1
|
Prisciandaro M, Santinelli E, Tomarchio V, Tafuri MA, Bonchi C, Palazzo G, Nobile C, Marinucci A, Mele M, Annibali O, Rigacci L, Vacca M. Stem Cells Collection and Mobilization in Adult Autologous/Allogeneic Transplantation: Critical Points and Future Challenges. Cells 2024; 13:586. [PMID: 38607025 PMCID: PMC11011310 DOI: 10.3390/cells13070586] [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: 02/05/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Achieving successful hematopoietic stem cell transplantation (HSCT) relies on two fundamental pillars: effective mobilization and efficient collection through apheresis to attain the optimal graft dose. These cornerstones pave the way for enhanced patient outcomes. The primary challenges encountered by the clinical unit and collection facility within a transplant program encompass augmenting mobilization efficiency to optimize the harvest of target cell populations, implementing robust monitoring and predictive strategies for mobilization, streamlining the apheresis procedure to minimize collection duration while ensuring adequate yield, prioritizing patient comfort by reducing the overall collection time, guaranteeing the quality and purity of stem cell products to optimize graft function and transplant success, and facilitating seamless coordination between diverse entities involved in the HSCT process. In this review, we aim to address key questions and provide insights into the critical aspects of mobilizing and collecting hematopoietic stem cells for transplantation purposes.
Collapse
Affiliation(s)
- Michele Prisciandaro
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Enrico Santinelli
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
- Program in Immunology, Molecular Medicine and Applied Biotechnologies, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Tomarchio
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Maria Antonietta Tafuri
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Cecilia Bonchi
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Gloria Palazzo
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Carolina Nobile
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Alessandra Marinucci
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| | - Marcella Mele
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Ombretta Annibali
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Luigi Rigacci
- Operative Research Unit of Hematology and Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (E.S.); (V.T.); (M.A.T.); (M.M.); (O.A.); (L.R.)
| | - Michele Vacca
- Operative Research Unit of Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy; (C.B.); (G.P.); (C.N.); (A.M.)
| |
Collapse
|
2
|
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
|
3
|
Kharya G, Sapkota S, Teotia N, Chaudhary M, Swathymon KK, Chodan P, Peters S, Anthony A, Sharma B, Yadav H, Bakane A, Joseph M. Thiotepa-based reduced toxicity conditioning in combination with post-transplant cyclophosphamide and mTOR inhibitor for heavily transfused acquired severe aplastic anemia in children and young adults: encouraging outcomes of a pilot study. Bone Marrow Transplant 2023; 58:233-236. [PMID: 36434270 DOI: 10.1038/s41409-022-01876-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Gaurav Kharya
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India.
| | - Sudhir Sapkota
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Neeraj Teotia
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Mohit Chaudhary
- Department of Transfusion Medicine, Indraprastha Apollo Hospitals, Delhi, India
| | - K K Swathymon
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Pema Chodan
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Sherin Peters
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Anju Anthony
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Bharti Sharma
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Himshikha Yadav
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Atish Bakane
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Manju Joseph
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, 110076, India
| |
Collapse
|
4
|
Zhuang L, Lauro D, Wang S, Yuan S. Addition of plerixafor in poorly mobilized allogeneic stem cell donors. J Clin Apher 2022; 37:388-394. [PMID: 35633513 PMCID: PMC9539930 DOI: 10.1002/jca.21992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 12/02/2022]
Abstract
Background Peripheral blood stem cells (PBSCs) are the predominant graft source for adult allogeneic hematopoietic stem cell transplantation (HSCT). In poorly mobilized autologous donors, plerixafor improves collection outcomes. We examine plerixafor use in allogeneic donors who mobilize poorly with granulocyte colony‐stimulating factor (G‐CSF) in those who are healthy and those with pre‐existing medical conditions, and determine the optimal threshold to add plerixafor. Study Design/Methods We retrospectively examined all allogeneic PBSC collections from January 2013 to October 2020 at our center. Donors received G‐CSF 10 mcg/kg daily for 4 days before undergoing apheresis collection on day 5. Plerixafor was added based on poor CD34+ cell collection yield after the first or second collection day. Results Of the 1008 allogeneic donors, 41 (4.1%) received one dose of plerixafor in addition to G‐CSF due to poor collection yield. After starting plerixafor there was a 0.75‐ to 7.74‐fold (median 2.94) increase in CD34+ yield from the previous day. No donors with G‐CSF‐only mobilization who collected <2.0 × 106 CD34+ cells/kg recipient weight on day one achieved the goal of ≥4.0 × 106 CD34+ cells/kg recipient weight total over 2 days but 59.2% of donors who used rescue plerixafor did. Conclusion Donors both healthy and those with pre‐existing disease responded well to plerixafor with minimal side effects. If the first‐day collection yield is less than ~63% of the collection goal, addition of plerixafor may be necessary to reach the collection goal and limit the number of collection days in allogeneic donors.
