1
|
Konuma T, Fujioka M, Fuse K, Hosoi H, Masamoto Y, Doki N, Uchida N, Tanaka M, Sawa M, Nishida T, Ishikawa J, Asada N, Nakamae H, Hasegawa Y, Onizuka M, Maeda T, Fukuda T, Kawamura K, Kanda Y, Ohbiki M, Atsuta Y, Itonaga H. Clinical effects of granulocyte colony-stimulating factor administration and the timing of its initiation on allogeneic hematopoietic cell transplantation outcomes for myelodysplastic syndrome. Transplant Cell Ther 2025:S2666-6367(25)01099-1. [PMID: 40107381 DOI: 10.1016/j.jtct.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Granulocyte colony-stimulating factor (G-CSF) accelerates neutrophil recovery after allogeneic hematopoietic cell transplantation (HCT). However, the optimal use of G-CSF and the timing of its initiation after allogeneic HCT for myelodysplastic syndrome (MDS) according to graft type have not been determined. This retrospective study aimed to investigate the effects of using G-CSF administration and the timing of its initiation on transplant outcomes in adult patients with MDS undergoing allogeneic HCT. Using Japanese registry data, we retrospectively investigated the effects of G-CSF administration and the timing of its initiation on transplant outcomes among 4140 adults with MDS after bone marrow transplantation (BMT), peripheral blood stem cell transplantation (PBSCT), or single-unit cord blood transplantation (CBT) between 2013 and 2022. Multivariate analysis showed that early (days 0 to 4) and late (days 5 to 10) G-CSF administration significantly accelerated neutrophil recovery compared with no G-CSF administration following BMT, PBSCT, and CBT, but there was no benefit of early G-CSF initiation for early neutrophilic recovery regardless of graft type. Late G-CSF initiation was significantly associated with a higher risk of overall chronic GVHD following PBSCT (hazard ratio [HR], 1.63; 95% confidence interval [CI], 1.18 to 2.24; P = .002) and CBT (HR, 2.09; 95% CI, 1.21 to 3.60; P = .007) compared with no G-CSF administration. Late G-CSF initiation significantly improved OS compared with no G-CSF administration only following PBSCT (HR, 0.74; 95% CI, 0.58 to 0.94; P = .015). However, G-CSF administration and the timing of its initiation did not affect acute GVHD, relapse, or non-relapse mortality, irrespective of graft type. These results suggest that G-CSF administration significantly accelerated neutrophil recovery after BMT, PBSCT, and CBT, but increased risk of overall chronic GVHD after PBSCT and CBT. However, the effect of early and late G-CSF initiation on transplant outcomes needs further study in adult patients with MDS.
Collapse
Affiliation(s)
- Takaaki Konuma
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Machiko Fujioka
- Department of Hematology, Sasebo City General Hospital, Sasebo, Japan
| | - Kyoko Fuse
- Faculty of Medicine, Department of Hematology, Endocrinology and Metabolism, Niigata University, Niigata, Japan
| | - Hiroki Hosoi
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Yosuke Masamoto
- Department of Cell Therapy and Transplantation Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Naoyuki Uchida
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Masatsugu Tanaka
- Department of Hematology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Anjo, Japan
| | - Tetsuya Nishida
- Department of Hematology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan
| | - Jun Ishikawa
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yuta Hasegawa
- Department of Hematology, Hokkaido University Hospital, Sapporo, Japan
| | - Makoto Onizuka
- Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Takeshi Maeda
- Department of Hematology and oncology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Koji Kawamura
- Department of Hematology, Tottori University Hospital, Yonago, Tottori, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Jichi Medical University, Shimotsuke, Japan
| | - Marie Ohbiki
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan; Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Nagakute, Japan; Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan; Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hidehiro Itonaga
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, Japan
| |
Collapse
|
2
|
Alnughmush A, Sayyed A, Remberger M, AL-Shaibani E, Chen C, Chiarello C, Pasic I, Novitzky-Basso I, Law AD, Lam W, Kim D(DH, Gerbitz A, Viswabandya A, Kumar R, Michelis FV, Mattsson J. Impact of Granulocyte Colony-Stimulating Factor (G-CSF) on Clinical Outcomes in Allogeneic Hematopoietic Cell Transplantation: Does Speeding Up Neutrophil Engraftment Make a Difference? Transplant Direct 2025; 11:e1753. [PMID: 39802196 PMCID: PMC11723673 DOI: 10.1097/txd.0000000000001753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025] Open
Abstract
Background Despite decades of post-allogeneic hematopoietic cell transplantation (HCT) growth factor utilization, its role remains undefined, leading to ongoing debates and research. The theoretical impacts of growth factors have been challenged in numerous studies. Methods In this retrospective cohort study conducted at the Princess Margaret Cancer Centre, we analyzed the clinical outcomes of 509 patients who underwent allogeneic HCT between May 1, 2019, and May 31, 2022. This study aimed to assess the impact of granulocyte colony-stimulating factor (G-CSF) administration posttransplantation on neutrophil and platelet engraftment, incidence of bloodstream infections (BSIs), graft-versus-host disease, engraftment syndrome (ES), and survival metrics including overall survival, nonrelapse mortality, and graft-versus-host disease-free/relapse-free survival. Results Our findings indicate that G-CSF administration expedited neutrophil engraftment (16 versus 18 d, P = 0.009) and was associated with a decreased incidence of BSI (9.4% versus 31.3%, P = 0.014). However, this benefit was counterbalanced by a significant delay in platelet engraftment (21 versus 17 d, P < 0.001). Multivariate logistic regression analysis identified mismatched donors (odds ratio, 1.72; 95% confidence interval, 1.03-2.88; P = 0.038) and the duration of G-CSF therapy (odds ratio, 1.04; 95% confidence interval, 1.00-1.09; P = 0.038) as independent predictors for the development of ES. Despite these hematological impacts, there was no observed advantage in overall survival, nonrelapse mortality, or graft-versus-host disease-free/relapse-free survival among patients who received G-CSF compared with those who did not. Conclusions Although G-CSF post-HCT expedited neutrophil engraftment and reduced BSI risk, it did not result in a survival advantage. The association with ES necessitates careful consideration.
Collapse
Affiliation(s)
- Ahmed Alnughmush
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Hematology, Stem Cell Transplant and Cellular Therapy, Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ayman Sayyed
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Division of Clinical Sciences, Department of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Mats Remberger
- KFUE, Uppsala University Hospital, and IMV, Uppsala University, Uppsala, Sweden
| | - Eshrak AL-Shaibani
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Carol Chen
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Caden Chiarello
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ivan Pasic
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Igor Novitzky-Basso
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Arjun Datt Law
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wilson Lam
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Dennis (Dong Hwan) Kim
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Armin Gerbitz
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Auro Viswabandya
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Rajat Kumar
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Fotios V. Michelis
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jonas Mattsson
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Araki D, Chen V, Redekar N, Salisbury-Ruf C, Luo Y, Liu P, Li Y, Smith RH, Dagur P, Combs C, Larochelle A. Post-transplant G-CSF impedes engraftment of gene-edited human hematopoietic stem cells by exacerbating p53-mediated DNA damage response. Cell Stem Cell 2025; 32:53-70.e8. [PMID: 39536761 PMCID: PMC11698648 DOI: 10.1016/j.stem.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 05/06/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Granulocyte-colony-stimulating factor (G-CSF) is commonly used to accelerate recovery from neutropenia following chemotherapy and autologous transplantation of hematopoietic stem and progenitor cells (HSPCs) for malignant disorders. However, its utility after ex vivo gene therapy in human HSPCs remains unexplored. We show that administering G-CSF from day 1 to 14 post-transplant impedes engraftment of CRISPR-Cas9 gene-edited human HSPCs in murine xenograft models. G-CSF affects gene-edited HSPCs through a cell-intrinsic mechanism, causing proliferative stress and amplifying the early p53-mediated DNA damage response triggered by Cas9-mediated DNA double-strand breaks. This underscores a threshold mechanism where p53 activation must reach a critical level to impair cellular function. Transiently inhibiting p53 or delaying the initiation of G-CSF treatment to day 5 post-transplant attenuates its negative impact on gene-edited HSPCs. The potential for increased HSPC toxicity associated with post-transplant G-CSF administration in CRISPR-Cas9 autologous HSPC gene therapy warrants consideration in clinical trials.
Collapse
Affiliation(s)
- Daisuke Araki
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Vicky Chen
- Integrated Data Science Services (IDSS), National Institutes of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Neelam Redekar
- Integrated Data Science Services (IDSS), National Institutes of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Christi Salisbury-Ruf
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Yan Luo
- DNA Sequencing and Genomics Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Poching Liu
- DNA Sequencing and Genomics Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Yuesheng Li
- DNA Sequencing and Genomics Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Richard H Smith
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Pradeep Dagur
- Flow Cytometry Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Christian Combs
- Light Microscopy Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Andre Larochelle
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
4
|
Konuma T, Kameda K, Morita K, Kondo T, Kimura F, Nakasone H, Ouchi F, Uchida N, Tanaka M, Nishida T, Fukuda T, Hasegawa Y, Sakata‐Yanagimoto M, Onizuka M, Sawa M, Ota S, Asada N, Fujiwara S, Yoshihara S, Ishimaru F, Yoshimitsu M, Kanda Y, Ohbiki M, Atsuta Y, Yanada M. Different impacts of granulocyte colony-stimulating factor administration on allogeneic hematopoietic cell transplant outcomes for adult acute myeloid leukemia according to graft type. Am J Hematol 2025; 100:66-77. [PMID: 39564683 PMCID: PMC11625993 DOI: 10.1002/ajh.27521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024]
Abstract
We retrospectively evaluated the impacts of using granulocyte colony-stimulating factor (G-CSF) and its timing on posttransplant outcomes for 9766 adults with acute myeloid leukemia (AML) between 2013 and 2022 using a Japanese database. We separately evaluated three distinct cohorts based on graft type: 3248 received bone marrow transplantation (BMT), 3066 received peripheral blood stem cell transplantation (PBSCT), and 3452 received single-unit cord blood transplantation (CBT). Multivariate analysis showed that G-CSF administration significantly accelerated neutrophil recovery after BMT, PBSCT, and CBT. However, it was associated with a higher risk of grades II-IV acute graft-versus-host disease (GVHD) across all graft types. Moreover, an increased incidence of overall chronic GVHD was observed with G-CSF administration in BMT and CBT patients, but not in PBSCT patients. G-CSF administration significantly improved overall survival (OS) and leukemia-free survival (LFS) only following CBT. Regarding the timing of G-CSF, in comparison with late initiation of G-CSF (Days 5-10), early initiation (Days 0-4) did not provide benefits for hematopoietic recovery regardless of graft type. In contrast, late initiation was significantly associated with a lower risk of grades II-IV acute GVHD and better OS and LFS in CBT patients. These data demonstrated that G-CSF administration accelerated neutrophil recovery and increased the risk of grades II-IV acute GVHD across all graft types, but significantly improved survival outcomes but only following CBT. Therefore, routine use of G-CSF should be considered for CBT in adult patients with AML.
