1
|
Stiehl T. Stem cell graft dose and composition could impact on the expansion of donor-derived clones after allogeneic hematopoietic stem cell transplantation - a virtual clinical trial. Front Immunol 2024; 15:1321336. [PMID: 39737169 PMCID: PMC11682905 DOI: 10.3389/fimmu.2024.1321336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 09/10/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Hematopoietic stem cell transplantation is a potentially curative intervention for a broad range of diseases. However, there is evidence that malignant or pre-malignant clones contained in the transplant can expand in the recipient and trigger donor-derived malignancies. This observation has gained much attention in the context of clonal hematopoiesis, a medical condition where significant amounts of healthy blood cells are derived from a small number of hematopoietic stem cell clones. In many cases the dominating clones carry mutations conferring a growth advantage and thus could undergo malignant transformation in the recipient. Since clonal hematopoiesis exists in a significant proportion of potential stem cell donors, a more detailed understanding of its role for stem cell transplantation is required. Methods We propose mechanistic computational models and perform virtual clinical trials to investigate clonal dynamics during and after allogenic hematopoietic stem cell transplantation. Different mechanisms of clonal expansion are considered, including mutation-related changes of stem cell proliferation and self-renewal, aberrant response of mutated cells to systemic signals, and self-sustaining chronic inflammation triggered by the mutated cells. Results Model simulations suggest that an aberrant response of mutated cells to systemic signals is sufficient to explain the frequently observed quick expansion of the mutated clone shortly after transplantation which is followed by a stabilization of the mutated cell number at a constant value. In contrary, a mutation-related increase of self-renewal or self-sustaining chronic inflammation lead to ongoing clonal expansion. Our virtual clinical trials suggest that a low number of transplanted stem cells per kg of body weight increases the transplantation-related expansion of donor-derived clones, whereas the transplanted progenitor dose or growth factor support after transplantation have no impact on clonal dynamics. Furthermore, in our simulations the change of the donors' variant allele frequencies in the year before stem cell donation is associated with the expansion of donor-derived clones in the recipient. Discussion This in silico study provides insights in the mechanisms leading to clonal expansion and identifies questions that could be addressed in future clinical trials.
Collapse
Affiliation(s)
- Thomas Stiehl
- Aachen Medical School, Institute for Computational Biomedicine & Disease Modeling,
RWTH Aachen University, Aachen, Germany
- Department for Science and Environment, Roskilde University,
Roskilde, Denmark
| |
Collapse
|
2
|
Patel SA, Spiegel JY, Dahiya S. Second Primary Cancer After Chimeric Antigen Receptor-T-Cell Therapy: A Review. JAMA Oncol 2024:2827886. [PMID: 39666320 DOI: 10.1001/jamaoncol.2024.5412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Importance The commercialization of chimeric antigen receptor-T-cell (CAR-T) therapy has changed the landscape of treatment of hematological cancers. Numerous studies from the early 2000s paved the way for cell-based targeted therapeutics, which have been established as practice-changing therapies in lymphoma, leukemia, and multiple myeloma. However, there has been some recent concern about the risk for second primary cancers (SPCs). Observations Multiple cases of SPCs arising after CAR-T therapy have been reported to the US Food and Drug Administration. Most SPCs have been negative for the chimeric antigen receptor transgene, with rare reports of transgene-positive cancers. This review summarizes the most salient literature on epidemiology and pathobiology of SPCs after CAR-T therapy. Additionally, a discussion is provided on potential mitigation strategies for SPCs after CAR-T therapies. Conclusions and Relevance The results of this review suggest that there are limited data to suggest that inadvertent transgene insertion is associated with SPCs in the post-CAR-T setting. Nonetheless, evidence-based practical solutions and scientific strategies for risk mitigation can be implemented. These include optimization of T-cell manufacturing, application of safer synthetic immunobiology, and implementation of high-fidelity genomic testing, including baseline screening for clonal hematopoiesis. These strategies may inform optimal design of the next generation of CAR-T products that confer minimal risk for SPCs such that the risk-benefit profile remains favorable to proceed with CAR-T administration for eligible patients.
Collapse
Affiliation(s)
- Shyam A Patel
- Division of Hematology and Oncology, Department of Medicine, Center for Clinical and Translational Science, UMass Chan Medical School, Worcester
| | - Jay Y Spiegel
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Saurabh Dahiya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
3
|
Xie Y, Kazakova V, Weeks LD, Gerber JM, Tai J, Zhang TY, Lowsky R, Wu X, Yang C, Patel SA. Effects of donor-engrafted clonal hematopoiesis in allogeneic and autologous stem cell transplantation: a systematic review and meta-analysis. Bone Marrow Transplant 2024; 59:1585-1593. [PMID: 39183321 PMCID: PMC11531373 DOI: 10.1038/s41409-024-02403-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Donor stem cell health may be critically important to the success of hematopoietic stem cell transplantation (HSCT). Herein, we performed this systematic review and meta-analysis including meta-regression to assess the impact of donor-engrafted clonal hematopoiesis (CH) in allogeneic HSCT (allo-HSCT) and impact of pre-transplant CH in autologous HSCT (auto-HSCT). We applied random-effects models to analyze 5 allo-HSCT studies with 3192 donor-recipient pairs and 9 auto-HSCT studies with 2854 patients. We found that donor-engrafted CH after allo-HSCT decreased the risk of disease relapse [Hazard Ratio (HR) = 0.79, 95% Confidence Interval (CI): (0.67, 0.93)], but did not affect overall survival (OS) [HR = 0.91, 95% CI: (0.75, 1.11)], progression-free survival (PFS) [HR = 0.94, 95% CI: (0.63, 1.41)], or non-relapse mortality [HR = 1.06, 95% CI: (0.81, 1.39)]. In contrast, pre-transplant CH in auto-HSCT recipients resulted in inferior OS [HR = 1.30, 95% CI: (1.16, 1.46)], inferior PFS [HR = 1.35, 95% CI: (1.18, 1.54)], and higher risk for therapy-related myeloid neoplasm [HR = 4.85, 95% CI: (2.39, 9.82)] when compared to auto-HSCT recipients without CH. This study sheds light onto the debate about prospective "CHIP screening" for stem cell donors and addresses the impact of CH as a transmissible phenomenon.
Collapse
Affiliation(s)
- Yiyu Xie
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA
| | - Vera Kazakova
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA
| | - Lachelle D Weeks
- Center for Early Detection and Interception of Blood Cancers, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jonathan M Gerber
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA
- Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA, USA
| | - Jesse Tai
- Dept. of Medicine - Division of Hematology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Tian Y Zhang
- Dept. of Medicine - Division of Hematology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert Lowsky
- Dept. of Medicine - Division of Blood & Marrow Transplantation & Cellular Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaojin Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chengwu Yang
- Measurement, Outcome, and Design Section, Division of Biostatistics and Health Service Research, Department of Population and Quantitative Health Sciences, T.H. Chan School of Medicine, UMass Chan Medical School, Worcester, MA, USA.
- Department of Obstetrics & Gynecology, T.H. Chan School of Medicine, UMass Chan Medical School, Worcester, MA, USA.
| | - Shyam A Patel
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA.
- Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Oshima MU, Higgins J, Jenkins I, Randolph T, Smith T, Valentine C, Salk J, Yeung C, Beppu L, Campbell J, Carpenter PA, Lee SJ, Flowers ME, Radich JP, Storb R. Characterization of clonal dynamics using duplex sequencing in donor-recipient pairs decades after hematopoietic cell transplantation. Sci Transl Med 2024; 16:eado5108. [PMID: 39441907 DOI: 10.1126/scitranslmed.ado5108] [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: 02/07/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
After allogeneic hematopoietic cell transplantation (HCT), a very small number of donor stem cells reconstitute the recipient hematopoietic system, whereas the donor is left with a near-normal pool of stem cells. We hypothesized that the increased replicative stress on transplanted donor cells in the recipient could lead to the disproportionate proliferation of clonal hematopoiesis (CH) variants. We obtained blood samples from 16 related donor-recipient pairs at a median of 33.8 years (range: 6.6 to 45.7) after HCT, including the longest surviving HCT recipients in the world. For 11 of 16 pairs, a donor sample from the time of HCT was available for comparison. We performed ultrasensitive duplex sequencing of genes recurrently mutated in myeloid malignancies and CH, as well as a set of functionally neutral genomic regions representative of human genomic content at large. CH variants were observed in all donors, even those as young as 12 years old. Where donor pre-HCT sample was available, the average mutation rate in donors compared to recipients post-HCT was similar (2.0% versus 2.6% per year, respectively) within genes recurrently mutated in myeloid malignancies. Twenty-two (5.6%) of the 393 variants shared between paired donors and recipients post-HCT showed ≥10-fold higher variant allele frequency (VAF) in the recipient. A longer time since HCT was positively associated with the expansion of shared variant VAFs in the recipient. In conclusion, even decades after HCT, there does not appear to be widespread accelerated clonal expansion in the transplanted cells, highlighting the immense regenerative capacity of the human hematopoietic system.
Collapse
Affiliation(s)
- Masumi Ueda Oshima
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Isaac Jenkins
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Timothy Randolph
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | | | | | - Jesse Salk
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- TwinStrand Biosciences, Seattle, WA 98121, USA
| | - Cecilia Yeung
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lan Beppu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Judy Campbell
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Mary E Flowers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jerald P Radich
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Rainer Storb
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Hematology & Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
5
|
Hamad L, Chekar CK, Anthias C, Machin L. Facilitating the ethical sourcing of donor hematopoietic stem cells for cell and gene therapy research and development. Regen Med 2024; 19:317-326. [PMID: 38889086 PMCID: PMC11346549 DOI: 10.1080/17460751.2024.2357930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Aim: Unrelated stem cell donor registries (DRs) are increasingly engaging in the field of cell and gene therapy (CGT). This study aims to explore the values, concerns, needs and expectations of donors and members of the public on donating hematopoietic stem cells (HSCs) for CGT.Methods: Seven focus groups were conducted in 2019 with members of the public, prospective donors and donors on the Anthony Nolan DR in the UK.Results: Participants expressed concerns over increased frequency of donation and incidental findings and required more information on the type of research including the purpose and possible outcomes.Conclusion: Addressing donors' concerns, needs and expectations on donating cellular materials for CGT research and development is essential to maintaining the highest standards for donor care and safety within this rapidly emerging field.
Collapse
Affiliation(s)
- Lina Hamad
- Lancaster Medical School, Lancaster University, Lancaster, UK
| | | | - Chloe Anthias
- Anthony Nolan, London, UK
- The Institute of Cancer Research, London, UK
| | - Laura Machin
- Lancaster Medical School, Lancaster University, Lancaster, UK
- Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
6
|
Munshi PN, McCurdy SR. Age barriers in allogeneic hematopoietic cell transplantation: Raising the silver curtain. Am J Hematol 2024; 99:922-937. [PMID: 38414188 DOI: 10.1002/ajh.27228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/13/2023] [Accepted: 01/01/2024] [Indexed: 02/29/2024]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is no longer exclusively for the young. With an aging population, development of non-intensive remission-inducing strategies for hematologic malignancies, and novel graft-versus-host disease-prevention platforms, an older population of patients is pursuing HCT. The evolving population of HCT recipients requires an overhaul in the way we risk-stratify and optimize patients prior to HCT. Here, we review the history and current state of HCT for older adults and propose an assessment and intervention flow to bridge the gaps in today's clinical guidelines.
Collapse
Affiliation(s)
- Pashna N Munshi
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shannon R McCurdy
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Hoffmann M, Banz Y, Halter J, Schoumans J, Tchinda J, Bacher U, Pabst T. True Donor Cell Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation: Diagnostic and Therapeutic Considerations-Brief Report. Curr Oncol 2024; 31:2067-2075. [PMID: 38668056 PMCID: PMC11049222 DOI: 10.3390/curroncol31040153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Donor cell leukemia (DCL) is a rare complication after allogeneic hematopoietic stem cell transplantation (HSCT) accounting for 0.1% of relapses and presenting as secondary leukemia of donor origin. Distinct in phenotype and cytogenetics from the original leukemia, DCL's clinical challenge lies in its late onset. Its origin is affected by donor cell anomalies, transplant environment, and additional mutations. A 43-year-old woman, treated for early stage triple-negative breast cancer, developed mixed-phenotype acute leukemia (MPAL), 12 years later. Following induction chemotherapy, myeloablative conditioning, and allo-HSCT from her fully HLA-matched brother, she exhibited multiple cutaneous relapses of the original leukemia, subsequently evolving into DCL of the bone marrow. Cytogenetic analysis revealed a complex male karyotype in 20 out of 21 metaphases, however, still showing the MPAL phenotype. DCL diagnosis was confirmed by 90.5% XY in FISH analysis and the male karyotype. Declining further intensive chemotherapy including a second allo-HSCT, she was subsequently treated with repeated radiotherapy, palliative systemic therapies, and finally venetoclax and navitoclax but died seven months post-DCL diagnosis. This case underlines DCL's complexity, characterized by unique genetics, further complicating diagnosis. It highlights the need for advanced diagnostic techniques for DCL identification and underscores the urgency for early detection and better prevention and treatment strategies.
