1
|
Nadaf R, Lee H, Bonney D, Hanasoge-Nataraj R, Senthil S, Horgan C, Guiver M, Poulton K, Wynn R. Mixed T-Cell Chimerism Following Hematopoietic Cell Transplantation for Non-Malignant Disorders Is Common, Facilitates Anti-Viral Immunity, and Is Not Associated with Graft Failure in Pediatric Patients. Cells 2024; 13:2119. [PMID: 39768209 PMCID: PMC11727161 DOI: 10.3390/cells13242119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/13/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Myeloid chimerism better reflects donor stem cell engraftment than whole-blood chimerism in assessing graft function following allogeneic hematopoietic stem cell transplant (HCT). We describe our experience with 130 patients aged younger than 18 years, treated with allogeneic HCT using bone marrow or PBSC from HLA-matched donors for non-malignant diseases, whose pre-transplant conditioning therapy included alemtuzumab and who were monitored with lineage-specific chimerism after transplant. At 6 years post-transplant, overall survival (OS) was 91.1% and event-free survival (EFS) was 81.5%, with no grade III-IV acute GvHD or chronic GVHD observed. Recipient T-cells did not contribute to graft loss. Mixed T-cell chimerism (MC) did not affect EFS, and there was no connection between T-cell chimerism and myeloid chimerism in patients with MC or graft loss. MC significantly correlated with virus infection; more children with MC were CMV seropositive than those with complete chimerism (CC). Additionally, MC was more common in patients with CMV viramia post-transplant. CD8 T-cell reconstitution was affected by viral reactivation, including CMV, with CD8 T-cell counts higher in the MC group than in the CC group. Mixed T-cell chimerism is due to autologous, virus-specific, predominantly CD8, T-cell expansion, and is protective and not deleterious to the recipient.
Collapse
Affiliation(s)
- Rubiya Nadaf
- Departments of Blood and Marrow Transplant, Royal Manchester Children’s Hospital, Manchester M13 9WL, UK; (D.B.); (R.H.-N.); (S.S.); (C.H.)
| | - Helena Lee
- Departments of Transplantation Laboratories, Manchester Royal Infirmary, Manchester M13 9WL, UK; (H.L.); (K.P.)
| | - Denise Bonney
- Departments of Blood and Marrow Transplant, Royal Manchester Children’s Hospital, Manchester M13 9WL, UK; (D.B.); (R.H.-N.); (S.S.); (C.H.)
| | - Ramya Hanasoge-Nataraj
- Departments of Blood and Marrow Transplant, Royal Manchester Children’s Hospital, Manchester M13 9WL, UK; (D.B.); (R.H.-N.); (S.S.); (C.H.)
| | - Srividhya Senthil
- Departments of Blood and Marrow Transplant, Royal Manchester Children’s Hospital, Manchester M13 9WL, UK; (D.B.); (R.H.-N.); (S.S.); (C.H.)
| | - Claire Horgan
- Departments of Blood and Marrow Transplant, Royal Manchester Children’s Hospital, Manchester M13 9WL, UK; (D.B.); (R.H.-N.); (S.S.); (C.H.)
| | - Malcolm Guiver
- Departments of Virology Laboratories, Manchester Royal Infirmary, Manchester M13 9WL, UK
| | - Kay Poulton
- Departments of Transplantation Laboratories, Manchester Royal Infirmary, Manchester M13 9WL, UK; (H.L.); (K.P.)
| | - Robert Wynn
- Departments of Blood and Marrow Transplant, Royal Manchester Children’s Hospital, Manchester M13 9WL, UK; (D.B.); (R.H.-N.); (S.S.); (C.H.)