Collapse
Affiliation(s)
- Takaaki Konuma
- Department of Hematology/Oncology, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Kazuaki Kameda
- Division of HematologyJichi Medical University Saitama Medical CenterSaitamaJapan
| | - Kaoru Morita
- Division of HematologyJichi Medical UniversityShimotsukeJapan
| | - Tadakazu Kondo
- Department of HematologyKobe City Medical Center General HospitalKobeJapan
| | - Fumihiko Kimura
- Division of Hematology, Department of Internal MedicineNational Defense Medical CollegeTokorozawaJapan
| | - Hideki Nakasone
- Division of HematologyJichi Medical University Saitama Medical CenterSaitamaJapan
- Division of Stem Cell Regulation, Center for Molecular MedicineJichi Medical UniversityShimotsukeJapan
| | - Fumihiko Ouchi
- Hematology DivisionTokyo Metropolitan Cancer and Infectious Diseases Center, Komagome HospitalTokyoJapan
| | | | | | - Tetsuya Nishida
- Department of HematologyJapanese Red Cross Aichi Medical Center Nagoya Daiichi HospitalNagoyaJapan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell TransplantationNational Cancer Center HospitalTokyoJapan
| | - Yuta Hasegawa
- Department of HematologyHokkaido University HospitalSapporoJapan
| | | | - Makoto Onizuka
- Department of Hematology and OncologyTokai University School of MedicineIseharaJapan
| | - Masashi Sawa
- Department of Hematology and OncologyAnjo Kosei HospitalAnjoJapan
| | - Shuichi Ota
- Department of HematologySapporo Hokuyu HospitalSapporoJapan
| | - Noboru Asada
- Department of Hematology and OncologyOkayama University HospitalOkayamaJapan
| | | | - Satoshi Yoshihara
- Department of HematologyHyogo Medical University HospitalNishinomiyaJapan
| | - Fumihiko Ishimaru
- Technical DepartmentJapanese Red Cross Society Blood Service HeadquartersTokyoJapan
| | - Makoto Yoshimitsu
- Department of Hematology and RheumatologyKagoshima University HospitalKagoshimaJapan
| | - Yoshinobu Kanda
- Division of HematologyJichi Medical UniversityShimotsukeJapan
| | - Marie Ohbiki
- Japanese Data Center for Hematopoietic Cell TransplantationNagakuteJapan
- Department of Registry Science for Transplant and Cellular TherapyAichi Medical University School of MedicineNagakuteJapan
- Department of Hematology and OncologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell TransplantationNagakuteJapan
- Department of Registry Science for Transplant and Cellular TherapyAichi Medical University School of MedicineNagakuteJapan
| | - Masamitsu Yanada
- Department of Hematology and OncologyNagoya City University East Medical CenterNagoyaJapan
| |
Collapse
|
5
|
Escribano-Serrat S, Pedraza A, Suárez-Lledó M, Charry P, De Moner B, Martinez-Sanchez J, Ramos A, Ventosa-Capell H, Moreno C, Guardia L, Monge-Escartín I, Riu G, Carcelero E, Cid J, Lozano M, Gómez P, García E, Martín L, Carreras E, Fernández-Avilés F, Martínez C, Rovira M, Salas MQ, Díaz-Ricart M. Safety and efficacy of G-CSF after allogeneic hematopoietic cell transplantation using post-transplant cyclophosphamide: clinical and in vitro examination of endothelial activation. Bone Marrow Transplant 2024; 59:1466-1476. [PMID: 39117736 DOI: 10.1038/s41409-024-02388-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Since 2021 the use of G-CSF was implemented in allo-HCT with PTCY-based prophylaxis with the aim of shortening the aplastic phase and reducing infectious complications. This study investigates the effectiveness of this change in protocol performed at our institution. One-hundred forty-six adults undergoing allo-HCT with PTCY-based prophylaxis were included, and among them, 58 (40%) received G-CSF. The median of days to neutrophil engraftment was shorter in the G-CSF group (15 vs. 20 days, p < 0.001). Patients receiving G-CSF had a lower incidence of day +30 bacterial bloodstream infections (BSI) than the rest (20.7% vs. 47.7%, p < 0.001). GVHD, SOS, and TA-TMA incidences were comparable between groups, and using G-CSF did not impact on survival. Endothelial activation was investigated using EASIX and by the measurement of soluble biomarkers in cryopreserved plasma samples obtained on days 0, +7, +14 and +21 of 39 consecutive patients (10 received G-CSF) included in the study. EASIX, VWF:Ag, sVCAM-1, sTNFRI, ST2, REG3α, TM and NETs medians values were comparable in patients receiving G-CSF and those who did not. Compared with allo-HCT performed without G-CSF, the addition of G-CSF to PTCY-based allo-HCT accelerated neutrophil engraftment contributing on decreasing BSI incidence, and without inducing additional endothelial activation.
Collapse
Affiliation(s)
- Silvia Escribano-Serrat
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alexandra Pedraza
- Blood Bank Department, Biomedical Diagnostic Center, Banc de Sang i Teixits, Hospital Clínic de Barcelona, Barcelona, Spain
| | - María Suárez-Lledó
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopoietic Transplantation Unit, Hematology Department, (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Paola Charry
- Apheresis and Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, (ICAMS), Hospital Clínic Barcelona, Barcelona, Spain
| | - Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundacio i Institut de Recerca Josep Carreras Contra la Leucemia (Campus Clínic), Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Alex Ramos
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Cristina Moreno
- Hematopoietic Transplantation Unit, Hematology Department, (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Laia Guardia
- Hematopoietic Transplantation Unit, Hematology Department, (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Gisela Riu
- Pharmacy Clinic Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Esther Carcelero
- Pharmacy Clinic Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Joan Cid
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Apheresis and Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, (ICAMS), Hospital Clínic Barcelona, Barcelona, Spain
| | - Miquel Lozano
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Apheresis and Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, (ICAMS), Hospital Clínic Barcelona, Barcelona, Spain
| | - Pilar Gómez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Estefanía García
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lidia Martín
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Enric Carreras
- Fundacio i Institut de Recerca Josep Carreras Contra la Leucemia (Campus Clínic), Barcelona, Spain
| | - Francesc Fernández-Avilés
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopoietic Transplantation Unit, Hematology Department, (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Carmen Martínez
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopoietic Transplantation Unit, Hematology Department, (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Montserrat Rovira
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Hematopoietic Transplantation Unit, Hematology Department, (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - María Queralt Salas
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Hematopoietic Transplantation Unit, Hematology Department, (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain.
| | - Maribel Díaz-Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
- Research Biomedical Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
6
|
Aydın Kaynar L, Özkurt ZN. The Effect of Granulocyte Colony-Stimulating Factor (G-CSF) on Early Complications and Graft-Versus-Host Disease (GVHD) in Allogeneic Stem Cell Transplantation (ASCT) Recipients. Cureus 2023; 15:e46105. [PMID: 37779681 PMCID: PMC10534265 DOI: 10.7759/cureus.46105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 10/03/2023] Open
Abstract
Objectives Granulocyte colony-stimulating factor (G-CSF) is commonly used to accelerate neutrophil recovery after allogeneic stem cell transplantation (ASCT) in most transplant centers. There was no consensus on the optimal use of G-CSF after ASCT. Although we use G-CSF to minimize morbidity and mortality, G-CSF can increase the risk of graft-versus-host disease (GVHD). In our study, we want to show the effect of prophylactic G-CSF on infection frequency, neutrophil and platelet engraftment, the duration of neutropenia, the development of GVHD, hospitalization time, and transplant-related mortality (TRM) after ASCT. Materials and methods One hundred (71 males and 29 females) patients who did not receive G-CSF and 100 (58 males and 42 females) patients who received prophylactic G-CSF were included in the study. Results Age, diagnosis, the time between diagnosis and transplantation, preparation regimen, donor type, and the number of infused cluster of differentiation (CD) 34+ cells were not different in both groups (p>0.05). The frequency of female patients was higher in the group receiving G-CSF. Febrile neutropenia was more frequent in patients who did not receive G-CSF. Neutrophil engraftment and platelet engraftment were detected longer in patients not receiving G-CSF. The frequency of veno-occlusive disease (VOD) and hyperacute, chronic, and acute GVHD was not different in both groups (p>0.05). One hundred-day TRM and five-year overall survival (OS) were similar in the two groups (p>0.05). Conclusions Our study supports that G-CSF usage does not cause an increase in the frequency of GVHD and has a positive effect on the process by accelerating myeloid engraftment. In light of the data in our study, we can say that the use of G-CSF should be investigated in a randomized controlled clinical trial.
Collapse
|
7
|
Araki D, Chen V, Redekar N, Salisbury-Ruf C, Luo Y, Liu P, Li Y, Smith RH, Dagur P, Combs C, Larochelle A. Post-Transplant Administration of G-CSF Impedes Engraftment of Gene Edited Human Hematopoietic Stem Cells by Exacerbating the p53-Mediated DNA Damage Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547089. [PMID: 37425704 PMCID: PMC10327043 DOI: 10.1101/2023.06.29.547089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Granulocyte colony stimulating factor (G-CSF) is commonly used as adjunct treatment to hasten recovery from neutropenia following chemotherapy and autologous transplantation of hematopoietic stem and progenitor cells (HSPCs) for malignant disorders. However, the utility of G-CSF administration after ex vivo gene therapy procedures targeting human HSPCs has not been thoroughly evaluated. Here, we provide evidence that post-transplant administration of G-CSF impedes engraftment of CRISPR-Cas9 gene edited human HSPCs in xenograft models. G-CSF acts by exacerbating the p53-mediated DNA damage response triggered by Cas9- mediated DNA double-stranded breaks. Transient p53 inhibition in culture attenuates the negative impact of G-CSF on gene edited HSPC function. In contrast, post-transplant administration of G-CSF does not impair the repopulating properties of unmanipulated human HSPCs or HSPCs genetically engineered by transduction with lentiviral vectors. The potential for post-transplant G-CSF administration to aggravate HSPC toxicity associated with CRISPR-Cas9 gene editing should be considered in the design of ex vivo autologous HSPC gene editing clinical trials.
