1
|
Shapiro RM, Kim HT, Dulery R, Liney D, Garrity HM, Panaro K, Au C, Gervais C, Little JS, Ho VT, Cutler CS, Koreth J, Gooptu M, Antin JH, Kelkar AH, Romee R, Wu CJ, Ritz J, Soiffer RJ, Nikiforow S. Active infection at the time of CD34+ selected stem cell boost is associated with treatment failure and poor overall survival. Blood Adv 2024; 8:4729-4737. [PMID: 38748871 PMCID: PMC11413676 DOI: 10.1182/bloodadvances.2023012418] [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: 01/02/2024] [Accepted: 04/30/2024] [Indexed: 09/11/2024] Open
Abstract
ABSTRACT The use of CD34+ selected stem cell boost (SCB) after allogeneic hematopoietic cell transplant (allo-HCT) has been increasing. Predictors of treatment failure after SCB, both in the context of poor graft function (PGF) or other settings, are not well characterized. We report among the largest single-center retrospective experiences of the use of SCB and evaluate potential predictors of response and outcomes. A total of 58 patients who underwent HCT between 2015 and 2022 and who received SCB, were identified. The indication for SCB was predominantly PGF, defined as the presence of ≥2 cytopenias for at least 2 consecutive weeks beyond day +14 after allo-HCT in the presence of ≤30% bone marrow cellularity and ≥90% donor myeloid chimerism in the absence of morphologic disease. The median dose of infused CD34+ selected SCB products was 3.88 × 106 CD34+ cells per kg (range, 0.99 × 106 to 9.92 × 106). The median 2-year overall survival and nonrelapse mortality after SCB was 47% and 38%, respectively. The cumulative incidences of 6-month grade 3 to 4 acute and 2-year moderate-severe chronic graft-versus-host disease after SCB were 3.4% and 12%, respectively. Overall response (complete response + partial response) was attained in 36 of 58 patients (62%) and in 69% of patients with PGF. On multivariable analysis, an active infection at the time of SCB was the greatest predictor of poor response and survival (P = .013) after SCB. SCB can restore hematopoiesis in the majority of patients, particularly for those with PGF and in whom there is no active infection at the time of infusion.
Collapse
Affiliation(s)
- Roman M. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Haesook T. Kim
- Department of Data Science, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, MA
| | - Remy Dulery
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Sorbonne University, Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, Assistance Publique - Hôpitaux de Paris, Inserm UMRs 938, Centre de recherche Saint-Antoine, Paris, France
| | - Deborah Liney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Heather M. Garrity
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Kevin Panaro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Chloe Au
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Casey Gervais
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jessica S. Little
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Vincent T. Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Corey S. Cutler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - John Koreth
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mahasweta Gooptu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Joseph H. Antin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Amar H. Kelkar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Rizwan Romee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Robert J. Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Sarah Nikiforow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Tian JS, Tay A. Progress on Electro-Enhancement of Cell Manufacturing. SMALL METHODS 2024; 8:e2301281. [PMID: 38059759 DOI: 10.1002/smtd.202301281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/09/2023] [Indexed: 12/08/2023]
Abstract
With the long persistence of complex, chronic diseases in society, there is increasing motivation to develop cells as living medicine to treat diseases ranging from cancer to wounds. While cell therapies can significantly impact healthcare, the shortage of starter cells meant that considerable raw materials must be channeled solely for cell expansion, leading to expensive products with long manufacturing time which can prevent accessibility by patients who either cannot afford the treatment or have highly aggressive diseases and cannot wait that long. Over the last three decades, there has been increasing knowledge on the effects of electrical modulation on proliferation, but to the best of the knowledge, none of these studies went beyond how electro-control of cell proliferation may be extended to enhance industrial scale cell manufacturing. Here, this review is started by discussing the importance of maximizing cell yield during manufacturing before comparing strategies spanning biomolecular/chemical/physical to modulate cell proliferation. Next, the authors describe how factors governing invasive and non-invasive electrical stimulation (ES) including capacitive coupling electric field may be modified to boost cell manufacturing. This review concludes by describing what needs to be urgently performed to bridge the gap between academic investigation of ES to industrial applications.
Collapse
Affiliation(s)
- Johann Shane Tian
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
3
|
Miura S, Ueda K, Minakawa K, Nollet KE, Ikeda K. Prospects and Potential for Chimerism Analysis after Allogeneic Hematopoietic Stem Cell Transplantation. Cells 2024; 13:993. [PMID: 38891125 PMCID: PMC11172215 DOI: 10.3390/cells13110993] [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: 05/21/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Chimerism analysis after allogeneic hematopoietic stem cell transplantation serves to confirm engraftment, indicate relapse of hematologic malignancy, and attribute graft failure to either immune rejection or poor graft function. Short tandem repeat PCR (STR-PCR) is the prevailing method, followed by quantitative real-time PCR (qPCR), with detection limits of 1-5% and 0.1%, respectively. Chimerism assays using digital PCR or next-generation sequencing, both of which are more sensitive than STR-PCR, are increasingly used. Stable mixed chimerism is usually not associated with poor outcomes in non-malignant diseases, but recipient chimerism may foretell relapse of hematologic malignancies, so higher detection sensitivity may be beneficial in such cases. Thus, the need for and the type of intervention, e.g., immunosuppression regimen, donor lymphocyte infusion, and/or salvage second transplantation, should be guided by donor chimerism in the context of the feature and/or residual malignant cells of the disease to be treated.
Collapse
Affiliation(s)
- Saori Miura
- Department of Clinical Laboratory Sciences, Fukushima Medical University School of Health Sciences, Fukushima 960-8516, Japan
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Koki Ueda
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Keiji Minakawa
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kenneth E. Nollet
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kazuhiko Ikeda
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| |
Collapse
|
4
|
Kilic Gunes E, Yigit Kaya S, Yaman F, Yeniay MK, Vural K, Comert M, Sevindik OG, Andic N, Dagdas S, Nizam Ozen I, Kaynar L, Yavasoglu F, Ozet G, Karakus V, Ayli M. Eltrombopag treatment in thrombocytopenia following hematopoietic stem cell transplantation: A multicenter real-world experience. Leuk Res 2024; 140:107484. [PMID: 38520796 DOI: 10.1016/j.leukres.2024.107484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
INTRODUCTION Thrombocytopenia is among the most common complications following hematopoietic stem cell transplantation and is associated with increased mortality and morbidity with no standard treatment yet. In this multicenter and retrospective study, we aim to present our multi-center experience of Eltrombopag treatment in patients with isolated thrombocytopenia following HSCT. MATERIAL-METHOD A total of 73 patients from 5 centers who underwent autologous or allogeneic stem cell transplantation, had no primary disease relapse, all of whom had neutrophil engraftment, complete chimerism, and who were diagnosed with Prolonged Isolated Thrombocytopenia (PIT) or Secondary Failure Of Platelet Recovery (SFPR) were included in the study. The patients were initiated on Eltrombopag at a dose of 50-150 mg. Complete response was defined as a platelet count >50×109/L for 7 consecutive days with no transfusion support. RESULTS A total of 50.3% of the patients underwent Autologous and 49.7% Allogeneic Stem Cell Transplantation, 54.8% were diagnosed with PIT, and 45.2% were diagnosed with SFPR, and the treatment with 50-150 mg/day Eltrombopag was initiated on the median day +42. Complete response was achieved in 71.2% of these patients on the median day 23 of the treatment. No significant effects of the initial dose (50-150 mg/day) were detected in the Complete Response in the multivariate analysis on response. An insufficient number of Megakaryocytes in the bone marrow before Eltrombopag treatment was determined as an independent risk factor in determining the response (OR 3.57, 95% CI 1.21-10.55). The overall survival of the patients who did not respond to Eltrombopag was found to be significantly worse than that of patients who responded (p=0.022, HR:2.74, 95% CI 1.12-6.54). CONCLUSION As a result of the present study, Eltrombopag treatment was found to be effective and safe in thrombocytopenia that develops following hematopoietic stem cell transplantation. It was concluded that its use may be more effective in patients with sufficient bone marrow megakaryocytes before the treatment and an initial dose of 50 mg/day may be appropriate in terms of cost, effectiveness, and toxicity. Large-scale randomized and controlled prospective studies are needed to determine the roles of Eltrombopag treatment in patients with post-transplant PIT and SFPR.
Collapse
Affiliation(s)
- Ebru Kilic Gunes
- University of Health and Sciences, Gulhane Training and Research Hospital, Department of Hematology, Ankara, Turkiye.
| | - Sureyya Yigit Kaya
- Istanbul Medipol University, Faculty of Medicine, Department of Hematology, Istanbul, Turkiye
| | - Fatih Yaman
- Eskisehir Osmangazi University, Faculty of Medicine, Department of Hematology, Eskisehir, Turkiye
| | - Mustafa Kemal Yeniay
- University of Health and Sciences, Ankara Bilkent City Hospital, Department of Hematology, Ankara, Turkiye
| | - Kurtulus Vural
- University of Health and Sciences, Antalya Training and Research Hospital, Department of Hematology, Antalya, Turkiye
| | - Melda Comert
- University of Health and Sciences, Gulhane Training and Research Hospital, Department of Hematology, Ankara, Turkiye
| | - Omur Gokmen Sevindik
- Istanbul Medipol University, Faculty of Medicine, Department of Hematology, Istanbul, Turkiye
| | - Neslihan Andic
- Eskisehir Osmangazi University, Faculty of Medicine, Department of Hematology, Eskisehir, Turkiye
| | - Simten Dagdas
- University of Health and Sciences, Ankara Bilkent City Hospital, Department of Hematology, Ankara, Turkiye
| | - Ilknur Nizam Ozen
- University of Health and Sciences, Antalya Training and Research Hospital, Department of Hematology, Antalya, Turkiye
| | - Leylagul Kaynar
- Istanbul Medipol University, Faculty of Medicine, Department of Hematology, Istanbul, Turkiye
| | - Filiz Yavasoglu
- Eskisehir Osmangazi University, Faculty of Medicine, Department of Hematology, Eskisehir, Turkiye
| | - Gulsum Ozet
- University of Health and Sciences, Ankara Bilkent City Hospital, Department of Hematology, Ankara, Turkiye; Ankara Yildirim Beyazit University, Faculty of Medicine, Department of Hematology, Ankara Bilkent City Hospital, Ankara, Turkiye
| | - Volkan Karakus
- University of Health and Sciences, Antalya Training and Research Hospital, Department of Hematology, Antalya, Turkiye
| | - Meltem Ayli
- University of Health and Sciences, Gulhane Training and Research Hospital, Department of Hematology, Ankara, Turkiye
| |
Collapse
|
5
|
De Togni E, Cole O, Abboud R. Janus kinase inhibition in the treatment and prevention of graft-versus-host disease. Front Immunol 2024; 15:1304065. [PMID: 38380328 PMCID: PMC10877010 DOI: 10.3389/fimmu.2024.1304065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
Graft-versus-host disease (GVHD) is a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). For many years, corticosteroids have been the mainstay treatment for GVHD, but cases of steroid-refractory GVHD and the severe adverse effects of high-dose corticosteroids have increased the need for preventative and therapeutic strategies for GVHD. Due to the nature of alloreactive T cells, GVHD is inherently linked to the graft-versus-leukemia (GVL) effect, the therapeutic driving force behind stem cell transplantation. A considerable clinical challenge is to preserve GVL while suppressing GVHD. The field of GVHD research has greatly expanded over the past decades, including advancements in T cell modulation and depletion, antibody therapies, chemotherapeutics, cellular therapies, and Janus kinase inhibition. In this review, we discuss current approaches and advances in the prophylaxis and treatment of GVHD with a focus on new emerging advancements in Janus kinase inhibitor therapy.