Collapse
Affiliation(s)
- Michèle Hoffmann
- Department of Medical Oncology, Inselspital Bern, Bern University Hospital, University of Bern, 3012 Bern, Switzerland;
| | - Yara Banz
- Institute of Tissue Medicine and Pathology, University of Bern, 3012 Bern, Switzerland;
| | - Jörg Halter
- Division of Hematology, University Hospital Basel, 4031 Basel, Switzerland;
| | - Jacqueline Schoumans
- Oncogenomic Laboratory, Service and Central Laboratory of Hematology, University Hospital Lausanne (CHUV), 1005 Lausanne, Switzerland;
| | - Joëlle Tchinda
- Laboratory for Oncology, University Children’s Hospital Zürich, 8032 Zürich, Switzerland;
- Department of Hematology and Central Laboratory, Inselspital Bern, Bern University Hospital, University of Bern, 3012 Bern, Switzerland;
| | - Ulrike Bacher
- Department of Hematology and Central Laboratory, Inselspital Bern, Bern University Hospital, University of Bern, 3012 Bern, Switzerland;
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital Bern, Bern University Hospital, University of Bern, 3012 Bern, Switzerland;
| |
Collapse
|
8
|
Gibson CJ, Lindsley RC, Gondek LP. Clonal hematopoiesis in the setting of hematopoietic cell transplantation. Semin Hematol 2024; 61:9-15. [PMID: 38429201 PMCID: PMC10978245 DOI: 10.1053/j.seminhematol.2024.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 03/03/2024]
Abstract
Clonal hematopoiesis (CH) in autologous transplant recipients and allogeneic transplant donors has genetic features and clinical associations that are distinct from each other and from non-cancer populations. CH in the setting of autologous transplant is enriched for mutations in DNA damage response pathway genes and is associated with adverse outcomes, including an increased risk of therapy-related myeloid neoplasm and inferior overall survival. Studies of CH in allogeneic transplant donors have yielded conflicting results but have generally shown evidence of potentiated alloimmunity in recipients, with some studies showing an association with favorable recipient outcomes.
Collapse
Affiliation(s)
| | - R Coleman Lindsley
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Lukasz P Gondek
- Division of Hematologic Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
9
|
Pryce A, Van Eerden E, Cody M, Oakes J, DeSalvo A, Bannon S, Burlton C, Pawson R, Fingrut W, Barriga F, Ward J, Ingram C, Walsh M, El-Ghariani K, Ocheni S, Machin L, Allan D, Mengling T, Anthias C. Genetic Findings of Potential Donor Origin following Hematopoietic Cell Transplantation: Recommendations on Donor Disclosure and Genetic Testing from the World Marrow Donor Association. Transplant Cell Ther 2024; 30:143-154. [PMID: 38056629 DOI: 10.1016/j.jtct.2023.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/23/2023] [Accepted: 11/23/2023] [Indexed: 12/08/2023]
Abstract
Following hematopoietic cell transplantation (HCT), recipients are subjected to extensive genetic testing to monitor the efficacy of the transplantation and identify relapsing malignant disease. This testing is increasingly including the use of large gene panels, which may lead to incidental identification of genetic and molecular information of potential donor origin. Deciphering whether variants are of donor origin, and if so, whether there are clinical implications for the donor can prove challenging. In response to queries from donor registries and transplant centers regarding best practices in managing donors when genetic mutations of potential donor origin are identified, the Medical Working Group of the World Marrow Donor Association established an expert group to review available evidence and develop a framework to aid decision making. These guidelines aim to provide recommendations on predonation consenting, postdonation testing of recipients, and informing and managing donors when findings of potential donor origin are identified in recipients post-transplantation. It is recognized that registries will have different access to resources and financing structures, and thus whenever possible, we have made suggestions on how recommendations can be adapted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jane Ward
- South African Bone Marrow Registry, Cape Town, South Africa
| | | | | | | | | | - Laura Machin
- Lancaster University, United Kingdom; Imperial College London, London, United Kingdom
| | | | | | - Chloe Anthias
- Anthony Nolan, London, United Kingdom; Royal Marsden Hospital, London, United Kingdom.
| |
Collapse
|
10
|
Kim KH, Kim T, Novitzky-Basso I, Lee H, Yoo Y, Ahn JS, Pasic I, Law A, Lam W, Michelis FV, Gerbitz A, Viswabandya A, Lipton J, Kumar R, Mattsson J, Zhang Z, Kaushansky N, Brilon Y, Chapal-Ilani N, Biezuner T, Shlush LI, Kim DDH. Clonal hematopoiesis in the donor does not adversely affect long-term outcomes following allogeneic hematopoietic stem cell transplantation: result from a 13-year follow-up. Haematologica 2023; 108:1817-1826. [PMID: 36727396 PMCID: PMC10316278 DOI: 10.3324/haematol.2022.281806] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Donor clonal hematopoiesis may be transferred to the recipient through allogeneic hematopoietic stem cell transplantation (HSCT), but the potential for adverse long-term impact on transplant outcomes remains unknown. A total of 744 samples from 372 recipients who received HSCT and the corresponding donors were included. Bar-coded error-corrected sequencing using a modified molecular inversion probe capture protocol was performed, which targeted 33 genes covering mutations involved in clonal hematopoiesis with indeterminate potential (CHIP) and other acute myeloid leukemia-related mutations. A total of 30 mutations were detected from 25 donors (6.7%): the most frequently mutated gene was TET2 (n=7, 28%), followed by DNMT3A (n=4, 16%), SMC3 (n=3, 12%) and SF3B1 (n=3, 12%). With a median follow-up duration of 13 years among survivors, the presence of CHIP in the donor was not associated with recipient overall survival (P=0.969), relapse incidence (P=0.600) or non-relapse mortality (P=0.570). Donor CHIP did not impair neutrophil (P=0.460) or platelet (P=0.250) engraftment, the rates of acute (P=0.490), or chronic graft-versus-host disease (P=0.220). No significant difference was noted for secondary malignancy following HSCT between the two groups. The present study suggests that the presence of CHIP in allogeneic stem donors does not adversely affect transplant outcomes after HSCT. Accordingly, further study is warranted to reach a clearer conclusion on whether molecular profiling to determine the presence of CHIP mutations is necessary for the pretransplant evaluation of donors prior to stem cell donation.
Collapse
Affiliation(s)
- Kyoung Ha Kim
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Hospital, Seoul
| | - TaeHyung Kim
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Computer Science, University of Toronto, Toronto, ON, Canada; The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON
| | - Igor Novitzky-Basso
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Hyewon Lee
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada; Division of Rare and Refractory Cancer, Division of Hemato-Oncology, and Center for Hematologic Malignancy Research Institute and Hospital National Cancer Center
| | - Youngseok Yoo
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Jae-Sook Ahn
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada; Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University, Gwangju
| | - Ivan Pasic
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Arjun Law
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Wilson Lam
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Fotios V Michelis
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Armin Gerbitz
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Auro Viswabandya
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Jeffrey Lipton
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Rajat Kumar
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto
| | - Jonas Mattsson
- Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation
| | - Zhaolei Zhang
- Department of Computer Science, University of Toronto, Toronto, ON, Canada; The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON
| | | | - Yardena Brilon
- Department of Immunology, Weizmann Institute of Science, Rehovot
| | - Noa Chapal-Ilani
- Department of Immunology, Weizmann Institute of Science, Rehovot
| | - Tamir Biezuner
- Department of Immunology, Weizmann Institute of Science, Rehovot
| | - Liran I Shlush
- Department of Immunology, Weizmann Institute of Science, Rehovot.
| | - Dennis Dong Hwan Kim
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada; Institute for Medical Science, Faculty of Medicine, University of Toronto, Toronto.
| |
Collapse
|
11
|
Khattab A, Patruni S, Patrus G, Samhouri Y, Fazal S, Lister J. Donor Cell Leukemia Following Allogeneic Hematopoietic Stem Cell Transplantation. J Hematol 2023; 12:138-144. [PMID: 37435412 PMCID: PMC10332864 DOI: 10.14740/jh1124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/12/2023] [Indexed: 07/13/2023] Open
Abstract
Approximately 25,000 allogeneic transplants are performed annually worldwide; a figure that has steadily increased over the past three decades. The study of transplant recipient survivorship has become a cogent topic and post-transplant donor cell pathology warrants further study. Donor cell leukemia (DCL) is a rare but serious complication of allogeneic stem cell transplantation (SCT) where the recipient develops a form leukemia originating from the donor cells used for transplantation. Detection of abnormalities predicting donor cell pathology might inform donor selection, and the design of survivorship programs for early detection of these abnormalities might allow therapeutic intervention earlier in the disease course. We present four recipients of allogeneic hematopoietic stem cell transplant (HSCT) from our institution who developed donor cell abnormalities allogeneic SCT, highlighting their clinical characteristics and challenges.