Collapse
Affiliation(s)
- Lefan Zhuang
- Division of Transfusion Medicine, Department of Pathology, City of Hope National Medical Center, Duarte, California, USA
| | - Deisen Lauro
- Division of Transfusion Medicine, Department of Pathology, City of Hope National Medical Center, Duarte, California, USA
| | - Shirong Wang
- Division of Transfusion Medicine, Department of Pathology, City of Hope National Medical Center, Duarte, California, USA
| | - Shan Yuan
- Division of Transfusion Medicine, Department of Pathology, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
5
|
Kurnikova E, Trakhtman P, Balashov D, Garloeva J, Kumukova I, Khismatullina R, Pershin D, Shelikhova L, Novichkova G, Maschan A. Efficacy and safety of a reduced dose of plerixafor in combination with granulocyte colony-stimulating factor in healthy haploidentical stem cell donors. Vox Sang 2022; 117:853-861. [PMID: 35332550 DOI: 10.1111/vox.13266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/14/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Implementation of the technique of immunomagnetic selection requires the procurement of a large number of CD34+ cells from haploidentical donors within a single apheresis procedure. The release of stem cells with granulocyte colony-stimulating factor (G-CSF) alone is unsatisfactory in a number of donors, and plerixafor, a CXCR4 chemokine receptor antagonist, could be used as an additional mobilization agent. The aim of our study was to examine whether a lower dose of plerixafor (0.12 mg/kg) can provide sufficient increase in CD34+ cells in the peripheral blood of allogeneic healthy donors in comparison with a historical control group. In addition, we assessed the risk of inability to provide the recipient with a transplant containing the optimal dose of 8-10 × 106 CD34+ cells/kg body weight of the recipient. MATERIALS AND METHODS In a prospective, single-arm study, we examined the results of 105 mobilizations in healthy adult haploidentical donors with G-CSF and plerixafor at a dose of 0.12 mg/kg. The historical control group consisted of 106 mobilizations with G-CSF and plerixafor at 0.24 mg/kg. RESULTS The median increase in the number of CD34+ cells from day 4 to day 5 of mobilization was 69 cells/μl (range, 28-240) versus 77 cells/μl (24-217) in the groups of 0.12 and 0.24 mg/kg of plerixafor, respectively (p-value 0.255). The apheresis products contained a median of 14.4 × 106 /kg recipient body weight CD34+ cells versus 12.9 × 106 /kg in the groups that received 0.12 and 0.24 mg/kg of plerixafor, respectively (p-value 0.118). The obtained differences were not significant, which means the application of a decreased dose of plerixafor did not affect the results of mobilization. CONCLUSION The obtained differences in collection were not significant, and thus the application of a decreased dose of plerixafor did not affect the results of mobilization.