Collapse
Affiliation(s)
- Elisa De Togni
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Oladipo Cole
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Ramzi Abboud
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
6
|
Al-Ramahi JS, Shahzad M, Nguyen A, Li K, Amin MK, Ahmed N, Lutfi F, DeJarnette S, Chaudhary SG, Bansal R, Abdelhakim H, Shune L, Abdallah AO, Singh AK, Abhyankar SH, McGuirk JP, Mushtaq MU. Favorable outcomes following CD34-selected stem cell boost for poor graft function after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2024; 59:134-137. [PMID: 37833526 DOI: 10.1038/s41409-023-02125-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Affiliation(s)
- Joe S Al-Ramahi
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Moazzam Shahzad
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrea Nguyen
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kevin Li
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Muhammad Kashif Amin
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nausheen Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Forat Lutfi
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shaun DeJarnette
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sibgha Gull Chaudhary
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Rajat Bansal
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Haitham Abdelhakim
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Leyla Shune
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Al-Ola Abdallah
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anurag K Singh
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sunil H Abhyankar
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Joseph P McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Muhammad Umair Mushtaq
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
7
|
He Y, Ma R, Wang HF, Zhang YY, Lyu M, Mo XD, Yan CH, Wang Y, Zhang XH, Xu LP, Liu KY, Huang XJ, Sun YQ. [Clinical analysis of 8 cases of refractory hematopoietic reconstitution after haploid hematopoietic stem cell transplantation treated with purified donor CD34-selected hematopoietic stem cells]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:1027-1031. [PMID: 38503527 PMCID: PMC10834869 DOI: 10.3760/cma.j.issn.0253-2727.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Indexed: 03/21/2024]
Affiliation(s)
- Y He
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - R Ma
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - H F Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Y Y Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - M Lyu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - X D Mo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - C H Yan
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Y Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - X H Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - L P Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - K Y Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - X J Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | - Y Q Sun
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| |
Collapse
|
8
|
Worel N, Ljungman P, Verheggen ICM, Hoogenboom JD, Knelange NS, Eikema DJ, Sánchez-Ortega I, Riillo C, Centorrino I, Averbuch D, Chabannon C, de la Camara R, Kuball J, Ruggeri A. Fresh or frozen grafts for allogeneic stem cell transplantation: conceptual considerations and a survey on the practice during the COVID-19 pandemic from the EBMT Infectious Diseases Working Party (IDWP) and Cellular Therapy & Immunobiology Working Party (CTIWP). Bone Marrow Transplant 2023; 58:1348-1356. [PMID: 37673982 DOI: 10.1038/s41409-023-02099-w] [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: 02/26/2023] [Revised: 08/12/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
The COVID-19 pandemic has had a significant impact on medical practices, including the delivery of allogeneic hematopoietic cell transplantation (HCT). In response, transplant centers have made changes to their procedures, including an increased use of cryopreservation for allogeneic haematopoietic progenitor cell (HPC) grafts. The use of cryopreserved grafts for allogeneic HCT has been reviewed and analysed in terms of potential benefits and drawbacks based on existing data on impact on cell subsets, hematological recovery, and clinical outcomes of approximately 2000 patients from different studies. A survey of European Society for Blood and Marrow Transplantation centers was also conducted to assess changes in practice during the pandemic and any unnecessary burdens on HPC donors. Before the pandemic, only 7.4% of transplant centers were routinely cryopreserving HPC products, but this percentage increased to 90% during the pandemic. The results of this review and survey suggest that cryopreservation of HPC grafts is a viable option for allogeneic HCT in certain situations, but further research is needed to determine long-term effects and ethical discussions are required to balance the needs of donors and patients when using frozen allografts.
Collapse
Affiliation(s)
- N Worel
- Medical University Vienna; Department. of Transfusion Medicine and Cell Therapy, Vienna, Austria.
| | - P Ljungman
- Department. of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge and Div. of Hematology, Department. of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - D-J Eikema
- EBMT Statistical Unit, Leiden, Netherlands
| | | | - C Riillo
- Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - I Centorrino
- Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - D Averbuch
- Faculty of Medicine, Hebrew University of Jerusalem; Hadassah Medical Center, Jerusalem, Israel
| | - C Chabannon
- Institut Paoli-Calmettes, Centre de Lutte Contre le Cancer; Centre d'Investigations Cliniques en Biothérapie, Université d'Aix-Marseille, Inserm CBT, 1409, Marseille, France
| | | | - J Kuball
- Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - A Ruggeri
- Ospedale San Raffaele s.r.l., Haematology and BMT, Milan, Italy
| |
Collapse
|
9
|
Arslan Davulcu E, Akad Soyer N, Vural F. Eltrombopag for the Treatment of Allogeneic Hematopoietic Stem Cell Transplantation-Related Poor Graft Function. Cureus 2023; 15:e44555. [PMID: 37790070 PMCID: PMC10544786 DOI: 10.7759/cureus.44555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 10/05/2023] Open
Abstract
INTRODUCTION Allogeneic stem cell transplantation (ASCT) is a crucial therapeutic strategy for hematological and non-hematological disorders. Poor graft function (PGF) after ASCT presents a critical challenge that does not have a standardized treatment approach. A thrombopoietin-mimetic oral drug eltrombopag shows promise in some bone failure syndromes. This study aimed to analyze the efficacy of eltrombopag in treating PGF after ASCT. METHODS Patients receiving eltrombopag for PGF after ASCT between 2017 and 2020 were retrospectively evaluated. Patients' characteristics, details for ASCT, timing, treatment, and possible contributors for PGF, response to eltrombopag treatment, and overall response rate (ORR) were analyzed. Results: Eighteen patients were assessed. Eltrombopag treatment yielded a favorable response in 11 patients, resulting in an ORR of 61%. The ORR in secondary PGF was better than that in primary PGF (83% and 17% respectively). There was a marked enhancement in platelet and hemoglobin levels following eltrombopag treatment (p=0.001 and p=0.030, respectively), while neutrophil values exhibited no significant change (p=0.8). Among the responding patients, four individuals (22%) underwent a tapering and discontinuation of eltrombopag. No toxicity was observed above grade one, and no patient discontinued eltrombopag because of intolerability or adverse events. CONCLUSION Our findings affirm that eltrombopag can treat poor graft function after allogeneic stem cell transplantation without significant toxicities. These results contribute to the growing body of evidence supporting the use of eltrombopag in poor graft function after allogeneic stem cell transplantation, providing insights into its potential benefits and limitations.
Collapse
Affiliation(s)
- Eren Arslan Davulcu
- Department of Hematology, University of Health Sciences, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, TUR
| | - Nur Akad Soyer
- Department of Hematology, Faculty of Medicine, Ege University, Izmir, TUR
| | - Filiz Vural
- Department of Hematology, Faculty of Medicine, Ege University, Izmir, TUR
| |
Collapse
|
10
|
Müskens KF, Lindemans CA, Dandis R, Nierkens S, Belderbos ME. Definitions, incidence and outcome of poor graft function after hematopoietic cell transplantation: A systematic review and meta-analysis. Blood Rev 2023; 60:101076. [PMID: 36990959 DOI: 10.1016/j.blre.2023.101076] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
Poor graft function (PGF) after allogeneic hematopoietic stem cell transplantation (HCT) is a serious complication with high morbidity and mortality. The reported incidence of PGF, its risk factors and outcome vary substantially between studies. This variability may be explained by heterogeneity in patient cohorts and HCT strategies, differences in the underlying causes of cytopenia, as well as by differences in PGF definition. In this systematic review and meta-analysis, we provide an overview of the various PGF definitions used and determined the impact of this variability on the reported incidence and outcome. We searched MEDLINE, EMBASE and Web of Science up to July 2022, for any study on PGF in HCT recipients. We performed random-effect meta-analyses for incidence and outcome and subgroup analyses based on different PGF criteria. Among 69 included studies (14.265 HCT recipients), we found 63 different PGF definitions, using various combinations of 11 common criteria. The median incidence of PGF was 7% (IQR: 5-11%, 22 cohorts). The pooled survival of PGF patients was 53% (95% CI: 45-61%, 23 cohorts). The most commonly reported risk factors associated with PGF were history of cytomegalovirus infection and prior graft-versus-host disease. Incidence was lower in studies with strict cytopenic cutoffs, while survival was lower for primary compared to secondary PGF. This work indicates that a standardized, quantitative definition of PGF is needed to facilitate clinical guideline development and to advance scientific progress.