| |
Collapse
|
2
|
Pandrowala A, Khan S, Kataria D, Kakunje M, Mishra V, Mamtora D, Mudaliar S, Bodhanwala M, Agarwal B, Hiwarkar P. The role of graft T-cell size in patients receiving alemtuzumab serotherapy for non-malignant disorders: results of an institutional protocol. Sci Rep 2024; 14:988. [PMID: 38200046 PMCID: PMC10781954 DOI: 10.1038/s41598-023-50416-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Although graft T cells assist in engraftment, mediate antiviral immune-reconstitution, and cause graft-versus-host disease, graft size is not determined by T-cell content of the graft. The conventional method of graft size determination based on CD34+ cells with alemtuzumab serotherapy is associated with delayed immune reconstitution, contributing to an increased risk of viral infections and graft failure. Alemtuzumab, a long half-life anti-CD52 monoclonal antibody is a robust T-cell depleting serotherapy, and relatively spares memory-effector T cells compared to naïve T cells. We therefore hypothesized that graft size based on T-cell content in patients receiving peripheral blood stem cell graft with alemtuzumab serotherapy would facilitate immune-reconstitution without increasing the risk of graft-versus-host disease. We retrospectively analysed twenty-six consecutive patients with non-malignant disorders grafted using alemtuzumab serotherapy and capping of graft T cells to a maximum of 600 million/kg. The graft T-cell capping protocol resulted in early immune-reconstitution without increasing the risk of severe graft-versus-host disease. Graft T-cell content correlated with CD4+ T-cell reconstitution and acute graft-versus-host disease. The course of CMV viraemia was predictable without recurrence and associated with early T-cell recovery. No patient developed chronic graft-versus-host disease. Overall survival at one year was 100% and disease-free survival was 96% at a median of 899 days (range: 243-1562). Graft size determined by peripheral blood stem cell graft T-cell content in patients receiving alemtuzumab serotherapy for non-malignant disorders is safe and leads to early T-cell immune-reconstitution with excellent survival outcomes.
Collapse
Affiliation(s)
- Ambreen Pandrowala
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, 400012, India
| | - Sanna Khan
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, 400012, India
| | - Darshan Kataria
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, 400012, India
| | - Manasa Kakunje
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, 400012, India
| | - Varsha Mishra
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, 400012, India
| | - Dhruv Mamtora
- Department of Pathology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Sangeeta Mudaliar
- Department of Paediatric Haematology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Minnie Bodhanwala
- Department of Paediatrics, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Bharat Agarwal
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, 400012, India
- Department of Paediatric Haematology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Prashant Hiwarkar
- Department of Blood and Marrow Transplantation, Bai Jerbai Wadia Hospital for Children, Mumbai, 400012, India.
| |
Collapse
|
3
|
Peggs KS, Albon SJ, Oporto Espuelas M, Irving C, Richardson R, Casanovas-Company J, Wallace R, Guvenel A, Ghorashian S, Collura A, Subramaniyam M, Flutter B, Popova B, Castro F, Lopes A, Champion K, Schofield O, Clifton-Hadley L, Taylor T, Farrell M, Adams S, Gilmour KC, Mackinnon S, Tholouli E, Amrolia PJ. Immunotherapy with CD25/CD71-allodepleted T cells to improve T-cell reconstitution after matched unrelated donor hematopoietic stem cell transplant: a randomized trial. Cytotherapy 2023; 25:82-93. [PMID: 36220712 DOI: 10.1016/j.jcyt.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/21/2022] [Accepted: 08/27/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND AIMS Delayed immune reconstitution is a major challenge after matched unrelated donor (MUD) stem cell transplant (SCT). In this randomized phase 2 multi-center trial, Adoptive Immunotherapy with CD25/71 allodepleted donor T cells to improve immunity after unrelated donor stem cell transplant (NCT01827579), the authors tested whether allodepleted donor T cells (ADTs) can safely be used to improve immune reconstitution after alemtuzumab-based MUD SCT for hematological malignancies. METHODS Patients received standard of care or up to three escalating doses of ADTs generated through CD25+/CD71+ immunomagnetic depletion. The primary endpoint of the study was circulating CD3+ T-cell count at 4 months post-SCT. Twenty-one patients were treated, 13 in the ADT arm and eight in the control arm. RESULTS The authors observed a trend toward improved CD3+ T-cell count at 4 months in the ADT arm versus the control arm (230/µL versus 145/µL, P = 0.18), and three ADT patients achieved normal CD3+ T-cell count at 4 months (>700/µL). The rates of significant graft-versus-host disease (GVHD) were comparable in both cohorts, with grade ≥2 acute GVHD in seven of 13 and four of eight patients and chronic GVHD in three of 13 and three of eight patients in the ADT and control arms, respectively. CONCLUSIONS These data suggest that adoptive transfer of ADTs is safe, but that in the MUD setting the benefit in terms of T-cell reconstitution is limited. This approach may be of more use in the context of more rigorous T-cell depletion.
