AML Patients. J Clin Med 2019; 9:jcm9010039. [PMID: 31878060 PMCID: PMC7019914 DOI: 10.3390/jcm9010039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) is the preferred curative treatment for several hematological malignancies. The efficacy of allo-SCT depends on the graft-versus-leukemia (GvL) effect. However, the prognosis of patients with relapsed acute myeloid leukemia (AML) following allo-SCT is poor. Donor lymphocyte infusion (DLI) is utilized after allo-SCT in this setting to prevent relapse, to prolong progression free survival, to establish full donor chimerism and to restore the GvL effect in patients with hematological malignancies. Thus, there are different options for the administration of DLI in AML patients. DLI is currently used prophylactically and in the setting of an overt relapse. In addition, in the minimal residual disease (MRD) setting, DLI may be a possibility to improve overall survival. However, DLI might increase the risk of severe life-threatening complications such as graft-versus-host disease (GvHD) as well as severe infections. The transfusion of lymphocytes has been tested not only for the treatment of hematological malignancies but also chronic infections. In this context, manipulated DLI in a prophylactic or therapeutic approach are an option, e.g., virus-specific DLI using different selection methods or antigen-specific DLI such as peptide-specific CD8+ cytotoxic T lymphocytes (CTLs). In addition, T cells are also genetically engineered, using both chimeric antigen receptor (CAR) genetically modified T cells and T cell receptor (TCR) genetically modified T cells. T cell therapies in general have the potential to enhance antitumor immunity, augment vaccine efficacy, and limit graft-versus-host disease after allo-SCT. The focus of this review is to discuss the different strategies to use donor lymphocytes after allo-SCT. Our objective is to give an insight into the functional effects of DLI on immunogenic antigen recognition for a better understanding of the mechanisms of DLI. To ultimately increase the GvL potency without raising the risk of GvHD at the same time.
Collapse
|
245
|
Abstract
PURPOSE OF REVIEW Transplant recipients are at risk for cytomegalovirus (CMV) infection and associated morbidity and mortality. We summarize recently introduced or currently investigated modalities for prevention and treatment of CMV infection in hematopoietic cell (HCT) and solid organ transplant (SOT) recipients. RECENT FINDINGS Letermovir was recently approved for CMV prevention in HCT recipients. Data from real world studies support its role to improve outcomes in this population. Letermovir is currently under investigation for broader patient populations and indications. Maribavir is in late stages of development for CMV treatment and may provide a safer alternative to currently available anti-CMV drugs. Promising CMV vaccine candidates and adoptive cell therapy approaches are under evaluation. CMV immune monitoring assays are predicted to play a more central role in our clinical decision making. In recent years, major advances have been made in CMV prevention and treatment in transplant recipients. Rigorous research is ongoing and is anticipated to further impact our ability to improve outcomes in this population.
Collapse
Affiliation(s)
- Anat Stern
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, NY1250 1st Avenue, New York, NY, 10065, USA
| | - Genovefa A Papanicolaou
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, NY1250 1st Avenue, New York, NY, 10065, USA.
| |
Collapse
|
246
|
Celilova S, Toret E, Adaklı Aksoy B, Ovalı E, Bozkurt C. CMV-specific T-Cells for Treatment of CMV Infection after Hematopoietic Stem Cell Transplantation in a Pediatric Case: First Application in Turkey. Turk J Haematol 2019; 37:65-67. [PMID: 31718117 PMCID: PMC7057740 DOI: 10.4274/tjh.galenos.2019.2019.0293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Sevil Celilova
- Altınbaş University Faculty of Medicine, Medicalpark Bahçelievler Hospital, Department of Pediatric Hematology-Oncology & Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Ersin Toret
- Altınbaş University Faculty of Medicine, Medicalpark Bahçelievler Hospital, Department of Pediatric Hematology-Oncology & Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Başak Adaklı Aksoy
- Altınbaş University Faculty of Medicine, Medicalpark Bahçelievler Hospital, Department of Pediatric Hematology-Oncology & Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Ercüment Ovalı
- Acıbadem University Faculty of Medicine, Altunizade Hospital, Department of Hematology, İstanbul, Turkey
| | - Ceyhun Bozkurt
- İstinye University Faculty of Medicine, Medicalpark Bahcelievler Hospital, Department of Pediatric Hematology-Oncology & Bone Marrow Transplantation Unit, İstanbul, Turkey
| |
Collapse
|
247
|
Schultze-Florey RE, Tischer-Zimmermann S, Heuft HG, Priesner C, Lamottke B, Heim A, Sauer M, Sykora KW, Blasczyk R, Eiz-Vesper B, Maecker-Kolhoff B. Transfer of Hexon- and Penton-selected adenovirus-specific T cells for refractory adenovirus infection after haploidentical stem cell transplantation. Transpl Infect Dis 2019; 22:e13201. [PMID: 31643129 DOI: 10.1111/tid.13201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/05/2019] [Accepted: 10/12/2019] [Indexed: 12/17/2022]
Abstract
Adenovirus (HAdV) infections confer a high risk of morbidity and mortality for immunocompromised patients after stem cell transplantation (SCT). Treatment with standard antiviral drugs is of limited efficacy and associated with a high rate of adverse effects. HAdV-specific T cells are crucial for sustained viral elimination and the efficacy of adoptive T-cell therapy with donor-derived HAdV-specific T cells has been reported by several investigators. Here, we report our experience with the transfer of HAdV-specific T cells specific for penton, which was recently identified as an immunodominant target of T cells, and hexon in a 14-year-old boy after T-cell-depleted haploidentical SCT for myelodysplastic syndrome (MDS). He developed severe HAdV-associated enteritis complicated by acute graft-versus-host disease (GvHD). The patient received ten infusions of allogeneic HAdV-specific T cells manufactured from the haploidentical stem cell donor using the CliniMacs Interferon-γ (IFN-γ) cytokine capture and immunomagnetic selection. Initially, T cells were generated against the immunodominant target hexon and in subsequent transfers dual antigen-specific T cells against hexon and penton were applied. T-cell transfers were scheduled individually tailored to current immunosuppressive treatment. Each transfer was followed by reduction of HAdV load in peripheral blood and clinical improvement. Importantly, T-cell responses to both penton and hexon pools emerged in patient blood after repetitive transfers. Unfortunately, the patient experienced bacterial sepsis, and in this context, severe GvHD requiring intensive immunosuppression followed by secondary progression of HAdV infection. The patient succumbed to multiorgan failure 283 days after SCT. This case demonstrates the feasibility of HAdV-specific T-cell transfer even in the presence of immunosuppressive treatment. Targeting of multiple immunodominant viral proteins may prove valuable in patients with complicated HAdV infections.