Collapse
Affiliation(s)
- Konradin F Müskens
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Caroline A Lindemans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Rana Dandis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Center for Translational Immunology, Utrecht University, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Mirjam E Belderbos
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands.
| |
Collapse
|
11
|
Bowman S, Stanek J, Bajwa R, Polishchuk V, Abu-Arja R, Rangarajan HG. CD34 Stem Cell Boost in Pediatric Allogeneic Stem Cell Transplant Recipients: A Case Series and Review of Literature. Clin Hematol Int 2023:10.1007/s44228-023-00042-w. [PMID: 37027103 DOI: 10.1007/s44228-023-00042-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Patients with poor graft function (PGF) or declining donor chimerism (DC) post allogeneic hematopoietic cell transplantation (HCT) may benefit from a CD34-selected stem cell boost (SCB). We retrospectively studied outcomes of fourteen pediatric patients (PGF: 12 and declining DC: 2), with a median age of 12.8 (range 0.08-20.6) years at HCT, who received a SCB. Primary and secondary endpoints included resolution of PGF or improvement in DC (≥ 15% increase), overall survival (OS) and transplant-related mortality (TRM), respectively. The median CD34 dose infused was 7.47 × 106/kg (range 3.51 × 106-3.39 × 107/kg). Among patients with PGF who survived ≥ 3 months post-SCB (n = 8), we observed a non-significant decrease in the cumulative median number of red cell transfusions, platelet transfusions, and GCSF but not intravenous immunoglobulin doses in the 3 months before and after SCB. Overall response rate (ORR) was 50%, with 29% complete and 21% partial responses. ORR was better in recipients who received lymphodepletion (LD) pre-SCB versus none (75% versus 40%; p = 0.56). The incidence of acute and chronic graft-versus-host-disease was 7% and 14%, respectively. The 1-year OS was 50% (95% CI 23-72%) and TRM was 29% (95% CI 8-58%). SCB was effective in half of our cohort with possible benefit of LD pre-SCB.
Collapse
Affiliation(s)
- Sara Bowman
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
| | - Joe Stanek
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Biostatistics Resource at Nationwide Children's Hospital, Columbus, OH, USA
| | - Rajinder Bajwa
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Veronika Polishchuk
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Rolla Abu-Arja
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Hemalatha G Rangarajan
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
12
|
Liu E, Fang P, Xin H, Li S, Liu Y, Xu Y, Chen Y. Efficacy and safety of avatrombopag for thrombocytopenia following allogeneic hematopoietic stem cell transplantation: A real-world data evaluation on 14 cases. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:376-385. [PMID: 37164921 PMCID: PMC10930078 DOI: 10.11817/j.issn.1672-7347.2023.220600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 05/12/2023]
Abstract
OBJECTIVES Thrombocytopenia following allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a common and serious complication that leads to an increased risk of bleeding and poor prognosis. Traditional strategies consist of platelet transfusion, glucocorticoid therapy, intravenous human immunoglobulin, recombinant human thrombopoietin injection, and CD34+-selected hematopoietic stem cell transplantation, but the effects of these treatments are not satisfactory and the treatment continues to be challenged. This study aims to determine the treating efficacy of avatrombopag, a novel thrombopoietin receptor agonist, on thrombocytopenia after allo-HSCT, and to increase the evidence-based medical evidence for the clinical use of this drug. METHODS Fourteen patients with thrombocytopenia after allo-HSCT underwent avatrom-bopag treatment from September 2020 to September 2021 were retrospectively studied. Of these patients, 8 patients had delayed platelet engraftment (DPE) and 6 cases had secondary failure of platelet recovery (SFPR). The efficacy and safety of the treatment and the survival of the patients were assessed. RESULTS The median treatment time of avatrombopag was 34 days, and no patients stopped treatment due to adverse reactions or drug intolerance. Compared with the treatment before, the levels of platelet count, megakaryocytes, and hemoglobin in patients were significantly increased (P=0.000 1, P=0.001 0, and P=0.001 7, respectively). The optimal platelet count of 13 patients reached the complete response standard after drug withdrawal. The median follow-up time of 14 patients was 371 days, and the 2-year overall survival rate was 78.6%. CONCLUSIONS Avatrombopag is effective on increasing platelet counts in patients with thrombocytopenia after allo-HSCT, with a good safety profile. It is a suitable therapeutic option for thrombocytopenia after allo-HSCT.
Collapse
Affiliation(s)
- Enyi Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008.
- Hunan Hematologic Neoplasms Clinical Medical Research Center, Changsha 410008.
- National Clinical Research Center for Hematologic Diseases, First Affiliated Hospital of Soochow University, Suzhou Jiangshu 215006, China.
| | - Peng Fang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
- Hunan Hematologic Neoplasms Clinical Medical Research Center, Changsha 410008
- National Clinical Research Center for Hematologic Diseases, First Affiliated Hospital of Soochow University, Suzhou Jiangshu 215006, China
| | - Hongya Xin
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
- Hunan Hematologic Neoplasms Clinical Medical Research Center, Changsha 410008
- National Clinical Research Center for Hematologic Diseases, First Affiliated Hospital of Soochow University, Suzhou Jiangshu 215006, China
| | - Shujun Li
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
- Hunan Hematologic Neoplasms Clinical Medical Research Center, Changsha 410008
- National Clinical Research Center for Hematologic Diseases, First Affiliated Hospital of Soochow University, Suzhou Jiangshu 215006, China
| | - Yi Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
- Hunan Hematologic Neoplasms Clinical Medical Research Center, Changsha 410008
- National Clinical Research Center for Hematologic Diseases, First Affiliated Hospital of Soochow University, Suzhou Jiangshu 215006, China
| | - Yajing Xu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008
- Hunan Hematologic Neoplasms Clinical Medical Research Center, Changsha 410008
- National Clinical Research Center for Hematologic Diseases, First Affiliated Hospital of Soochow University, Suzhou Jiangshu 215006, China
| | - Yan Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha 410008.
- Hunan Hematologic Neoplasms Clinical Medical Research Center, Changsha 410008.
- National Clinical Research Center for Hematologic Diseases, First Affiliated Hospital of Soochow University, Suzhou Jiangshu 215006, China.
| |
Collapse
|
13
|
Kırcalı E, Cengiz Seval G, Öztürk C, Yılmaz H, Koyun D, Civriz Bozdağ S, Toprak SK, Topçuoğlu P, Arslan Ö, Özcan M, Demirer T, İlhan O, Gürman G, Beksaç M, Kurt Yüksel M. Eltrombopag for the Treatment of Poor Graft Function Following Haematopoietic Cell Transplantation: Real-Life Data. Balkan Med J 2023; 40:51-56. [PMID: 36571427 PMCID: PMC9874246 DOI: 10.4274/balkanmedj.galenos.2022.2022-2-48] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Eltrombopag has an off-label indication for haematopoietic cell transplantation in patients experiencing delayed thrombocyte recovery and/or thrombocytopaenia. Aims To present our centre’s experience of using this agent not only for post- haematopoietic cell transplantation thrombocytopaenia but also for poor graft functioning in the post-haematopoietic cell transplantation setting. Study Design Retrospective cross-sectional study. Methods Thirty-nine patients who had persistent cytopaenia following haematopoietic cell transplantation and treated with eltrombopag at our centre between October 2011 and December 2021 were retrospectively identified. During this period, 9 (23.1%) and 30 (76.9%) patients who underwent allogeneic transplantations, respectively, received eltrombopag. Results The female-to-male ratio was 12:27, and the median transplant age was 49 (18-70) years. Eight (20.5%) patients had isolated thrombocytopaenia, 19 (49.4%) had bi-lineage cytopaenia and 12 (30.1%) had pancytopaenia. Patients received a median of 50 mg/day (25-150 mg/day) of eltrombopagfor a median duration of 82 (24-386) days. Nine (23.1%) patients had autologous haematopoietic cell transplantation, and 30 (76.9%) had allogeneic haematopoietic cell transplantation (14 unrelated, 9 sibling and 7 haploidentical). The median donor age was 32 (20-67) years. The median follow-up was 16.4 (1.8-84.3) months. The median pre-treatment platelet count was 11x109/l (1-23), which increased to 41x109/l (6-150). The median platelet count increment was 29.5x109/l (p = 0.001). The pre-treatment median neutrophil count was 1.19x109/l (0.39-5.1), which increased to 2.35 x109/l (0.1-5.33) (p = 0.05), and the pre-treatment median haemoglobin was 8.3 (6.2-14) g/dl, which increased to 10 (6.2-14) g/dl (p = 0.001) with eltrombopag. No eltrombopag-related hepatotoxicity occurred; however, 1 (2.6%) patient failed to continue treatment because of two consecutive episodes of deep venous thrombosis. Six (15.4%) patients were unresponsive to eltrombopag and dependent on blood product transfusions. After a median time of 82 days, 61.5% of the patients discontinued eltrombopag successfully. Conclusion The results confirmed that eltrombopag could provide a rapid, sustained response in patients with poor graft functioning after haematopoietic cell transplantation. This finding is essential given the high rate of non-relapse mortality caused by poor graft functioning after haematopoietic cell transplantation.
Collapse
Affiliation(s)
- Ekin Kırcalı
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey,* Address for Correspondence: Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey E-mail:
| | | | - Cemaleddin Öztürk
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Hülya Yılmaz
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Derya Koyun
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sinem Civriz Bozdağ
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Selami Koçak Toprak
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Pervin Topçuoğlu
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Önder Arslan
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Muhit Özcan
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Taner Demirer
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Osman İlhan
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Günhan Gürman
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Meral Beksaç
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Meltem Kurt Yüksel
- Department of Hematology, Ankara University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
14
|
Maslikova UV, Popova NN, Drokov MY, Khamaganova EG. Graft failure in allogeneic hematopoietic stem cell recipients: diagnosis and treatment. BULLETIN OF THE MEDICAL INSTITUTE "REAVIZ" (REHABILITATION, DOCTOR AND HEALTH) 2023. [DOI: 10.20340/vmi-rvz.2023.1.tx.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Graft failure is a group of complications after allogeneic hematopoietic stem cell transplantation, which occurs according to different data up to 30%. The group of complications includes primary and secondary graft failure, primary, secondary and transient poor graft function and graft rejection. Diagnostic difficulties consist in the lack of unified diagnostic criteria accepted in the transplantation community and in the dual interpretation of these complications according to the foreign literature. The purpose of this literature review was to identify the most common criteria of different types of graft failure and determine the tactics of diagnosis and treatment. In this review we analyzed data from various literature sources, gave definitions of graft failure and poor graft function. We analyzed the literature data on the methods used to treat these conditions.
