1
|
Oyama T, Fujiwara SI, Tominaga R, Yokoyama D, Noguchi A, Furuki S, Koyama S, Murahashi R, Nakashima H, Hyodo K, Ikeda T, Kawaguchi SI, Toda Y, Nagayama T, Umino K, Minakata D, Morita K, Ashizawa M, Yamamoto C, Hatano K, Sato K, Otsuki I, Ohmine K, Kanda Y. Effects of CD34 + cell dose on engraftment and long-term outcomes after allogeneic bone marrow transplantation. Clin Transplant 2024; 38:e15313. [PMID: 38581299 DOI: 10.1111/ctr.15313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND The number of CD34+ cells in the graft is generally associated with time to engraftment and survival in transplantation using cord blood or allogeneic peripheral blood stem cells. However, the significance of abundant CD34+ in bone marrow transplantation (BMT) remained unclear. METHODS We retrospectively reviewed 207 consecutive adult patients who underwent their first BMT at Jichi Medical University between January 2009 and June 2021. RESULTS The median nucleated cell count (NCC) and CD34+ cell dose were 2.17 × 108/kg (range .56-8.52) and 1.75 × 106/kg (.21-5.84), respectively. Compared with 104 patients in the low CD34+ group (below the median), 103 patients in the high CD34+ group (above the median) showed faster engraftment at day +28 in terms of neutrophil (84.6% vs. 94.2%; p = .001), reticulocyte (51.5% vs. 79.6%; p < .001), and platelet (39.4% vs. 72.8%; p < .001). There were no significant differences in overall survival, relapse, nonrelapse mortality, acute or chronic graft-versus-host disease, or infectious complications between the two groups in univariate and multivariate analyses. Low or high NCC had no significant effect on overall survival, nonrelapse mortality, cumulative incidence of relapse and graft-versus-host disease, either. While a positive correlation was observed between NCC and the CD34+ cell dose, a high CD34+ cell dose was associated with rapid hematopoietic recovery, even in patients with NCC below the median. CONCLUSION Measurement of CD34+ cell dose in addition to NCC was useful for predicting hematopoietic recovery, but seemed to have little influence on the long-term outcome in BMT.
Collapse
Affiliation(s)
- Takashi Oyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichiro Fujiwara
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
- Division of Cell Transplantation and Transfusion, Jichi Medical University, Tochigi, Japan
| | - Ryutaro Tominaga
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Daizo Yokoyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Atsuto Noguchi
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shuka Furuki
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shunsuke Koyama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Rui Murahashi
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Hirotomo Nakashima
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuki Hyodo
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takashi Ikeda
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shin-Ichiro Kawaguchi
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yumiko Toda
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takashi Nagayama
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
- Division of Cell Transplantation and Transfusion, Jichi Medical University, Tochigi, Japan
| | - Kento Umino
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Daisuke Minakata
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kaoru Morita
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masahiro Ashizawa
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Chihiro Yamamoto
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kaoru Hatano
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuya Sato
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Ikuko Otsuki
- Division of Cell Transplantation and Transfusion, Jichi Medical University, Tochigi, Japan
| | - Ken Ohmine
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
2
|
Mamo T, Sumstad D, DeFor TE, Cao Q, MacMillan ML, Brunstein C, Juckett M, McKenna DH. Harvest Quality, Nucleated Cell Dose and Clinical Outcomes in Bone Marrow Transplantation: A Retrospective Study. Transplant Cell Ther 2023; 29:638.e1-638.e8. [PMID: 37419326 PMCID: PMC10592389 DOI: 10.1016/j.jtct.2023.07.003] [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: 03/28/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/09/2023]
Abstract
Higher doses of infused nucleated cells (NCs) are associated with improved clinical outcomes in bone marrow transplantation (BMT) recipients. Most clinicians recommend infusing at least 2.0 × 108 NCs/kg. BMT clinicians request a target NC dose, but the harvested NC dose may be below the requested NC dose even before cell processing. We conducted this retrospective study to investigate the quality of bone marrow (BM) harvest and factors that influence infused NC doses at our institution. We also correlated infused NC doses with clinical outcomes. The study population included 347 BMT recipients (median age, 11 years; range, <1 to 75 years) at the University of Minnesota between 2009 and 2019. Underlying diagnoses mainly included 39% malignant and 61% nonmalignant diagnoses. Requested, harvested, and infused NC doses, as well as cell processing data, were obtained from the Cell Therapy Laboratory; clinical outcomes data were obtained from the University of Minnesota BMT Database. BM harvests were facilitated either by our institution (61%) or by the National Marrow Donor Program (39%). Associations of infused doses with baseline characteristics were assessed using the general Wilcoxon test/Pearson's correlation coefficient. The association of infused dose with neutrophil engraftment (absolute neutrophil count >500) by day 42, platelet engraftment (>20,000) by 6 months, acute graft-versus-host disease grade II-IV, and overall survival (OS) at 5 years were evaluated using regression and Kaplan-Meier curves. The median requested NC dose was 3.0 × 108/kg (range, 2 to 8 × 108/kg), and the median harvested and infused NC doses were 4.0 × 108/kg and 3.6 × 108/kg, respectively. Only 7% of donors had a harvested dose below the minimum requested dose. Moreover, the correlation between requested doses and harvested doses was adequate, with a harvested/requested dose ratio <.5 observed in only 5% of harvests. Additionally, the harvest volume and cell processing method were significantly correlated with the infused dose. Harvest volume exceeding the median of 948 mL was related to a significantly lower infused dose (P < .01). Moreover, hydroxyethyl starch (HES)/buffy coat processing (used to reduce RBCs with major ABO incompatibility) led to a significantly lower infused dose (P < .01). Donor age (median, 19 years; range, <1 to-70 years) and sex did not significantly influence the infused dose. Finally, the infused dose was significantly correlated with neutrophil and platelet engraftment (P < .05) but not with 5-year OS (P = .87) or aGVHD (P = .33). In our program's experience, BM harvesting is efficient and meets the requested minimum dose for 93% of recipients. Harvest volume and cell process play significant roles in determining the final infused dose. Minimizing harvest volume and cell processing could lead to increased infused dose and thus improved outcomes. Moreover, a higher infused dose leads to a better rate of neutrophil and platelet engraftment but not to improved OS, which may be linked to the sample size of our study.
Collapse
Affiliation(s)
- Theodros Mamo
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota.
| | - Darin Sumstad
- M Health, Fairview Cell Therapy Laboratory and Molecular & Cellular Therapeutics, University of Minnesota, St Paul, Minnesota
| | - Todd E DeFor
- Masonic Cancer Center Biostatistics Core, University of Minnesota, Minneapolis, Minnesota
| | - Qing Cao
- Masonic Cancer Center Biostatistics Core, University of Minnesota, Minneapolis, Minnesota
| | - Margaret L MacMillan
- Blood and Marrow Transplantation & Cellular Therapy Program, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Claudio Brunstein
- Blood and Marrow Transplantation & Cellular Therapy Program, University of Minnesota, Minneapolis, Minnesota; Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Mark Juckett
- Blood and Marrow Transplantation & Cellular Therapy Program, University of Minnesota, Minneapolis, Minnesota; Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - David H McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota; M Health, Fairview Cell Therapy Laboratory and Molecular & Cellular Therapeutics, University of Minnesota, St Paul, Minnesota
| |
Collapse
|
3
|
Canarutto D, Omer Javed A, Pedrazzani G, Ferrari S, Naldini L. Mobilization-based engraftment of haematopoietic stem cells: a new perspective for chemotherapy-free gene therapy and transplantation. Br Med Bull 2023; 147:108-120. [PMID: 37460391 PMCID: PMC10502445 DOI: 10.1093/bmb/ldad017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION In haematopoietic stem cell transplantation (HSCT), haematopoietic stem cells (HSCs) from a healthy donor replace the patient's ones. Ex vivo HSC gene therapy (HSC-GT) is a form of HSCT in which HSCs, usually from an autologous source, are genetically modified before infusion, to generate a progeny of gene-modified cells. In HSCT and HSC-GT, chemotherapy is administered before infusion to free space in the bone marrow (BM) niche, which is required for the engraftment of infused cells. Here, we review alternative chemotherapy-free approaches to niche voidance that could replace conventional regimens and alleviate the morbidity of the procedure. SOURCES OF DATA Literature was reviewed from PubMed-listed peer-reviewed articles. No new data are presented in this article. AREAS OF AGREEMENT Chemotherapy exerts short and long-term toxicity to haematopoietic and non-haematopoietic organs. Whenever chemotherapy is solely used to allow engraftment of donor HSCs, rather than eliminating malignant cells, as in the case of HSC-GT for inborn genetic diseases, non-genotoxic approaches sparing off-target tissues are highly desirable. AREAS OF CONTROVERSY In principle, HSCs can be temporarily moved from the BM niches using mobilizing drugs or selectively cleared with targeted antibodies or immunotoxins to make space for the infused cells. However, translation of these principles into clinically relevant settings is only at the beginning, and whether therapeutically meaningful levels of chimerism can be safely established with these approaches remains to be determined. GROWING POINTS In pre-clinical models, mobilization of HSCs from the niche can be tailored to accommodate the exchange and engraftment of infused cells. Infused cells can be further endowed with a transient engraftment advantage. AREAS TIMELY FOR DEVELOPING RESEARCH Inter-individual efficiency and kinetics of HSC mobilization need to be carefully assessed. Investigations in large animal models of emerging non-genotoxic approaches will further strengthen the rationale and encourage application to the treatment of selected diseases.
Collapse
Affiliation(s)
- Daniele Canarutto
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, MI, Italy
- Vita-Salute San Raffaele University, Via Olgettina, 60, 20132 Milano, MI, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, MI, Italy
| | - Attya Omer Javed
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, MI, Italy
| | - Gabriele Pedrazzani
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, MI, Italy
- Vita-Salute San Raffaele University, Via Olgettina, 60, 20132 Milano, MI, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, MI, Italy
- Vita-Salute San Raffaele University, Via Olgettina, 60, 20132 Milano, MI, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milano, MI, Italy
- Vita-Salute San Raffaele University, Via Olgettina, 60, 20132 Milano, MI, Italy
| |
Collapse
|
4
|
Wang Z, Liao Y, Wang C, Tang C, Fang C, Luo J, Liu H, Mo X, Wang Z, Shen L, Wang J, Chen X, Yin Z, Li J, Shen W. Stem cell-based therapeutic strategies for rotator cuff tendinopathy. J Orthop Translat 2023; 42:73-81. [PMID: 37664079 PMCID: PMC10470406 DOI: 10.1016/j.jot.2023.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 09/05/2023] Open
Abstract
Rotator cuff tendinopathy is a common musculoskeletal disorder that imposes significant health and economic burden. Stem cell therapy has brought hope for tendon healing in patients with final stage rotator cuff tendinopathy. Some clinical trials have confirmed the effectiveness of stem cell therapy for rotator cuff tendinopathy, but its application has not been promoted and approved. There are still many issues that should be solved prior to using stem cell therapy in clinical applications. The optimal source and dose of stem cells for rotator cuff tendinopathy should be determined. We also proposed novel prospective approaches that can overcome cell population heterogeneity and standardize patient types for stem cell applications. The translational potential of this article This review explores the optimal sources of stem cells for rotator cuff tendinopathy and the principles for selecting stem cell dosages. Key strategies are provided for stem cell population standardization and recipient selection.