Collapse
Affiliation(s)
- Ahmed Khattab
- Department of Medical Oncology and Hematology, Allegheny Health Network, Pittsburgh, PA, USA
- Division of Hematology and Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA
| | - Sunita Patruni
- Department of Medical Oncology and Hematology, Northwell Health, Lake Success, NY, USA
| | - Gina Patrus
- Division of Hematology and Cellular Therapy, Allegheny Health Network, Pittsburgh, PA, USA
| | - Yazan Samhouri
- Division of Hematology and Cellular Therapy, Allegheny Health Network, Pittsburgh, PA, USA
| | - Salman Fazal
- Division of Hematology and Cellular Therapy, Allegheny Health Network, Pittsburgh, PA, USA
| | - John Lister
- Division of Hematology and Cellular Therapy, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Lin C, Schwarzbach A, Sanz J, Montesinos P, Stiff P, Parikh S, Brunstein C, Cutler C, Lindemans CA, Hanna R, Koh LP, Jagasia MH, Valcarcel D, Maziarz RT, Keating AK, Hwang WYK, Rezvani AR, Karras NA, Fernandes JF, Rocha V, Badell I, Ram R, Schiller GJ, Volodin L, Walters MC, Hamerschlak N, Cilloni D, Frankfurt O, McGuirk JP, Kurtzberg J, Sanz G, Simantov R, Horwitz ME. Multicenter Long-Term Follow-Up of Allogeneic Hematopoietic Cell Transplantation with Omidubicel: A Pooled Analysis of Five Prospective Clinical Trials. Transplant Cell Ther 2023; 29:338.e1-338.e6. [PMID: 36775201 PMCID: PMC10149622 DOI: 10.1016/j.jtct.2023.01.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
Omidubicel is an umbilical cord blood (UCB)-derived ex vivo-expanded cellular therapy product that has demonstrated faster engraftment and fewer infections compared with unmanipulated UCB in allogeneic hematopoietic cell transplantation. Although the early benefits of omidubicel have been established, long-term outcomes remain unknown. We report on a planned pooled analysis of 5 multicenter clinical trials including 105 patients with hematologic malignancies or sickle cell hemoglobinopathy who underwent omidubicel transplantation at 26 academic transplantation centers worldwide. With a median follow-up of 22 months (range, .3 to 122 months), the 3-year estimated overall survival and disease-free survival were 62.5% and 54.0%, respectively. With up to 10 years of follow-up, omidubicel showed durable trilineage hematopoiesis. Serial quantitative assessments of CD3+, CD4+, CD8+, CD19+, CD116+CD56+, and CD123+ immune subsets revealed median counts remaining within normal ranges through up to 8 years of follow-up. Secondary graft failure occurred in 5 patients (5%) in the first year, with no late cases reported. One case of donor-derived myeloid neoplasm was reported at 40 months post-transplantation. This was also observed in a control arm patient who received only unmanipulated UCB. Overall, omidubicel demonstrated stable trilineage hematopoiesis, immune competence, and graft durability in extended follow-up.
Collapse
Affiliation(s)
- Chenyu Lin
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | | | - Jaime Sanz
- Hematology Department, Hospital Universitario y Polit
| | | | - Patrick Stiff
- Division of Hematology and Oncology, Loyola University Medical Center, Chicago, Illinois
| | | | - Claudio Brunstein
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota; Department of Hematology and Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, Ohio
| | - Corey Cutler
- Division of Stem Cell Transplantation and Cellular Therapies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Caroline A Lindemans
- Department of Pediatric Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rabi Hanna
- Department of Pediatric Hematology Oncology and Bone Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Liang Piu Koh
- Department of Hematology-Oncology, National University Cancer Institute, Singapore
| | - Madan H Jagasia
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David Valcarcel
- Department of Haematology and Haemotherapy, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Richard T Maziarz
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Amy K Keating
- Blood and Marrow Transplantation, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - William Y K Hwang
- Department of Haematology, National Cancer Centre Singapore, Singapore; Department of Haematology, Singapore General Hospital, Singapore; Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Andrew R Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Nicole A Karras
- Department of Pediatrics, City of Hope National Medical Center, Duarte, California
| | | | | | - Isabel Badell
- Pediatric Haematology and Stem Cell Transplantation Unit, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Ron Ram
- BMT Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gary J Schiller
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Leonid Volodin
- Division of Hematology and Oncology, University of Virginia, Charlottesville, Virginia
| | - Mark C Walters
- Benioff Children's Hospital, University of California San Francisco, Oakland, California
| | | | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Olga Frankfurt
- Division of Hematology and Oncology, Northwestern University, Chicago, Illinois
| | - Joseph P McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Joanne Kurtzberg
- Division of Hematology-Oncology, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Guillermo Sanz
- Hematology Department, Hospital Universitario y Polit; Health Reserach Institute La Fe, Valencia, Spain; CIBERONC, ISCIII, Madrid, Spain
| | | | - Mitchell E Horwitz
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
13
|
Xie Z, Zeidan AM. CHIPing away the progression potential of CHIP: A new reality in the making. Blood Rev 2023; 58:101001. [PMID: 35989137 DOI: 10.1016/j.blre.2022.101001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/15/2022]
Abstract
Over the past few years, we have gained a deeper understanding of clonal hematopoiesis of indeterminate potential (CHIP), especially with regard to the epidemiology, clinical sequelae, and mechanical aspects. However, interventional strategies to prevent or delay the potential negative consequences of CHIP remain underdeveloped. In this review, we highlight the latest updates on clonal hematopoiesis research, including molecular mechanisms and clinical implications, with a particular focus on the evolving strategies for the interventions that are being evaluated in ongoing observational and interventional trials. There remains an urgent need to formulate standardized and evidence-based recommendations and guidelines for evaluating and managing individuals with clonal hematopoiesis. In addition, patient-centric endpoints must be defined for clinical trials, which will enable us to continue the robust development of effective preventive strategies and improve clinical outcomes.