Collapse
|
15
|
Fraint E, Farooki S, Klein E, Mauguen A, Prockop SE, Scaradavou A, Curran K, Cancio M, Spitzer B, Boelens JJ, Oved J, Harris A, O'Reilly RJ, Kernan NA. Durable Engraftment and Excellent Overall Survival After CD34-Selected Peripheral Blood Stem Cell Boost in Pediatric Patients With Poor Graft Function Following Allogeneic Stem Cell Transplantation. Transplant Cell Ther 2023; 29:46.e1-46.e6. [PMID: 36210027 PMCID: PMC9825630 DOI: 10.1016/j.jtct.2022.09.027] [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: 08/03/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Poor graft function (PGF) is a life-threatening complication after allogeneic stem cell transplantation (alloSCT). Historically, outcomes of patients with PGF have been very poor, and there are no standardized approaches to treatment. Furthermore, few outcomes after CD34-selected stem cell boost (CD34+SCB) for PGF in pediatric alloSCT recipients have been reported. Here we report on a single center experience with CD34+SCB for PGF after alloSCT in patients treated on the Pediatric Transplant and Cellular Therapy Service at MSK Kids, Memorial Sloan Kettering Cancer Center. A retrospective analysis of patients transplanted for malignant and nonmalignant disorders who received a CD34+SCB between 2008 to 2020 for treatment of PGF defined as the need for granulocyte colony-stimulating factor (G-CSF) and/or packed red blood cell or platelet transfusion support with bone marrow donor chimerism ≥85%. Peripheral blood stem cells from the original donor were the source for CD34+SCB. Durable complete recovery (durable CR) was defined as recovery of peripheral blood counts without recurrent need for G-CSF or transfusion support. The main outcomes of interest were recovery of hematopoiesis and overall survival. Development of graft versus host disease (GVHD) was an additional outcome of interest. Fourteen patients with PGF received a boost. Six patients had no known infection, while 8 patients had PGF associated with an infection. The probability of CR at 60 days was 79% (95% confidence interval [CI], 57%-100%). The overall survival at both 2 and 5 years was 78% (95% CI, 56%-100%). One patient developed GVHD, which was fatal. No other CD34+SCB-related toxicities were observed. While including patients with PGF as recently defined by the American Society for Transplantation and Cellular Therapy, as well as PGF in patients with concomitant infections, we demonstrate that CD34+SCB is safe and can provide for durable trilineage hematopoietic recovery and long-term survival in pediatric patients after alloSCT.
Collapse
Affiliation(s)
- Ellen Fraint
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx New York
| | - Sana Farooki
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Division of Pediatric Hematology/Oncology, Charleston Area Medical Center, West Virginia University, Charleston, West Virginia
| | - Elizabeth Klein
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susan E Prockop
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York; Dana Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Andromachi Scaradavou
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Kevin Curran
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Maria Cancio
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Barbara Spitzer
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Jaap Jan Boelens
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Joseph Oved
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Andrew Harris
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Richard J O'Reilly
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Nancy A Kernan
- Department of Pediatrics, Stem Cell Transplantation and Cellular Therapies Service, MSK Kids, Memorial Sloan Kettering Cancer Service, New York, New York; Department of Pediatrics, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
16
|
Non-relapse cytopenias following allogeneic stem cell transplantation, a case based review. Bone Marrow Transplant 2022; 57:1489-1499. [DOI: 10.1038/s41409-022-01761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/08/2022]
|
17
|
Man Y, Lu Z, Yao X, Gong Y, Yang T, Wang Y. Recent Advancements in Poor Graft Function Following Hematopoietic Stem Cell Transplantation. Front Immunol 2022; 13:911174. [PMID: 35720412 PMCID: PMC9202575 DOI: 10.3389/fimmu.2022.911174] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/06/2022] [Indexed: 01/05/2023] Open
Abstract
Poor graft function (PGF) is a life-threatening complication that occurs after transplantation and has a poor prognosis. With the rapid development of haploidentical hematopoietic stem cell transplantation, the pathogenesis of PGF has become an important issue. Studies of the pathogenesis of PGF have resulted in some success in CD34+-selected stem cell boosting. Mesenchymal stem cells, N-acetyl-l-cysteine, and eltrombopag have also been investigated as therapeutic strategies for PGF. However, predicting and preventing PGF remains challenging. Here, we propose that the seed, soil, and insect theories of aplastic anemia also apply to PGF; CD34+ cells are compared to seeds; the bone marrow microenvironment to soil; and virus infection, iron overload, and donor-specific anti-human leukocyte antigen antibodies to insects. From this perspective, we summarize the available information on the common risk factors of PGF, focusing on its potential mechanism. In addition, the safety and efficacy of new strategies for treating PGF are discussed to provide a foundation for preventing and treating this complex clinical problem.
Collapse
Affiliation(s)
- Yan Man
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Zhixiang Lu
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xiangmei Yao
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Yuemin Gong
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Tonghua Yang
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China,*Correspondence: Tonghua Yang, ; Yajie Wang,
| | - Yajie Wang
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China,*Correspondence: Tonghua Yang, ; Yajie Wang,
| |
Collapse
|
18
|
Prabahran A, Koldej R, Chee L, Ritchie D. Clinical features, pathophysiology, and therapy of poor graft function post-allogeneic stem cell transplantation. Blood Adv 2022; 6:1947-1959. [PMID: 34492685 PMCID: PMC8941468 DOI: 10.1182/bloodadvances.2021004537] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/07/2021] [Indexed: 01/05/2023] Open
Abstract
Poor graft function (PGF), defined by the presence of multilineage cytopenias in the presence of 100% donor chimerism, is a serious complication of allogeneic stem cell transplant (alloSCT). Inducers or potentiators of alloimmunity such as cytomegalovirus reactivation and graft-versus-host disease are associated with the development of PGF, however, more clinical studies are required to establish further risk factors and describe outcomes of PGF. The pathophysiology of PGF can be conceptualized as dysfunction related to the number or productivity of the stem cell compartment, defects in bone marrow microenvironment components such as mesenchymal stromal cells and endothelial cells, or immunological suppression of post-alloSCT hematopoiesis. Treatment strategies focused on improving stem cell number and function and microenvironment support of hematopoiesis have been attempted with variable success. There has been limited use of immune manipulation as a therapeutic strategy, but emerging therapies hold promise. This review details the current understanding of the causes of PGF and methods of treatment to provide a framework for clinicians managing this complex problem.
Collapse
Affiliation(s)
- Ashvind Prabahran
- Department of Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Parkville, VIC, Australia
- Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, VIC, Australia; and
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Rachel Koldej
- Department of Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Parkville, VIC, Australia
- Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, VIC, Australia; and
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Lynette Chee
- Department of Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Parkville, VIC, Australia
- Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, VIC, Australia; and
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - David Ritchie
- Department of Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Parkville, VIC, Australia
- Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, VIC, Australia; and
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
19
|
Shahzad M, Siddiqui RS, Anwar I, Chaudhary SG, Ali T, Naseem M, Ahmed TF, Ahmed Z, Khurana S, Ahmed N, Balusu R, Singh AK, Hematti P, Callander NS, Abhyankar SH, McGuirk JP, Mushtaq MU. Outcomes with CD34-Selected Stem Cell Boost for Poor Graft Function after Allogeneic Hematopoietic Stem Cell Transplantation: A Systematic Review and Meta-Analysis. Transplant Cell Ther 2021; 27:877.e1-877.e8. [PMID: 34284148 DOI: 10.1016/j.jtct.2021.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/08/2021] [Accepted: 07/11/2021] [Indexed: 12/28/2022]
Abstract
Poor graft function (PGF) is a life-threatening complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT) characterized by multilineage cytopenia in the absence of mixed donor chimerism (<95% donor), relapse, or severe graft-versus-host disease (GVHD). We present a systemic review and meta-analysis aimed at assessing the outcomes with CD34-selected stem cell boost (SCB) for PGF in adult allo-HSCT recipients. We screened a total of 1753 records identified from 4 databases (PubMed, Embase, Cochrane, and ClinicalTrials.gov) following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, using the search terms "hematological malignancies," "hematopoietic stem cell transplantation," "CD34 antigen(s)," "graft failure," and "poor graft function," from the date of inception to January 2021. After excluding review, duplicate, and nonrelevant articles, we included 7 studies reporting outcomes following administration of CD34-selected SCB for PGF after allo-HSCT, including hematologic complete response (CR) and overall response rate (ORR), GVHD, and overall survival (OS). Quality evaluation was done using the National Institutes of Health quality assessment tool. Pooled analysis was done using the R 'meta' package, and proportions with 95% confidence intervals (CIs) were computed. The inter-study variance was calculated using the Der Simonian-Laird estimator. We identified 209 patients who received CD34-selected SCB for PGF after allo-HSCT. The median age was 49 years (range, 18 to 69 years), and 61% were men. Primary graft sources included peripheral blood stem cells (72%) and bone marrow (28%). Donor types were matched sibling (37%), matched unrelated (36%), mismatched unrelated (22%), and haploidentical donors (5%). The median time from allo-HSCT to SCB was 138 days (range, 113 to 450 days). The median SCB dose was 3.45 × 106 CD34 cells/kg (range, 3.1 to 4.9 × 106 cells/kg). CR and ORR were 72% (95% CI, 63% to 79%; I2 = 26%) and 80% (95% CI, 74% to 85%; I2 = 0%), respectively. After a median follow-up of 42 months (range, 30 to 77 months), the actuarial survival rate was 54% (95% CI, 47% to 61%; I2 = 0%). OS ranged from 80% at 1 year to 40% at 9 years. The incidences of acute and chronic GVHD after SCB were 17% (95% CI, 13% to 23%; I2 = 0%) and 18% (95% CI, 8% to 34%; I2 = 76%), respectively. Nonrelapse mortality was reported in 42 patients, with a pooled rate of 27% (95% CI, 17% to 40; I2 = 59%), and death due to relapse was reported in 25 patients, with a pooled rate of 17% (95% CI, 11% to 23%; I2 = 0%). Our data show that CD34-selected SCB improves outcomes after PGF post allo-HSCT with an acceptable toxicity profile. The literature lacks high-quality randomized evidence, and there remains an unmet need for prospective studies to address the optimal dosing and manipulation of SCB. © 2021 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.