Collapse
Affiliation(s)
- Zetao Wang
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Youguo Liao
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Binjiang Institute of Zhejiang University, Hangzhou, China
| | - Cailian Fang
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Junchao Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hengzhi Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianan Mo
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Zicheng Wang
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Lingfang Shen
- Air Force Health Care Center for Special Services, Hangzhou, China
| | | | - Xiao Chen
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zi Yin
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianyou Li
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Weiliang Shen
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- Department of Orthopedic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Orthopaedics Research Institute of Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Czarnogórski MC, Sakowska J, Maziewski M, Zieliński M, Piekarska A, Obuchowski I, Młyński M, Dutka M, Sadowska-Klasa A, Zarzycka E, Bieniaszewska M, Trzonkowski P, Witkowski JM, Hellmann A, Ruckemann-Dziurdzińska K, Zaucha JM. Ageing-resembling phenotype of long-term allogeneic hematopoietic cells recipients compared to their donors. Immun Ageing 2022; 19:51. [PMID: 36324179 PMCID: PMC9628063 DOI: 10.1186/s12979-022-00308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Ageing is a complex phenomenon that leads to decreased proliferative activity, loss of function of the cells, and cellular senescence. Senescence of the immune system exacerbates individual's immune response, both humoral and cellular but increases the frequency of infections. We hypothesized that physiological ageing of adaptive immune system occurs in recipients of allogeneic hematopoietic cells transplant (allo-HCT) at faster rate when compared to their respective donors since the small number of donor cells undergo immense proliferative stress restoring recipients hematopoiesis. We compared molecular characterizations of ageing between recipients and donors of allo-HCT: telomeric length and immunophenotypic changes in main lymphocyte subsets - CD4+, CD8+, CD19+, CD56+. RESULTS Median telomeric length (TL) of CD8+ lymphocytes was significantly longer in donors compared to recipients (on average 2,1 kb and 1,7 kb respectively, p = 0,02). Similar trends were observed for CD4+ and CD19+ although the results did not reach statistical significance. We have also found trends in the immunophenotype between recipients and donors in the subpopulations of CD4+ (naïve and effector memory), CD8+ Eomes+ and B-lymphocytes (B1 and B2). Lower infection risk recipients had also a significantly greater percentage of NK cells (22,3%) than high-risk patients (9,3%) p = 0,04. CONCLUSION Our data do not support the initial hypothesis of accelerated aging in the long term all-HCT recipients with the exception of the recipients lymphocytes (mainly CD8+) which present some molecular features, characteristic for physiological ageing (telomeric shortening, immunophenotype) when compared to their respective donors. However, a history of lower infection numbers in HCT recipients seems to be associated with increased percentage of NK cells. The history of GVHD seems not to affect the rate of ageing. Therefore, it is safe to conclude that the observed subtle differences between recipients' and donors' cells result mainly from the proliferative stress in the early period after allo-HCT and the difference between hosts' and recipients' microenvironments.
Collapse
Affiliation(s)
| | - Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Mateusz Maziewski
- Department of Physiopathology, Medical University of Gdańsk, Gdańsk, Poland
| | - Maciej Zieliński
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Agnieszka Piekarska
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Igor Obuchowski
- Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, University of Gdańsk, Gdańsk, Poland
| | - Mikołaj Młyński
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Magdalena Dutka
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Alicja Sadowska-Klasa
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ewa Zarzycka
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Maria Bieniaszewska
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Jacek M Witkowski
- Department of Physiopathology, Medical University of Gdańsk, Gdańsk, Poland
| | - Andrzej Hellmann
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Jan M Zaucha
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
6
|
Shang P, Yu L, Cao S, Guo C, Zhang W. An improved cell line-derived xenograft humanized mouse model for evaluation of PD-1/PD-L1 blocker BMS202-induced immune responses in colorectal cancer. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1497-1506. [PMID: 36269133 PMCID: PMC9827804 DOI: 10.3724/abbs.2022145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The establishment of an in vivo mouse model mimicking human tumor-immune environments provides a promising platform for immunotherapy assessment, drug discovery and clinical decision guidance. To this end, we construct humanized NCG mice by transplanting human hCD34 + hematopoietic progenitors into non-obese diabetic (NOD) Cg- Prkdc scidIL2rg tm1Wjl /Sz (null; NCG) mice and monitoring the development of human hematopoietic and immune systems (Hu-NCG). The cell line-derived xenograft (CDX) Hu-NCG mouse models are set up to assess the outcome of immunotherapy mediated by the small molecule BMS202. As a PD-1/PD-L1 blocker, BMS202 shows satisfactory antitumour efficacy in the HCT116 and SW480 xenograft Hu-NCG mouse models. Mechanistically, BMS202 exerts antitumour efficacy by improving the tumor microenvironment and enhancing the infiltration of hCD8 + T cells and the release of hIFNγ in tumor tissue. Thus, tumor-bearing Hu-NCG mice are a suitable and important in vivo model for preclinical study, particularly in cancer immunotherapy.
Collapse
Affiliation(s)
- Pengzhao Shang
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing210009China
| | - Liting Yu
- Department of PharmacyBinzhou Medical UniversityYantai264003China
| | - Shucheng Cao
- School of EngineeringChina Pharmaceutical UniversityNanjing210009China
| | - Changying Guo
- School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing210009China,Correspondence address. Tel: +86-15737957481; (W.Z.) / Tel: +86-18252099426; (C.G.) @cpu.edu.cn
| | - Wanheng Zhang
- Department of Pharmacythe First Affiliated Hospitaland College of Clinical Medicine of Henan University of Science and TechnologyLuoyang471003China,Correspondence address. Tel: +86-15737957481; (W.Z.) / Tel: +86-18252099426; (C.G.) @cpu.edu.cn
| |
Collapse
|
7
|
Li M, Morse B, Kassim S. Development and clinical translation considerations for the next wave of gene modified hematopoietic stem and progenitor cells therapies. Expert Opin Biol Ther 2022; 22:1177-1191. [PMID: 35833356 DOI: 10.1080/14712598.2022.2101361] [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: 11/04/2022]
Abstract
INTRODUCTION Consistent and reliable manufacture of gene modified hematopoietic stem and progenitor cell (HPSC) therapies will be of the utmost importance as they become more mainstream and address larger populations. Robust development campaigns will be needed to ensure that these products will be delivered to patients with the highest quality standards. AREAS COVERED Through publicly available manuscripts, press releases, and news articles - this review touches on aspects related to HSPC therapy, development, and manufacturing. EXPERT OPINION Recent advances in genome modification technology coupled with the longstanding clinical success of HSPCs warrants great optimism for the next generation of engineered HSPC-based therapies. Treatments for some diseases that have thus far been intractable now appear within reach. Reproducible manufacturing will be of critical importance in delivering these therapies but will be challenging due to the need for bespoke materials and methods in combination with the lack of off-the-shelf solutions. Continued progress in the field will manifest in the form of industrialization which currently requires attention and resources directed toward the custom reagents, a focus on closed and automated processes, and safer and more precise genome modification technologies that will enable broader, faster, and safer access to these life-changing therapies.
Collapse
Affiliation(s)
| | - Brent Morse
- Dark Horse Consulting Group, Walnut Creek, CA, USA
| | | |
Collapse
|
8
|
de Koning C, Tao W, Lacna A, van Veghel K, Horwitz ME, Sanz G, Jagasia MH, Wagner JE, Stiff PJ, Hanna R, Cilloni D, Valcárcel D, Peled T, Galamidi Cohen E, Goshen U, Pandit A, Lindemans CA, Jan Boelens J, Nierkens S. Lymphoid and myeloid immune cell reconstitution after nicotinamide-expanded cord blood transplantation. Bone Marrow Transplant 2021; 56:2826-2833. [PMID: 34312498 PMCID: PMC8563413 DOI: 10.1038/s41409-021-01417-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Omidubicel (nicotinamide-expanded cord blood) is a potential alternative source for allogeneic hematopoietic cell transplantation (HCT) when an HLA-identical donor is lacking. A phase I/II trial with standalone omidubicel HCT showed rapid and robust neutrophil and platelet engraftment. In this study, we evaluated the immune reconstitution (IR) of patients receiving omidubicel grafts during the first 6 months post-transplant, as IR is critical for favorable outcomes of the procedure. Data was collected from the omidubicel phase I-II international, multicenter trial. The primary endpoint was the probability of achieving adequate CD4+ T-cell IR (CD4IR: > 50 × 106/L within 100 days). Secondary endpoints were the recovery of T-cells, natural killer (NK)-cells, B-cells, dendritic cells (DC), and monocytes as determined with multicolor flow cytometry. LOESS-regression curves and cumulative incidence plots were used for data description. Thirty-six omidubicel recipients (median 44; 13-63 years) were included, and IR data was available from 28 recipients. Of these patients, 90% achieved adequate CD4IR. Overall, IR was complete and consisted of T-cell, monocyte, DC, and notably fast NK- and B-cell reconstitution, compared to conventional grafts. Our data show that transplantation of adolescent and adult patients with omidubicel results in full and broad IR, which is comparable with IR after HCT with conventional graft sources.
Collapse
Affiliation(s)
- Coco de Koning
- University Medical Center Utrecht, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Weiyang Tao
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Amelia Lacna
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Guillermo Sanz
- Hospital Universitario y Politécnico la Fe, València, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | - Caroline A Lindemans
- University Medical Center Utrecht, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jaap Jan Boelens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stefan Nierkens
- University Medical Center Utrecht, Utrecht, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Gul HG, Erkurt MA, Sarici A, Kuku I, Kaya E, Berber I, Merter M, Bicim S, Ozgul M. An innovation in stem cell harvesting: Heparin use. Transfus Apher Sci 2021; 60:103240. [PMID: 34404617 DOI: 10.1016/j.transci.2021.103240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVES Stem cell transplantation is a growing treatment strategy for most malignant and non- malignant hematological diseases. Plerixafor and granulocyte colony stimulating factor (G-CSF) are usually used in mobilization regimens to increase the CD34+ cell count in the harvest. Heparin is a sulphated glycosaminoglycated polymer with 12-15 kDa mass. Heparin inhibits the CXCR4/SDF1 axis, as does plerixafor. In this study, our aim was to investigate the effect of using heparin on stem cell mobilization and harvesting. MATERIALS AND METHODS We administered 5000 units of unfractioned heparin intravenously in 150 mL (mL) of isotonic sodium chloride solution, 15 min before the stem cell harvesting procedure to 141 patients who underwent bone marrow transplantation between the years of 2018 and 2019 at our Stem Cell Transplantation Unit. Thirty patients were included as a control group, and they were not given heparin. The study population included patients with multiple myeloma and lymphoma equally in each group. RESULTS In all patients hematopoeitic stem cells were successfully harvested in a single cycle of apheresis. In multiple myeloma patients who received heparin, the mean collected CD34+ cell number was 8 × 106/kg, and the mean CD34+ cell number yield was 12,555/μl. In the control group, the mean collected CD34+ cell number was 4,2 × 106/kg, and mean CD34+ cell number in yield was 492/μl. In lymphoma patients who received heparin, the mean collected CD34+ cell number was 6,8 × 106/kg, and the mean CD34+ cell number was 1421/μl. In the control group the mean collected CD34+ cell number was 4,3 × 106/kg, and the mean CD34+ cell number was 358/μl. The effect of heparin on the collected stem cell number in both myeloma and lymphoma patients was statistically significant (p < 0.01). CONCLUSIONS Our results have shown that heparin increases harvested stem cell numbers significantly. Heparin may be a promising agent for stem cell harvesting.