Collapse
Affiliation(s)
- Zhuoer Xie
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center and Smilow Cancer Hospital, Yale University School of Medicine, CT, United States.
| |
Collapse
|
14
|
Testa U, Castelli G, Pelosi E. Clonal Hematopoiesis: Role in Hematologic and Non-Hematologic Malignancies. Mediterr J Hematol Infect Dis 2022; 14:e2022069. [PMID: 36119457 PMCID: PMC9448266 DOI: 10.4084/mjhid.2022.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/18/2022] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cells (HSCs) ensure the coordinated and balanced production of all hematopoietic cell types throughout life. Aging is associated with a gradual decline of the self-renewal and regenerative potential of HSCs and with the development of clonal hematopoiesis. Clonal hematopoiesis of indeterminate potential (CHIP) defines the clonal expansion of genetically variant hematopoietic cells bearing one or more gene mutations and/or structural variants (such as copy number alterations). CHIP increases exponentially with age and is associated with cancers, including hematologic neoplasia, cardiovascular and other diseases. The presence of CHIP consistently increases the risk of hematologic malignancy, particularly in individuals who have CHIP in association with peripheral blood cytopenia.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
15
|
Ye B, Sheng Y, Zhang M, Hu Y, Huang H. Early detection and intervention of clonal hematopoiesis for preventing hematological malignancies. Cancer Lett 2022; 538:215691. [DOI: 10.1016/j.canlet.2022.215691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/03/2022] [Accepted: 04/17/2022] [Indexed: 12/17/2022]
|
16
|
Menendez-Gonzalez JB, Rodrigues NP. Exploring the Associations Between Clonal Hematopoiesis of Indeterminate Potential, Myeloid Malignancy, and Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:73-88. [PMID: 35237959 DOI: 10.1007/978-1-0716-1924-7_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Outgrowth of a mutated hematopoietic stem/progenitor clone and its descendants, also known as clonal hematopoiesis, has long been considered as either a potential forerunner to hematologic malignancy or as a clinically silent phase in leukemia that antedates symptomatic disease. That definition of clonal hematopoiesis has now been expanded to encompass patients who harbor specific genetic/epigenetic mutations that lead to clonal hematopoiesis of indeterminate potential (CHIP) and, with it, a relatively heightened risk for both myeloid malignancy and atherosclerosis during aging. In this review, we provide contemporary insights into the cellular and molecular basis for CHIP and explore the relationship of CHIP to myeloid malignancy and atherosclerosis. We also discuss emerging strategies to explore CHIP biology and clinical targeting of CHIP related malignancy and cardiovascular disease.
Collapse
Affiliation(s)
- Juan Bautista Menendez-Gonzalez
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Neil P Rodrigues
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
17
|
Gibson CJ, Kim HT, Zhao L, Murdock HM, Hambley B, Ogata A, Madero-Marroquin R, Wang S, Green L, Fleharty M, Dougan T, Cheng CA, Blumenstiel B, Cibulskis C, Tsuji J, Duran M, Gocke CD, Antin JH, Nikiforow S, DeZern AE, Chen YB, Ho VT, Jones RJ, Lennon NJ, Walt DR, Ritz J, Soiffer RJ, Gondek LP, Lindsley RC. Donor Clonal Hematopoiesis and Recipient Outcomes After Transplantation. J Clin Oncol 2022; 40:189-201. [PMID: 34793200 PMCID: PMC8718176 DOI: 10.1200/jco.21.02286] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Clonal hematopoiesis (CH) can be transmitted from a donor to a recipient during allogeneic hematopoietic cell transplantation. Exclusion of candidate donors with CH is controversial since its impact on recipient outcomes and graft alloimmune function is uncertain. PATIENTS AND METHODS We performed targeted error-corrected sequencing on samples from 1,727 donors age 40 years or older and assessed the effect of donor CH on recipient clinical outcomes. We measured long-term engraftment of 102 donor clones and cytokine levels in 256 recipients at 3 and 12 months after transplant. RESULTS CH was present in 22.5% of donors, with DNMT3A (14.6%) and TET2 (5.2%) mutations being most common; 85% of donor clones showed long-term engraftment in recipients after transplantation, including clones with a variant allele fraction < 0.01. DNMT3A-CH with a variant allele fraction ≥ 0.01, but not smaller clones, was associated with improved recipient overall (hazard ratio [HR], 0.79; P = .042) and progression-free survival (HR, 0.72; P = .003) after adjustment for significant clinical variables. In patients who received calcineurin-based graft-versus-host disease prophylaxis, donor DNMT3A-CH was associated with reduced relapse (subdistribution HR, 0.59; P = .014), increased chronic graft-versus-host disease (subdistribution HR, 1.36; P = .042), and higher interleukin-12p70 levels in recipients. No recipient of sole DNMT3A or TET2-CH developed donor cell leukemia (DCL). In seven of eight cases, DCL evolved from donor CH with rare TP53 or splicing factor mutations or from donors carrying germline DDX41 mutations. CONCLUSION Donor CH is closely associated with clinical outcomes in transplant recipients, with differential impact on graft alloimmune function and potential for leukemic transformation related to mutated gene and somatic clonal abundance. Donor DNMT3A-CH is associated with improved recipient survival because of reduced relapse risk and with an augmented network of inflammatory cytokines in recipients. Risk of DCL in allogeneic hematopoietic cell transplantation is driven by somatic myelodysplastic syndrome-associated mutations or germline predisposition in donors.
Collapse
Affiliation(s)
- Christopher J. Gibson
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Haesook T. Kim
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA
| | - Lin Zhao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
- Department of Hematology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - H. Moses Murdock
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Bryan Hambley
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Alana Ogata
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | | | - Shiyu Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Lisa Green
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Mark Fleharty
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Tyler Dougan
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Chi-An Cheng
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | | | - Carrie Cibulskis
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Junko Tsuji
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Madeleine Duran
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Christopher D. Gocke
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University, Baltimore, MD
| | - Joseph H. Antin
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Sarah Nikiforow
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Amy E. DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cell Therapy Program, Massachusetts General Hospital, Boston, MA
| | - Vincent T. Ho
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Richard J. Jones
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Niall J. Lennon
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - David R. Walt
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Jerome Ritz
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Robert J. Soiffer
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Lukasz P. Gondek
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - R. Coleman Lindsley
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
18
|
Gibson CJ, Kim HT, Zhao L, Murdock HM, Hambley B, Ogata A, Madero-Marroquin R, Wang S, Green L, Fleharty M, Dougan T, Cheng CA, Blumenstiel B, Cibulskis C, Tsuji J, Duran M, Gocke CD, Antin JH, Nikiforow S, DeZern AE, Chen YB, Ho VT, Jones RJ, Lennon NJ, Walt DR, Ritz J, Soiffer RJ, Gondek LP, Lindsley RC. Donor Clonal Hematopoiesis and Recipient Outcomes After Transplantation. J Clin Oncol 2022. [PMID: 34793200 DOI: 10.1200/jco.2021.39.15suppl.e16213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
PURPOSE Clonal hematopoiesis (CH) can be transmitted from a donor to a recipient during allogeneic hematopoietic cell transplantation. Exclusion of candidate donors with CH is controversial since its impact on recipient outcomes and graft alloimmune function is uncertain. PATIENTS AND METHODS We performed targeted error-corrected sequencing on samples from 1,727 donors age 40 years or older and assessed the effect of donor CH on recipient clinical outcomes. We measured long-term engraftment of 102 donor clones and cytokine levels in 256 recipients at 3 and 12 months after transplant. RESULTS CH was present in 22.5% of donors, with DNMT3A (14.6%) and TET2 (5.2%) mutations being most common; 85% of donor clones showed long-term engraftment in recipients after transplantation, including clones with a variant allele fraction < 0.01. DNMT3A-CH with a variant allele fraction ≥ 0.01, but not smaller clones, was associated with improved recipient overall (hazard ratio [HR], 0.79; P = .042) and progression-free survival (HR, 0.72; P = .003) after adjustment for significant clinical variables. In patients who received calcineurin-based graft-versus-host disease prophylaxis, donor DNMT3A-CH was associated with reduced relapse (subdistribution HR, 0.59; P = .014), increased chronic graft-versus-host disease (subdistribution HR, 1.36; P = .042), and higher interleukin-12p70 levels in recipients. No recipient of sole DNMT3A or TET2-CH developed donor cell leukemia (DCL). In seven of eight cases, DCL evolved from donor CH with rare TP53 or splicing factor mutations or from donors carrying germline DDX41 mutations. CONCLUSION Donor CH is closely associated with clinical outcomes in transplant recipients, with differential impact on graft alloimmune function and potential for leukemic transformation related to mutated gene and somatic clonal abundance. Donor DNMT3A-CH is associated with improved recipient survival because of reduced relapse risk and with an augmented network of inflammatory cytokines in recipients. Risk of DCL in allogeneic hematopoietic cell transplantation is driven by somatic myelodysplastic syndrome-associated mutations or germline predisposition in donors.