Collapse
Affiliation(s)
- Moazzam Shahzad
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas; Department of Medicine, St Mary's Medical Center, Huntington, West Virginia
| | - Raheel S Siddiqui
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas; Department of Medicine, Icahn School of Medicine at Mount Sinai/Queens, New York, New York
| | - Iqra Anwar
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Sibgha Gull Chaudhary
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Tayyaba Ali
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Masooma Naseem
- Department of Medicine, Icahn School of Medicine at Mount Sinai/Queens, New York, New York
| | - Tehniat F Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Zahoor Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Sharad Khurana
- Division of Hematology & Oncology, University of Arizona College of Medicine, Tucson, Arizona
| | - Nausheen Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Ramesh Balusu
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Anurag K Singh
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Peiman Hematti
- Division of Hematology & Oncology, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin
| | - Natalie S Callander
- Division of Hematology & Oncology, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin
| | - Sunil H Abhyankar
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Joseph P McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Muhammad Umair Mushtaq
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
20
|
Eltrombopag for the treatment of poor graft function following allogeneic stem cell transplant: a retrospective multicenter study. Int J Hematol 2021; 114:228-234. [PMID: 33886103 DOI: 10.1007/s12185-021-03153-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 10/21/2022]
Abstract
This retrospective study assessed the effectiveness of eltrombopag (EPAG), a thrombopoietin receptor agonist, in the treatment of poor graft function (PGF) following an allogeneic haemopoietic stem cell transplantation (HSCT). Complete response was defined as normalization of blood counts, whereas partial response was defined as transfusion independence. A total of 48 patients with full donor chimerism after HSCT, received EPAG for a median of 120 days (range 10-591). Patients with uni- bi- or tri-lineage cytopenia started treatment at a median of 95 days (range 17-877) after HSCT. The overall response rate was 75%: 24 patients had a complete response and 12 had a partial response. Positive predictors of response were an HLA-matched donor, a CD34+ dose at transplant > 4 × 106/kg, and starting EPAG treatment at least 90 days after HSCT. Patients with more than one positive predictor had a response rate of 92% for the overall patient cohort and 94% for patients with tri-lineage cytopenia. One-year survival was 89% for complete responders, 60% for partial responders and 20% for non-responders (p = 0.0004). EPAG improves peripheral blood counts in patients with poor graft function following HSCT. Response to EPAG can be predicted and has a significant impact on survival.
Collapse
|
21
|
Prabahran A, Koldej R, Chee L, Wong E, Ritchie D. Evaluation of risk factors for and subsequent mortality from poor graft function (PGF) post allogeneic stem cell transplantation. Leuk Lymphoma 2021; 62:1482-1489. [PMID: 33522344 DOI: 10.1080/10428194.2021.1872072] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Poor Graft Function (PGF) is defined by multi-lineage cytopenias with complete donor chimerism post allogeneic transplantation, Risk factors for and subsequent mortality from PGF were assessed in our transplant cohort. Non-sibling donor [OR 1.97; 95% CI 1.02-3.70], ICU admission [OR 5.28; 95% CI 2.29-11.88] or blood culture positivity within the first 30 days [OR 1.67; 95% CI 1.07-2.62], grade III-IV acute graft vs host disease (GVHD) [OR 4.082; 95% CI 2.31-7.16] and CMV viremia [OR 2.43; 95% CI 1.53-3.88] and were significantly associated with development of PGF. PGF patients without count recovery had a 2 year OS of 6%. Severe GVHD, thrombocytopenia and anemia portended inferior survival and were used to develop a prognostic score for mortality from PGF. This analysis identifies risk factors predictive of PGF and poor survival in those without recovery.
Collapse
Affiliation(s)
- Ashvind Prabahran
- Department, of Clinical Haematology, Peter MacCallum Cancer/Royal Melbourne Hospital, Parkville, Australia.,Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, Australia.,The University of Melbourne, Parkville, Australia
| | - Rachel Koldej
- Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, Australia.,The University of Melbourne, Parkville, Australia
| | - Lynette Chee
- Department, of Clinical Haematology, Peter MacCallum Cancer/Royal Melbourne Hospital, Parkville, Australia.,Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, Australia.,The University of Melbourne, Parkville, Australia
| | - Eric Wong
- Department, of Clinical Haematology, Peter MacCallum Cancer/Royal Melbourne Hospital, Parkville, Australia.,Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, Australia
| | - David Ritchie
- Department, of Clinical Haematology, Peter MacCallum Cancer/Royal Melbourne Hospital, Parkville, Australia.,Australian Cancer Research Fund Translational Research Laboratory, Royal Melbourne Hospital, Parkville, Australia.,The University of Melbourne, Parkville, Australia
| |
Collapse
|
22
|
Calmels B, Gautier É, Magnani A, Magrin É, Mamez AC, Vaissié A, Yakoub-Agha I, Baudoux É. Procédé de préparation, contrôles de qualité et spécifications des immunosélections CD34+ : recommandations de la Société francophone de greffe de moelle et de thérapie cellulaire (SFGM-TC). Bull Cancer 2020; 107:S185-S192. [DOI: 10.1016/j.bulcan.2020.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/08/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
|
23
|
Abboud R, Slade M, Abboud C, DiPersio JF. Can planned CD34+ stem cell boost prevent poor graft function after peripheral blood haploidentical hematopoietic transplantation? Leuk Lymphoma 2020; 62:749-751. [PMID: 33135522 DOI: 10.1080/10428194.2020.1839657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Ramzi Abboud
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Slade
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Camille Abboud
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - John F DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
24
|
Cuadrado MM, Szydlo RM, Watts M, Patel N, Renshaw H, Dorman J, Lowdell M, Ings S, Anthias C, Madrigal A, Mackinnon S, Kottaridis P, Carpenter B, Hough R, Morris E, Thomson K, Peggs KS, Chakraverty R. Predictors of recovery following allogeneic CD34+-selected cell infusion without conditioning to correct poor graft function. Haematologica 2020; 105:2639-2646. [PMID: 33131253 PMCID: PMC7604618 DOI: 10.3324/haematol.2019.226340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/20/2019] [Indexed: 11/09/2022] Open
Abstract
Poor graft function is a serious complication following allogeneic hematopoietic stem cell transplantation. Infusion of CD34+-selected stem cells without pre-conditioning has been used to correct poor graft function, but predictors of recovery are unclear. We report the outcome of 62 consecutive patients who had primary or secondary poor graft function who underwent a CD34+-selected stem cell infusion from the same donor without further conditioning. Forty-seven of 62 patients showed hematological improvement and became permanently transfusion and growth factor-independent. In multivariate analysis, parameters significantly associated with recovery were shared CMV seronegative status for recipient/donor, the absence of active infection and matched recipient/donor sex. Recovery was similar in patients with mixed and full donor chimerism. Five -year overall survival was 74.4% (95% CI 59-89) in patients demonstrating complete recovery, 16.7% (95% CI 3-46) in patients with partial recovery and 22.2% (CI 95% 5-47) in patients with no response. In patients with count recovery, those with poor graft function in 1-2 lineages had superior 5-year overall survival (93.8%, 95% CI 82-99) than those with tri-lineage failure (53%, 95% CI 34-88). New strategies including cytokine or agonist support, or second transplant need to be investigated in patients who do not recover.
Collapse
Affiliation(s)
| | - Richard M. Szydlo
- Anthony Nolan Research Institute
- Department of Haematology, Imperial College London
| | - Mike Watts
- Wolfson Cellular Therapy Unit, University College Hospital London NHS Trust
| | - Nishil Patel
- Department of Haematology, Royal Free London NHS Trust
| | - Hanna Renshaw
- Department of Haematology, Royal Free London NHS Trust
| | - Jude Dorman
- Department of Haematology, University College Hospital NHS Trust
| | - Mark Lowdell
- Centre for Cell, Gene & Tissue Therapeutics, Royal Free London NHS Trust
| | - Stuart Ings
- Wolfson Cellular Therapy Unit, University College Hospital London NHS Trust
| | | | | | | | | | - Ben Carpenter
- Department of Haematology, University College Hospital NHS Trust
| | - Rachael Hough
- Department of Haematology, University College Hospital NHS Trust
| | - Emma Morris
- Department of Haematology, University College Hospital NHS Trust
| | - Kirsty Thomson
- Department of Haematology, University College Hospital NHS Trust
| | - Karl S. Peggs
- Department of Haematology, University College Hospital NHS Trust
- Department of Hematology, Cancer Institute, University College London, London, UK
| | - Ronjon Chakraverty
- Department of Haematology, University College Hospital NHS Trust
- Department of Hematology, Cancer Institute, University College London, London, UK
| |
Collapse
|
25
|
Aydin S, Dellacasa C, Manetta S, Giaccone L, Godio L, Iovino G, Bruno B, Busca A. Rescue treatment with eltrombopag in refractory cytopenias after allogeneic stem cell transplantation. Ther Adv Hematol 2020; 11:2040620720961910. [PMID: 33194161 PMCID: PMC7594218 DOI: 10.1177/2040620720961910] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Patients with post-transplant cytopenias due to poor graft function or
primary engraftment failure show poor prognosis with a high mortality rate
mainly because of graft versus host disease (GVHD),
infection and/or bleeding. Treatment options are scarce and a CD34+ stem
cell boost or a second bone marrow transplantation may be required to
restore adequate haematopoiesis. Methods: In the present study patients with primary engraftment failure
(n = 1) and refractory poor graft function
(n = 11) were treated with eltrombopag in a single
centre. The reason for eltrombopag treatment was trilineage cytopenia in six
patients, bilineage cytopenia in three patients and single lineage cytopenia
in three patients. Eltrombopag was initiated at a median of 214 (range:
120–877) days after haematopoietic stem cell transplantation (HCST) and
administered for a median time of 114 (range: 12 days to >490) days. In
8/12 patients eltrombopag was introduced at a dose of 75 mg/day and then
increased to 150 mg/day after 1 week; 1 patient was given 50 mg eltrombopag
per day, and 3 patients received 75 mg daily. Results: In 10/12 patients eltrombopag significantly enhanced blood count values and
patients became transfusion independent. Once stable haematological response
was obtained, treatment was tapered until final discontinuation in 9/10
responding patients. No grade 3 or 4 toxicities were observed. At time of
last follow up, 3/12 patients were dead, 2 due to disease relapse, 1 due to
GVHD and pneumonia. All patients except one maintained their complete
response and remain transfusion independent at a median of 858 (range:
429–1119) days. Conclusion: These preliminary data confirm that eltrombopag is able to rescue
multilineage haematopoiesis in patients with treatment-refractory cytopenias
after allogeneic HSCT.