Collapse
Affiliation(s)
- Hacer Gozde Gul
- Inonu University, Deparment of Internal Medicine, Malatya, Turkey
| | - Mehmet Ali Erkurt
- Inonu University, Department of Hematology & Bone Marrow Transplantation Unit, Malatya, Turkey.
| | - Ahmet Sarici
- Inonu University, Department of Hematology & Bone Marrow Transplantation Unit, Malatya, Turkey
| | - Irfan Kuku
- Inonu University, Department of Hematology & Bone Marrow Transplantation Unit, Malatya, Turkey
| | - Emin Kaya
- Inonu University, Department of Hematology & Bone Marrow Transplantation Unit, Malatya, Turkey
| | - Ilhami Berber
- Inonu University, Department of Hematology & Bone Marrow Transplantation Unit, Malatya, Turkey
| | - Mustafa Merter
- Elazig University, Deparment of Hematology, Elazig, Turkey
| | - Soykan Bicim
- Inonu University, Department of Hematology & Bone Marrow Transplantation Unit, Malatya, Turkey
| | - Mustafa Ozgul
- Inonu University, Department of Hematology & Bone Marrow Transplantation Unit, Malatya, Turkey
| |
Collapse
|
10
|
Rennert W, Sobh L, Cormier K, Smith J, Gonzalez C. The impact of donor total estimated blood volume on nucleated cell yield in bone marrow harvests for hematopoietic stem cell transplantation. Transfusion 2021; 61:1533-1541. [PMID: 33768535 DOI: 10.1111/trf.16374] [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: 10/26/2020] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Nucleated cell yields of marrow harvests depend on factors related to donors, the procedure itself, and the volume of marrow harvested. Few attempts have been made to relate donor characteristics to harvest volume. We hypothesize that the percentage of total donor blood volume accessed for harvesting impacts the nucleated cell yield per ml of marrow collected. METHODS AND MATERIALS We investigated 481 consecutive unrelated marrow harvests from a single center. Donor characteristics including weight, body mass index (BMI), white blood cells (WBCs), hemoglobin (Hgb), and platelet counts, as well as estimated total blood volume, were recorded and compared with nucleated cell yields and harvest volumes. RESULTS The percentage of donor blood volume accessed for marrow harvesting was inversely related to nucleated cell yields (r = -0.57). The donor-recipient weight differential impacted cell yields as well (r = 0.35), with heavier recipients requiring increased marrow volumes from smaller donors to satisfy their nucleated cell needs. 3.73 × 108 /kg of recipient weight could be collected with 95% certainty when harvest volumes did not exceed 16.1% of donor total blood volume. In a stepwise multiple regression analysis, 45.4% of cell yield variance was explained by blood volume percentage accessed for harvesting, donor weight, and WBC. Donor sex, BMI, and platelet counts did not contribute further to cell yield variance. Smokers had higher cell yields than nonsmokers (20.4 vs. 18.3 × 106 /ml; 95% confidence interval 0.62, 3.47) independent of other parameters. CONCLUSION Establishing the relationship between percentage of estimated donor total blood volume and recipient cell needs can facilitate donor selection for successful hematopoietic cell (HPC) transplants.
Collapse
Affiliation(s)
- Wolfgang Rennert
- Blood and Marrow Collection Program, Georgetown University Medical Center, Washington, DC, USA
| | - Lina Sobh
- Blood and Marrow Collection Program, Georgetown University Medical Center, Washington, DC, USA
| | - Katie Cormier
- Blood and Marrow Collection Program, Georgetown University Medical Center, Washington, DC, USA
| | - Jenna Smith
- Blood and Marrow Collection Program, Georgetown University Medical Center, Washington, DC, USA
| | - Corina Gonzalez
- Blood and Marrow Collection Program, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
11
|
Elmariah H, Naqvi SMH, Kim J, Nishihori T, Mishra A, Perez L, Faramand R, Lazaryan A, Liu HD, Khimani F, Nieder M, Anasetti C, Pidala J, Bejanyan N. Impact of infused CD34+ stem cell dosing for allogeneic peripheral blood stem cell transplantation with post-transplant cyclophosphamide. Bone Marrow Transplant 2021; 56:1683-1690. [PMID: 33658647 DOI: 10.1038/s41409-021-01219-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/23/2020] [Accepted: 01/11/2021] [Indexed: 01/25/2023]
Abstract
Higher infused total nucleated cell dose (TNC) in allogeneic bone marrow transplant (BMT) with post-transplant cyclophosphamide (PTCy) is associated with improved overall survival. As many centers prefer peripheral blood stem cell grafts (PBSCT) with PTCy, the effect of cell dose on outcomes with this platform also requires elucidation. We retrospectively evaluated 144 consecutive adult patients who received allogeneic T-cell replete PBSCT with PTCy-based graft-versus-host disease (GVHD) prophylaxis for a hematologic malignancy from 2012-2018. The infused CD34+ cell dose was stratified into low (<5 × 106/kg), intermediate (5-10 × 106/kg) and high (>10 × 106/kg) dose level groups. In multivariate analysis, the low CD34+ cell dose group had worse non-relapse mortality (HR = 4.51, 95% CI: 1.92-10.58, p < 0.001), progression- free survival (HR = 4.11, 95% CI: 2.07-8.15, p < 0.001), and overall survival (HR = 4.06, 95% CI: 2.00-8.25, p ≤ 0.001) compared to the intermediate group. Clinical outcomes between the intermediate and high CD34+ cell dose groups were similar. TNC and CD3+ cell dose had no significant impacts on outcomes. These findings suggest that, in patients receiving allogeneic PBSCT with PTCy, infused CD34+ cell doses >5 × 106 cells/kg may result in improved survival. Thus, this study supports targeting a CD34+ cell dose of >5 × 106 cells/kg for allogeneic PBSCT with PTCy.
Collapse
Affiliation(s)
- Hany Elmariah
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| | | | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Taiga Nishihori
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Asmita Mishra
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Lia Perez
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Rawan Faramand
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Aleksandr Lazaryan
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Hien D Liu
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Farhad Khimani
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Michael Nieder
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Claudio Anasetti
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Joseph Pidala
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Nelli Bejanyan
- Department of Bone Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
12
|
Arcuri LJ, Schirmer M, Colares M, Maradei S, Tavares R, Moreira MCR, Araujo RDC, Lerner D, Pacheco AGF. Impact of Anti-CMV IgG Titers and CD34 Count Prior to Hematopoietic Stem Cell Transplantation from Alternative Donors on CMV reactivation. Biol Blood Marrow Transplant 2020; 26:e275-e279. [DOI: 10.1016/j.bbmt.2020.07.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 01/10/2023]
|
13
|
External validation of models for KIR2DS1/KIR3DL1-informed selection of hematopoietic cell donors fails. Blood 2020; 135:1386-1395. [PMID: 31932846 DOI: 10.1182/blood.2019002887] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/11/2019] [Indexed: 01/13/2023] Open
Abstract
Several studies suggest that harnessing natural killer (NK) cell reactivity mediated through killer cell immunoglobulin-like receptors (KIRs) could reduce the risk of relapse after allogeneic hematopoietic cell transplantation. Based on one promising model, information on KIR2DS1 and KIR3DL1 and their cognate ligands can be used to classify donors as KIR-advantageous or KIR-disadvantageous. This study was aimed at externally validating this model in unrelated donor hematopoietic cell transplantation. The impact of the predictor on overall survival (OS) and relapse incidence was tested in a Cox regression model adjusted for patient age, a modified disease risk index, Karnofsky performance status, donor age, HLA match, sex match, cytomegalovirus match, conditioning intensity, type of T-cell depletion, and graft type. Data from 2222 patients with acute myeloid leukemia or myelodysplastic syndrome were analyzed. KIR genes were typed by using high-resolution amplicon-based next-generation sequencing. In univariable analyses and subgroup analyses, OS and the cumulative incidence of relapse of patients with a KIR-advantageous donor were comparable to patients with a KIR-disadvantageous donor. The adjusted hazard ratio from the multivariable Cox regression model was 0.99 (Wald test, P = .93) for OS and 1.04 (Wald test, P = .78) for relapse incidence. We also tested the impact of activating donor KIR2DS1 and inhibition by KIR3DL1 separately but found no significant impact on OS and the risk of relapse. Thus, our study shows that the proposed model does not universally predict NK-mediated disease control. Deeper knowledge of NK-mediated alloreactivity is necessary to predict its contribution to graft-versus-leukemia reactions and to eventually use KIR genotype information for donor selection.
Collapse
|
14
|
Arcuri LJ, Nabhan SK, Cunha R, Nichele S, Ribeiro AAF, Fernandes JF, Daudt LE, Rodrigues ALM, Arrais-Rodrigues C, Seber A, Atta EH, de Oliveira JSR, Funke VAM, Loth G, Junior LGD, Paz A, Calixto RF, Gomes AA, Araujo CES, Colturato V, Simoes BP, Hamerschlak N, Flowers ME, Pasquini R, Rocha V, Bonfim C. Impact of CD34 Cell Dose and Conditioning Regimen on Outcomes after Haploidentical Donor Hematopoietic Stem Cell Transplantation with Post-Transplantation Cyclophosphamide for Relapsed/Refractory Severe Aplastic Anemia. Biol Blood Marrow Transplant 2020; 26:2311-2317. [PMID: 32949751 DOI: 10.1016/j.bbmt.2020.09.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/31/2022]
Abstract
Severe aplastic anemia (SAA) is a life-threatening disease that can be cured with allogeneic cell transplantation (HCT). Haploidentical donor transplantation with post-transplantation cyclophosphamide (haplo-PTCy) is an option for patients lacking an HLA-matched donor. We analyzed 87 patients who underwent haplo-PTCy between 2010 and 2019. The median patient age was 14 years (range, 1 to 69 years), most were heavily transfused, and all received previous immunosuppression (25% without antithymocyte globulin). Almost two-thirds (63%) received standard fludarabine (Flu)/cyclophosphamide (Cy) 29/total body irradiation (TBI) 200 cGy conditioning, and the remaining patients received an augmented conditioning: Flu/Cy29/TBI 300-400 (16%), Flu/Cy50/TBI 200 (10%), or Flu/Cy50/TBI 400 (10%). All patients received PTCy-based graft-versus-host disease (GVHD) prophylaxis. Most grafts (93%) were bone marrow (BM). The median duration of follow-up was 2 years and 2 months. The median time to neutrophil recovery was 17 days. Primary graft failure occurred in 15% of the patients, and secondary or poor graft function occurred in 5%. The incidences of grade II-IV acute GVHD was 14%, and that of chronic GVHD was 9%. Two-year overall survival and event-free survival (EFS) were 79% and 70%, respectively. EFS was higher for patients who received augmented Flu/Cy/TBI (hazard ratio [HR], .28; P = .02), and those who received higher BM CD34 cell doses (>3.2 × 10E6/kg) (HR, .29; P = .004). The presence of donor-specific antibodies before HSCT was associated with lower EFS (HR, 3.92; P = .01). Graft failure (HR, 7.20; P < .0001) was associated with an elevated risk of death. Cytomegalovirus reactivation was frequent (62%). Haploidentical HCT for SAA is a feasible procedure; outcomes are improved with augmented conditioning regimens and BM grafts with higher CD34 cell doses.