Collapse
|
8
|
Sarıcı A, Erkurt MA, Kuku İ, Kaya E, Berber İ, Biçim S, Hidayet E, Kaya A, Keser MF, Bahçecioğlu ÖF, Uysal A. The effect of G-CSF used after allogeneic hematopoietic stem cell transplantation on engraftment times and platelet suspension replacement numbers. Transfus Apher Sci 2022; 61:103482. [DOI: 10.1016/j.transci.2022.103482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/25/2022] [Accepted: 06/04/2022] [Indexed: 10/18/2022]
|
9
|
Hatch RV, Freyer CW, Carulli A, Redline G, Babushok DV, Frey NV, Gill SI, Hexner EO, Luger SM, Martin ME, McCurdy SR, Perl AE, Porter DL, Pratz KW, Stadtmauer EA, Loren AW. Day 4 vs. day 12 G-CSF administration following reduced intensity peripheral blood allogeneic stem cell transplant. J Oncol Pharm Pract 2022; 28:892-897. [PMID: 35191732 DOI: 10.1177/10781552221080710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Granulocyte colony-stimulating factor (G-CSF) hastens neutrophil engraftment and reduces infections after allogeneic hematopoietic cell transplant (alloHCT), yet the optimal start date is unknown. Additionally, concurrent G-CSF and methotrexate for graft-vs-host disease (GVHD) prophylaxis may potentiate myelosuppression, and prolonged G-CSF is costly. Our institution changed from day + 4 to day + 12 G-CSF initiation following reduced intensity (RIC) alloHCT with methotrexate GVHD prophylaxis. METHODS We retrospectively compared day + 4 and day + 12 G-CSF initiation after RIC alloHCT from 2017-2021. The primary endpoint was the time to neutrophil engraftment. Secondary endpoints included length of stay (LOS) and the time to platelet engraftment as well as the incidence of infectious events, acute GVHD (aGVHD), and mucositis. RESULTS Thirty-two patients were included in each group with similar baseline characteristics. We observed faster neutrophil engraftment (median 12 vs. 15 days, p = 0.01) and platelet engraftment (median 13 vs. 15 days, p = 0.026) with day + 4 vs. day + 12 G-CSF initiation. Median LOS was 23 days (range, 19-32) with day + 4 initiation vs. 24 days (21-30) with day + 12 (p = 0.046). The incidence of culture-negative febrile neutropenia (p = 0.12), any grade aGVHD (p = 0.58), and grade 2-4 mucositis (p = 0.8) were similar between groups. CONCLUSION Compared to day + 4, day + 12 G-CSF initiation following RIC alloHCT had a longer time to neutrophil and platelet engraftment. Day + 12 initiation also resulted in longer LOS, which while statistically significant, was potentially of limited clinical significance. These findings are hypothesis generating.
Collapse
Affiliation(s)
- Rachel V Hatch
- Department of Pharmacy, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Craig W Freyer
- Department of Pharmacy, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Alison Carulli
- Department of Pharmacy, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Gretchen Redline
- Department of Pharmacy, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Daria V Babushok
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Noelle V Frey
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Saar I Gill
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Elizabeth O Hexner
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Selina M Luger
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Mary Ellen Martin
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Shannon R McCurdy
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Alexander E Perl
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - David L Porter
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Keith W Pratz
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Edward A Stadtmauer
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Alison W Loren
- Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and Oncology, 21798Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
10
|
Goedhart M, Slot E, Pascutti MF, Geerman S, Rademakers T, Nota B, Huveneers S, van Buul JD, MacNamara KC, Voermans C, Nolte MA. Bone Marrow Harbors a Unique Population of Dendritic Cells with the Potential to Boost Neutrophil Formation upon Exposure to Fungal Antigen. Cells 2021; 11:55. [PMID: 35011617 PMCID: PMC8750392 DOI: 10.3390/cells11010055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 02/01/2023] Open
Abstract
Apart from controlling hematopoiesis, the bone marrow (BM) also serves as a secondary lymphoid organ, as it can induce naïve T cell priming by resident dendritic cells (DC). When analyzing DCs in murine BM, we uncovered that they are localized around sinusoids, can (cross)-present antigens, become activated upon intravenous LPS-injection, and for the most part belong to the cDC2 subtype which is associated with Th2/Th17 immunity. Gene-expression profiling revealed that BM-resident DCs are enriched for several c-type lectins, including Dectin-1, which can bind beta-glucans expressed on fungi and yeast. Indeed, DCs in BM were much more efficient in phagocytosis of both yeast-derived zymosan-particles and Aspergillus conidiae than their splenic counterparts, which was highly dependent on Dectin-1. DCs in human BM could also phagocytose zymosan, which was dependent on β1-integrins. Moreover, zymosan-stimulated BM-resident DCs enhanced the differentiation of hematopoietic stem and progenitor cells towards neutrophils, while also boosting the maintenance of these progenitors. Our findings signify an important role for BM DCs as translators between infection and hematopoiesis, particularly in anti-fungal immunity. The ability of BM-resident DCs to boost neutrophil formation is relevant from a clinical perspective and contributes to our understanding of the increased susceptibility for fungal infections following BM damage.
Collapse
Affiliation(s)
- Marieke Goedhart
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Edith Slot
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Maria F. Pascutti
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Sulima Geerman
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Timo Rademakers
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (T.R.); (S.H.); (J.D.v.B.)
| | - Benjamin Nota
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (T.R.); (S.H.); (J.D.v.B.)
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jaap D. van Buul
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (T.R.); (S.H.); (J.D.v.B.)
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Katherine C. MacNamara
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA;
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Martijn A. Nolte
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands;
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
11
|
Singh V, Jang H, Kim S, Ayash L, Alavi A, Ratanatharathorn V, Uberti JP, Deol A. G-CSF use post peripheral blood stem cell transplant is associated with faster neutrophil engraftment, shorter hospital stay and increased incidence of chronic GVHD. Leuk Lymphoma 2020; 62:446-453. [PMID: 33043743 DOI: 10.1080/10428194.2020.1827244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The use of G-CSF post allogeneic transplant has become a common practice to accelerate neutrophil engraftment. There is some controversy in its use. To further evaluate the effectiveness, we compared outcomes in patients who underwent PBSCT, either with or without the planned use of G-CSF post SCT. Among consecutive 162 patients from October 2012 to October 2014, 65 patients received G-CSF post-PBSCT, and 97 did not. More patients in G-CSF group received MAC (78% vs. 55%). Patients who received G-CSF had earlier neutrophil engraftment (median days 11 vs. 14) and shorter post-transplant hospital stay (median days 16 vs. 20, p = 0.001). G-CSF use was associated with a higher rate of extensive chronic GVHD (44.3% vs.61.5%, p = 0.027). G-CSF cost the equivalent of 0.25 hospital days but shortened the initial transplant admission by 4 days. Early cost-benefit may be later offset by the economic burden of chronic GVHD and associated complications.
Collapse
Affiliation(s)
- Vijendra Singh
- Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Hyejeong Jang
- Biostatistics Core, Karmanos Cancer Institute, Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Seongho Kim
- Biostatistics Core, Karmanos Cancer Institute, Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Lois Ayash
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Asif Alavi
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Voravit Ratanatharathorn
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Joseph P Uberti
- Department of Oncology, Co-Director, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| | - Abhinav Deol
- Department of Oncology, Blood and Marrow Stem Cell Transplant Program, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| |
Collapse
|
12
|
George G, Martin AS, Chhabra S, Eapen M. The Effect of Granulocyte Colony-Stimulating Factor Use on Hospital Length of Stay after Allogeneic Hematopoietic Cell Transplantation: A Retrospective Multicenter Cohort Study. Biol Blood Marrow Transplant 2020; 26:2359-2364. [PMID: 32818554 DOI: 10.1016/j.bbmt.2020.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 10/23/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) is administered after allogeneic hematopoietic cell transplantation (HCT) to aid neutrophil recovery. We compared the effect of empiric G-CSF administration on the duration of index inpatient hospitalization stay after HCT for patients aged ≥18 years with a hematologic malignancy. G-CSF was considered empiric if administered between day -3 and day +6 in relation to graft infusion. We studied 3562 HCTs (1487 HLA-matched sibling donor HCTs and 2075 HLA-matched unrelated donor HCTs) between 2007 and 2016. Three hundred and thirteen (21%) recipients of HLA-matched sibling donor HCT and 417 (20%) recipients of HLA-matched unrelated donor HCT received empiric G-CSF therapy. The effect of G-CSF therapy on the index hospitalization stay was examined in generalized linear models (GLMs) with adjustment for other patient, disease, and transplantation characteristics and acute graft-versus-host disease and infection post-transplantation. The duration of index hospitalization by treatment group did not differ for HLA-matched sibling donor HCT but was shorter with G-CSF for HLA-matched unrelated donor HCT (15 days versus 19 days; P < .001). Our GLMs confirmed shorter hospitalization with the use of G-CSF therapy for HLA-matched unrelated donor HCT (P = .01). G-CSF therapy was not associated with early survival for either donor type, and there was no benefit or disadvantage of giving G-CSF to promote neutrophil recovery.
Collapse
Affiliation(s)
- Gemlyn George
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Andrew St Martin
- Center for International Blood and Bone Marrow Transplant Registry, Department of Medicine, Milwaukee, Wisconsin
| | - Saurabh Chhabra
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Bone Marrow Transplant Registry, Department of Medicine, Milwaukee, Wisconsin
| | - Mary Eapen
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Bone Marrow Transplant Registry, Department of Medicine, Milwaukee, Wisconsin
| |
Collapse
|
13
|
Filgrastim following HLA-Identical Allogeneic Bone Marrow Transplantation: Long-Term Outcomes of a Randomized Trial. Biol Blood Marrow Transplant 2018; 24:2459-2465. [PMID: 30036571 DOI: 10.1016/j.bbmt.2018.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 07/15/2018] [Indexed: 11/23/2022]
Abstract
Human recombinant granulocyte colony stimulating factor reduces the duration of neutropenia following HLA-identical allogeneic bone marrow transplantation. However, its use remains controversial due to the risk of increasing the incidence of acute graft-versus-host disease (GVHD) and slower platelet recovery. To clarify these risks, we conducted a prospective randomized placebo-controlled trial of filgrastim 5 µg/kg/day i.v. from day 7 post-transplant until neutrophil recovery in 145 consecutive adults undergoing HLA-identical allogeneic bone marrow transplantation, with cyclosporine and methotrexate as GVHD prophylaxis. The primary endpoint was the incidence of acute GVHD; hematological recovery, nonrelapse mortality, and post-transplant complications were secondary endpoints. Filgrastim had no significant effect on the incidence of acute GVHD, platelet recovery, platelet transfusion requirements, chronic GVHD, or survival. Filgrastim accelerated granulocyte recovery significantly (with absolute neutrophil counts >.5 × 109/L achieved after a median of 16 days versus 23 days for placebo; P < .0001), and reduced both early nonrelapse mortality (2.9% versus 10.5%; P = .042) and the duration of i.v. antibiotic therapy (18 days versus 26 days; P = .001) and hospitalization (27 versus 34 days; P = .017). In conclusion, in this setting, filgrastim reduced significantly the duration of neutropenia, i.v. antibiotic therapy, hospitalization, and early nonrelapse mortality, without increasing the risk of acute and chronic GVHD or relapse, or delaying platelet recovery.