Collapse
Affiliation(s)
- Semra Aydin
- A.O.U. Città della Salute e della Scienza, Dipartimento di Oncologia, Ematologia, Corso Bramante 88, Turin, 10126, Italy
| | - Chiara Dellacasa
- A.O.U. Città della Salute e della Scienza, Dipartimento di Oncologia, SSD Trapianto allogenico di cellule staminali, Turin, Italy
| | - Sara Manetta
- A.O.U. Città della Salute e della Scienza, Dipartimento di Oncologia, SSD Trapianto allogenico di cellule staminali, Turin, Italy
| | - Luisa Giaccone
- A.O.U. Città della Salute e della Scienza, Dipartimento di Oncologia, SSD Trapianto allogenico di cellule staminali, Turin, Italy
| | - Laura Godio
- A.O.U. Città della Salute e della Scienza, Anatomia Patologica, Turin, and University of Turin, Italy
| | - Giorgia Iovino
- A.O.U. Città della Salute e della Scienza, Dipartimento di Oncologia, Ematologia, Turin, Italy
| | - Benedetto Bruno
- A.O.U. Città della Salute e della Scienza, Dipartimento di Oncologia, SSD Trapianto allogenico di cellule staminali, Turin, Italy
| | - Alessandro Busca
- A.O.U. Città della Salute e della Scienza, Dipartimento di Oncologia, SSD Trapianto allogenico di cellule staminali, Turin, Italy
| |
Collapse
|
26
|
Chen J, Wang H, Zhou J, Feng S. Advances in the understanding of poor graft function following allogeneic hematopoietic stem-cell transplantation. Ther Adv Hematol 2020; 11:2040620720948743. [PMID: 32874483 PMCID: PMC7436797 DOI: 10.1177/2040620720948743] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
Poor graft function (PGF) following allogeneic hematopoietic stem-cell transplantation (allo-HSCT) is a life-threatening complication and is characterized by bilineage or trilineage blood cell deficiency and hypoplastic marrow with full chimerism. With the rapid development of allo-HSCT, especially haploidentical-HSCT, PGF has become a growing concern. The most common risk factors illustrated by recent studies include low dose of infused CD34+ cells, donor-specific antibody, cytomegalovirus infection, graft versus host disease (GVHD), iron overload and splenomegaly, among others. Because of the poor prognosis of PGF, it is crucial to uncover the underlying mechanism, which remains elusive. Recent studies have suggested that the bone marrow microenvironment may play an important role in the pathogenesis of PGF. Deficiency and dysfunction of endothelial cells and mesenchymal stem cells, elevated reactive oxygen species (ROS) levels, and immune abnormalities are believed to contribute to PGF. In this review, we also discuss recent clinical trials that evaluate the safety and efficacy of new strategies in patients with PGF. CD34+-selected stem-cell boost (SCB) is effective with an acceptable incidence of GVHD, despite the need for a second donation. Alternative strategies including the applications of mesenchymal stem cells, N-acetyl-l-cysteine (NAC), and eltrombopag have shown favorable outcomes, but further large-scale studies are needed due to the small sample sizes of the recent clinical trials.
Collapse
Affiliation(s)
- Juan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, China
| | - Hongtao Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, China
| | - Jiaxi Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Heping District, Tianjin, 300020, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, 300020, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Heping District, Tianjin, 300020, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences and Department of Stem Cells and Regenerative Medicine, Peking Union Medical College, Tianjin, 300020, China
| |
Collapse
|
27
|
Prabahran AA, Ritchie DS. Poor graft function, a significant and emerging clinical challenge post allogeneic stem cell transplantation. Leuk Lymphoma 2020; 61:2786-2787. [PMID: 32762478 DOI: 10.1080/10428194.2020.1803301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Ashvind A Prabahran
- Department of Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia.,Australian Cancer Research Fund Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,University of Melbourne, Melbourne
| | - David S Ritchie
- Department of Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia.,Australian Cancer Research Fund Laboratory, Royal Melbourne Hospital, Melbourne, Australia.,University of Melbourne, Melbourne
| |
Collapse
|
28
|
Gao F, Zhou X, Shi J, Luo Y, Tan Y, Fu H, Lai X, Yu J, Huang H, Zhao Y. Eltrombopag treatment promotes platelet recovery and reduces platelet transfusion for patients with post-transplantation thrombocytopenia. Ann Hematol 2020; 99:2679-2687. [PMID: 32519094 DOI: 10.1007/s00277-020-04106-2] [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: 04/03/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023]
Abstract
Post-transplantation thrombocytopenia (PT) is a common and severe complication which usually leads to poor prognosis. Eltrombopag (EPAG), a novel oral thrombopoietin (TPO) receptor agonist, has shown promising effects in thrombocytopenia due to immune thrombocytopenic purpura (ITP) and refractory severe aplastic anemia (rSAA), while the effectiveness of EPAG for PT patients still needs to be evaluated. A total of 32 PT patients receiving EPAG were retrospectively analyzed between September 2017 and July 2019, including 15 patients with poor graft function (PGF) and 17 patients with secondary failure of platelet recovery (SFPR). To date, 21 (65.6%) patients achieved overall recovery (OR) and 14 (43.8%) patients achieved complete recovery (CR). Among responders, 18 (85.7%) patients discontinued or tapered the drug and 16 (76.2%) patients successfully maintained their best response. During the EPAG treatment, responders received much lower median platelet transfusion units than non-responders (11 vs. 95, P < 0.001). After a median follow-up time of 364 days (range, 24-842), the overall survival in these patients was 78.1% (100% for responders and 36.4% for non-responders, P < 0.001). In the univariate and multivariate analysis, PGF was identified as the independent risk factor for OR (P = 0.041, HR = 5.333). Megakaryocyte (Megk) amounts (P = 0.025, HR = 14.638) and splenomegaly (P = 0.042, HR = 11.278) were identified as independent risk factors for CR. Besides, PGF patients tended to take a longer time to achieve PR and CR than SFPR patients. In conclusion, our data suggest that EPAG can promote platelet recovery and reduce platelet transfusion in PT patients.
Collapse
Affiliation(s)
- Fei Gao
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaoyu Zhou
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - Jimin Shi
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - Yi Luo
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - Yamin Tan
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - Huarui Fu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaoyu Lai
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - Jian Yu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China. .,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China.
| | - Yanmin Zhao
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, People's Republic of China. .,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
29
|
Dagdas S, Ucar MA, Ceran F, Gunes AK, Falay M, Ozet G. Comparison of allogenic stem cell transplantations performed with frozen or fresh stem cell products with regard to GVHD and mortality. Transfus Apher Sci 2020; 59:102742. [PMID: 32171686 DOI: 10.1016/j.transci.2020.102742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/05/2020] [Accepted: 02/20/2020] [Indexed: 11/30/2022]
Abstract
PURPOSE Stem cells are collected from donors and infused to the recipient in allogenic peripheral stem cell transplantations. The use of frozen stem cells can promote donor compatibility, and overcoming possible problems due to insufficient stem cell mobilization will also be easier. Nevertheless, studies about the use of frozen peripheral stem cells in allogenic transplantation are extremely rare. In this study, we aimed to compare the clinical outcomes of allogenic stem cell transplants from frozen or fresh stem cell products. MATERIALS AND METHODS This retrospective analysis was conducted between April 2004 and September 2018 in the bone marrow transplantation unit of Ankara Numune Training and Research Hospital. Clinical data of patients who received allogenic peripheral stem cell transplantations from fully matched sibling donors were compared for 42 fresh and 30 frozen stem cell transplants. RESULTS While the platelet engraftment period, febrile neutropenia period, hospitalization period, and 100-day mortality rates did not show any differences, the neutrophil engraftment period was longer in the frozen group (mean: 14 days vs. 16 days, p = 0.006). Acute and chronic graftversus-host disease (GVHD) rates were similar in both groups; however, the rate of grade 3 or4 chronic liver GVHD was slightly higher in transplants performed with fresh stem cells compared to the frozen group (p = 0.046). Overall survival was similar between the groups (p = 0.700). CONCLUSION The use of frozen peripheral stem cells in allogenic stem cell transplantation may be a reasonable option that can be applied without causing a significant change in clinical results.
Collapse
Affiliation(s)
- Simten Dagdas
- Ankara Numune Training and Research Hospital, Department of Hematology, Ankara, Turkey.
| | - Mehmet Ali Ucar
- Ankara Numune Training and Research Hospital, Department of Hematology, Ankara, Turkey.
| | - Funda Ceran
- Ankara Numune Training and Research Hospital, Department of Hematology, Ankara, Turkey.
| | - Ahmet Kursad Gunes
- Ankara Numune Training and Research Hospital, Department of Hematology, Ankara, Turkey.
| | - Mesude Falay
- Ankara Numune Training and Research Hospital, Department of Hematology, Ankara, Turkey.
| | - Gulsum Ozet
- Ankara Numune Training and Research Hospital, Department of Hematology, Ankara, Turkey.
| |
Collapse
|
30
|
Shafa M, Walsh T, Panchalingam KM, Richardson T, Menendez L, Tian X, Suresh Babu S, Dadgar S, Beller J, Yang F, Baghbaderani BA. Long-Term Stability and Differentiation Potential of Cryopreserved cGMP-Compliant Human Induced Pluripotent Stem Cells. Int J Mol Sci 2019; 21:ijms21010108. [PMID: 31877913 PMCID: PMC6982271 DOI: 10.3390/ijms21010108] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 01/08/2023] Open
Abstract
The clinical effectiveness of human induced pluripotent stem cells (iPSCs) is highly dependent on a few key quality characteristics including the generation of high quality cell bank, long-term genomic stability, post-thaw viability, plating efficiency, retention of pluripotency, directed differentiation, purity, potency, and sterility. We have already reported the establishment of iPSC master cell banks (MCBs) and working cell banks (WCBs) under current good manufacturing procedure (cGMP)-compliant conditions. In this study, we assessed the cellular and genomic stability of the iPSC lines generated and cryopreserved five years ago under cGMP-compliant conditions. iPSC lines were thawed, characterized, and directly differentiated into cells from three germ layers including cardiomyocytes (CMs), neural stem cells (NSCs), and definitive endoderm (DE). The cells were also expanded in 2D and 3D spinner flasks to evaluate their long-term expansion potential in matrix-dependent and feeder-free culture environment. All three lines successfully thawed and attached to the L7TM matrix, and formed typical iPSC colonies that expressed pluripotency markers over 15 passages. iPSCs maintained their differentiation potential as demonstrated with spontaneous and directed differentiation to the three germ layers and corresponding expression of specific markers, respectfully. Furthermore, post-thaw cells showed normal karyotype, negative mycoplasma, and sterility testing. These cells maintained both their 2D and 3D proliferation potential after five years of cryopreservation without acquiring karyotype abnormality, loss of pluripotency, and telomerase activity. These results illustrate the long-term stability of cGMP iPSC lines, which is an important step in establishing a reliable, long-term source of starting materials for clinical and commercial manufacturing of iPSC-derived cell therapy products.