Collapse
Affiliation(s)
- Leonardo Javier Arcuri
- Hospital Israelita Albert Einstein, Bone Marrow Transplantation Unit, Sao Paulo, Brazil.
| | - Samir Kanaan Nabhan
- Universidade Federal do Parana, Bone Marrow Transplantation Unit, Curitiba, Brazil
| | - Renato Cunha
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Bone Marrow Transplantation Unit, Ribeirao Preto, Brazil
| | - Samantha Nichele
- Universidade Federal do Parana, Bone Marrow Transplantation Unit, Curitiba, Brazil
| | | | - Juliana Folloni Fernandes
- Hospital Israelita Albert Einstein, Bone Marrow Transplantation Unit, Sao Paulo, Brazil; Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Bone Marrow Transplantation Unit, Sao Paulo, Brazil
| | - Liane Esteves Daudt
- Hospital das Clinicas de Porto Alegre, Bone Marrow Transplantation Unit, Porto Alegre, Brazil
| | | | | | - Adriana Seber
- Hospital Samaritano, Bone Marrow Transplantation Unit, Sao Paulo, Brazil
| | - Elias Hallack Atta
- Instituto Nacional de Cancer, Bone Marrow Transplantation Unit, Rio de Janeiro, Brazil
| | | | | | - Gisele Loth
- Universidade Federal do Parana, Bone Marrow Transplantation Unit, Curitiba, Brazil
| | - Luiz Guilherme Darrigo Junior
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Bone Marrow Transplantation Unit, Ribeirao Preto, Brazil
| | - Alessandra Paz
- Hospital das Clinicas de Porto Alegre, Bone Marrow Transplantation Unit, Porto Alegre, Brazil
| | - Rodolfo Froes Calixto
- Real Hospital Portugues de Beneficencia em Pernambuco, Bone Marrow Transplantation Unit, Recife, Brazil
| | | | - Carlos Eduardo Sa Araujo
- Instituto de Cardiologia do Distrito Federal, Bone Marrow Transplantation Unit, Brasilia, Brazil
| | | | - Belinda Pinto Simoes
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Bone Marrow Transplantation Unit, Ribeirao Preto, Brazil
| | - Nelson Hamerschlak
- Hospital Israelita Albert Einstein, Bone Marrow Transplantation Unit, Sao Paulo, Brazil
| | | | - Ricardo Pasquini
- Universidade Federal do Parana, Bone Marrow Transplantation Unit, Curitiba, Brazil
| | - Vanderson Rocha
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Bone Marrow Transplantation Unit, Sao Paulo, Brazil; Rede D'or, Bone Marrow Transplantation Unit, Sao Paulo, Brazil
| | - Carmem Bonfim
- Universidade Federal do Parana, Bone Marrow Transplantation Unit, Curitiba, Brazil
| |
Collapse
|
15
|
Maffini E, Labopin M, Blaise D, Ciceri F, Gülbas Z, Deconinck E, Leblond V, Chevallier P, Sociè G, Araujo MC, Koc Y, Savani BN, Gorin NC, Lanza F, Nagler A, Mohty M. CD34+ cell dose effects on clinical outcomes after T-cell replete haploidentical allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia using peripheral blood stem cells. A study from the acute leukemia working Party of the European Society for blood and marrow transplantation (EBMT). Am J Hematol 2020; 95:892-899. [PMID: 32303111 DOI: 10.1002/ajh.25826] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/23/2020] [Accepted: 04/08/2020] [Indexed: 12/15/2022]
Abstract
Previous observations have reported controversial conclusions regarding cell dose and survival endpoints after allogeneic hematopoietic stem cell transplantation (HSCT). We conducted a retrospective analysis on 414 adult patients (median age 54 years, range, 18-74 years) with acute myeloid leukemia (AML) in first and second complete remission. They received a T-cell replete allogeneic HSCT from haploidentical donors, using peripheral blood stem cells, between 2006-2018. Median number of infused CD34+ was 6.58 × 106 /kg (range, 2.2-31.2 × 106 /kg). Graft-vs-host disease (GVHD) prophylaxis was post-transplant cyclophosphamide in 293 patients and anti-lymphocyte serum in 121 patients. Conditioning was myeloablative in 179 patients and reduced-intensity in 235 patients. After a median follow-up of 23.3 months (range, 12.1-41.8 months), 2-year overall survival (OS) was 64.5% (95% CI 59.3%-69.7%) with leukemia-free survival (LFS) of 57.3% (95% CI 51.8%-62.7%) and non-relapse mortality (NRM) of 23.3% (95% CI 19%-27.7%). Grades III-IV acute GVHD day+100 incidence was 14.6% while extensive chronic GVHD was 14.4% at 2-years. Thirteen (3.2%) patients experienced graft failure. We found the optimal CD34+/kg threshold defining high (n = 334) vs low cell dose (n = 80) at 4.96 × 106 . Recipients of >4.96 × 106 /kg CD34+ cells experienced less NRM (Hazard ratio [HR] 0.48; 95% CI 0.30-0.76) and prolonged LFS (HR 0.63; 95% CI 0.43-0.91) and OS (HR 0.60; 95% CI 0.40-0.88) compared to those in the lower cell dose cohort. Larger cohort studies are needed to confirm these findings.
Collapse
Affiliation(s)
| | - Myriam Labopin
- Acute Leukemia Working Party OfficeHospital Saint Antoine Paris France
- Assistance Publique‐Hopitaux de ParisHospital Saint Antoine Paris France
- University Pierre et Marie Curie Paris France
- Institut National de la Santè et de la Recherche Médicale Unitè Mixte de Recherche en Santè Paris France
| | - Didier Blaise
- Programme de Transplantation & Therapie CellulaireCentre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes Marseille France
| | - Fabio Ciceri
- Ospedale San RaffaeleHaematology and BMT Milan Italy
| | - Zafer Gülbas
- Anadolu Medical Center HospitalBone Marrow Transplantation Department Kocaeli Turkey
| | - Eric Deconinck
- Hopital Jean MinjozService d'Hématologie Besancon France
| | - Veronique Leblond
- Universite Paris IVHopital la Pitié‐Salpêtrière, Hematologie Clinique Paris France
| | - Patrick Chevallier
- Centre Hospitalier Universitaire NantesDept. D'Hematologie Nantes France
| | - Gerard Sociè
- Hopital Saint‐LouisService d'Hematologie – BMT Paris France
| | - Mercedes C. Araujo
- Hospital U. Marqués de ValdecillaServicio de Hematología‐Hemoterapia Santander Spain
| | - Yener Koc
- Medical Park HospitalsStem Cell Transplant Unit Antalya Turkey
| | - Bipin N. Savani
- Division of Hematology and Medical OncologyVanderbilt University Medical Center Nashville Tennessee USA
| | | | | | - Arnon Nagler
- Hematology and Bone Marrow Transplantation DivisionChaim Sheba Medical Center Tel‐Hashomer Israel
- Sackler School of MedicineTel Aviv University Tel Aviv Israel
| | - Mohamad Mohty
- Saint‐Antoine Hospital Paris France
- Sorbonne University Paris France
- INSERM UMRs 938 Paris France
| |
Collapse
|
16
|
Simantirakis E, Tsironis I, Vassilopoulos G. FV Vectors as Alternative Gene Vehicles for Gene Transfer in HSCs. Viruses 2020; 12:E332. [PMID: 32204324 PMCID: PMC7150843 DOI: 10.3390/v12030332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/08/2020] [Accepted: 03/15/2020] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic Stem Cells (HSCs) are a unique population of cells, capable of reconstituting the blood system of an organism through orchestrated self-renewal and differentiation. They play a pivotal role in stem cell therapies, both autologous and allogeneic. In the field of gene and cell therapy, HSCs, genetically modified or otherwise, are used to alleviate or correct a genetic defect. In this concise review, we discuss the use of SFVpsc_huHSRV.13, formerly known as Prototype Foamy Viral (PFV or FV) vectors, as vehicles for gene delivery in HSCs. We present the properties of the FV vectors that make them ideal for HSC delivery vehicles, we review their record in HSC gene marking studies and their potential as therapeutic vectors for monogenic disorders in preclinical animal models. FVs are a safe and efficient tool for delivering genes in HSCs compared to other retroviral gene delivery systems. Novel technological advancements in their production and purification in closed systems, have allowed their production under cGMP compliant conditions. It may only be a matter of time before they find their way into the clinic.
Collapse
Affiliation(s)
- Emmanouil Simantirakis
- Gene Therapy Lab, Biomedical Research Foundation of the Academy of Athens, Division of Genetics and Gene Therapy, Basic Research II, 11527 Athens, Greece; (E.S.); (I.T.)
| | - Ioannis Tsironis
- Gene Therapy Lab, Biomedical Research Foundation of the Academy of Athens, Division of Genetics and Gene Therapy, Basic Research II, 11527 Athens, Greece; (E.S.); (I.T.)
| | - George Vassilopoulos
- Gene Therapy Lab, Biomedical Research Foundation of the Academy of Athens, Division of Genetics and Gene Therapy, Basic Research II, 11527 Athens, Greece; (E.S.); (I.T.)
- Division of Hematology, University of Thessaly Medical School, 41500 Larissa, Greece
| |
Collapse
|
17
|
Salas MQ, Atenafu EG, Bautista MR, Prem S, Lam W, Datt Law A, Shaibani ZA, Kim DDH, Michelis FV, Lipton JH, Viswabandya A, Mattsson J, Kumar R. Impact of CD34+ cell dose on reduced intensity conditioning regimen haploidentical hematopoietic stem cell transplantation. Eur J Haematol 2019; 104:36-45. [PMID: 31549435 DOI: 10.1111/ejh.13332] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Haploidentical hematopoietic stem cell transplant (haplo-SCT) has been associated with higher rates of graft rejection, and a higher dose of CD34+ cell dose is frequently requested. We aim to explore the impact of CD34+ cell dose in peripheral blood stem cell (PBSC) grafts using reduced intensity conditioning (RIC) in haplo-SCT. METHODS Sixty-eight consecutive haplo-SCT in adult patients were included. Graft-vs-host disease (GVHD) prophylaxis consisted on ATG, PTCy, and CsA. The cohort was divided in two groups using CD34+ dose of ≥ 9 × 106 CD34+/Kg as cutoff point. Median follow-up was 8.9 months. RESULTS Median cell dose infused was 9.32 × 106 CD34+/Kg. Forty (58.8%) recipients received grafts with CD34+ cells ≥9 × 106 /kg. The infusion ≥ 9 × 106 CD34+/Kg cell dose had a negative impact in overall survival (P = .03) after adjusting for age at transplant. The cumulative incidence of acute GVHD and graft failure were not significantly influenced per CD34+ cell dose. Only four recipients had grade III aGVHD, and all of them received grafts with a CD34+ cell dose ≥ 9 × 106 . CONCLUSION In RIC haplo-SCT, recipients may not benefit from PBSC grafts with a CD34+/kg cell dose higher than 9 × 106 cells/kg, as it can have an adverse impact in post-transplant outcome.