Collapse
Affiliation(s)
- Karl S Peggs
- Department of Hematology, University College London Hospital, London, UK
| | - Sarah J Albon
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Macarena Oporto Espuelas
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK.
| | - Catherine Irving
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Rachel Richardson
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Joan Casanovas-Company
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Rebecca Wallace
- Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK; Molecular Hematology Section, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Aleks Guvenel
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Sara Ghorashian
- Molecular Hematology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Department of Hematology, Great Ormond Street Hospital for Children, London, UK
| | - Angela Collura
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Meera Subramaniyam
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK
| | - Barry Flutter
- Gene and Cell Therapy, Great Ormond Street Hospital for Children, London, UK; Molecular Hematology Section, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Bilyana Popova
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Fernanda Castro
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Andre Lopes
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Kim Champion
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Oliver Schofield
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Laura Clifton-Hadley
- Cancer Research UK and University College London Cancer Trials Center, London, UK
| | - Thomas Taylor
- Department of Hematology, University College London Hospital, London, UK
| | - Maria Farrell
- Department of Hematology, Manchester Royal Infirmary, Manchester, UK
| | - Stuart Adams
- Department of Hematology, Great Ormond Street Hospital for Children, London, UK
| | - Kimberly C Gilmour
- Cell Therapy and Immunology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children, London, UK
| | - Stephen Mackinnon
- Department of Hematology, University College London Hospital, London, UK
| | - Eleni Tholouli
- Department of Hematology, Manchester Royal Infirmary, Manchester, UK
| | - Persis J Amrolia
- Molecular and Cellular Immunology Section, University College London Great Ormond Street Institute of Child Health, London, UK; Department of Bone Marrow Transplantation, Great Ormond Street Hospital for Children, London, UK.
| |
Collapse
|
4
|
Wang Z, Yang S, Zhu Z, Lei P, Yang J, Zhang P, Sun K. A Case of Successful Allogeneic Hematopoietic Stem Cell Transplantation for HHV8-Positive Castleman's Disease with a Review of the Literature. Cell Transplant 2021; 29:963689720943571. [PMID: 32713191 PMCID: PMC7563820 DOI: 10.1177/0963689720943571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objective: To investigate the long-term clinical efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for patients with human herpes virus 8 (HHV8)-positive multicentric Castleman’s disease (MCD). Methods: A 17-year-old female patient was admitted to Henan Provincial People’s Hospital with the complaint of febrile for half a month, headache, and enlarged superficial lymph nodes on October 5, 2010. HHV8-positive mixed cellular Castleman’s disease was found by pathological diagnosis of lymph nodes biopsy. After the administration of CHOP and Hyper-CVAD-B, the patient was still febrile, we administrated the followed COAP, two courses of VAD(Vincristine, Adriamycin, Dexamethasone), the patient received CR. Six months after CR, the patient relapsed, we administrated VAD and two courses of bortezomide+dexamethasone chemotherapy, and then the patient received PR. After that, the patient underwent allo-HSCT from his human leukocyte antigen (HLA)-matched unrelated donor after conditioning with Bu/Cy+Etoposide+Smoustin.graft-vs-host disease (GVHD) prophylaxis, which consisted of ATG (7.5 mg/kg, qd, ivdrip) from d-5 to d-2, cyclosporine (3 mg/kg/d, qd, ivdrip, for 24 h) started from day-1, MMF(0.5 g, tid, po.) started from day+1 to +28, and MTX (15 mg per time, ivdrip, d+1,+4,+7,+11). She received 3.5×106/L CD34+cells and 8.1×108/LMNC. Results: Granulocyte engraftment occurred on day+12, platelet engrafted on day+14. Bone marrow biopsy showed normalization of trilineage hematopoiesis on day+33, chimerism: 97.6%. The transplantation was successful and followed up for 7 years with CR. Conclusion: Allo-HSCT might cure patients with refractory/relapsed HHV8+ MCD.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Hematology, Henan University People’s Hospital, School of Clinical Medicine, Henan University, Zhengzhou, Henan, PR China
- Department of Hematology, Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital Zhengzhou, PR China
- Both the authors contributed equally to this article
| | - Shiwei Yang
- Institute of Hematology Henan Provincial People’s Hospital, Zhengzhou, Henan, PR China
- Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People’s Hospital, Zhengzhou, Henan, PR China
- Both the authors contributed equally to this article
| | - Zunmin Zhu
- Department of Hematology, Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital Zhengzhou, PR China
- Institute of Hematology Henan Provincial People’s Hospital, Zhengzhou, Henan, PR China
- Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People’s Hospital, Zhengzhou, Henan, PR China
| | - Pingchong Lei
- Department of Hematology, Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital Zhengzhou, PR China
| | - Jing Yang
- Department of Hematology, Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital Zhengzhou, PR China
| | - Ping Zhang
- Department of Hematology, Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital Zhengzhou, PR China
| | - Kai Sun
- Department of Hematology, Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital Zhengzhou, PR China
- Kai Sun, Department of Hematology, Henan Provincial People’s Hospital and Zhengzhou University People’s Hospital Zhengzhou 450003, PR China. Emails: ;
| |
Collapse
|
5
|
Reduced-intensity single-unit unrelated cord blood transplant with optional immune boost for nonmalignant disorders. Blood Adv 2021; 4:3041-3052. [PMID: 32634238 DOI: 10.1182/bloodadvances.2020001940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/28/2020] [Indexed: 12/24/2022] Open
Abstract
Children with many inherited nonmalignant disorders can be cured or their condition alleviated by hematopoietic stem cell transplantation (HSCT). Umbilical cord blood (UCB) units are a rapidly available stem cell source and offer great flexibility in HLA matching, allowing nearly uniform access to HSCT. Although reduced-intensity conditioning (RIC) regimens promise decreased treatment-related morbidity and mortality, graft failure and infections have limited their use in chemotherapy-naive patients. We prospectively evaluated a novel RIC regimen of alemtuzumab, hydroxyurea, fludarabine, melphalan, and thiotepa with a single-unit UCB graft in 44 consecutive patients with inborn errors of metabolism, immunity, or hematopoiesis. In addition, 5% of the UCB graft was re-cryopreserved and reserved for cord donor leukocyte infusion (cDLI) posttransplant. All patients engrafted at a median of 15 days posttransplant, and chimerism was >90% donor in the majority of patients at 1-year posttransplant with only 1 secondary graft failure. The incidence of grade II to IV graft-versus-host disease (GVHD) was 27% (95% confidence interval [CI], 17-43) with no extensive chronic GVHD. Overall survival was 95% (95% CI, 83-99) and 85% (95% CI, 64-93) at 1 and 5 years posttransplant, respectively. No significant end-organ toxicities were observed. The use of cDLI did not affect GVHD and showed signals of efficacy for infection control or donor chimerism. This RIC transplant regimen using single-unit UCB graft resulted in outstanding survival and remarkably low rates of graft failure. Implementation of the protocol not requiring pharmacokinetic monitoring would be feasible and applicable worldwide for children with inherited disorders of metabolism, immunity, or hematopoiesis. This trial was registered at www.clinicaltrials.gov as #NCT01962415.
Collapse
|
6
|
Stenger W, Künkele A, Niemann M, Todorova K, Pruß A, Schulte JH, Eggert A, Oevermann L. Donor selection in a pediatric stem cell transplantation cohort using PIRCHE and HLA-DPB1 typing. Pediatr Blood Cancer 2020; 67:e28127. [PMID: 31850671 DOI: 10.1002/pbc.28127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 11/07/2022]
Abstract
BACKGROUND New strategies to optimize donor selection for hematopoietic stem cell transplantation (HSCT) have mainly been evaluated in adults, but the disease spectrum requiring HSCT differs significantly in children and has consequences for the risk of complications, such as graft-versus-host disease (GvHD). PROCEDURES Here we evaluated whether HLA-DPB1 and Predicted Indirectly ReCognizable HLA-Epitope (PIRCHE) matching can improve donor selection and minimize risks specific for a pediatric cohort undergoing HSCT in Berlin between 2014 and 2016. RESULTS The percentage of HLA-DPB1-mismatched HSCT in the pediatric cohort was in line with the general distribution among matched unrelated donor HSCT. Nonpermissive HLA-DPB1 mismatches were not associated with a higher incidence of GvHD, but the incidence of relapse was higher in patients undergoing HSCT from HLA-DPB1-matched transplantations. High PIRCHE-I scores were associated with a significantly higher risk for developing GvHD in patients undergoing HSCT from nine of ten matched unrelated donors. This finding persisted after including HLA-DPB1 into the PIRCHE analysis. CONCLUSIONS Implementing PIRCHE typing in the donor selection process for HSCT in children could particularly benefit children with nonmalignant diseases and support further validation of PIRCHE-based donor selection in a larger number of children treated at different sites.