Collapse
|
31
|
Chang YJ, Zhao XY, Huang XJ. Granulocyte Colony-Stimulating Factor-Primed Unmanipulated Haploidentical Blood and Marrow Transplantation. Front Immunol 2019; 10:2516. [PMID: 31749802 PMCID: PMC6842971 DOI: 10.3389/fimmu.2019.02516] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/09/2019] [Indexed: 12/25/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF), a growth factor for neutrophils, has been successfully used for stem cell mobilization and T cell immune tolerance induction. The establishment of G-CSF-primed unmanipulated haploidentical blood and marrow transplantation (The Beijing Protocol) has achieved outcomes for the treatment of acute leukemia, myelodysplastic syndrome, and severe aplastic anemia with haploidentical allografts comparable to those of human leukocyte antigen (HLA)-matched sibling donor transplantation. Currently, G-CSF-mobilized bone marrow and/or peripheral blood stem cell sources have been widely used in unmanipulated haploidentical transplant settings. In this review, we summarize the roles of G-CSF in inducing T cell immune tolerance. We discuss the recent advances in the Beijing Protocol, mainly focusing on strategies that have been used to improve transplant outcomes in cases of poor graft function, virus infections, and relapse. The application of G-CSF-primed allografts in other haploidentical modalities is also discussed.
Collapse
Affiliation(s)
- Ying-Jun Chang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Xiang-Yu Zhao
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Xiao-Jun Huang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University Institute of Hematology, Peking University People's Hospital, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
32
|
Velier M, Granata A, Bramanti S, Calmels B, Furst S, Legrand F, Harbi S, Faucher C, Devillier R, Blaise D, Mfarrej B, Lemarie C, Chabannon C. A matched-pair analysis reveals marginally reduced CD34+ cell mobilization on second occasion in 27 related donors who underwent peripheral blood stem cell collection twice at the same institution. Transfusion 2019; 59:3442-3447. [PMID: 31625183 DOI: 10.1111/trf.15545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/01/2019] [Accepted: 07/08/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND In a small proportion of cases, hematopoietic function is insufficient after allogeneic hematopoietic stem cell transplantation, as a result of poor graft function or graft failure. These complications are common indications of re-mobilization of the initial donor, either for a second allograft or for an infusion of CD34+ Selected stem Cell Boost (SCB). METHODS AND MATERIALS We retrospectively reviewed the results of two cycles of CD34+ cell mobilization and collection. CD34+ cells mobilized and collected at each cycle were compared. When CD34+ cell selection from the collected allogeneic mononuclear cells was indicated, it was performed with the Clinimacs Plus® medical device, and results from in-process and final quality checks were analyzed. To assess the efficacy of CD34+ SCB, transfusion needs before and after the infusion of selected CD34+ cells were calculated. RESULTS The median peripheral blood concentration of CD34+ cells/μL was marginally reduced during the second cycle (35.6 vs 33.8, p < 0.05); results revealed a strong correlation between paired values (r = 0.85). The cumulative number of collected CD34+ cells were similar for both cycles; the total processed blood volume was higher during the second cycle (p = 0.023). For CD34+ immune-selection procedures, CD34+ cell recovery and purity were respectively 57% and 95%, with a median T-cell depletion of 6.7 log. Recipients' needs for platelet and red blood cell transfusions were significantly reduced after CD34+ SCB. CONCLUSION This study confirms the feasibility of a second cycle of mobilization in healthy related donors and the benefits of CD34+ SCB on hematopoietic reconstitution.
Collapse
Affiliation(s)
- Melanie Velier
- Institut Paoli-Calmettes, Centre de Thérapie Cellulaire, Cell Collection & Cell Processing Facility, Marseille, France.,Inserm, Centre d'Investigations Cliniques de Marseille, Module Biothérapies, Marseille, France
| | - Angela Granata
- Institut Paoli-Calmettes, Centre de Thérapie Cellulaire, Cell Collection & Cell Processing Facility, Marseille, France.,Institut Paoli-Calmettes, Oncohématologie, Marseille, France
| | | | - Boris Calmels
- Institut Paoli-Calmettes, Centre de Thérapie Cellulaire, Cell Collection & Cell Processing Facility, Marseille, France.,Inserm, Centre d'Investigations Cliniques de Marseille, Module Biothérapies, Marseille, France
| | - Sabine Furst
- Institut Paoli-Calmettes, Oncohématologie, Marseille, France
| | - Faewzeh Legrand
- Institut Paoli-Calmettes, Oncohématologie, Marseille, France
| | - Samia Harbi
- Institut Paoli-Calmettes, Oncohématologie, Marseille, France
| | | | - Raynier Devillier
- Institut Paoli-Calmettes, Oncohématologie, Marseille, France.,Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Didier Blaise
- Institut Paoli-Calmettes, Oncohématologie, Marseille, France.,Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Bechara Mfarrej
- Institut Paoli-Calmettes, Centre de Thérapie Cellulaire, Cell Collection & Cell Processing Facility, Marseille, France.,Inserm, Centre d'Investigations Cliniques de Marseille, Module Biothérapies, Marseille, France
| | - Claude Lemarie
- Institut Paoli-Calmettes, Centre de Thérapie Cellulaire, Cell Collection & Cell Processing Facility, Marseille, France.,Inserm, Centre d'Investigations Cliniques de Marseille, Module Biothérapies, Marseille, France
| | - Christian Chabannon
- Institut Paoli-Calmettes, Centre de Thérapie Cellulaire, Cell Collection & Cell Processing Facility, Marseille, France.,Inserm, Centre d'Investigations Cliniques de Marseille, Module Biothérapies, Marseille, France.,Institut Paoli-Calmettes, Oncohématologie, Marseille, France.,Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| |
Collapse
|
33
|
Panch SR, Reddy OL, Li K, Bikkani T, Rao A, Yarlagadda S, Highfill S, Fowler D, Childs RW, Battiwalla M, Barrett J, Larochelle A, Mackall C, Shah N, Stroncek DF. Robust Selections of Various Hematopoietic Cell Fractions on the CliniMACS Plus Instrument. Clin Hematol Int 2019; 1:161-167. [PMID: 34595426 PMCID: PMC8432366 DOI: 10.2991/chi.d.190529.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/26/2019] [Indexed: 11/30/2022] Open
Abstract
Cell separation technologies play a vital role in the graft engineering of hematopoietic cellular fractions, particularly with the rapid expansion of the field of cellular therapeutics. The CliniMACS Plus Instrument (Miltenyi Biotec) utilizes immunomagnetic techniques to isolate hematopoietic progenitor cells (HPCs), T cells, NK cells, and monocytes. These products are ultimately used for HPC transplantation and for the manufacture of adoptive immunotherapies. We evaluated the viable cell recovery and cell purity of selections and depletions performed on the CliniMACS Plus over a 10-year period at our facility, specifically assessing for the isolation of CD34+, CD4+, CD3+/CD56+, CD4+/CD8+, and CD25+ cells. Additionally, patient- and instrument-related factors affecting these parameters were examined. Viable cell recovery ranged from 32.3 ± 10.2% to 65.4 ± 15.4%, and was the highest for CD34+ selections. Cell purity ranged from 86.3 ± 7.2% to 99.0 ± 1.1%, and was the highest for CD4+ selections. Undesired cell fractions demonstrated a range of 1.2 ± 0.45 to 5.1 ± 0.4 log reductions. Red cell depletions averaged 2.12 ± 0.68 logs, while platelets were reduced by an average of 4.01 ± 1.57 logs. Donor characteristics did not impact viable cell recovery or cell purity for CD34+ or CD4+ cell enrichments; however, these were affected by manufacturing variables, including tubing size, bead quantity, and whether preselection platelet washes were performed. Our data demonstrate the efficient recovery of hematopoietic cellular fractions on the CliniMACS Plus that may be optimized by adjusting manufacturing variables.