Collapse
|
14
|
Clemmons AB, Gandhi AS, Albrecht B, Jacobson S, Pantin J. Impact of fluoroquinolone prophylaxis on infectious-related outcomes after hematopoietic cell transplantation. J Oncol Pharm Pract 2017; 25:326-332. [PMID: 29059026 DOI: 10.1177/1078155217735153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Patients immediately post-hematopoietic cell transplantation are at high risk for bacteremia. Judicious prophylactic antimicrobial utilization must balance anticipated benefits (reduction infections) versus risk (bacterial resistance, Clostridium difficile) . OBJECTIVE To compare infectious outcomes (primary: incidence bacteremia; secondary: febrile neutropenia, C. difficile, susceptibility of bacteremia, time to discharge and 30-day mortality) between hematopoietic cell transplantation who received fluoroquinolone prophylaxis to those who did not. METHODS A local institutional review board-approved retrospective study was conducted on all hematopoietic cell transplantation patients ( n = 171) comparing those who received fluoroquinolone prophylaxis ( n = 105) to those who did not ( n = 66). Data included infectious outcomes and mortality for the first 30 days post-hematopoietic cell transplantation. Chi-squared was performed for categorical variables (GraphPad Software Inc., 2015). Secondary analysis compared outcomes within autologous and allogeneic sub-groups. RESULTS Bacteremia was significantly lower for the overall cohort receiving fluoroquinolone (median duration eight days) versus those without fluoroquinolone (15.2% vs. 31.8%; P < 0.01). No difference was seen in C. difficile infection ( P = 0.81) or 30-day mortality (2.9% vs. 4.5%; P = 0.67). In the autologous sub-group ( n = 115), bacteremia was significantly lower in the fluoroquinolone cohort (8.5% vs. 27.3%; P = 0.0069), while no differences were seen in C. difficile infection ( P = 1) or 30-day mortality ( P = 1). In the allogeneic sub-group ( n = 56), there was no difference between those with and without fluoroquinolone in bacteremia (29.4% vs. 40.9%; P = 0.4) or C. difficile ( P = 0.72); however, there was a trend toward improved 30-day mortality (2.9% vs. 9.1%; P = 0.55). CONCLUSIONS Fluoroquinolone prophylaxis reduces incidence of bacteremia in autologous hematopoietic cell transplantation without increasing C. difficile after hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Amber B Clemmons
- 1 College of Pharmacy, University of Georgia, Augusta, GA USA.,2 Georgia Cancer Center, Augusta, GA, USA.,3 Department of Pharmacy, Augusta University Medical Center, Augusta, GA USA
| | - Arpita S Gandhi
- 1 College of Pharmacy, University of Georgia, Augusta, GA USA.,3 Department of Pharmacy, Augusta University Medical Center, Augusta, GA USA
| | | | - Stephanie Jacobson
- 2 Georgia Cancer Center, Augusta, GA, USA.,4 Department of Medicine, Augusta University Medical Center, Augusta, GA USA
| | - Jeremy Pantin
- 2 Georgia Cancer Center, Augusta, GA, USA.,4 Department of Medicine, Augusta University Medical Center, Augusta, GA USA
| |
Collapse
|
15
|
Cutino-Moguel MT, Eades C, Rezvani K, Armstrong-James D. Immunotherapy for infectious diseases in haematological immunocompromise. Br J Haematol 2017; 177:348-356. [PMID: 28369798 DOI: 10.1111/bjh.14595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Opportunistic infections remain a major problem across a broad spectrum of immunocompromised haematological patient groups, with viruses, bacteria, fungi and protozoa all presenting significant challenges. Given the major difficulties in treating many of these infections with the currently available antimicrobial chemotherapeutic arsenal, and the rapid emergence of antimicrobial resistance amongst all of the microbial kingdoms, novel strategies that enable host control or elimination of infection are urgently required. Recently, major progress has been made in our understanding of host immunocompromise in the haematological patient. In addition, a wide range of novel immunomodulatory strategies for infectious diseases have been developed. Here we discuss the major and wide-ranging areas of progress that have been made for host-directed immunotherapies in the context of infectious diseases, with relevance to haematological immunocompromise.
Collapse
Affiliation(s)
| | - Chris Eades
- Department of Infectious Diseases, Imperial College Healthcare NHS Trust, London, UK
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Centre, Houston, TX, USA
| | - Darius Armstrong-James
- Fungal Pathogens Laboratory, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
16
|
O'Rafferty C, O'Brien M, Smyth E, Keane S, Robinson H, Lynam P, O'Marcaigh A, Smith OP. Administration of G-CSF from day +6 post-allogeneic hematopoietic stem cell transplantation in children and adolescents accelerates neutrophil engraftment but does not appear to have an impact on cost savings. Pediatr Transplant 2016; 20:432-7. [PMID: 26841203 DOI: 10.1111/petr.12670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2015] [Indexed: 11/28/2022]
Abstract
G-CSF post-allogeneic HSCT accelerates neutrophil engraftment, but evidence that it impacts on cost-related outcomes is lacking. We performed a retrospective child and adolescent single-center cohort study examining G-CSF administration from Day +6 of allogeneic HSCT vs. ad hoc G-CSF use where clinically indicated. Forty consecutive children and adolescents undergoing allogeneic HSCT were included. End-points were as follows: time to engraftment; incidence of acute and chronic GvHD; number of patients alive at Day +100; 180-day TRM; post-transplant days in hospital; and cost of antimicrobials, TPN, and G-CSF usage. Neutrophil engraftment occurred earlier in the group that received G-CSF from Day +6. There was no difference between groups in any of the other end-points with the following exception: the cost of GCSF was significantly higher in the D + 6 G-CSF group. However, median G-CSF cost in this group amounted to only €280. There was a trend towards reduced cost of antimicrobials in the D + 6 G-CSF group, although this did not reach significance (p = 0.13). The median cost per patient of antimicrobial agents between groups differed by €1116. This study demonstrated the administration of G-CSF on Day +6 in pediatric HSCT to be safe. A further study using a larger cohort of patients is warranted to ascertain its true clinico-economic value.
Collapse
Affiliation(s)
- Ciara O'Rafferty
- Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Mairead O'Brien
- Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Elaine Smyth
- Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Sinead Keane
- Department of Pharmacy, Our Lady's Children's Hospital, Dublin, Ireland
| | - Hillary Robinson
- Department of Dietetics, Our Lady's Children's Hospital, Dublin, Ireland
| | - Paul Lynam
- Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | - Aengus O'Marcaigh
- Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland.,Trinity College, Dublin, Ireland
| | - Owen P Smith
- Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland.,Trinity College, Dublin, Ireland
| |
Collapse
|
17
|
Smith TJ, Bohlke K, Lyman GH, Carson KR, Crawford J, Cross SJ, Goldberg JM, Khatcheressian JL, Leighl NB, Perkins CL, Somlo G, Wade JL, Wozniak AJ, Armitage JO. Recommendations for the Use of WBC Growth Factors: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol 2015; 33:3199-212. [PMID: 26169616 DOI: 10.1200/jco.2015.62.3488] [Citation(s) in RCA: 615] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To update the 2006 American Society of Clinical Oncology guideline on the use of hematopoietic colony-stimulating factors (CSFs). METHODS The American Society of Clinical Oncology convened an Update Committee and conducted a systematic review of randomized clinical trials, meta-analyses, and systematic reviews from October 2005 through September 2014. Guideline recommendations were based on the review of the evidence by the Update Committee. RESULTS Changes to previous recommendations include the addition of tbo-filgrastim and filgrastim-sndz, moderation of the recommendation regarding routine use of CSFs in older patients with diffuse aggressive lymphoma, and addition of recommendations against routine dose-dense chemotherapy in lymphoma and in favor of high-dose-intensity chemotherapy in urothelial cancer. The Update Committee did not address recommendations regarding use of CSFs in acute myeloid leukemia or myelodysplastic syndromes in adults. RECOMMENDATIONS Prophylactic use of CSFs to reduce the risk of febrile neutropenia is warranted when the risk of febrile neutropenia is approximately 20% or higher and no other equally effective and safe regimen that does not require CSFs is available. Primary prophylaxis is recommended for the prevention of febrile neutropenia in patients who are at high risk on the basis of age, medical history, disease characteristics, and myelotoxicity of the chemotherapy regimen. Dose-dense regimens that require CSFs should only be used within an appropriately designed clinical trial or if supported by convincing efficacy data. Current recommendations for the management of patients exposed to lethal doses of total-body radiotherapy, but not doses high enough to lead to certain death as a result of injury to other organs, include the prompt administration of CSFs.
Collapse
Affiliation(s)
- Thomas J Smith
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Kari Bohlke
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Gary H Lyman
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Kenneth R Carson
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Jeffrey Crawford
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Scott J Cross
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - John M Goldberg
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - James L Khatcheressian
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Natasha B Leighl
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Cheryl L Perkins
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - George Somlo
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - James L Wade
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - Antoinette J Wozniak
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | - James O Armitage
- Thomas J. Smith, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Kari Bohlke, American Society of Clinical Oncology, Alexandria; Scott J. Cross, Virginia Oncology Associates, Norfolk; James L. Khatcheressian, Virginia Cancer Institute, Richmond, VA; Gary H. Lyman, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA; Kenneth R. Carson, Washington University, St Louis, MO; Jeffrey Crawford, Duke Medicine, Durham, NC; John M. Goldberg, University of Miami Miller School of Medicine, Miami, FL; Natasha B. Leighl, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Cheryl L. Perkins, patient representative, Dallas, TX; George Somlo, City of Hope National Medical Center, Duarte, CA; James L. Wade, Cancer Care Specialists of Central Illinois, Decatur, IL; Antoinette J. Wozniak, Karmanos Cancer Institute, Detroit, MI; and James O. Armitage, University of Nebraska Medical Center, Omaha, NE
| | | |
Collapse
|
18
|
Ricci MJ, Medin JA, Foley RS. Advances in haplo-identical stem cell transplantation in adults with high-risk hematological malignancies. World J Stem Cells 2014; 6:380-390. [PMID: 25258660 PMCID: PMC4172667 DOI: 10.4252/wjsc.v6.i4.380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 04/24/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Abstract
Allogeneic bone marrow transplant is a life-saving procedure for adults and children that have high-risk or relapsed hematological malignancies. Incremental advances in the procedure, as well as expanded sources of donor hematopoietic cell grafts have significantly improved overall rates of success. Yet, the outcomes for patients for whom suitable donors cannot be found remain a significant limitation. These patients may benefit from a hematopoietic cell transplant wherein a relative donor is fully haplotype mismatched. Previously this procedure was limited by graft rejection, lethal graft-versus-host disease, and increased treatment-related toxicity. Recent approaches in haplo-identical transplantation have demonstrated significantly improved outcomes. Based on years of incremental pre-clinical research into this unique form of bone marrow transplant, a range of approaches have now been studied in patients in relatively large phase II trials that will be summarized in this review.