Collapse
Affiliation(s)
- Maria Queralt Salas
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Eshetu G Atenafu
- Department of Biostatistics, Princes Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Maria Rhida Bautista
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Shruti Prem
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wilson Lam
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Arjun Datt Law
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Zeyad-Al Shaibani
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Dennis Dong Hwan Kim
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Fotios V Michelis
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jeffrey Howard Lipton
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Auro Viswabandya
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jonas Mattsson
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Rajat Kumar
- Section of Medical Oncology and Hematology, Department of Internal Medicine, University of Toronto, ON, Canada.,Hans Messner Allogeneic Blood and Marrow Transplantation Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
18
|
Saad A, Lamb L, Wang T, Hemmer MT, Spellman S, Couriel D, Alousi A, Pidala J, Abdel-Azim H, Agrawal V, Aljurf M, Beitinjaneh AM, Bhatt VR, Buchbinder D, Byrne M, Cahn JY, Cairo M, Castillo P, Chhabra S, Diaz MA, Farhan S, Floisand Y, Frangoul HA, Gadalla SM, Gajewski J, Gale RP, Gandhi M, Gergis U, Hamilton BK, Hematti P, Hildebrandt GC, Kamble RT, Kanate AS, Khandelwal P, Lazaryan A, MacMillan M, Marks DI, Martino R, Mehta PA, Nishihori T, Olsson RF, Patel SS, Qayed M, Rangarajan HG, Reshef R, Ringden O, Savani BN, Schouten HC, Schultz KR, Seo S, Shaffer BC, Solh M, Teshima T, Urbano-Ispizua A, Verdonck LF, Vij R, Waller EK, William B, Wirk B, Yared JA, Yu LC, Arora M, Hashmi S. Impact of T Cell Dose on Outcome of T Cell-Replete HLA-Matched Allogeneic Peripheral Blood Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1875-1883. [PMID: 31085303 PMCID: PMC7071947 DOI: 10.1016/j.bbmt.2019.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 01/24/2023]
Abstract
Data on whether the T cell dose of allogeneic peripheral blood stem cell (PBSC) products influences transplantation outcomes are conflicting. Using the Center for International Blood and Marrow Transplant Research database, we identified 2736 adult patients who underwent first allogeneic PBSC transplantation for acute leukemia or myelodysplastic syndrome between 2008 and 2014 using an HLA-matched sibling donor (MSD) or an 8/8-matched unrelated donor (MUD). We excluded ex vivo and in vivo T cell-depleted transplantations. Correlative analysis was performed between CD3+ T cell dose and the risk of graft-versus-host-disease (GVHD), relapse, nonrelapse mortality (NRM), disease-free survival (DFS), and overall survival (OS). Using maximum likelihood estimation, we identified CD3+ T cell dose cutoff that separated the risk of acute GVHD (aGVHD) grade II-IV in both the MSD and MUD groups. A CD3+ T cell dose cutoff of 14 × 107 cells/kg identified MSD/low CD3+ (n = 223) and MSD/high CD3+ (n = 1214), and a dose of 15 × 107 cells/kg identified MUD/low CD3+ (n = 197) and MUD/high CD3+ (n = 1102). On univariate analysis, the MSD/high CD3+ group had a higher cumulative incidence of day +100 aGVHD grade II-IV compared with the MSD/low CD3+ group (33% versus 25%; P = .009). There were no differences between the 2 groups in engraftment rate, risk of aGVHD grade III-IV or chronic GVHD (cGVHD), NRM, relapse, DFS, or OS. The MUD/high CD3+ group had a higher cumulative incidence of day +100 aGVHD grade II-IV compared with the MUD/low CD3+ group (49% versus 41%; P = .04). There were no differences between the 2 groups in engraftment rate, risk of severe aGVHD or cGVHD, NRM, relapse, DFS, or OS. Multivariate analysis of the MSD and MUD groups failed to show an association between CD3+ T cell dose and the risk of either aGVHD grade II-IV (P = .10 and .07, respectively) or cGVHD (P = .80 and .30, respectively). Subanalysis of CD4+ T cells, CD8+ T cells, and CD4+/CD8+ ratio failed to identify cutoff values predictive of transplantation outcomes; however, using the log-rank test, the sample size was suboptimal for identifying a difference at this cutoff cell dose. In this registry study, the CD3+ T cell dose of PBSC products did not influence the risk of aGVHD or cGVHD or other transplantation outcomes when using an MSD or an 8/8-matched MUD. Subset analyses of CD4+ and CD8+ T cell doses were not possible given our small sample size.
Collapse
Affiliation(s)
- Ayman Saad
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Lawrence Lamb
- University of Alabama at Birmingham, Birmingham, Alabama
| | - Tao Wang
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael T Hemmer
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Stephen Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be the Match, Minneapolis, Minnesota
| | - Daniel Couriel
- Utah Blood and Marrow Transplant Program, Salt Lake City, Utah
| | - Amin Alousi
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Hisham Abdel-Azim
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital of Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Vaibhav Agrawal
- Division of Hematology-Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mahmoud Aljurf
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Vijaya Raj Bhatt
- The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - David Buchbinder
- Division of Pediatric Hematology, Children's Hospital of Orange County, Orange, California
| | - Michael Byrne
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jean-Yves Cahn
- Department of Hematology, CHU Grenoble Alpes, Grenoble, France
| | - Mitchell Cairo
- Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatrics, New York Medical College, New York, New York
| | - Paul Castillo
- UF Health Shands Children's Hospital, Gainesville, Florida
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Miguel Angel Diaz
- Department of Hematology/Oncology, Hospital Infantil Universitario Nino Jesus, Madrid, Spain
| | - Shatha Farhan
- Henry Ford Hospital Bone Marrow Transplant Program, Detroit, Michigan
| | | | - Hadar A Frangoul
- Children's Hospital at TriStar Centennial and Sarah Cannon Research Institute, Nashville, Tennessee
| | - Shahinaz M Gadalla
- Division of Cancer Epidemiology & Genetics, Clinical Genetics Branch, National Cancer Institute, Rockville, Maryland
| | | | - Robert Peter Gale
- Hematology Research Center, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Manish Gandhi
- Division of Transfusion Medicine, Mayo Clinic, Rochester, Minnesota
| | - Usama Gergis
- Hematologic Malignancies & Bone Marrow Transplant, Department of Medical Oncology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - Betty Ky Hamilton
- Blood & Marrow Transplant Program, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Peiman Hematti
- Division of Hematology/Oncology/Bone Marrow Transplantation, Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, Wisconsin
| | | | - Rammurti T Kamble
- Division of Hematology and Oncology, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Abraham S Kanate
- Osborn Hematopoietic Malignancy and Transplantation Program, West Virginia University, Morgantown, West Virginia
| | - Pooja Khandelwal
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Aleksandr Lazaryan
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Margaret MacMillan
- University of Minnesota Blood and Marrow Transplant Program, Pediatrics, Minneapolis, Minnesota
| | - David I Marks
- Adult Bone Marrow Transplant, University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | - Rodrigo Martino
- Division of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Parinda A Mehta
- Division of Bone Marrow Transplant and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Richard F Olsson
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden; Centre for Clinical Research Sormland, Uppsala University, Uppsala, Sweden
| | - Sagar S Patel
- Blood and Marrow Transplant Program, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Muna Qayed
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Hemalatha G Rangarajan
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplantation, Nationwide Children's Hospital, Columbus, Ohio
| | - Ran Reshef
- Blood and Marrow Transplantation Program and Columbia Center for Translational Immunobiology, Columbia University Medical Center, New York, New York
| | - Olle Ringden
- Translational Cell Therapy Research, Karolinska Institute, Stockholm, Sweden
| | - Bipin N Savani
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Harry C Schouten
- Department of Hematology, Academische Ziekenhuis, Maastricht, Netherlands
| | - Kirk R Schultz
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, British Columbia's Children's Hospital, The University of British Columbia, Vancouver, Britich Columbia, Canada
| | - Sachiko Seo
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | | | - Melhem Solh
- The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia
| | | | - Alvaro Urbano-Ispizua
- Department of Hematology, Hospital Clinic, University of Barcelona, IDIBAPS, and Josep Carreras Institute of Research, Barcelona, Spain
| | - Leo F Verdonck
- Department of Hematology/Oncology, Isala Clinic, Zwolle, The Netherlands
| | - Ravi Vij
- Division of Hematology and Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Edmund K Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Basem William
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Baldeep Wirk
- Division of Bone Marrow Transplant, Seattle Cancer Care Alliance, Seattle, Washington
| | - Jean A Yared
- Blood & Marrow Transplantation Program, Division of Hematology/Oncology, Department of Medicine, Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Lolie C Yu
- Division of Hematology/Oncology and HSCT, Center for Cancer and Blood Disorders, Children's Hospital/Louisiana State University Medical Center, New Orleans, Louisiana
| | - Mukta Arora
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, Minnesota.
| | - Shahrukh Hashmi
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia; Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
19
|
Evans J, Needle JJ, Hirani SP. Early outcomes of gastrostomy feeding in paediatric allogenic bone marrow transplantation: A retrospective cohort study. Clin Nutr ESPEN 2019; 31:71-79. [DOI: 10.1016/j.clnesp.2019.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 02/02/2023]
|
20
|
de Kruijf EJFM, Fibbe WE, van Pel M. Cytokine-induced hematopoietic stem and progenitor cell mobilization: unraveling interactions between stem cells and their niche. Ann N Y Acad Sci 2019; 1466:24-38. [PMID: 31006885 PMCID: PMC7217176 DOI: 10.1111/nyas.14059] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/15/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
Peripheral blood hematopoietic stem and progenitor cells (HSPCs), mobilized by granulocyte colony‐stimulating factor, are widely used as a source for both autologous and allogeneic stem cell transplantation. The use of mobilized HSPCs has several advantages over traditional bone marrow–derived HSPCs, including a less invasive harvesting process for the donor, higher HSPC yields, and faster hematopoietic reconstitution in the recipient. For years, the mechanisms by which cytokines and other agents mobilize HSPCs from the bone marrow were not fully understood. The field of stem cell mobilization research has advanced significantly over the past decade, with major breakthroughs in the elucidation of the complex mechanisms that underlie stem cell mobilization. In this review, we provide an overview of the events that underlie HSPC mobilization and address the relevant cellular and molecular components of the bone marrow niche. Furthermore, current and future mobilizing agents will be discussed.
Collapse
Affiliation(s)
- Evert-Jan F M de Kruijf
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem E Fibbe
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Melissa van Pel
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
21
|
Lara-Weisshaupt E, Tistl I, Hale K, Torosian T, Schlaphoff T, Manor S, Jindra P, Schöffel-Weiß S, Audat F, Abress L, Pingel J. Audits of collection and apheresis centers: guidelines by the World Marrow Donor Association Working Group Quality and Regulation. Bone Marrow Transplant 2018; 54:244-257. [PMID: 30108327 DOI: 10.1038/s41409-018-0252-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/27/2018] [Accepted: 05/17/2018] [Indexed: 11/09/2022]
Abstract
According to the Standards of the World Marrow Donor Association (WMDA), unrelated stem cell donor registries and donor centers are responsible for compliance of their collection and apheresis centers with these Standards. To ensure high stem cell product quality and high standards for safety and satisfaction of voluntary unrelated stem cell donors, we here present guidelines for audits of collection and apheresis centers that can be used by new and established donor registries, as well as by collection centers in preparation of audits. We define the general requirements and recommendations for collaboration with the collection and apheresis centers and define critical procedures for the collection of the stem cell product, such as information session, medical assessment, product collection, quality controls, product handover for transportation, and donor follow-up. The specific guidelines are accompanied by detailed checklists and forms that can be found in Supplementary Information and may be used during an initial or follow-up on-site or paper-based audit.
Collapse
Affiliation(s)
| | - Ingrid Tistl
- Zentrales Knochenmarkspender-Register Deutschland (ZKRD), Ulm, Germany
| | - Kuchen Hale
- National Marrow Donor Program, Minneapolis, USA
| | | | | | - Sigal Manor
- Ezer Mizion Bone Marrow Donor Registry, Petah-Tikva, Israel
| | - Pavel Jindra
- Czech National Marrow Donors Registry, Pilsen, Czech Republic
| | | | | | | | | |
Collapse
|
22
|
Yamamoto C, Ogawa H, Fukuda T, Igarashi A, Okumura H, Uchida N, Hidaka M, Nakamae H, Matsuoka KI, Eto T, Ichinohe T, Atsuta Y, Kanda Y. Impact of a Low CD34 + Cell Dose on Allogeneic Peripheral Blood Stem Cell Transplantation. Biol Blood Marrow Transplant 2017; 24:708-716. [PMID: 29196077 DOI: 10.1016/j.bbmt.2017.10.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/14/2017] [Indexed: 11/29/2022]
Abstract
Although the CD34+ cell dose in allogeneic peripheral blood stem cell transplantation (PBSCT) is considered to be associated with transplantation outcomes, a lower acceptable threshold has not been defined. We retrospectively analyzed 2919 adult patients with hematologic malignancies who underwent related PBSCT in Japan between 2001 and 2014. According to the number of CD34+ cells in the graft, we categorized 2494 patients in the standard group (2 to 5 × 106 cells/kg), 377 patient in the low group (1 to 2 × 106 cells/kg), and 48 patients in the very low group (<1 × 106 cells/kg). Compared with the standard group, the low and very low groups showed delayed neutrophil recovery (93.8%, 89.5%, and 78.3%, respectively at day +28; P < .001) and platelet recovery (69.3%, 53.0%, and 45.5%, respectively at day +28; P < .001). The 2-year overall survival (OS) in the 3 groups was 45.5%, 45.3%, and 29.8%, respectively, with inferior survival in the very low group. However, a higher percentage of high-risk patients may account for the inferior survival in the very low group, and no significant difference in OS was found in a multivariate analysis. There were no differences in relapse, nonrelapse mortality, or the development of graft-versus-host disease among the 3 groups. In conclusion, allogeneic PBSCT with low CD34+ cell doses of 1 to 2 × 106 cells/kg gives acceptable results, whereas further investigations are needed to evaluate the effects of lower doses of <1 × 106 cells/kg owing to the smaller number and the higher percentage of patients with adverse prognostic factors in this cohort.