Collapse
Affiliation(s)
- Christopher J Gibson
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Haesook T Kim
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA
| | - Lin Zhao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD.,Department of Hematology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - H Moses Murdock
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Bryan Hambley
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Alana Ogata
- Department of Pathology, Brigham and Women's Hospital, Boston, MA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | | | - Shiyu Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Lisa Green
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Mark Fleharty
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Tyler Dougan
- Department of Pathology, Brigham and Women's Hospital, Boston, MA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Chi-An Cheng
- Department of Pathology, Brigham and Women's Hospital, Boston, MA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | | | - Carrie Cibulskis
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Junko Tsuji
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Madeleine Duran
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Christopher D Gocke
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD.,Division of Molecular Pathology, Department of Pathology, Johns Hopkins University, Baltimore, MD
| | - Joseph H Antin
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Sarah Nikiforow
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cell Therapy Program, Massachusetts General Hospital, Boston, MA
| | - Vincent T Ho
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Richard J Jones
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Niall J Lennon
- Genomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - David R Walt
- Department of Pathology, Brigham and Women's Hospital, Boston, MA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA
| | - Jerome Ritz
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Robert J Soiffer
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Lukasz P Gondek
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - R Coleman Lindsley
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
19
|
Roloff GW, Drazer MW, Godley LA. Inherited Susceptibility to Hematopoietic Malignancies in the Era of Precision Oncology. JCO Precis Oncol 2022; 5:107-122. [PMID: 34994594 DOI: 10.1200/po.20.00387] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
As germline predisposition to hematopoietic malignancies has gained increased recognition and attention in the field of oncology, it is important for clinicians to use a systematic framework for the identification, management, and surveillance of patients with hereditary hematopoietic malignancies (HHMs). In this article, we discuss strategies for identifying individuals who warrant diagnostic evaluation and describe considerations pertaining to molecular testing. Although a paucity of prospective data is available to guide clinical monitoring of individuals harboring pathogenic variants, we provide recommendations for clinical surveillance based on consensus opinion and highlight current advances regarding the risk of progression to overt malignancy in HHM variant carriers. We also discuss the prognosis of HHMs and considerations surrounding the utility of allogeneic stem-cell transplantation in these individuals. We close with an overview of contemporary issues at the intersection of HHMs and precision oncology.
Collapse
Affiliation(s)
- Gregory W Roloff
- Department of Medicine, Loyola University Medical Center, Maywood, IL
| | - Michael W Drazer
- Section of Hematology/Oncology, Department of Medicine and the Department of Human Genetics, the University of Chicago, Chicago, IL
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine and the Department of Human Genetics, the University of Chicago, Chicago, IL
| |
Collapse
|
20
|
Kusne Y, Xie Z, Patnaik MM. Clonal Hematopoiesis: Molecular and Clinical Implications. Leuk Res 2022; 113:106787. [DOI: 10.1016/j.leukres.2022.106787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 11/16/2022]
|
21
|
Newell LF, Dunlap J, Gatter K, Bagby GC, Press RD, Cook RJ, Fletcher L, Leonard JT, Leong KM, Bubalo JS, Olyaei A, Deloughery TG, Maziarz RT, Maynard E, Orloff SL, Enestvedt CK. Graft-versus-host disease after liver transplantation is associated with bone marrow failure, hemophagocytosis, and DNMT3A mutations. Am J Transplant 2021; 21:3894-3906. [PMID: 33961341 DOI: 10.1111/ajt.16635] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/16/2021] [Accepted: 04/30/2021] [Indexed: 01/25/2023]
Abstract
Graft-versus-host disease after liver transplantation (LT-GVHD) is rare, frequently fatal, and associated with bone marrow failure (BMF), cytopenias, and hyperferritinemia. Given hyperferritinemia and cytopenias are present in hemophagocytic lymphohistiocytosis (HLH), and somatic mutations in hematopoietic cells are associated with hyperinflammatory responses (clonal hematopoiesis of indeterminate potential, CHIP), we identified the frequency of hemophagocytosis and CHIP mutations in LT-GVHD. We reviewed bone marrow aspirates and biopsies, quantified blood/marrow chimerism, and performed next-generation sequencing (NGS) with a targeted panel of genes relevant to myeloid malignancies, CHIP, and BMF. In all, 12 marrows were reviewed from 9 LT-GVHD patients. In all, 10 aspirates were evaluable for hemophagocytosis; 7 had adequate DNA for NGS. NGS was also performed on marrow from an LT cohort (n = 6) without GVHD. Nine of 10 aspirates in LT-GVHD patients showed increased hemophagocytosis. Five (71%) of 7 with LT-GVHD had DNMT3A mutations; only 1 of 6 in the non-GVHD LT cohort demonstrated DNMT3A mutation (p = .04). Only 1 LT-GVHD patient survived. BMF with HLH features was associated with poor hematopoietic recovery, and DNMT3A mutations were over-represented, in LT-GVHD patients. Identification of HLH features may guide prognosis and therapeutics. Further studies are needed to clarify the origin and impact of CHIP mutations on the hyperinflammatory state.