Collapse
Affiliation(s)
- Rebecca E Schultze-Florey
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany.,Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany.,Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Hans-Gert Heuft
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Christoph Priesner
- Institute for Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Britta Lamottke
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Albert Heim
- Institute for Virology, Hannover Medical School, Hannover, Germany
| | - Martin Sauer
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Karl-Walter Sykora
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany.,Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany.,Integrated Research and Treatment Center Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| |
Collapse
|
248
|
Tunkel AR, Baron EL, Buch KA, Marty FM, Martinez-Lage M. Case 31-2019: A 45-Year-Old Woman with Headache and Somnolence. N Engl J Med 2019; 381:1459-1470. [PMID: 31597024 DOI: 10.1056/nejmcpc1904045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Allan R Tunkel
- From the Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI (A.R.T.); the Department of Medicine, Newton-Wellesley Hospital, Newton, MA (E.L.B.); and the Departments of Radiology (K.A.B.) and Pathology (M.M.-L.), Massachusetts General Hospital, the Department of Medicine, Brigham and Women's Hospital (F.M.M.), and the Departments of Radiology (K.A.B.), Medicine (F.M.M.), and Pathology (M.M.-L.), Harvard Medical School - all in Boston
| | - Elinor L Baron
- From the Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI (A.R.T.); the Department of Medicine, Newton-Wellesley Hospital, Newton, MA (E.L.B.); and the Departments of Radiology (K.A.B.) and Pathology (M.M.-L.), Massachusetts General Hospital, the Department of Medicine, Brigham and Women's Hospital (F.M.M.), and the Departments of Radiology (K.A.B.), Medicine (F.M.M.), and Pathology (M.M.-L.), Harvard Medical School - all in Boston
| | - Karen A Buch
- From the Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI (A.R.T.); the Department of Medicine, Newton-Wellesley Hospital, Newton, MA (E.L.B.); and the Departments of Radiology (K.A.B.) and Pathology (M.M.-L.), Massachusetts General Hospital, the Department of Medicine, Brigham and Women's Hospital (F.M.M.), and the Departments of Radiology (K.A.B.), Medicine (F.M.M.), and Pathology (M.M.-L.), Harvard Medical School - all in Boston
| | - Francisco M Marty
- From the Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI (A.R.T.); the Department of Medicine, Newton-Wellesley Hospital, Newton, MA (E.L.B.); and the Departments of Radiology (K.A.B.) and Pathology (M.M.-L.), Massachusetts General Hospital, the Department of Medicine, Brigham and Women's Hospital (F.M.M.), and the Departments of Radiology (K.A.B.), Medicine (F.M.M.), and Pathology (M.M.-L.), Harvard Medical School - all in Boston
| | - Maria Martinez-Lage
- From the Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI (A.R.T.); the Department of Medicine, Newton-Wellesley Hospital, Newton, MA (E.L.B.); and the Departments of Radiology (K.A.B.) and Pathology (M.M.-L.), Massachusetts General Hospital, the Department of Medicine, Brigham and Women's Hospital (F.M.M.), and the Departments of Radiology (K.A.B.), Medicine (F.M.M.), and Pathology (M.M.-L.), Harvard Medical School - all in Boston
| |
Collapse
|
249
|
Ilic D, Liovic M, Noli L. Industry updates from the field of stem cell research and regenerative medicine in June 2019. Regen Med 2019. [DOI: 10.2217/rme-2019-0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The latest developments in the field of stem cell research and regenerative medicine compiled from publicly available information and press releases from nonacademic institutions in June 2019.
Collapse
Affiliation(s)
- Dusko Ilic
- Stem Cell Laboratories, Guy’s Assisted Conception Unit, Department of Women & Children’s Health, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - Mirjana Liovic
- Medical Center for Molecular Biology, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Laila Noli
- Department of Pathological Sciences, Fakeeh College for Medical Sciences, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
250
|
Haemorrhagic cystitis, preventive and treatment interventions in patients undergoing haematopoietic stem cell transplantation: A scoping review. Eur J Oncol Nurs 2019; 42:50-62. [DOI: 10.1016/j.ejon.2019.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 07/03/2019] [Accepted: 07/11/2019] [Indexed: 12/29/2022]
|