Collapse
Affiliation(s)
- Chihiro Yamamoto
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Hiroyasu Ogawa
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Aiko Igarashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hirokazu Okumura
- Department of Internal Medicine (Hematology), Toyama Prefectural Central Hospital, Toyama, Japan
| | - Naoyuki Uchida
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Michihiro Hidaka
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka City University Hospital, Osaka, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan; Nagoya University Graduate School of Medicine, Department of Healthcare Administration, Nagoya, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan.
| |
Collapse
|
23
|
Strategies before, during, and after hematopoietic cell transplantation to improve T-cell immune reconstitution. Blood 2016; 128:2607-2615. [PMID: 27697775 DOI: 10.1182/blood-2016-06-724005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/28/2016] [Indexed: 12/11/2022] Open
Abstract
T-cell immune reconstitution (IR) after allogeneic hematopoietic cell transplantation (allo-HCT) is highly variable between patients and may take several months to even years. Patients with delayed or unbalanced T-cell IR have a higher probability of developing transplantation-related morbidity, mortality, and relapse of disease. Hence, there is a need for strategies to better predict and improve IR to reduce these limitations of allo-HCT. In this review, we provide an update of current and in-near-future clinically relevant strategies before, during, and after transplantation to achieve successful T-cell IR. Potent strategies are choosing the right HCT source (eg, donor-recipient matching, cell dose, graft manipulation), individualized conditioning and serotherapy (eg, antithymocyte globulin), nutritional status, exercise, home care, modulation of microbiota, enhancing homeostatic peripheral expansion, promoting thymopoiesis, and the use of adjuvant-targeted cellular immunotherapies. Strategies to prevent graft-versus-host disease are important as well because this complication and the subsequent need for immunosuppression affects T-cell IR and function. These options aim for personalized precision transplantation, where allo-HCT therapy is designed to boost a well-balanced T-cell IR and limit complications in individual patients, resulting in overall lower morbidity and higher survival chances.
Collapse
|
24
|
Guo ZP, Wang T, Xu LP, Zhang XH, Wang Y, Huang XJ, Chang YJ. Factors affecting the CD34 + cell yields from the second donations of healthy donors: The steady-state lymphocyte count is a good predictive factor. Transfus Apher Sci 2016; 55:311-317. [PMID: 27665155 DOI: 10.1016/j.transci.2016.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 08/02/2016] [Accepted: 08/16/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND A second allogeneic hematopoietic stem-cell transplantation and donor lymphocyte infusion using cells from the same donor is a therapeutic option in the case of stem-cell graft failure or disease relapse, but little is known about the factors associated with the CD34+ cell yields from second donations. METHODS One-hundred healthy donors who underwent a second mobilization treatment and peripheral blood stem-cell (PBSC) collection were studied. For both mobilization processes, 5 µg of granulocyte colony-stimulating factor per kg per day was administered. The blood counts of the donors were monitored during the processes. RESULTS The second donations from the same donors provided lower apheresis yields than did the initial collections. The number of CD34+ cells collected from normal donors after a second cycle of PBSC mobilization was associated with their steady-state lymphocyte counts and the intertransplantation interval. Female sex negatively affected the CD34+ cell yields. The cutoff value for the steady-state absolute lymphocyte count was 2.055 × 109/L. CONCLUSION To harvest greater numbers of CD34+ cells from second collections, male donors and those with intervals of longer than 9 months between donations should be selected. The lymphocyte counts prior to the first donations may predict the content of CD34+ cells in the allografts prepared using the second donations.
Collapse
Affiliation(s)
- Zhi-Ping Guo
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing 100044, China
| | - Tao Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing 100044, China; Department of Hematology, Shanxi Da Yi Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, China
| | - Lan-Ping Xu
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing 100044, China
| | - Xiao-Hui Zhang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing 100044, China
| | - Yu Wang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing 100044, China
| | - Xiao-Jun Huang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing 100044, China
| | - Ying-Jun Chang
- Peking University Institute of Hematology, Peking University People's Hospital, Beijing 100044, China; Collaborative Innovation Center of Hematology, Peking University, Beijing 100044, China.
| |
Collapse
|
25
|
Thurman-Newell JA, Petzing JN, Williams DJ. Quantification of biological variation in blood-based therapy--a summary of a meta-analysis to inform manufacturing in the clinic. Vox Sang 2015; 109:394-402. [PMID: 26174339 PMCID: PMC5016773 DOI: 10.1111/vox.12288] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/18/2015] [Accepted: 03/27/2015] [Indexed: 11/28/2022]
Abstract
Background and Objectives Biological raw materials, the basis for cellular therapies such as stem cells, have a significantly greater degree of complexity than their traditional pharmaceutical counterparts. This can be attributed to the inherent variation of its source – human beings. Currently, cell therapies are made in small, ad hoc batches, but larger scale production is a prerequisite to meeting future demand and will require a quality‐by‐design approach to manufacturing that will be designed around, or be robust to this variation. Quantification of variation will require understanding of the current baseline and stratification of its sources. Materials and Methods Haematopoietic stem cell therapy was chosen as a case study to explore this variation, and a PRISMA‐guided (Preferred Reporting Items for Systematic Reviews and Meta‐Analyses) systematic meta‐analysis was carried out for a number of predetermined cell measurements. Results From this data set, it appears that the extent of variation in therapeutic dose (in terms of transplanted total nucleated cells and CD34+ cells per kilogram) for HSCT is between one and four orders of magnitude of the median. Conclusions This is tolerated under the practice of medicine but would be unmanageable from a biomanufacturing perspective and raises concerns about comparable levels of efficacy and treatment. A number of sources that will contribute towards this variation are also reported, as is the direction of travel for 4 greater clarity of the scale of this challenge.
Collapse
Affiliation(s)
- J A Thurman-Newell
- Healthcare Engineering Group, Centre for Biological Engineering, Loughborough University, Loughborough, UK
| | - J N Petzing
- Healthcare Engineering Group, Centre for Biological Engineering, Loughborough University, Loughborough, UK
| | - D J Williams
- Healthcare Engineering Group, Centre for Biological Engineering, Loughborough University, Loughborough, UK
| |
Collapse
|
26
|
Goker H, Etgul S, Buyukasik Y. Optimizing mobilization strategies in difficult-to-mobilize patients: The role of plerixafor. Transfus Apher Sci 2015; 53:23-9. [PMID: 26099666 DOI: 10.1016/j.transci.2015.05.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Peripheral blood stem cell collection is currently the most widely used source for hematopoietic autologous transplantation. Several factors such as advanced age, previous chemotherapy, disease and marrow infiltration at the time of mobilization influence the efficacy of CD34(+) progenitor cell mobilization. Despite the safety and efficiency of the standard mobilization protocols (G-CSF ± chemotherapy), there is still a significant amount of mobilization failure rate (10-40%), which necessitate novel agents for effective mobilization. Plerixafor, is a novel agent, has been recently approved for mobilization of hematopoietic stem cells (HSCs). The combination of Plerixafor with G-CSF provides the collection of large numbers of stem cells in fewer apheresis sessions and can salvage those who fail with standard mobilization regimens. The development and optimization of practical algorithms for the use Plerixafor is crucial to make hematopoietic stem cell mobilization more efficient in a cost-effective way. This review is aimed at summarizing how to identify poor mobilizers, and define rational use of Plerixafor for planning mobilization in hard-to-mobilize patients.
Collapse
Affiliation(s)
- Hakan Goker
- Hematology Department, Hacettepe University School of Medicine, Ankara, Turkey.
| | - Sezgin Etgul
- Hematology Department, Hacettepe University School of Medicine, Ankara, Turkey
| | - Yahya Buyukasik
- Hematology Department, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
27
|
Deotare U, Al-Dawsari G, Couban S, Lipton JH. G-CSF-primed bone marrow as a source of stem cells for allografting: revisiting the concept. Bone Marrow Transplant 2015; 50:1150-6. [PMID: 25915812 DOI: 10.1038/bmt.2015.80] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/23/2022]
Abstract
The source of hematopoietic stem cells (HSCs) for allogeneic transplantation has evolved over the last decades, from the sole use of unstimulated bone marrow (BM) to the use of G-CSF (filgrastim)-mobilized peripheral blood, G-CSF-primed BM (G-BM) and cord blood. G-CSF-mobilized PBSC has replaced BM as the most commonly used source of allogeneic stem cells. G-BM is a source of HSCs, with studies demonstrating the safety and feasibility of this strategy with the potential for reducing GvHD, while retaining the speed of engraftment. Although the G-BM had lost its use as the optimal source of stem cells, after the widespread use of haploidentical transplantation, their use has resurfaced in 2010. This source can still be used in today's world of transplantation in aplastic anemia and other benign diseases, as well as in children donors. This study intends to review the evidence for this approach and whether this approach still has merit in the ever-evolving field of allogenic HSC transplantation. The merit of G-BM is its ability to offer speed of engraftment with reduced GvHD.
Collapse
Affiliation(s)
- U Deotare
- Allogeneic Bone Marrow Transplant Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - G Al-Dawsari
- Allogeneic Bone Marrow Transplant Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - S Couban
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - J H Lipton
- Allogeneic Bone Marrow Transplant Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Binsfeld M, Beguin Y, Belle L, Otjacques E, Hannon M, Briquet A, Heusschen R, Drion P, Zilberberg J, Bogen B, Baron F, Caers J. Establishment of a murine graft-versus-myeloma model using allogeneic stem cell transplantation. PLoS One 2014; 9:e113764. [PMID: 25415267 PMCID: PMC4240591 DOI: 10.1371/journal.pone.0113764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 10/29/2014] [Indexed: 11/18/2022] Open
Abstract
Background Multiple myeloma (MM) is a malignant plasma cell disorder with poor long-term survival and high recurrence rates. Despite evidence of graft-versus-myeloma (GvM) effects, the use of allogeneic hematopoietic stem cell transplantation (allo-SCT) remains controversial in MM. In the current study, we investigated the anti-myeloma effects of allo-SCT from B10.D2 mice into MHC-matched myeloma-bearing Balb/cJ mice, with concomitant development of chronic graft-versus-host disease (GvHD). Methods and results Balb/cJ mice were injected intravenously with luciferase-transfected MOPC315.BM cells, and received an allogeneic (B10.D2 donor) or autologous (Balb/cJ donor) transplant 30 days later. We observed a GvM effect in 94% of the allogeneic transplanted mice, as the luciferase signal completely disappeared after transplantation, whereas all the autologous transplanted mice showed myeloma progression. Lower serum paraprotein levels and lower myeloma infiltration in bone marrow and spleen in the allogeneic setting confirmed the observed GvM effect. In addition, the treated mice also displayed chronic GvHD symptoms. In vivo and in vitro data suggested the involvement of effector memory CD4 and CD8 T cells associated with the GvM response. The essential role of CD8 T cells was demonstrated in vivo where CD8 T-cell depletion of the graft resulted in reduced GvM effects. Finally, TCR Vβ spectratyping analysis identified Vβ families within CD4 and CD8 T cells, which were associated with both GvM effects and GvHD, whereas other Vβ families within CD4 T cells were associated exclusively with either GvM or GvHD responses. Conclusions We successfully established an immunocompetent murine model of graft-versus-myeloma. This is the first murine GvM model using immunocompetent mice that develop MM which closely resembles human MM disease and that are treated after disease establishment with an allo-SCT. Importantly, using TCR Vβ spectratyping, we also demonstrated the presence of GvM unique responses potentially associated with the curative capacity of this immunotherapeutic approach.