Collapse
|
19
|
Filippini P, Rutella S. Recent advances on cellular therapies and immune modulators for graft-versus-host disease. Expert Rev Clin Immunol 2014; 10:1357-74. [PMID: 25196777 DOI: 10.1586/1744666x.2014.955475] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The efficacy of allogeneic hematopoietic stem cell transplantation is counterbalanced by the occurrence of life-threatening immune-mediated complications, such as graft-versus-host disease (GVHD), a multistep disease which is reportedly fatal to approximately 15% of transplant recipients. It is now established that T-cell-dendritic cell interactions, T-cell activation, release of proinflammatory cytokines and T-cell trafficking partake in GVHD pathogenesis. This article will focus on the most recent strategies aimed at preventing/treating GVHD by manipulating components of the innate and adaptive immune response from both the donor and the host.
Collapse
Affiliation(s)
- Perla Filippini
- Department of Systems Medicine, IRCCS San Raffaele Pisana, Rome, Italy
| | | |
Collapse
|
20
|
Pichler H, Witt V, Winter E, Boztug H, Glogova E, Pötschger U, Matthes-Martin S, Fritsch G. No Impact of Total or Myeloid Cd34+ Cell Numbers on Neutrophil Engraftment and Transplantation-Related Mortality after Allogeneic Pediatric Bone Marrow Transplantation. Biol Blood Marrow Transplant 2014; 20:676-83. [DOI: 10.1016/j.bbmt.2014.01.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/26/2014] [Indexed: 01/30/2023]
|
21
|
Al-Kadhimi Z, Gul Z, Chen W, Smith D, Abidi M, Deol A, Ayash L, Lum L, Waller EK, Ratanatharathorn V, Uberti J. High incidence of severe acute graft-versus-host disease with tacrolimus and mycophenolate mofetil in a large cohort of related and unrelated allogeneic transplantation patients. Biol Blood Marrow Transplant 2014; 20:979-85. [PMID: 24709007 DOI: 10.1016/j.bbmt.2014.03.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/16/2014] [Indexed: 01/01/2023]
Abstract
Both acute and chronic graft-versus-host disease (GVHD) are major causes of morbidity and mortality in patients undergoing allogeneic hematopoietic stem cell transplantation (AHSCT). The optimal pharmacological regimen for GVHD prophylaxis is unclear, but combinations of a calcineurin inhibitor (cyclosporin or tacrolimus [Tac]) and an antimetabolite (methotrexate or mycophenolate mofetil [MMF]) are typically used. We retrospectively evaluated the clinical outcomes of 414 consecutive patients who underwent AHSCT from sibling (SD) or unrelated donors (UD) with Tac/MMF combination, between January 2005 and August 2010. The median follow-up was 60 months. Less than one third of the patients received a reduced-intensity chemoregimen. The incidence of grades III and IV acute GVHD was 22.3% and 36.5% in SD and UD groups, respectively (P = .0007). The incidence of chronic GVHD was 47.1% and 52.7% in the SD and UD groups, respectively. Nonrelapse mortality (NRM) at 60 months was 33.3% and 46.5% in the SD and UD groups, respectively (P = .0016). The incidence of relapse was 22.4% for UD and 28.8% for SD. Five-year overall survival was 43% and 34% in the SD and UD groups, respectively (P = .0183). GVHD was the leading cause of death for the entire cohort. Multivariable analysis showed that 8/8 HLA match, patient's age < 60, and low-risk disease were associated with better survival. The use of Tac/MMF for GVHD prophylaxis was associated with a relatively high incidence of severe acute GVHD and NRM in AHSCT from sibling and unrelated donors.
Collapse
Affiliation(s)
- Zaid Al-Kadhimi
- Department of Hematology and Medical Oncology, Emory University and Winship Cancer Center, Atlanta, Georgia.
| | - Zartash Gul
- Division of Hematology/BMT, Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Wei Chen
- Biostatistics Core, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| | - Daryn Smith
- Biostatistics Core, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| | - Muneer Abidi
- Blood and Marrow Program, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| | - Abhinav Deol
- Blood and Marrow Program, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| | - Lois Ayash
- Blood and Marrow Program, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| | - Lawrence Lum
- Blood and Marrow Program, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Emory University and Winship Cancer Center, Atlanta, Georgia
| | - Voravit Ratanatharathorn
- Blood and Marrow Program, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| | - Joseph Uberti
- Blood and Marrow Program, Department of Oncology, Wayne State University/Karmanos Cancer Center, Detroit, Michigan
| |
Collapse
|
22
|
Liu H, Chen Y, Xuan L, Wu X, Zhang Y, Fan Z, Huang F, Zhang X, Jiang Q, Sun J, Liu Q. Soluble human leukocyte antigen G molecule expression in allogeneic hematopoietic stem cell transplantation: good predictor of acute graft-versus-host disease. Acta Haematol 2013; 130:160-8. [PMID: 23711991 DOI: 10.1159/000350488] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 02/28/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND Graft-versus-host disease (GVHD) remains a main complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Human leukocyte antigen G (HLA-G) is a non-classical class I molecule exerting multiple immunoregulatory functions. The aim of this study was to explore the relationship between soluble HLA-G (sHLA-G) and GVHD after allo-HSCT. METHODS The sHLA-G levels were examined using enzyme-linked immunosorbent assay in patients with hematological malignancies (n = 106) before transplantation, on days +15 and +30 after transplantation, as well as healthy volunteers (n = 10). RESULTS The levels of sHLA-G5, sHLA-G6 and sHLA-G7 in patients on days +15 and +30 after transplantation were all significantly higher than those before transplantation (all p ≤ 0.001). The increased levels of sHLA-G5 on days +15 and +30 after transplantation were both significantly higher in patients with grade 0-I acute GVHD (aGVHD) compared to those with grade II-IV aGVHD (both p < 0.001). The increased levels of sHLA-G5 on days +15 and +30 after transplantation were both negatively correlated with the severity of aGVHD (both p < 0.001). CONCLUSION sHLA-G5 might be a predictor of the occurrence and severity of aGVHD, which may help to establish individual prophylaxis against aGVHD and improve the survival for patients after allo-HSCT.
Collapse
Affiliation(s)
- Hui Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Graft failure may contribute to increased morbidity and mortality after allogeneic hematopoietic SCT (allo-HSCT). Here, we present risk factors for graft failure in all first allo-HSCTs performed at our center from 1995 to mid-2010 (n=967). Graft failure was defined as >95% recipient cells any time after engraftment with no signs of relapse, or re-transplantation because of primary or secondary neutropenia (<0.5 × 10(9)/L) and/or thrombocytopenia (<30 × 10(9)/L). Fifty-four patients (5.6%) experienced graft failure. The majority were because of autologous reconstitution (n=43), and only a few patients underwent re-transplantation because of primary (n=6) or secondary (n=5) graft failures. In non-malignant disorders, graft failure had no effect on survival, whereas in malignant disease graft failure was associated with reduced 5-year survival (22 vs 53%, P<0.01). In multivariate analysis, ex vivo T-cell depletion (relative risk (RR) 8.82, P<0.001), HLA-mismatched grafts (RR 7.64, P<0.001), non-malignant disorders (RR 3.32, P<0.01) and reduced-intensity conditioning (RR 2.58, P<0.01) increased the risk for graft failure, whereas graft failures were prevented by total nucleated cell doses of ≥ 2.5 × 10(8)/kg (RR 0.36, P<0.01). In conclusion, graft failure was only associated with inferior survival in malignant disease. Non-malignant disorders, HLA match, conditioning intensity, immunosuppression regimen and cell dose all influenced graft failure risk.
Collapse
|
24
|
A novel conditioning regimen improves outcomes in β-thalassemia major patients using unrelated donor peripheral blood stem cell transplantation. Blood 2012; 120:3875-81. [DOI: 10.1182/blood-2012-03-417998] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Abstract
We used a novel NF-08-TM transplant protocol based on intravenous busulfan, cyclophosphamide, fludarabine, and thiotepa in 82 consecutive patients with β-thalassemia major (TM), including 52 with allogeneic peripheral blood stem cell transplantation (PBSCT) from unrelated donors (UDs) with well-matched human leukocyte antigens and 30 with hematopoietic stem cell transplantation (HSCT) from matched sibling donors (MSDs). The median age at transplantation was 6.0 years (range, 0.6-15.0 years), and the ratio of male-to-female patients was 56:26. The median follow-up time was 24 months (range, 12-39 months). The estimated 3-year overall survival and TM-free survival were 92.3% and 90.4% in the UD-PBSCT group and 90.0% and 83.3% in the MSD-HSCT group. The cumulative incidences of graft rejection and grades III-IV acute graft-versus-host disease were 1.9% and 9.6%, respectively, in the UD-PBSCT group and 6.9% and 3.6%, respectively, in the MSD-HSCT group. The cumulative incidence of transplant-related mortality was 7.7% in the UD-PBSCT group and 10.0% in the MSD-HSCT group. In conclusion, UD-PBSCTs using the well-tolerated NF-08-TM protocol show similar results to MSD-HSCTs and can be used to treat β-thalassemia patients in the absence of MSDs.
Collapse
|
25
|
Ringdén O, Labopin M, Gorin NC, Volin L, Torelli GF, Attal M, Jouet JP, Milpied N, Socié G, Cordonnier C, Michallet M, Atienza AI, Hermine O, Mohty M. Growth factor-associated graft-versus-host disease and mortality 10 years after allogeneic bone marrow transplantation. Br J Haematol 2012; 157:220-9. [PMID: 22299728 DOI: 10.1111/j.1365-2141.2012.09034.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 12/12/2011] [Indexed: 11/29/2022]
Abstract
This study analysed the effects of growth factor on outcome after haematopoietic stem-cell transplantation (HSCT) with >9 years follow-up. Of 1887 adult patients with acute leukaemia who received bone marrow from human leucocyte antigen (HLA)-identical siblings and were treated with myeloablative conditioning, 459 (24%) were treated with growth factor. Growth factor hastened engraftment of neutrophils (P < 0·0001), but reduced platelet counts (P = 0·0002). Graft-versus-host disease (GVHD)-free survival (no acute GVHD grade II-IV or chronic GVHD) at 10 years was 12 ± 2% (±SE) in the growth factor group, as opposed to 17 ± 2% in the controls [hazard ratio (HR) 0·81, P = 0·001]. Similar differences in GVHD-free survival were seen in patients with or without conditioning with total body irradiation (TBI). Non-relapse mortality (NRM) was higher in the growth factor group irrespective of whether or not TBI conditioning was included [HR = 1·48; 95% confidence interval (CI): 1·15-1·9; P = 0·002; HR = 1·59; 95% CI: 1·07-2·37; P = 0·02, respectively]. Both groups had similar probabilities of leukaemic relapse (HR = 0·96; 95% CI: 0·78-1·18; P = 0·71). Leukaemia-free survival (LFS) at 10 years was 35 ± 2% in those receiving growth factor prophylaxis, as opposed to 44 ± 1% in the controls (HR = 0·70; 95% CI: 0·60-0·82; P = 0·00001). Prophylaxis with growth factor increases the risk of GVHD, does not affect relapse, increases NRM and reduces LFS > 10 years after HSCT, regardless of conditioning with TBI.