Collapse
Affiliation(s)
- Laura F Newell
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Jennifer Dunlap
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Ken Gatter
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Grover C Bagby
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Richard D Press
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Rachel J Cook
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Luke Fletcher
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Jessica T Leonard
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Kelli M Leong
- Department of Pharmacy, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph S Bubalo
- Department of Pharmacy, Oregon Health & Science University, Portland, Oregon, USA
| | - Ali Olyaei
- Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, Oregon, USA
| | - Thomas G Deloughery
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Richard T Maziarz
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, USA
| | - Erin Maynard
- Department of Surgery, Division of Abdominal Organ Transplantation, Oregon Health & Science University, Portland, Oregon, USA
| | - Susan L Orloff
- Department of Surgery, Division of Abdominal Organ Transplantation, Oregon Health & Science University, Portland, Oregon, USA
| | - C Kristian Enestvedt
- Department of Surgery, Division of Abdominal Organ Transplantation, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
22
|
Köhnke T, Majeti R. Clonal Hematopoiesis: From Mechanisms to Clinical Intervention. Cancer Discov 2021; 11:2987-2997. [PMID: 34407958 PMCID: PMC8854454 DOI: 10.1158/2159-8290.cd-21-0901] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022]
Abstract
Our knowledge of how clonal hematopoiesis relates to diverse health conditions has grown vastly over the past years, touching upon many specialties beyond cancer medicine. Given that clonal hematopoiesis can act as a precursor to overt disease in many settings, the promise of early intervention has garnered much attention. In this review, we discuss the state of clonal hematopoiesis research and outline the challenges in developing clinical trials of early interventions. We anticipate that incidental findings of clonal hematopoiesis will become more common in the near future, but evidence-based efforts of how to manage these findings is currently lacking. SIGNIFICANCE Our knowledge regarding the relevance of clonal hematopoiesis has increased drastically over the past years. However, evidence of how to manage these findings is currently lacking. In this review, we summarize the current state of clonal hematopoiesis research and outline the challenges of developing clinical trials in this field. We anticipate that incidental findings of clonal hematopoiesis will become more common in the near future and argue that there is urgency to start designing and conducting prospective trials.
Collapse
Affiliation(s)
- Thomas Köhnke
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Lead contact
| |
Collapse
|
23
|
Kennedy VE, Olin RL. Haematopoietic stem-cell transplantation in older adults: geriatric assessment, donor considerations, and optimisation of care. LANCET HAEMATOLOGY 2021; 8:e853-e861. [PMID: 34624239 DOI: 10.1016/s2352-3026(21)00231-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
Haematopoietic stem-cell transplantation (HSCT) has seen substantial growth among older adults. Chronological age is no longer viewed as an absolute barrier to HSCT, and alternative methods for assessing pre-transplantation fitness are increasingly used. In this Series paper, we summarise the metrics for pre-transplantation risk assessment in older adults, including both traditional metrics and geriatric assessment, and the ability of these metrics to predict post-transplantation outcomes. We also discuss strategies to broaden the utility of geriatric assessment, including in chronologically younger HSCT candidates and to guide individualised pre-transplantation interventions. Finally, we discuss donor considerations in older adults, including use of older sibling donors, haploidentical donors, and emerging data for donor-associated clonal haematopoiesis of indeterminate potential.
Collapse
Affiliation(s)
- Vanessa E Kennedy
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Rebecca L Olin
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Barabino SML, Citterio E, Ronchi AE. Transcription Factors, R-Loops and Deubiquitinating Enzymes: Emerging Targets in Myelodysplastic Syndromes and Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13153753. [PMID: 34359655 PMCID: PMC8345071 DOI: 10.3390/cancers13153753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The advent of DNA massive sequencing technologies has allowed for the first time an extensive look into the heterogeneous spectrum of genes and mutations underpinning myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML). In this review, we wish to explore the most recent advances and the rationale for the potential therapeutic interest of three main actors in myelo-leukemic transformation: transcription factors that govern myeloid differentiation; RNA splicing factors, which ensure proper mRNA maturation and whose mutations increase R-loops formation; and deubiquitinating enzymes, which contribute to genome stability in hematopoietic stem cells (HSCs). Abstract Myeloid neoplasms encompass a very heterogeneous family of diseases characterized by the failure of the molecular mechanisms that ensure a balanced equilibrium between hematopoietic stem cells (HSCs) self-renewal and the proper production of differentiated cells. The origin of the driver mutations leading to preleukemia can be traced back to HSC/progenitor cells. Many properties typical to normal HSCs are exploited by leukemic stem cells (LSCs) to their advantage, leading to the emergence of a clonal population that can eventually progress to leukemia with variable latency and evolution. In fact, different subclones might in turn develop from the original malignant clone through accumulation of additional mutations, increasing their competitive fitness. This process ultimately leads to a complex cancer architecture where a mosaic of cellular clones—each carrying a unique set of mutations—coexists. The repertoire of genes whose mutations contribute to the progression toward leukemogenesis is broad. It encompasses genes involved in different cellular processes, including transcriptional regulation, epigenetics (DNA and histones modifications), DNA damage signaling and repair, chromosome segregation and replication (cohesin complex), RNA splicing, and signal transduction. Among these many players, transcription factors, RNA splicing proteins, and deubiquitinating enzymes are emerging as potential targets for therapeutic intervention.
Collapse
|
25
|
Burns SS, Kapur R. Clonal Hematopoiesis of Indeterminate Potential as a Novel Risk Factor for Donor-Derived Leukemia. Stem Cell Reports 2021; 15:279-291. [PMID: 32783925 PMCID: PMC7419737 DOI: 10.1016/j.stemcr.2020.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a critical treatment modality for many hematological and non-hematological diseases that is being extended to treat older individuals. However, recent studies show that clonal hematopoiesis of indeterminate potential (CHIP), a common, asymptomatic condition characterized by the expansion of age-acquired somatic mutations in blood cell lineages, may be a risk factor for the development of donor-derived leukemia (DDL), unexplained cytopenias, and chronic graft-versus-host disease. CHIP may contribute to the pathogenesis of these significant transplant complications via various cell-autonomous and non-cell-autonomous mechanisms, and the clinical presentation of DDL may be broader than anticipated. A more comprehensive understanding of the contributions of CHIP to DDL may have important implications for the screening of donors and will improve the safety of HSCT. The objective of this review is to discuss studies linking DDL and CHIP and to explore potential mechanisms by which CHIP may contribute to DDL.