Collapse
Affiliation(s)
- Marilène Binsfeld
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Ludovic Belle
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Eléonore Otjacques
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Muriel Hannon
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Alexandra Briquet
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Roy Heusschen
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | | | - Jenny Zilberberg
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey, United States of America
| | - Bjarne Bogen
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- KG Jebsen centre for research on influenza vaccines, Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
29
|
Waller EK, Logan BR, Harris WAC, Devine SM, Porter DL, Mineishi S, McCarty JM, Gonzalez CE, Spitzer TR, Krijanovski OI, Linenberger ML, Woolfrey A, Howard A, Wu J, Confer DL, Anasetti C. Improved survival after transplantation of more donor plasmacytoid dendritic or naïve T cells from unrelated-donor marrow grafts: results from BMTCTN 0201. J Clin Oncol 2014; 32:2365-72. [PMID: 24982459 DOI: 10.1200/jco.2013.54.4577] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To characterize relationships between specific immune cell subsets in bone marrow (BM) or granulocyte colony-stimulating factor-mobilized peripheral blood (PB) stem cells collected from unrelated donors and clinical outcomes of patients undergoing transplantation in BMTCTN 0201. PATIENTS AND METHODS Fresh aliquots of 161 BM and 147 PB stem-cell allografts from North American donors randomly assigned to donate BM or PB stem cells and numbers of transplanted cells were correlated with overall survival (OS), relapse, and graft-versus-host disease (GvHD). RESULTS Patients with evaluable grafts were similar to all BMTCTN 0201 patients. The numbers of plasmacytoid dendritic cells (pDCs) and naïve T cells (Tns) in BM allografts were independently associated with OS in multivariable analyses including recipient and donor characteristics, such as human leukocyte antigen mismatch, age, and use of antithymocyte globulin. BM recipients of > median number of pDCs, naïve CD8(+) T cells (CD8Tns), or naïve CD4(+) T cells (CD4Tns) had better 3-year OS (pDCs, 56% v 35%; P = .025; CD8Tns, 56% v 37%; P = .012; CD4Tns, 55% v 37%; P = .009). Transplantation of more BM Tns was associated with less grade 3 to 4 acute GvHD but similar rates of relapse. Transplantation of more BM pDCs was associated with fewer deaths resulting from GvHD or from graft rejection. Analysis of PB grafts did not identify a donor cell subset significantly associated with OS, relapse, or GvHD. CONCLUSION Donor immune cells in BM but not PB stem-cell grafts were associated with survival after unrelated-donor allogeneic hematopoietic stem-cell transplantation. The biologic activity of donor immune cells in allogeneic transplantation varied between graft sources. Donor grafts with more BM-derived Tns and pDCs favorably regulated post-transplantation immunity in allogeneic hematopoietic stem-cell transplantation.
Collapse
Affiliation(s)
- Edmund K Waller
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL.
| | - Brent R Logan
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Wayne A C Harris
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Steven M Devine
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - David L Porter
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Shin Mineishi
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - John M McCarty
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Corina E Gonzalez
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Thomas R Spitzer
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Oleg I Krijanovski
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Michael L Linenberger
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Ann Woolfrey
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Alan Howard
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Juan Wu
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Dennis L Confer
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Claudio Anasetti
- Edmund K. Waller and Wayne A.C. Harris, Emory University, Atlanta, GA; Brent R. Logan, Medical College of Wisconsin, Milwaukee, WI; Steven M. Devine, Ohio State University, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Shin Mineishi, University of Alabama at Birmingham, Birmingham, AL; John M. McCarty, Medical College of Virginia, Richmond, VA; Corina E. Gonzalez, Georgetown University Hospital, Washington, DC; Thomas R. Spitzer, Massachusetts General Hospital, Boston, MA; Oleg I. Krijanovski, Sutter East Bay Medical Foundation, Berkeley, CA; Michael L. Linenberger and Ann Woolfrey, Fred Hutchinson Cancer Research Center, Seattle, WA; Alan Howard and Dennis L. Confer, National Marrow Donor Program, Minneapolis, MN; Juan Wu, EMMES Corporation, Rockville, MD; and Claudio Anasetti, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
30
|
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]
|
31
|
Human umbilical cord blood mononuclear cells in a double-hit model of bronchopulmonary dysplasia in neonatal mice. PLoS One 2013; 8:e74740. [PMID: 24069341 PMCID: PMC3778007 DOI: 10.1371/journal.pone.0074740] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/02/2013] [Indexed: 02/01/2023] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) presents a major threat of very preterm birth and treatment options are still limited. Stem cells from different sources have been used successfully in experimental BPD, induced by postnatal hyperoxia. Objectives We investigated the effect of umbilical cord blood mononuclear cells (MNCs) in a new double-hit mouse model of BPD. Methods For the double-hit, date mated mice were subjected to hypoxia and thereafter the offspring was exposed to hyperoxia. Human umbilical cord blood MNCs were given intraperitoneally by day P7. As outcome variables were defined: physical development (auxology), lung structure (histomorphometry), expression of markers for lung maturation and inflammation on mRNA and protein level. Pre- and postnatal normoxic pups and sham treated double-hit pups served as control groups. Results Compared to normoxic controls, sham treated double-hit animals showed impaired physical and lung development with reduced alveolarization and increased thickness of septa. Electron microscopy revealed reduced volume density of lamellar bodies. Pulmonary expression of mRNA for surfactant proteins B and C, Mtor and Crabp1 was reduced. Expression of Igf1 was increased. Treatment with umbilical cord blood MNCs normalized thickness of septa and mRNA expression of Mtor to levels of normoxic controls. Tgfb3 mRNA expression and pro-inflammatory IL-1β protein concentration were decreased. Conclusion The results of our study demonstrate the therapeutic potential of umbilical cord blood MNCs in a new double-hit model of BPD in newborn mice. We found improved lung structure and effects on molecular level. Further studies are needed to address the role of systemic administration of MNCs in experimental BPD.
Collapse
|
32
|
Servais S, Beguin Y, Baron F. Emerging drugs for prevention of graft failure after allogeneic hematopoietic stem cell transplantation. Expert Opin Emerg Drugs 2013; 18:173-92. [DOI: 10.1517/14728214.2013.798642] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
33
|
Extensive chronic GVHD is associated with donor blood CD34+ cell count after G-CSF mobilization in non-myeloablative allogeneic PBSC transplantation. Bone Marrow Transplant 2012; 47:1564-8. [PMID: 22609881 DOI: 10.1038/bmt.2012.75] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The correlation between the incidence of GVHD and the number of infused CD34(+) cells remains controversial for PBSC transplantation after a reduced-intensity-conditioning (RIC) regimen. We evaluated 99 patients transplanted with an HLA-identical sibling after the same RIC (2-Gy-TBI/fludarabine). Donor and recipient characteristics, donor's blood G-CSF-mobilized CD34(+) cell count, and number of infused CD34(+) and CD3(+) cells were analyzed as risk factors for acute and chronic GVHD There was a trend for an increased incidence of extensive chronic GVHD in the quartile of patients receiving more than 10 × 10(6) CD34(+) cells/kg (P = 0.05). Interestingly, the number of donor's blood CD34(+) cells at day 5 of G-CSF mobilization was closely associated with the incidence of extensive chronic GVHD, that is, 48% (95% CI: 28-68) at 24-months in the quartile of patients whose donors had the highest CD34(+) cell counts versus 24.3% (95% CI: 14-34) in the other patients (P = 0.007). In multivariate analysis, the only factor correlating with extensive chronic GVHD (cGVHD) was the donor's blood CD34(+) cell count after G-CSF (HR 2.49; 95% CI: 1.16-5.35, P = 0.019). This study shows that the incidence of cGVHD is more strongly associated with the donor's ability to mobilize CD34(+) cells than with the number of infused CD34(+) cells.
Collapse
|
34
|
Abstract
This review evaluates the latest information on the mobilisation of haemopoietic stem cells for transplantation, with the focus on what is the current best practice and how new understanding of the bone marrow stem cell niche provides new insights into optimising mobilisation regimens. The review then looks at the mobilisation of mesenchymal stromal cells, immune cells as well as malignant cells and what clinical implications there are.
Collapse
|
35
|
Abstract
Abstract
Transplantation with 2-5 × 106 mobilized CD34+cells/kg body weight lowers transplantation costs and mortality. Mobilization is most commonly performed with recombinant human G-CSF with or without chemotherapy, but a proportion of patients/donors fail to mobilize sufficient cells. BM disease, prior treatment, and age are factors influencing mobilization, but genetics also contributes. Mobilization may fail because of the changes affecting the HSC/progenitor cell/BM niche integrity and chemotaxis. Poor mobilization affects patient outcome and increases resource use. Until recently increasing G-CSF dose and adding SCF have been used in poor mobilizers with limited success. However, plerixafor through its rapid direct blockage of the CXCR4/CXCL12 chemotaxis pathway and synergy with G-CSF and chemotherapy has become a new and important agent for mobilization. Its efficacy in upfront and failed mobilizers is well established. To maximize HSC harvest in poor mobilizers the clinician needs to optimize current mobilization protocols and to integrate novel agents such as plerixafor. These include when to mobilize in relation to chemotherapy, how to schedule and perform apheresis, how to identify poor mobilizers, and what are the criteria for preemptive and immediate salvage use of plerixafor.
Collapse
|
36
|
Implications of CD34+ cell dose on clinical and haematological outcome of allo-SCT for acquired aplastic anaemia. Bone Marrow Transplant 2009; 45:886-94. [DOI: 10.1038/bmt.2009.267] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Abstract
PURPOSE OF REVIEW Granulocyte colony-stimulating factor-mobilized peripheral blood stem cells are widely used to reconstitute hematopoiesis; however, preclinical and clinical studies show that improvements to this mobilization can be achieved. We discuss the development of new mobilizing regimens and evaluation of new findings on mobilized stem cell populations that may improve the utility and convenience of peripheral blood stem cell transplant. RECENT FINDINGS Chemokines and their receptors regulate leukocyte trafficking, and altering chemokine signaling pathways mobilizes stem cells. In recent trials, combination use of the chemokine (C-X-C motif) receptor 4 antagonist AMD3100 and granulocyte colony-stimulating factor mobilized more CD34 cells in fewer days than granulocyte colony-stimulating factor alone and allowed more patients to proceed to autotransplant. In preclinical studies the chemokine GRObeta synergizes with granulocyte colony-stimulating factor and when used alone or with granulocyte colony-stimulating factor mobilizes more primitive hematopoietic stem cells with less apoptosis, higher integrin activation, lower CD26 expression and enhanced marrow homing compared with granulocyte colony-stimulating factor. Hematopoietic stem cells mobilized by GRObeta or AMD3100 demonstrate superior engraftment and contribution to chimerism in primary and secondary transplant studies in mice, and peripheral blood stem cells mobilized by AMD3100 and granulocyte colony-stimulating factor in patients demonstrate enhanced engraftment capabilities in immunodeficient mice. SUMMARY Alternate regimens differentially mobilize stem cell populations with unique intrinsic properties with the potential to expand the utility of hematopoietic transplantation. Continued mechanistic evaluation will be critical to our understanding of mechanisms of mobilization and their use in regenerative medicine.