Collapse
Affiliation(s)
- Olle Ringdén
- Centre for Allogeneic Stem Cell Transplantation and Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Increased costs after allogeneic haematopoietic SCT are associated with major complications and re-transplantation. Bone Marrow Transplant 2011; 47:706-15. [DOI: 10.1038/bmt.2011.162] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Remberger M, Ackefors M, Berglund S, Blennow O, Dahllöf G, Dlugosz A, Garming-Legert K, Gertow J, Gustafsson B, Hassan M, Hassan Z, Hauzenberger D, Hägglund H, Karlsson H, Klingspor L, Kumlien G, Le Blanc K, Ljungman P, Machaczka M, Malmberg KJ, Marschall HU, Mattsson J, Olsson R, Omazic B, Sairafi D, Schaffer M, Svahn BM, Svenberg P, Swartling L, Szakos A, Uhlin M, Uzunel M, Watz E, Wernerson A, Wikman A, Wikström AC, Winiarski J, Ringdén O. Improved survival after allogeneic hematopoietic stem cell transplantation in recent years. A single-center study. Biol Blood Marrow Transplant 2011; 17:1688-97. [PMID: 21620989 DOI: 10.1016/j.bbmt.2011.05.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 05/03/2011] [Indexed: 10/18/2022]
Abstract
We analyzed the outcome of allogeneic hematopoietic stem cell transplantation (HSCT) over the past 2 decades. Between 1992 and 2009, 953 patients were treated with HSCT, mainly for a hematologic malignancy. They were divided according to 4 different time periods of treatment: 1992 to 1995, 1996 to 2000, 2001 to 2005, and 2006 to 2009. Over the years, many factors have changed considerably regarding patient age, diagnosis, disease stage, type of donor, stem cell source, genomic HLA typing, cell dose, type of conditioning, treatment of infections, use of granulocyte-colony stimulating factor (G-CSF), use of mesenchymal stem cells, use of cytotoxic T cells, and home care. When we compared the last period (2006-2009) with earlier periods, we found slower neutrophil engraftment, a higher incidence of acute graft-versus-host disease (aGVHD) of grades II-IV, and less chronic GVHD (cGHVD). The incidence of relapse was unchanged over the 4 periods (22%-25%). Overall survival (OS) and transplant-related mortality (TRM) improved significantly in the more recent periods, with the best results during the last period (2006-2009) and a 100-day TRM of 5.5%. This improvement was also apparent in a multivariate analysis. When correcting for differences between the 4 groups, the hazard ratio for mortality in the last period was 0.59 (95% confidence interval [CI]: 0.44-0.79; P < .001) and for TRM it was 0.63 (CI: 0.43-0.92; P = .02). This study shows that the combined efforts to improve outcome after HSCT have been very effective. Even though we now treat older patients with more advanced disease and use more alternative HLA nonidentical donors, OS and TRM have improved. The problem of relapse still has to be remedied. Thus, several different developments together have resulted in significantly lower TRM and improved survival after HSCT over the last few years.
Collapse
Affiliation(s)
- Mats Remberger
- Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sun ZM, Liu HL, Geng LQ, Wang XB, Yao W, Liu X, Ding KY, Han YS, Yang HZ, Tang BL, Tong J, Zhu WB, Wang ZY. HLA-matched sibling transplantation with G-CSF mobilized PBSCs and BM decreases GVHD in adult patients with severe aplastic anemia. J Hematol Oncol 2010; 3:51. [PMID: 21194460 PMCID: PMC3023734 DOI: 10.1186/1756-8722-3-51] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 12/31/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an effective treatment for severe aplastic anemia (SAA). However, graft failure and graft-versus-host disease (GVHD) are major causes of the early morbidity in Allo-HSCT. METHODS To reduce graft failure and GVHD, we treated fifteen patients with SAA using high- dose of HSCT with both G-CSF mobilized PB and BMSCs from HLA-identical siblings to treat patients with SAA. RESULTS All patients had successful bone marrow engraftment. Only one patient had late rejection. Median time to ANC greater than 0.5 × 10(9)/L and platelet counts greater than 20 × 10(9)/L was 12 and 16.5 days, respectively. No acute GVHD was observed. The incidence of chronic GVHD was 6.67%. The total three-year probability of disease-free survival was 79.8%. CONCLUSION HSCT with both G-CSF mobilized PB and BMSCs is a promising approach for heavily transfused and/or allo-immunized patients with SAA.
Collapse
|
29
|
Norlin AC, Remberger M. A comparison of Campath and Thymoglobulin as part of the conditioning before allogeneic hematopoietic stem cell transplantation. Eur J Haematol 2010; 86:57-66. [DOI: 10.1111/j.1600-0609.2010.01537.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Granulocyte colony-stimulating factor induced acute and chronic graft-versus-host disease. Transplantation 2010; 90:1022-9. [PMID: 20814354 DOI: 10.1097/tp.0b013e3181f585c7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND A recent experimental study in mice shed new light on the controversy as to whether granulocyte colony-stimulating factor (G-CSF) increases graft-versus-host disease (GVHD). Total body irradiation and bone marrow were found to be prerequisites for acute GVHD. This study encouraged us to perform a retrospective clinical study. METHODS We compared 260 patients given G-CSF prophylaxis after allogeneic hematopoietic stem-cell transplantation with 205 controls transplanted between 1993 and 2003. RESULTS G-CSF hastened the engraftment of neutrophils, but that of platelets was delayed (P<0.0001). The proportion of acute GVHD of grades II to IV was 29% in the G-CSF group and 19% in the controls (P<0.01) and that of chronic GVHD was 54% and 43%, respectively (P=0.019). G-CSF increased acute and chronic GVHD in patients preferentially conditioned with chemotherapy. Unexpectedly, it exacerbated acute GVHD in recipients of peripheral blood stem cells and enhanced chronic GVHD in bone marrow recipients. A multivariable analysis showed that acute GVHD (hazards ratio=1.52, P=0.03) and chronic GVHD (hazards ratio=1.51, P=0.004) were associated with G-CSF. There was no significant difference between study groups regarding nonrelapse mortality, relapse, or survival. CONCLUSION G-CSF increased acute and chronic GVHD in patients treated with chemotherapy but did not affect relapse or survival.
Collapse
|
31
|
Rapid engraftment at a cost? Transplantation 2010; 90:949-50. [PMID: 20802399 DOI: 10.1097/tp.0b013e3181f585ee] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Manga K, Serban G, Schwartz J, Slotky R, Patel N, Fan J, Bai X, Chari A, Savage D, Suciu-Foca N, Colovai AI. Increased adenosine triphosphate production by peripheral blood CD4+ cells in patients with hematologic malignancies treated with stem cell mobilization agents. Hum Immunol 2010; 71:652-8. [PMID: 20381567 DOI: 10.1016/j.humimm.2010.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 03/16/2010] [Accepted: 03/29/2010] [Indexed: 10/19/2022]
Abstract
Hematopoietic stem cell (HSC) transplantation is an important therapeutic option for patients with hematologic malignancies. To explore the immunomodulatory effects of HSC mobilization agents, we studied the function and phenotype of CD4(+) T cells from 16 adult patients with hematologic malignancies undergoing HSC mobilization treatment for autologous transplantation. Immune cell function was determined using the Immuknow (Cylex) assay by measuring the amount of adenosine triphosphate (ATP) produced by CD4(+) cells from whole blood. ATP activity measured in G-CSF-treated patients was significantly higher than that measured in healthy individuals or "nonmobilized" patients. In patients treated with G-CSF, CD4(+) T cells were predominantly CD25(low)FOXP3(low), consistent with an activated phenotype. However, T-cell depletion did not abrogate ATP production in blood samples from G-CSF-treated patients, indicating that CD4(+) myeloid cells contributed to the increased ATP levels observed in these patients. There was a significant correlation between ATP activity and patient survival, suggesting that efficient activation of CD4(+) cells during mobilization treatment predicts a low risk of disease relapse. Monitoring immune cell reactivity using the Immuknow assay may assist in the clinical management of patients with hematologic malignancies and optimization of HSC mobilization protocols.
Collapse
Affiliation(s)
- Kiran Manga
- Department of Pathology, Columbia University Medical Center, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
During the past three decades, allogeneic stem cell transplantation (ASCT) has developed from being an experimental therapy in patients with endstage leukemia into a well-established therapy in patients with a range of disorders of the immunohematopoietic system. Graft-versus-host disease (GVHD), acute or chronic, attacking host tissue is a major threat. However, donor immunocompetent T cells have a potent graft-versus-leukemia effect. A combination of calcineurin inhibitors and methotrexate is the standard therapy to prevent GVHD. Modulation of the immunosuppressive regimen may induce mild acute and mild chronic GVHD, reduce the risk of relapse, and improve long-term survival. Natural killer cells also play a role in this context. Killer cell immunoglobulin-like receptor incompatibility between recipient and donor may reduce the risk of relapse in patients with myeloid leukemia. Relapse of leukemia is a major cause of death after ASCT. Minimal residual disease and recipient leukemia lineage-specific chimerism are sensitive techniques for early detection of leukemic relapse. Donor lymphocyte infusions can enhance the antitumor effect, especially for patients with molecular relapse. The allogeneic graft-versus-cancer effect has been demonstrated in patients with metastatic breast, renal, colorectal, ovarian, prostatic, and pancreatic carcinoma. Mesenchymal stem cells have immunomodulatory properties and may be used for immunomodulation of GVHD and tissue repair. All things considered, the future looks promising for ASCT.