Collapse
Affiliation(s)
- Sandhya R Panch
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Opal L Reddy
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Katherine Li
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Thejaswi Bikkani
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Anusha Rao
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Swathi Yarlagadda
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Steven Highfill
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Daniel Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard W Childs
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Minocher Battiwalla
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John Barrett
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andre Larochelle
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Crystal Mackall
- Cancer Immunology and Immunotherapy Program, Stanford Cancer Institute, Palo Alto, California, USA
| | - Nirali Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - David F Stroncek
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| |
Collapse
|
34
|
Virus reactivation and low dose of CD34+ cell, rather than haploidentical transplantation, were associated with secondary poor graft function within the first 100 days after allogeneic stem cell transplantation. Ann Hematol 2019; 98:1877-1883. [DOI: 10.1007/s00277-019-03715-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/06/2019] [Indexed: 12/17/2022]
|
35
|
Fei XH, He JB, Cheng HY, Yin YM, Zhang WJ, Zhang SQ, Wang XC, Wang JB. [Effects of CD34(+) selected stem cells for the treatment of poor graft function after allogeneic stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 39:828-832. [PMID: 30369204 PMCID: PMC7348294 DOI: 10.3760/cma.j.issn.0253-2727.2018.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
目的 观察输注纯化供者CD34+细胞治疗单倍型造血干细胞移植后移植物功能不良的疗效和安全性。 方法 对2014年1月至2018年3月期间接受供者纯化CD34+细胞治疗的12例单倍型造血干细胞移植后移植物功能不良患者进行回顾性分析。 结果 12例患者中男9例、女3例,中位年龄26(14~54)岁;急性淋巴细胞白血病4例,急性髓系白血病5例,慢性髓性白血病2例,慢性粒-单核细胞白血病1例。分选产物CD34+细胞纯度为92.0%(44.0%~97.0%),回收率为55.0%(45.0%~96.7%),回输CD34+细胞的中位数为1.9(0.9~4.4)×106/kg,CD3+细胞中位数为0.6(0.3~2.0)×104/kg。中性粒细胞恢复中位时间为18(14~39)d,血小板恢复中位时间为29(16~153)d,红细胞恢复中位时间为60(9~124)d。12例患者输注过程中未发生严重不良反应,10例获得完全缓解,1例患者获得部分缓解,1例患者无效,未发生重症感染和重度GVHD。 结论 回输供者纯化CD34+细胞是单倍型造血干细胞移植后植入功能不良的一种安全、有效治疗方法。
Collapse
Affiliation(s)
- X H Fei
- Aerospace Center Hospital of Peking University, Beijing 100049, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Yafour N, Couturier MA, Azarnoush S, Girault S, Hermet E, Masouridi Levrat S, Schmidt A, Michallet M, Etancelin P, Guillaume T, Malard F, Sirvent A, Yakoub-Agha I, Poiré X. [Second allogeneic hematopoietic stem cell transplant: Guidelines from the francophone Society of bone marrow transplantation and cellular therapy (SFGM-TC)]. Bull Cancer 2018; 106:S40-S51. [PMID: 30409466 DOI: 10.1016/j.bulcan.2018.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/31/2018] [Indexed: 01/10/2023]
Abstract
Disease recurrence and graft dysfunction after allogeneic hematopoietic stem cell transplantation (allo-HSCT) currently remain among the major causes of treatment failure in malignant and non-malignant hematological diseases. A second allo-HSCT is a valuable therapeutic option to salvage those situations. During the 8th annual harmonization workshops of the french Society of bone marrow transplantation and cellular therapy (SFGM-TC), a designated working group reviewed the literature in order to elaborate unified guidelines on feasibility, indications, donor choice and conditioning in the case of a second allo-HSCT. In case of relapse, a second allo-HSCT with reduced intensity or non-myeloablative conditioning is a reasonable option, particularly in patients with a good performance status (Karnofsky/Lansky>80%), low co-morbidity score (EBMT score≤3), a longer remission duration after the first allo-HSCT (>6 months), and who present low disease burden at the time of second allo-HSCT. Matched related donors tend to be associated with better outcomes. In the presence of graft dysfunction (primary and secondary graft rejection), an immunoablative conditioning regimen is recommended. A donor change remains a valid option, especially in the absence of graft-versus-host disease after the first allo-HSCT.
Collapse
Affiliation(s)
- Nabil Yafour
- Établissement Hospitalier et Universitaire 1er-Novembre 1954, service d'hématologie et de thérapie cellulaire, BP 4166, 31000 Ibn Rochd, Oran, Algérie; Université d'Oran 1, Ahmed Ben Bella, faculté de médecine, Oran, Algérie.
| | - Marie Anne Couturier
- Hôpital Morvan, institut cancérologie-hématologie, CHRU Brest, 2, avenue Foch, 29200 Brest, France
| | - Saba Azarnoush
- Université Paris Diderot, hôpital Robert-Debré, service d'immuno-hématologie pédiatrique, 48, boulevard Sérurier, 75019 Paris, France
| | - Stéphane Girault
- CHU Limoges, hématologie clinique et thérapie cellulaire, 2, avenue Martin-Luther-King, 87042 Limoges, France
| | - Eric Hermet
- Université d'Auvergne EA3846, CIC-501, CHU Estaing, service de thérapie cellulaire et d'hématologie clinique adulte, Clermont-Ferrand, 58, rue Montalembert, 63000 Clermont-Ferrand, France
| | - Stavroula Masouridi Levrat
- Geneva university hospitals, division of hematology, department of medical specialties, rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Suisse
| | - Aline Schmidt
- CHU d'Angers, maladies du sang, 4, rue Larrey, 49100 Angers, France; Université d'Angers, Inserm U892/CNRS 6299, 49035 Angers, France
| | - Mauricette Michallet
- Centre hospitalier Lyon Sud, hématologie clinique, 165, chemin du Grand-Revoyet, 69495 Pierre-Bénite cedex Lyon, France
| | - Pascaline Etancelin
- Centre Henri-Becquerel, laboratoire de génétique oncologique, rue d'Amiens, 76000 Rouen, France
| | - Thierry Guillaume
- CHU de Nantes, Hôtel-Dieu, service d'hématologie, 1, place Ricordeau, 44000 Nantes, France
| | - Florent Malard
- AP-HP, hôpital Saint-Antoine, service d'hématologie et de thérapie cellulaire, 75012 Paris, France
| | - Anne Sirvent
- CHU Montpellier, hôpital Arnaud-de-Villeneuve, service de l'onco-hématologie pédiatrique, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France
| | - Ibrahim Yakoub-Agha
- CHU de Lille, maladies du sang, unité d'Allogreffe de CSH, 59000 Lille, France; Université de Lille 2, LIRIC, Inserm U995, 59000 Lille, France
| | - Xavier Poiré
- Cliniques universitaires Saint-Luc, service d'hématologie, 10, avenue Hippocrate, 1200 Bruxelles, Belgique
| |
Collapse
|
37
|
Platelet number and graft function predict intensive care survival in allogeneic stem cell transplantation patients. Ann Hematol 2018; 98:491-500. [PMID: 30406350 DOI: 10.1007/s00277-018-3538-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 10/26/2018] [Indexed: 12/18/2022]
Abstract
Despite significant advances in the treatment of complications requiring intensive care unit (ICU) admission, ICU mortality remains high for patients after allogeneic stem cell transplantation. We evaluated the role of thrombocytopenia and poor graft function in allogeneic stem cell recipients receiving ICU treatments along with established prognostic ICU markers in order to identify patients at risk for severe complications. At ICU admission, clinical and laboratory data of 108 allogeneic stem cell transplanted ICU patients were collected and retrospectively analyzed. Platelet counts (≤ 50,000/μl, p < 0.0005), hemoglobin levels (≤ 8.5 mg/dl, p = 0.019), and leukocyte count (≤ 1500/μl, p = 0.025) along with sepsis (p = 0.002) and acute myeloid leukemia (p < 0.0005) correlated significantly with survival. Multivariate analysis confirmed thrombocytopenia (hazard ratio (HR) 2.79 (1.58-4.92, 95% confidence interval (CI)) and anemia (HR 1.82, 1.06-3.11, 95% CI) as independent mortality risk factors. Predominant ICU diagnoses were acute respiratory failure (75%), acute kidney injury (47%), and septic shock (30%). Acute graft versus host disease was diagnosed in 42% of patients, and 47% required vasopressors. Low platelet (≤ 50,000/μl) and poor graft function are independent prognostic factors for impaired survival in critically ill stem cell transplanted patients. The underlying pathophysiology of poor graft function is not fully understood and currently under investigation. High-risk patients may be identified and ICU treatments stratified according to allogeneic stem cell patients' individual risk profiles. In contrast to previous studies involving medical or surgical ICU patients, the fraction of thrombocytopenic patients was larger and low platelets were a better differentiating factor in multivariate analysis than any other parameter.
Collapse
|
38
|
Tang C, Chen F, Kong D, Ma Q, Dai H, Yin J, Li Z, Chen J, Zhu X, Mao X, Wu D, Tang X. Successful treatment of secondary poor graft function post allogeneic hematopoietic stem cell transplantation with eltrombopag. J Hematol Oncol 2018; 11:103. [PMID: 30115080 PMCID: PMC6097332 DOI: 10.1186/s13045-018-0649-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/08/2018] [Indexed: 12/01/2022] Open
Abstract
Poor graft function (PGF) is a life-threatening complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Current treatment strategies include the use of growth factors, CD34+-selected stem cell boost, mesenchymal stem cell transfusion, and second allo-HSCT, but these treatments are not effective in all patients. Eltrombopag, an oral thrombopoietin receptor agonist, which showed promising results in severe aplasia anemia, may be an alternative choice for PGF patients. Therefore, we treated 12 patients who responded poorly to standard treatments for secondary PGF after allo-HSCT with eltrombopag. The median duration was 116 (35–1000) days from transplantation to PGF diagnosis and 59 (30–180) days from PGF diagnosis to eltrombopag treatment. Eltrombopag was started at a dose of 25 mg/d for 3 days and then increased to 50 or 75 mg/d. Median treatment duration was 8 (2–23) weeks. Ten patients (83.3%) responded to the treatment: 8 achieved complete response (CR), and the remaining 2 achieved partial response. In the 10 responding subjects, median platelet count was 18 (5–27) × 109/L vs 74 (30–117) × 109/L prior to and after treatment. Neutrophil count was 0.51 (0.28–0.69) × 109/L vs 1.84 (0.78–4.90) × 109/L. Hemoglobin was 88 (63–123) vs 101 (78–134) g/L. In the 8 patients who achieved CR, the time from eltrombopag initiation to achieving CR was 29 (10–49) days; the response lasted until the last follow-up in all 8 CR subjects (10–18 months). The 12-month overall survival rate was 83.3%. There was no treatment-related mortality and no evidence of cataract, thrombosis, or any other grade 3/4 toxicities.
Collapse
Affiliation(s)
- Cen Tang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Feng Chen
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Danqing Kong
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Qinfen Ma
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haiping Dai
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jia Yin
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zheng Li
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jia Chen
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiaming Zhu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China.,Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xinliang Mao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China. .,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou, 510405, China.
| | - Depei Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China. .,Institute of Blood and Marrow Transplantation, Suzhou, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Xiaowen Tang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology, Suzhou, China. .,Institute of Blood and Marrow Transplantation, Suzhou, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|