Collapse
Affiliation(s)
- Louis M Pelus
- Department of Microbiology and Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
38
|
Robin M, Kernèis S, Porcher R, de Latour RP, Traineau R, Ribaud P, Rocha V, Devergie A, Marolleau JP, Benburan M, Socié G, Larghero J. Influence of bone marrow nucleated red blood cell dose on outcome after allogeneic haematopoietic stem cell transplantation. Br J Haematol 2008; 140:725-7. [PMID: 18302719 DOI: 10.1111/j.1365-2141.2008.07009.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
39
|
Cashen AF, Lazarus HM, Devine SM. Mobilizing stem cells from normal donors: is it possible to improve upon G-CSF? Bone Marrow Transplant 2007; 39:577-88. [PMID: 17369869 DOI: 10.1038/sj.bmt.1705616] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Currently, granulocyte colony stimulating factor (G-CSF) remains the standard mobilizing agent for peripheral blood stem cell (PBSC) donors, allowing the safe collection of adequate PBSCs from the vast majority of donors. However, G-CSF mobilization can be associated with some significant side effects and requires a multi-day dosing regimen. The other cytokine approved for stem cell mobilization, granulocyte-macrophage colony stimulating factor (GM-CSF), alters graft composition and may reduce the development of graft-versus-host disease, but a significant minority of donors fails to provide sufficient CD34+ cells with GM-CSF and some experience unacceptable toxicity. AMD3100 is a promising new mobilizing agent, which may have several advantages over G-CSF for donor mobilization. As it is a direct antagonist of the interaction between the chemokine stromal-derived factor-1 and its receptor CXCR4, AMD3100 mobilizes PBSCs within hours rather than days. It is also well tolerated, with no significant side effects reported in any of the clinical trials to date. Studies of autologous and allogeneic transplantation of AMD3100 mobilized grafts have demonstrated prompt and stable engraftment. Here, we review the current state of stem cell mobilization in normal donors and discuss novel strategies for donor stem cell mobilization.
Collapse
Affiliation(s)
- A F Cashen
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | | |
Collapse
|
40
|
Dhédin N, Chamakhi I, Perreault C, Roy DC, Sauvageau G, Ducruet T, Busque L, Fish D, Bélanger R, Roy J. Evidence that donor intrinsic response to G-CSF is the best predictor of acute graft-vs-host disease following allogeneic peripheral blood stem cell transplantation. Exp Hematol 2006; 34:107-14. [PMID: 16413397 DOI: 10.1016/j.exphem.2005.09.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Revised: 09/19/2005] [Accepted: 09/19/2005] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Risk factors of acute graft-vs-host disease (GVHD) following allogeneic bone marrow transplantation have been well described before. In this study, we tested the hypothesis that acute GVHD after allogeneic peripheral blood stem cell (PBSC) transplant might be associated with donors' responsiveness to granulocyte colony-stimulating factor (G-CSF), rather than the dose of CD34(+) cells infused. PATIENTS AND METHODS We retrospectively analyzed mobilization and transplant data (demographic characteristics, donor blood cell subsets after G-CSF, graft composition) in 149 consecutive HLA-identical donor/recipient pairs in order to identify acute GVHD risk factors. RESULTS In 25% of donors, G-CSF mobilization led to an outstanding response, defined as greater than 117 x 10(6) CD34(+) cells/L. Overall, incidence of grades II-IV acute GVHD was 20.1% (95% CI: 16.6-23.6). Following univariate analysis, the incidence increased significantly in recipients receiving greater than 10 x 10(6) CD34(+) cells/kg (35% vs 15%; p = 0.007), and those transplanted from outstanding mobilizers (41% vs 12%, p < 0.0001). In multivariate analysis, only transplantation from outstanding mobilizers remained significant (p = 0.02). Donor or recipient demographic characteristics and lymphocyte subsets reinfused in the graft had no impact. CONCLUSIONS We demonstrate for the first time that donor responsiveness to G-CSF is associated with acute GVHD following PBSC transplantation. If confirmed, this correlation will help to identify recipients who could potentially benefit from improved prophylaxis. As further corollary, decreasing the dose of CD34(+) cells infused is unlikely to prevent acute GVHD. Future studies should focus on the molecular bases of interindividual discrepancies in response to G-CSF.
Collapse
Affiliation(s)
- Nathalie Dhédin
- Division of Hematology, Hôpital Maisonneuve-Rosemont, and Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Li K, Chuen CKY, Lee SM, Law P, Fok TF, Ng PC, Li CK, Wong D, Merzouk A, Salari H, Gu GJS, Yuen PMP. Small peptide analogue of SDF-1alpha supports survival of cord blood CD34+ cells in synergy with other cytokines and enhances their ex vivo expansion and engraftment into nonobese diabetic/severe combined immunodeficient mice. Stem Cells 2005; 24:55-64. [PMID: 16123381 DOI: 10.1634/stemcells.2005-0082] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The SDF-1/CXCR4 axis has been implicated in the chemotaxis, homing, mobilization, and expansion of hematopoietic stem and progenitor cells. We studied the effects of a SDF-1 peptide analogue CTCE-0214 on the survival of cord blood CD34+ cells in culture, expansion, and engraftment of expanded cells in the nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model. Our results demonstrated that CTCE-0214 synergized with thrombopoietin (TPO), stem cell factor (SCF), or flt-3 ligand (FL) on the survival of stem and progenitor cells in culture. Adding CTCE-0214 at a low concentration (0.01 ng/ml) for 4 days together with TPO, SCF, and FL significantly enhanced ex vivo expansion of CD34+ cells to subsets of primitive (CD34+CD38- cells, colony-forming unit-mixed [CFU-GEMMs]), erythroid (CFU-Es), myeloid (CFU-GMs), and megakaryocytic (CD61+CD41+ cells, CFU-MKs) progenitors, as well as their multilineage engraftment in NOD/SCID mice. Interestingly, the short exposure of expanded cells to CTCE-0214 (100 and 500 ng/ml) for 4 hours did not increase the quantity of progenitor cells but enhanced their engraftment capacity. The proportion of CD34+ cells expressing surface CXCR4 was decreased, but the overall number of this population increased upon expansion. The small peptide analogue of SDF-1 could be developed for ex vivo expansion and improving engraftment of cord blood transplantation.
Collapse
Affiliation(s)
- Karen Li
- Department of Paediatrics, The Chinese University of Hong Kong, 6th Floor, Clinical Sciences Block, Prince of Wales Hospital, Shatin, NT, Hong Kong, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Devine SM, Brown RA, Mathews V, Trinkaus K, Khoury H, Adkins D, Vij R, Sempek D, Graubert T, Tomasson M, Goodnough LT, DiPersio JF. Reduced risk of acute GVHD following mobilization of HLA-identical sibling donors with GM-CSF alone. Bone Marrow Transplant 2005; 36:531-8. [PMID: 16025152 DOI: 10.1038/sj.bmt.1705091] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We retrospectively reviewed the results of transplanting peripheral blood progenitor cell (PBPC) allografts from HLA-matched sibling donors mobilized using various hematopoietic cytokines. Patients had received allografts mobilized with Granulocyte colony-stimulating factor (G-CSF) (G, N = 65) alone, G plus Granulocyte-macrophage colony stimulating factor (GM-CSF) (G/GM, N = 70), or GM-CSF alone at 10 or 15 microg/kg/day (GM, N = 10 at 10 microg/kg/day and 21 at 15 microg/kg/day). The CD34+ and CD3+ cell content of grafts were significantly lower following GM alone compared to G alone (P < 0.001 and 0.04, respectively). Nonhematopoietic toxicity observed in donors precluded dose escalation of GM-CSF beyond 10 microg/kg/day. Hematopoietic recovery was similar among all three groups. Grades II-IV acute graft-versus-host disease (GVHD) was observed in only 13% of patients in the GM alone group compared to 49 and 69% in the G alone or G/GM groups, respectively (P < 0.001). In a multivariate analysis, receipt of PBPC mobilized with GM alone was associated with a lower risk of grades II-IV acute GVHD (hazard ratio 0.21; 95% CI 0.073, 0.58) compared to G alone or G/GM. There were no differences in relapse risk or overall survival among the groups. Donor PBPC grafts mobilized with GM-CSF alone result in prompt hematopoietic engraftment despite lower CD34+ cell doses and may reduce the risk of grades II-IV acute GVHD following HLA-matched PBPC transplantation.
Collapse
Affiliation(s)
- S M Devine
- Siteman Cancer Center and Department of Medicine, Division of Oncology, Section of Stem Cell Transplantation, Leukemia, and Stem Cell Biology, Washington University School of Medicine, St Louis, MO, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Baron F, Maris MB, Storer BE, Sandmaier BM, Panse JP, Chauncey TR, Sorror M, Little MT, Maloney DG, Storb R, Heimfeld S. High doses of transplanted CD34+ cells are associated with rapid T-cell engraftment and lessened risk of graft rejection, but not more graft-versus-host disease after nonmyeloablative conditioning and unrelated hematopoietic cell transplantation. Leukemia 2005; 19:822-8. [PMID: 15772701 DOI: 10.1038/sj.leu.2403718] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This report examines the impact of graft composition on outcomes in 130 patients with hematological malignancies given unrelated donor granulocyte-colony-stimulating-factor-mobilized peripheral blood mononuclear cells (G-PBMC) (n = 116) or marrow (n = 14) transplantation after nonmyeloablative conditioning with 90 mg/m(2) fludarabine and 2 Gy TBI. The median number of CD34(+) cells transplanted was 6.5 x 10(6)/kg. Higher numbers of grafted CD14(+) (P = 0.0008), CD3(+) (P = 0.0007), CD4(+) (P = 0.001), CD8(+) (P = 0.004), CD3(-)CD56(+) (P = 0.003), and CD34(+) (P = 0.0001) cells were associated with higher levels of day 28 donor T-cell chimerism. Higher numbers of CD14(+) (P = 0.01) and CD34(+) (P = 0.0003) cells were associated with rapid achievement of complete donor T-cell chimerism, while high numbers of CD8(+) (P = 0.005) and CD34(+) (P = 0.01) cells were associated with low probabilities of graft rejection. When analyses were restricted to G-PBMC recipients, higher numbers of grafted CD34(+) cells were associated with higher levels of day 28 donor T-cell chimerism (P = 0.01), rapid achievement of complete donor T-cell chimerism (P = 0.02), and a trend for lower risk for graft rejection (P = 0.14). There were no associations between any cell subsets and acute or chronic GVHD nor relapse/progression. These data suggest more rapid engraftment of donor T cells and reduced rejection rates could be achieved by increasing the doses of CD34(+) cells in unrelated grafts administered after nonmyeloablative conditioning.
Collapse
Affiliation(s)
- F Baron
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cao TM, Shizuru JA, Wong RM, Sheehan K, Laport GG, Stockerl-Goldstein KE, Johnston LJ, Stuart MJ, Grumet FC, Negrin RS, Lowsky R. Engraftment and survival following reduced-intensity allogeneic peripheral blood hematopoietic cell transplantation is affected by CD8+ T-cell dose. Blood 2004; 105:2300-6. [PMID: 15572597 DOI: 10.1182/blood-2004-04-1473] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The influence of graft composition on clinical outcomes after reduced-intensity conditioning is not well-characterized. In this report we prospectively enumerated CD34+, CD3+, CD4+, and CD8+ cell doses in granulocyte colony-stimulating factor-mobilized peripheral blood mononuclear cell (G-PBMC) allografts in 63 patients who received transplants following non-myeloablative conditioning with total body irradiation 200 cGy plus fludarabine as treatment for malignant diseases. Donors were HLA-identical siblings (n = 38) or HLA-matched unrelated individuals (n = 25). By univariate analyses G-PBMC CD8+ T-cell dose in at least the 50th percentile favorably correlated with full donor blood T-cell chimerism (P = .03), freedom from progression (P = .001), and overall survival (P = .01). No G-PBMC cell dose influenced grade II to IV acute or extensive chronic graft-versus-host disease. In multivariate analysis only G-PBMC CD8+ T-cell dose (P = .003; RR = 0.2, 95% CI = 0.1-0.6) was associated with improved freedom from progression. Infusion of low G-PBMC CD8+ T-cell dose for reduced-intensity allografting may adversely affect T-cell engraftment and survival outcome.
Collapse
Affiliation(s)
- Thai M Cao
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University Schol of Medicine, 300 Pasteur Dr, H3249, MC 5623, Stanford, CA 94305-5623, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Novel strategies for hematopoietic stem cell mobilization. Curr Opin Organ Transplant 2004. [DOI: 10.1097/00075200-200403000-00012] [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]
|