Collapse
Affiliation(s)
- Olle Ringdén
- Division of Clinical Immunology, Karolinska Institutet, Karolinska University, Hospital, Huddinge, SE-141 86 Stockholm, Sweden
| |
Collapse
|
34
|
Waxman IM, Militano O, Baldinger L, Roman E, Qualter E, Morris E, Garvin J, Bradley MB, Bhatia M, Satwani P, George D, Del Toro G, Hawks R, Wolownik K, Foley S, Cheung YK, Schwartz J, van de Ven C, Baxter-Lowe LA, Cairo MS. Sequential administration of sargramostim and filgrastim in pediatric allogeneic stem cell transplantation recipients undergoing myeloablative conditioning. Pediatr Transplant 2009; 13:464-74. [PMID: 18785912 DOI: 10.1111/j.1399-3046.2008.01000.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
G-CSF and GM-CSF both hasten myeloid engraftment post-MA-alloSCT; however, GM-CSF is earlier acting and less expensive. The objective was to evaluate efficacy/safety of sequential administration of GM-CSF followed by G-CSF in children post-MA-alloSCT. From January 2001 to June 2005, 31 children received 32 MA-alloSCT: mean age 6.65 yr; MRD BM or PBSC vs. related or unrelated UCB 11:21; malignant vs. non-malignant disorders 22:10. GM-CSF (250 microg/m(2) IV QD) began on day of stem cell infusion. GM-CSF was switched to G-CSF (10 microg/kg IV QD) when WBC >or= 300/mm(3) x 2 days. G-CSF continued until ANC >or= 2500/mm(3) x 2 days, then tapered to maintain ANC >or= 1000/mm(3). Median time to myeloid engraftment (ANC >or= 500/mm(3) x 3 days) was 17 days [13 days vs. 24 days, MRD BM/PBSC vs. UCB (p < 0.0001)], occurring at a median time of two days after switch to G-CSF. Clinically relevant adverse events were bone pain (n = 8) and large pleural effusion (n = 1). It was estimated that sequential GM-CSF/G-CSF was cost-effective compared with G-CSF alone [cost-savings of $1311/patient ($41,952/study), 2007 Red Book Average Wholesale Price]. In summary, it was demonstrated that sequential administration of GM-CSF/G-CSF post-MA-alloSCT was safe, cost-effective and resulted in prompt myeloid engraftment.
Collapse
Affiliation(s)
- Ian M Waxman
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
After hematopoietic SCT (HSCT), G-CSF is commonly used to enhance stem cell engraftment to minimize the morbidity and mortality associated with prolonged neutropenia. However, there is no consensus on the optimal use of G-CSF after high-dose chemotherapy followed by HSCT. This review was performed to evaluate the evidence regarding the use of G-CSF after autologous and allogeneic HSCT. Studies investigating the use of G-CSF in comparison to control (observation or placebo), early vs delayed initiation of G-CSF, and other approaches driven by patient-specific parameters to identify optimal use of G-CSF have been reviewed. Various outcomes such as neutrophil and platelet engraftment, post-transplant length of hospital stay, post-transplant complications such as infection and GVHD, and survival have been assessed. Finally, we provide the level of evidence for each of the outcomes analyzed while evaluating strategies for using G-CSF in patients undergoing autologous or allogeneic HSCT.
Collapse
|
36
|
Hauzenberger D, Schaffer M, Ringdén O, Hassan Z, Omazic B, Mattsson J, Wikström AC, Remberger M. Outcome of haematopoietic stem cell transplantation in patients transplanted with matched unrelated donorsvsallele-mismatched donors: a single centre study. ACTA ACUST UNITED AC 2008; 72:549-58. [DOI: 10.1111/j.1399-0039.2008.01148.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
37
|
Ernst P, Bacigalupo A, Ringdén O, Ruutu T, Kolb HJ, Lawrinson S, Skacel T. A Phase 3, Randomized, Placebo-controlled Trial of Filgrastim in Patients with Haematological Malignancies Undergoing Matched-related Allogeneic Bone Marrow Transplantation. ACTA ACUST UNITED AC 2008; 1:89-96. [PMID: 19639030 PMCID: PMC2710993 DOI: 10.1111/j.1753-5174.2008.00013.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction Recombinant granulocyte colony-stimulating factor (G-CSF) may aid engraftment post high-dose chemo-/radiotherapy in patients with haematological malignancies undergoing allogeneic bone marrow transplantation (BMT); however, the effects of G-CSF on graft-versus-host disease (GvHD), relapse, and survival are not well defined. Methods In this double-blind, randomized, placebo-controlled, multicentre, phase 3 study, the effects of the G-CSF Filgrastim on neutrophil and platelet recovery, and on clinical outcomes were evaluated. Patients (12–55 years) receiving an allogeneic BMT for a haematological malignancy were randomized to receive Filgrastim 5 µg/kg or placebo. Study treatment was continued until patients achieved an absolute neutrophil count (ANC) ≥0.5 × 109/L, or until day 42. Results Fifty-one patients (Filgrastim, N = 25; placebo, N = 26) were evaluable. Patients treated with Filgrastim had significantly faster engraftment with ANC ≥0.5 × 109/L being achieved after a median (range) of 15.0 (1.0–22.0) days vs. 19.0 (15.0–28.0) days for placebo (P< 0.0001). The incidence of GvHD was comparable for both groups. During the limited follow-up (2 years), Filgrastim had no adverse effect on mortality and possibly reduced the rate of relapse.
Collapse
Affiliation(s)
- Peter Ernst
- Division of Haematology, Haukeland University HospitalBergen, Norway
| | | | | | - Tapani Ruutu
- Department of Medicine, Helsinki University Central HospitalHelsinki, Finland
| | | | | | - Tomas Skacel
- Medical Affairs, Amgen (Europe) GmbHZug, Switzerland
- Department of Internal Medicine—Hemato-onkology, University HospitalBrno, Czech Republic
| |
Collapse
|
38
|
Unrelated cord blood and mismatched unrelated volunteer donor transplants, two alternatives in patients who lack an HLA-identical donor. Bone Marrow Transplant 2008; 42:643-8. [DOI: 10.1038/bmt.2008.239] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Remberger M, Mattsson J, Hausenberger D, Schaffer M, Svahn BM, Ringdén O. Genomic tissue typing and optimal antithymocyte globuline dose using unrelated donors results in similar survival and relapse as HLA-identical siblings in haematopoietic stem-cell transplantation for leukaemia. Eur J Haematol 2008; 80:419-28. [DOI: 10.1111/j.1600-0609.2008.01047.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
40
|
A prospective randomized study using N-acetyl-L-cysteine for early liver toxicity after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2008; 41:785-90. [DOI: 10.1038/sj.bmt.1705969] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
41
|
Allogeneic stem cell transplantation: low immunoglobulin levels associated with decreased survival. Bone Marrow Transplant 2007; 41:267-73. [PMID: 17994123 DOI: 10.1038/sj.bmt.1705892] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The aim of this study was to evaluate the effects and kinetics of IgG levels after allogeneic stem cell transplantation (SCT). This study retrospectively examines 179 consecutive patients undergoing SCT between 1995 and 2002. Diagnoses included acute and chronic leukemia (n=136), solid tumors (n=11), other malignancies (n=16) and non-malignant diseases (n=16). Standard myeloablative conditioning was given to 146 patients, and 33 patients received reduced intensity conditioning. Serum samples for measurement of IgG levels were collected 3, 6 and 12 months after SCT, and then yearly. IgG levels increased after SCT throughout the study period. Factors that were associated with low IgG levels after SCT were acute graft-versus-host disease (GVHD), patient age < or =30 years, female donor-to-male recipient, not receiving anti-thymocyte globulin and type of GVHD prophylaxis. Compared to patients with moderately low or normal levels as measured twice during the first year after transplantation, patients with low IgG levels (<4 g/l) showed a decreased survival rate (54 vs 71%, P=0.04) and an increased incidence of transplant-related mortality (27 vs 9%, P<0.01). IgG levels generally increase after SCT. Persistent low levels of IgG are a risk factor for death after SCT.
Collapse
|
42
|
Remberger M, Mattsson J, Hassan Z, Karlsson N, LeBlanc K, Omazic B, Okas M, Sairafi D, Ringdén O. Risk factors for acute graft-versus-host disease grades II-IV after reduced intensity conditioning allogeneic stem cell transplantation with unrelated donors: a single centre study. Bone Marrow Transplant 2007; 41:399-405. [PMID: 17982493 DOI: 10.1038/sj.bmt.1705913] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We analysed factors associated with moderate to severe acute GVHD in 111 patients treated with fludarabin-based reduced intensity conditioning (RIC) and allogeneic haematopoietic stem cell transplantation (HSCT). Most patients had a haematological malignancy. Donors were 97 HLA-A, -B and -DRbeta1 identical unrelated and 14 HLA-A, -B or -DRbeta1 allele mismatched unrelated donors. In the univariate analysis, we found ten factors associated with acute GVHD. These were diagnosis (P=0.06), GVHD prophylaxis with combinations other than CsA+MTX (P=0.006), graft nucleated (P<0.001) and CD34 (P<0.001) cell-dose, bidirectional ABO mismatch (P=0.001), conditioning (P=0.002), hospital vs home-care (P=0.06), ATG dose (P<0.001), donor herpes virus serology (P=0.07) and an immunized female donor to male recipient (P=0.05). In the multivariate analysis, three factors remained significant: a high CD34 cell dose (P<0.001), low dose (4 mg/kg) ATG (P<0.001), and an immunized female donor to male recipient (P<0.01). Patients receiving a CD34 cell dose > or =17.0 x 10(6) per kg had a higher incidence of GVHD, 53.7%, compared to 22.3% in patients receiving a lower dose (P=0.002). In patients without any of these risk factors (n=70), the incidence of acute GVHD was 14.1%, while it was 38.0 and 85.0% in patients with one (n=29) or two (n=10) risk factors (P<0.001). We concluded that risk factors for acute GVHD using RIC are similar as using myeloablative conditioning.
Collapse
Affiliation(s)
- M Remberger
- Department of Clinical Immunology and Centre for Allogeneic Stem Cell Transplantation, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Littlewood TJ, Collins GP. Granulocyte and erythropoietic stimulating proteins after high-dose chemotherapy for myeloma. Bone Marrow Transplant 2007; 40:1147-55. [PMID: 17846601 DOI: 10.1038/sj.bmt.1705845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
High-dose chemotherapy is an established treatment for patients with myeloma. In randomized trials it has been shown to prolong disease-free survival by around 1 year compared to patients receiving chemotherapy alone. Physically and psychologically high-dose therapy takes its toll on the patient who may be in hospital for around 3 weeks and take some weeks or months to convalesce after discharge. Granulocyte colony stimulating factors and erythropoietic stimulating agents will speed neutrophil and red cell recovery, respectively, when used at an appropriate time after the high-dose chemotherapy. The clinical value of these laboratory findings is uncertain and the role of these agents after high-dose chemotherapy remains a subject for debate.
Collapse
Affiliation(s)
- T J Littlewood
- Department of Haematology, John Radcliffe Hospital, Oxford, UK.
| | | |
Collapse
|
44
|
Abstract
In recent years, investigators have unraveled a previously unrecognized role for granulocyte colony-stimulating factor (G-CSF) in the regulation of T-cell and dendritic cell functions. The experimental evidence in favor of G-CSF-mediated immune regulation includes the ability to skew T-cell cytokine secretion to T-helper type 2 responses, and to promote regulatory T-cell and tolerogenic dendritic cell differentiation. Accordingly, beneficial effects of G-CSF have been detected in animal models of immune-mediated diseases, including posttransplantation graft-versus-host disease, experimental autoimmune encephalomyelitis, lupus nephritis, inflammatory bowel disease, and diabetes. The growing body of evidence supporting a novel role for G-CSF in the induction of T-cell tolerance is reviewed herein.