Collapse
Affiliation(s)
- Wiebke Stenger
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Annette Künkele
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Kremena Todorova
- Center for Transfusion Medicine and Cell Therapies Berlin, Berlin, Germany
| | - Axel Pruß
- Center for Transfusion Medicine and Cell Therapies Berlin, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena Oevermann
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
7
|
Fleischer LC, Spencer HT, Raikar SS. Targeting T cell malignancies using CAR-based immunotherapy: challenges and potential solutions. J Hematol Oncol 2019; 12:141. [PMID: 31884955 PMCID: PMC6936092 DOI: 10.1186/s13045-019-0801-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/09/2019] [Indexed: 12/23/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been successful in treating B cell malignancies in clinical trials; however, fewer studies have evaluated CAR T cell therapy for the treatment of T cell malignancies. There are many challenges in translating this therapy for T cell disease, including fratricide, T cell aplasia, and product contamination. To the best of our knowledge, no tumor-specific antigen has been identified with universal expression on cancerous T cells, hindering CAR T cell therapy for these malignancies. Numerous approaches have been assessed to address each of these challenges, such as (i) disrupting target antigen expression on CAR-modified T cells, (ii) targeting antigens with limited expression on T cells, and (iii) using third party donor cells that are either non-alloreactive or have been genome edited at the T cell receptor α constant (TRAC) locus. In this review, we discuss CAR approaches that have been explored both in preclinical and clinical studies targeting T cell antigens, as well as examine other potential strategies that can be used to successfully translate this therapy for T cell disease.
Collapse
Affiliation(s)
- Lauren C Fleischer
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, GA, USA
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
| | - H Trent Spencer
- Molecular and Systems Pharmacology Graduate Program, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University School of Medicine, Atlanta, GA, USA
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA
| | - Sunil S Raikar
- Cell and Gene Therapy Program, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
8
|
Kosulin K, Pichler H, Lawitschka A, Geyeregger R, Lion T. Diagnostic Parameters of Adenoviremia in Pediatric Stem Cell Transplant Recipients. Front Microbiol 2019; 10:414. [PMID: 30853954 PMCID: PMC6396503 DOI: 10.3389/fmicb.2019.00414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Despite recent progress in the diagnostic risk assessment of human adenovirus (HAdV) infections in immunocompromised patients, clinical complications mediated by these viruses continue contributing to significant morbidity and mortality, particularly in the pediatric hematopoietic allogeneic stem cell transplant (HSCT) setting. Current data highlight the importance of monitoring stool samples to assess the risk of invasive HAdV infections in children undergoing HSCT. The advent of novel, more effective antiviral treatment options might permit successful virus control even at the stage of systemic infection, thus increasing the interest in optimized HAdV monitoring in peripheral blood (PB). We have screened over 300 pediatric HCST recipients by serial monitoring of stool and PB specimens, and identified 31 cases of invasive HAdV infection by quantitative pan-adenovirus RQ-PCR analysis of consecutive PB specimens. The diagnostic parameters assessed included HAdV peak levels (PL) and the time-averaged area under the curve (AAUC) of virus copy numbers. The predictive value for patient outcome reflected by non-relapse and HAdV-related mortality was determined. The patients were assigned to quartiles based on their PL and AAUC, and the readouts were highly correlated (p < 0.0001). Non-relapse mortality in patients by AAUC quartile (lowest to highest) was 26, 50, 75, and 86%, respectively, and AAUC was strongly correlated with non-relapse mortality (p < 0.0001), while the association between PL and non-relapse mortality was less pronounced (p = 0.013). HAdV-related mortality was absent or very low in patients within the two lower quartiles of both PL and AAUC, and increased to ≥70% in the upper two quartiles. Despite the significant correlation of PL and AAUC with patient outcome, it is necessary to consider that the risk of non-relapse mortality even within the lowest quartile was still relatively high, and it might be difficult therefore to translate the results into differential treatment approaches. By contrast, the correlation with HAdV-related mortality might permit the identification of a low-risk patient subset. Nevertheless, the well-established correlation of HAdV shedding into the stool and intestinal expansion of the virus with the risk of invasive infection will expectedly remain an essential diagnostic parameter in the pediatric HSCT setting.