Collapse
Affiliation(s)
- Sarah S Burns
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Reuben Kapur
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Molecular Biology and Biochemistry, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
26
|
Sibling donor-derived myeloid sarcoma after hematopoietic stem cell transplant. HUMAN PATHOLOGY: CASE REPORTS 2021. [DOI: 10.1016/j.ehpc.2021.200512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
27
|
Newell LF, Williams T, Liu J, Yu Y, Chen Y, Booth GC, Knight RJ, Goslee KR, Cook RJ, Leonard J, Meyers G, Traer E, Press RD, Fan G, Wang Y, Raess PW, Maziarz RT, Dunlap J. Engrafted Donor-Derived Clonal Hematopoiesis after Allogenic Hematopoietic Cell Transplantation is Associated with Chronic Graft-versus-Host Disease Requiring Immunosuppressive Therapy, but no Adverse Impact on Overall Survival or Relapse. Transplant Cell Ther 2021; 27:662.e1-662.e9. [PMID: 33901720 DOI: 10.1016/j.jtct.2021.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 11/19/2022]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is an age-associated condition defined by the presence of a somatic mutation in a leukemia-associated gene in individuals who otherwise have no evidence of a hematologic malignancy. In the allogeneic hematopoietic cell transplantation (HCT) setting, clonal hematopoiesis (CH) mutations present in donor stem cells can be transferred to recipients at the time of HCT. Given that the consequences of donor-derived CH in HCT recipients are not entirely clear, we sought to investigate clinical outcomes in patients with engrafted donor-derived CH using a matched cohort analysis of both related and unrelated donors. Of 209 patients with next-generation sequencing performed before and after HCT, donor-derived CH mutations were detected in 15 (5.2%). DNMT3A was the most commonly mutated gene (9 of 15; 60%); mutations in SF3B1, CSF3R, STAT3, CBLB, TET2, and ASXL1 were also identified. Donor-derived CH was not associated with delayed neutrophil or platelet engraftment, and there was no impact on conversion to full donor chimerism. No patients with donor-derived CH experienced relapse, in contrast to 15.6% (7 of 45) in the matched control cohort without CH (P = .176). Donor-derived CH was not associated with worse overall survival; however, patients with donor-derived CH were more likely to develop chronic graft-versus-host disease (GVHD) necessitating systemic immunosuppressive therapy (IST) (P = .045) and less likely to discontinue IST (P = .03) compared with controls without donor-derived CH. We conclude that donor-derived CH does not have an adverse impact on relapse, survival, or engraftment outcomes but may potentiate a graft-versus-leukemia effect, as reflected by increased chronic GVHD necessitating IST.
Collapse
Affiliation(s)
- Laura F Newell
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Todd Williams
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - James Liu
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - Yun Yu
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Yiyi Chen
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Georgeann C Booth
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Rebekah J Knight
- Cellular Therapy Laboratory, Hospital and Clinics, Oregon Health & Science University, Portland, Oregon
| | - Kelli R Goslee
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Rachel J Cook
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Jessica Leonard
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Gabrielle Meyers
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Elie Traer
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Richard D Press
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon; Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - Guang Fan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - Ying Wang
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - Philipp W Raess
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - Richard T Maziarz
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Jennifer Dunlap
- Department of Pathology, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
28
|
Hammond D, Loghavi S. Clonal haematopoiesis of emerging significance. Pathology 2021; 53:300-311. [PMID: 33685721 DOI: 10.1016/j.pathol.2021.02.005] [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: 11/08/2020] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/17/2022]
Abstract
Clonal haematopoiesis (CH) is a ubiquitous feature of aging and provides mechanistic insight into the inextricable relationship between chronic inflammation and age-related diseases. Although CH confers a cumulative risk of subsequent haematological malignancy, particularly myeloid neoplasms, that risk is heavily mutation- and context-specific. Individuals with mutations in DNA damage response pathway genes receiving select cytotoxic therapies for solid tumours are among the highest risk groups for subsequent development of myeloid neoplasms. Multiple lines of evidence suggest that TET2-mutated macrophages causally contribute to cardiometabolic disease through the generation of proinflammatory cytokines. It is speculated that such CH-related inflammation is a shared driver of several other chronic diseases. Whether we can intervene in individuals with CH to diminish the risk of subsequent haematological malignancy or non-haematological disease remains to be seen. However, precision anti-cytokine therapies are a rational starting point to break the feedforward loop between clonal myeloid expansion, inflammation, and end-organ damage.
Collapse
Affiliation(s)
- Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
Kurosawa S, Iwama A. Aging and leukemic evolution of hematopoietic stem cells under various stress conditions. Inflamm Regen 2020; 40:29. [PMID: 33292805 PMCID: PMC7643313 DOI: 10.1186/s41232-020-00138-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have self-renewal capacity and differentiation potential into all lineages of blood cells throughout the lifetime of an organism. The function of HSCs gradually changes during aging. To date, various stress factors influencing HSC aging have been identified. The increased production of reactive oxygen species and DNA damage responses are causatively attributed to HSC aging. The increased apolarity is a prominent feature of aged HSCs, whereas it is less obvious in young HSCs. The bone marrow (BM) microenvironment niche is a crucial factor for HSC aging. Mesenchymal stem cells show skewed differentiation during aging, which leads to decreased bone formation and increased adipogenesis. The accumulation of adipocytes confers negative effects on hematopoiesis. Loss of sympathetic nerve fibers or adrenoreceptor β3 signaling induces premature HSC and niche aging. Epigenetic regulators such as polycomb group proteins and the sirtuin family of proteins act to prevent premature aging. Targeting these factors, several rejuvenation strategies for aged HSCs have been employed in mice. However, we still do not know whether these strategies can be extrapolated to human HSCs. Aging is frequently accompanied by the development of clonal hematopoiesis, which is called age-related clonal hematopoiesis (ARCH) or clonal hematopoiesis of indeterminate potential (CHIP). Most ARCH/CHIP mutations occur in genes encoding epigenetic regulators including DNMT3A, TET2, and ASXL1, which suggests the relevance of epigenetic drift during the aging process. ARCH/CHIP is a strong risk factor for subsequent hematologic cancer. Notably, it also has an impact on the development of non-malignant disorders such as coronary heart disease. Further studies are warranted to decipher the complete picture of molecular crosstalk that regulates HSC aging.
Collapse
Affiliation(s)
- Shuhei Kurosawa
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
30
|
Steensma DP, Bolton KL. What to tell your patient with clonal hematopoiesis and why: insights from 2 specialized clinics. Blood 2020; 136:1623-1631. [PMID: 32736381 PMCID: PMC7530645 DOI: 10.1182/blood.2019004291] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/24/2020] [Indexed: 12/19/2022] Open
Abstract
Acquired genetic mutations in hematopoietic stem or progenitor cells can lead to clonal expansion and imbalanced blood cell production. Clonal hematopoiesis is exceptionally common with human aging, confers a risk of evolution to overt hematologic malignancy, and increases all-cause mortality and the risk of cardiovascular disease. The degree of risk depends on the specific mutant allele driving clonal expansion, number of mutations, mutant allele burden, and concomitant nongenetic risk factors (eg, hypertension or cigarette smoking). People with clonal hematopoiesis may come to clinical attention in a variety of ways, including during the evaluation of a possible hematologic malignancy, as an incidental discovery during molecular analysis of a nonhematologic neoplasm, after hematopoietic cell transplantation, or as a result of germline testing for inherited variants. Even though the risk of clonal progression or a cardiovascular event in an individual patient with clonal hematopoiesis may be low, the possibility of future clinical consequences may contribute to uncertainty and worry, because it is not yet known how to modify these risks. This review summarizes clinical considerations for patients with clonal hematopoiesis, including important points for hematologists to consider discussing with affected persons who may understandably be anxious about having a mutation in their blood that predisposes them to develop a malignancy, but which is significantly more likely to result in a myocardial infarction or stroke. The increasing frequency with which people with clonal hematopoiesis are discovered and the need for counseling these patients is driving many institutions to create specialized clinics. We describe our own experience with forming such clinics.
Collapse
|