Collapse
Affiliation(s)
- Sergio Rutella
- Department of Hematology, Laboratory of Immunology, Catholic University Medical School, Largo A. Gemelli 8, 00168 Rome, Italy.
| |
Collapse
|
45
|
Seggewiss R, Einsele H. Hematopoietic Growth Factors Including Keratinocyte Growth Factor in Allogeneic and Autologous Stem Cell Transplantation. Semin Hematol 2007; 44:203-11. [PMID: 17631184 DOI: 10.1053/j.seminhematol.2007.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The aim of hematopoietic stem cell transplantation (HSCT) is to cure patients of malignancies, autoimmune diseases, and immunodeficiency disorders by redirecting the immune system: the often described graft-versus-leukemia (GVL) or graft-versus-tumor (GVT) effects. Unfortunately, fulfillment of this goal is often hampered by relapse of the underlying disease, graft-versus-host disease (GVHD), or severe opportunistic infections, which account for the majority of post-transplantation deaths. Moreover, studies of long-term survivors of transplantation indicate an accelerated immune aging due to the transplantation procedure itself, preceding chemo- or radiotherapy, and acute and chronic GVHD. Significant advances have been made towards overcoming these obstacles by enhancing immune reconstitution with hematopoietic growth factors (HGFs) such as granulocyte colony-stimulating factor (G-CSF) or erythropoietin (EPO) or through the application of cytokines. In addition, there are approaches to promote the thymic-dependent development of naive T cells, which are prepared for the interaction with a multitude of pathogens. Examples are the application of keratinocyte growth factor (KGF), neuroendocrine hormones such as growth hormone or prolactin, sex hormone ablation, or the invention of a three-dimensional artificial thymus based on a cytomatrix. Might these measures result in a higher rate of healthy and fully recovered patients? Here we review progress in each of these areas.
Collapse
Affiliation(s)
- Ruth Seggewiss
- Department of Internal Medicine II, Julius-Maximilians-University, Würzburg, Germany
| | | |
Collapse
|
46
|
Remberger M, Watz E, Ringdén O, Mattsson J, Shanwell A, Wikman A. Major ABO blood group mismatch increases the risk for graft failure after unrelated donor hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2007; 13:675-82. [PMID: 17531777 DOI: 10.1016/j.bbmt.2007.01.084] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Accepted: 01/30/2007] [Indexed: 01/28/2023]
Abstract
Two hundred twenty-four patients with leukemia transplanted with an unrelated donor between 1991 and 2003 at the Karolinska University Hospital were analyzed according to association between graft failure and ABO, RhD, MNSs, and Kidd blood group antigen compatibility. Median age was 29 years (range: 0-55). Conditioning consisted of total-body irradiation or busulfan-based myeloablative conditioning. A bone marrow graft was given to 152 patients, and 72 patients received peripheral blood stem cells. Most patients received graft-versus-host disease prophylaxis with cyclosporine and MTX. Graft failure (GF) was seen in 6 (2.7%) patients. In the multivariate analysis major ABO mismatch (odds ratio [OR] 14.9, 95% confidence interval [CI] 2.01-110, P = .008) and HLA-allele mismatch (6.42, 1.19-34.8, P = .03) was significantly associated to GF. In patients with and without major ABO mismatch the incidence of GF was 7.5% and 0.6% (P = .02), respectively. Using an ABO major mismatched graft increases the risk for GF after unrelated donor hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Mats Remberger
- Department of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
47
|
Uzunel M, Remberger M, Sairafi D, Hassan Z, Mattsson J, Omazic B, Barkholt L, Ringdén O. Unrelated versus related allogeneic stem cell transplantation after reduced intensity conditioning. Transplantation 2006; 82:913-9. [PMID: 17038906 DOI: 10.1097/01.tp.0000233865.20232.51] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The use of reduced intensity conditioning (RIC) regimens in allogeneic hematopoietic stem cell transplantation (HSCT) has increased over the past five years. PATIENTS In this study, involving 137 patients, we compared the outcome after RIC in patients receiving grafts from matched unrelated donors (MUD; n=74) and sibling donors (n=63). The MUD and sibling groups were comparable regarding diagnosis, including solid tumors and hematological malignancies, and conditioning regimens. RESULTS Engraftment was successful in most patients (88%), with no significant difference between MUD and sibling transplants. Cytomegalovirus (CMV) infection was more common in the MUD group (65%) than in the sibling group (46%) (P=0.04). No difference in severe acute graft-versus-host disease (GVHD) was found between the groups. However, the incidence of chronic GVHD was higher after sibling transplants. This was probably due to higher donor age in this group, since this was the only significant risk factor for chronic GVHD in multivariate analysis. The incidence of transplant related mortality (TRM) was significantly higher after MUD transplantation (40%) than after sibling transplantation (16%) (P<0.01). Because relapse/disease progression was more common after sibling transplantation, there was no significant difference in overall survival between the two groups. CONCLUSION Using unrelated donors after RIC is feasible, but it resulted in more CMV infection and increased transplant-related mortality. Survival was comparable to that of sibling transplants.
Collapse
Affiliation(s)
- Mehmet Uzunel
- Department of Clinical Immunology, Karolinska Institute at Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Dekker A, Bulley S, Beyene J, Dupuis LL, Doyle JJ, Sung L. Meta-Analysis of Randomized Controlled Trials of Prophylactic Granulocyte Colony-Stimulating Factor and Granulocyte-Macrophage Colony-Stimulating Factor After Autologous and Allogeneic Stem Cell Transplantation. J Clin Oncol 2006; 24:5207-15. [PMID: 17114653 DOI: 10.1200/jco.2006.06.1663] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The primary objective of our meta-analysis was to determine whether prophylactic hematopoietic colony-stimulating factors (CSFs) after hematopoietic autologous and allogeneic stem-cell transplantation (SCT) reduced documented infections. Our secondary objectives were to determine whether prophylactic CSFs affected other outcomes including parenteral antibiotic therapy duration, infection-related mortality, graft-versus-host disease (GVHD), or treatment-related mortality. Methods We included studies if there was random assignment between CSFs and placebo/no therapy and CSFs were given after SCT and before recovery of neutrophils. From 3,778 reviewed study articles, 34 were included based on predefined inclusion criteria. All analyses were conducted using a random effects model. Results CSFs reduced the risk of documented infections (relative risk [RR] 0.87; 95% CI, 0.76 to 1.00; P = .05) and duration of parenteral antibiotics (weighted mean difference, −1.39 days, 95% CI, −2.56 to −0.22; P = .02) but did not reduce infection-related mortality (RR, 0.76; 95% CI, 0.41 to 1.44; P = .4). CSFs did not increase grade 2 to 4 acute GVHD (RR, 1.03; 95% CI, 0.81 to 1.31; P = .8) or treatment-related mortality (RR, 1.00; 95% CI, 0.78 to 1.29; P = .98). Conclusion CSFs were associated with a small reduction in the risk of documented infections but did not affect infection or treatment-related mortality.
Collapse
Affiliation(s)
- Allison Dekker
- Department of Public Health Sciences, Health Policy Management and Evaluation, and Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
49
|
Svahn BM, Alvin O, Ringdén O, Gardulf A, Remberger M. Costs of allogeneic hematopoietic stem cell transplantation. Transplantation 2006; 82:147-53. [PMID: 16858272 DOI: 10.1097/01.tp.0000226171.43943.d3] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND This study aims to determine the total costs after allogeneic hematopoietic stem cell transplantation (ASCT) and factors associated with increases or decreases in costs. METHODS We collected all in- and outpatient costs during 5 years in 93 patients who had undergone ASCT in 1998 and 1999 at our unit. The inpatient costs included all those related to a patient from the first day of admission until discharge and then all costs of readmission in the Stockholm area. RESULTS The total median cost of five posttransplant years was 139,414 (52,095-345,640) euros (euros) or 167,296 US dollars (the rate of 1 euro is approximately 1.2 US dollars). The costs were highest during the first year-median inpatient and outpatient costs 100,650 euros and 13,066 euros, respectively. The total costs during the first year were higher in patients with acute graft-versus-host disease grades III-IV (relative hazards [RH] 1.35, P = 0.003), bacteremia (RH 1.33, P = 0.005), veno-occlusive disease of the liver (RH 1.32, P = 0.005), prophylaxis with granulocyte colony-stimulating factor (G-CSF; RH 1.31, P = 0.01), acute leukemia (RH 1.32, P = 0.008), and treatment in hospital instead of at home (RH 1.20, P < 0.07). During the early transplant period, a second transplantation (RH 1.28, P = 0.014) and hemorrhagic cystitis (HC; RH 1.24, P = 0.03) were also associated with higher costs. The total 5-year cost declined with longer survival rates (r = 0.4028, P < 0.001) and reduced intensity conditioning (RH 0.79, P=0.024). CONCLUSION Higher costs of ASCT were associated with retransplantation, acute leukemia, G-CSF prophylaxis, hospital care, myeloablative conditioning, and major transplant-related complications.
Collapse
Affiliation(s)
- Britt-Marie Svahn
- Center for Allogeneic Stem Cell Transplantation, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
50
|
Ringdén O, Le Blanc K. Allogeneic hematopoietic stem cell transplantation: state of the art and new perspectives. APMIS 2006; 113:813-30. [PMID: 16480452 DOI: 10.1111/j.1600-0463.2005.apm_336.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (ASCT) is a well-established therapy for leukemias and other immunohematopoietic disorders. In more recent years, bone marrow as stem cell source has been replaced by peripheral blood stem cells, which results in faster engraftment. Cord blood grafts are increasingly used. Conditioning prior to transplant may be myeloablative or nonmyeloablative. The latter is used preferentially in patients with high age or organ impairment. Isolation in the hospital during posttransplant pancytopenia has been challenged by promising results using home care. PCR diagnosis and new antifungal and antiviral treatment have reduced morbidity and mortality. The major threat to a successful outcome after ASCT is leukemic relapse. PCR techniques to detect recipient cells in the leukemic cell lineage or minimal residual disease enable early detection of leukemic cells. Donor lymphocyte infusions have an antileukemic effect. ASCT has shown an antitumor effect in metastatic cancers from breast, kidney, colon, ovaries, prostate and pancreas. Mesenchymal stem cells may be derived from bone marrow and have the capacity to differentiate into several mesenchymal tissues, such as bone, cartilage and fat. They seem to escape the immune system and have immunomodulatory effects in vitro and in vivo. To conclude, ASCT is a potent immunotherapy.
Collapse
Affiliation(s)
- Olle Ringdén
- Karolinska Institutet, Division of Clinical Immunology and Center for Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| | | |
Collapse
|