Collapse
Affiliation(s)
- Karin Kosulin
- Molecular Microbiology, Children's Cancer Research Institute, Vienna, Austria
| | - Herbert Pichler
- Stem Cell Transplant Unit, St. Anna Children's Hospital, Vienna, Austria
| | - Anita Lawitschka
- Stem Cell Transplant Unit, St. Anna Children's Hospital, Vienna, Austria
| | - René Geyeregger
- Molecular Microbiology, Children's Cancer Research Institute, Vienna, Austria
| | - Thomas Lion
- Molecular Microbiology, Children's Cancer Research Institute, Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Guilcher GMT, Shah R, Shenoy S. Principles of alemtuzumab immunoablation in hematopoietic cell transplantation for non-malignant diseases in children: A review. Pediatr Transplant 2018; 22. [PMID: 29352515 DOI: 10.1111/petr.13142] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/29/2017] [Indexed: 12/19/2022]
Abstract
Alemtuzumab is a humanized mAb targeted to CD52. Alemtuzumab is highly immunosuppressive with the ability to deplete T and B cells (in addition to other immune cell lines). A growing understanding of the PKs, dosing, and timing of administration of alemtuzumab has allowed for the study of its use as a conditioning agent for allogeneic HCT. The highly immunosuppressive properties of the drug are particularly appealing in the setting of non-malignant HCT, where GVHD provides no clinical benefit and relapse of malignancy is not applicable. In addition, the degree of immune suppression achieved with alemtuzumab has allowed for a reduction in the intensity of myeloablative cytotoxic agents included in some HCT conditioning regimens, allowing for fewer acute and late toxicities. This review paper will provide a comprehensive summary of the mechanism of action, PKs, dosing, and timing of alemtuzumab, a brief description of its use in various allogeneic HCT protocols for non-malignant conditions and a summary of the data regarding its use for GVHD therapy. The goal of this review was to provide an understanding as to how alemtuzumab might be safely incorporated into HCT conditioning regimens for children with non-malignant disease, allowing for expanded access to curative HCT therapy.
Collapse
Affiliation(s)
- Gregory M T Guilcher
- Section of Paediatric Oncology/BMT, Departments of Oncology and Paediatrics, University of Calgary, Calgary, AB, Canada
| | - Ravi Shah
- Department of Paediatric Haematology/BMT, Great Ormond Street Hospital, NHS Foundation Trust, London, UK
| | - Shalini Shenoy
- Division of Pediatric Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Lu L, Zhang G, Li R, Zhao Z, Li W, Liu T, Fu W. Molecular Chimeric Recipient Precursor T Cells Promote Cardiac Allograft Survival in Mice. Transplant Proc 2015; 47:2978-84. [PMID: 26707325 DOI: 10.1016/j.transproceed.2015.09.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/31/2015] [Accepted: 09/17/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Molecular chimerism has become a potential method to induce donor-specific transplant tolerance. We researched the prolongation of cardiac allograft survival by recipient mouse molecular chimeric precursor T cells (pre-T cells) or hematopoietic stem cells (HSCs) infusion in vivo. METHODS The donor C57BL/6 mouse MHC-I gene (H-2K(b) and H-2D(b) gene) were amplified by RT-PCR. The identified recipient BALB/c mouse pre-T cells and HSCs were transduced by the pMSCVneo retroviral vector of C57BL/6 mouse MHC-I gene (pMSCVneo-H-2D(b)/H-2K(b)). Then the molecular chimeric cells were transfused back to the BALB/c mice. Allogeneic T-lymphocyte proliferation was assessed in mixed lymphocyte reactions (MLR). A mouse model of heterotopic abdominal heart transplantation was performed to evaluate survival times and histological grade of acute rejection at 7 days after transplantation. RESULTS BALB/c mice molecular chimeric pre-T cells and HSCs were cultured successfully after pMSCV-H-2D(b)/H-2K(b) transduction. After the molecular chimeric pre-T cell treatment, the result of MLR showed that the stimulating index of allogeneic T lymphocyte had a statistically significant decrease, which also exhibited a significant reduction after molecular chimeric HSC treatment. The survival time of cardiac allograft was prolonged after chimeric pre-T cell or HSC infusion; meanwhile, pathologic rejection grade decreased significantly. Nevertheless, molecular chimeric pre-T cells exhibited a longer median survival time. CONCLUSION The molecular chimeric recipient mouse pre-T cell or HSC infusion reduced spleen T cells' response to allogeneic T cells in vitro and delayed cardiac allograft rejection in vivo. Pre-T cells have more advantages than HSCs on the prolongation of mouse cardiac allograft survival.
Collapse
Affiliation(s)
- L Lu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - G Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - R Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Z Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - W Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - T Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - W Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|