Collapse
Affiliation(s)
- Elena Kurnikova
- Transfusion Medicine Service, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Pavel Trakhtman
- Transfusion Medicine Service, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Dmitry Balashov
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Juliya Garloeva
- Transfusion Medicine Service, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Irina Kumukova
- Transfusion Medicine Service, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Rimma Khismatullina
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Dmitriy Pershin
- Transplantation Immunology and Immunotherapy Laboratory, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Larisa Shelikhova
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Galina Novichkova
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexey Maschan
- Department of Hematopoietic Stem Cell Transplantation, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
6
|
HLA-haploidentical TCRαβ+/CD19+-depleted stem cell transplantation in children and young adults with Fanconi anemia. Blood Adv 2021; 5:1333-1339. [PMID: 33656536 DOI: 10.1182/bloodadvances.2020003707] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
We report on the outcome of 24 patients with Fanconi anemia (FA) lacking an HLA matched related or unrelated donor, given an HLA-haploidentical T-cell receptor αβ (TCRαβ+) and CD19+ cell-depleted hematopoietic stem cell transplantation (HSCT) in the context of a prospective, single-center phase 2 trial. Sustained primary engraftment was achieved in 22 (91.6%) of 24 patients, with median time to neutrophil recovery of 12 days (range, 9-15 days) and platelet recovery of 10 days (range, 7-14 days). Cumulative incidences of grade 1 to 2 acute graft-versus-host disease (GVHD) and chronic GVHD were 17.4% (95% confidence interval [CI], 5.5%-35.5%) and 5.5% (95% CI, 0.8%-33.4%), respectively. The conditioning regimen, which included fludarabine, low-dose cyclophosphamide and, in most patients, single-dose irradiation was well tolerated; no fatal transplant-related toxicity was observed. With a median follow-up of 5.2 years (range, 0.3-8.7 years), the overall and event-free survival probabilities were 100% and 86.3% (95% CI, 62.8%-95.4%), respectively (2 graft failures and 1 case of poor graft function were considered as events). The 2 patients who experienced primary graft failure underwent a subsequent successful HSCT from the other parent. This is the first report of FA patients given TCRαβ+/CD19+-depleted haplo-HSCT in the context of a prospective trial, and the largest series of T-cell-depleted haplo-HSCT in FA reported to date. This trial was registered at www.clinicaltrials.gov as #NCT01810120.
Collapse
|
7
|
Bakane A, Rauthan A, Madasu A, Kharya G. Successful T-cell-depleted haploidentical hematopoietic stem cell transplant for a novel NF-E2 mutation causing inherited thrombocytopenia. Pediatr Blood Cancer 2021; 68:e28792. [PMID: 33283958 DOI: 10.1002/pbc.28792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Atish Bakane
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, India
| | - Archana Rauthan
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, India
| | - Anjan Madasu
- Pediatric Hematology Oncology Unit, Dubai Hospital, Dubai, UAE
| | - Gaurav Kharya
- Center for Bone Marrow Transplant and Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, India
| |
Collapse
|
8
|
Kurnikova E, Trakhtman P, Pershin D, Ilyushina M, Khismatullina R, Maschan M, Novichkova G, Maschan A. Plerixafor added to G-CSF allows mobilization of a sufficient number of hematopoietic progenitors without impacting the efficacy of TCR-alpha/beta depletion in pediatric haploidentical and genoidentical donors failing to mobilize with G-CSF alone. J Clin Apher 2021; 36:547-552. [PMID: 33682959 DOI: 10.1002/jca.21891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/21/2020] [Accepted: 02/22/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND Collection of a large number of early hematopoietic progenitors is essential for allogeneic apheresis products intended for TCR-alpha/beta depletion. MATERIALS AND METHODS We added plerixafor 0.24 mg/kg body weight (bw) on day 4 of high-dose filgrastim mobilization 10 hours prior to apheresis in 16 (30.5%) pediatric allogeneic donors who failed to recover a sufficient number of CD34+ cells. RESULTS On day 4 of G-CSF, the median CD34+ cell count in peripheral blood was 6 per μL (range 4-9 per μL) in 6 poor mobilizers and 16 per μL (range 12-19 per μL) in insufficient mobilizers. In all donors, the threshold of 50 CD34+ cells/μL was achieved, and the median increase was 14.8-fold in poor mobilizers and 6.5-fold in insufficient mobilizers, whereas it was 3.45-fold increase in those mobilized with G-CSF alone. DISCUSSION In all donors, a predefined number of >10 × 106 CD34+ cells/kg of recipient bw before depletion was reached in the apheresis product. The use of plerixafor did not affect the purity of further TCR-alpha/beta depletion. Side effects were mild to moderate and consisted of nausea and vomiting. CONCLUSION Thus, the safety and high efficacy of plerixafor was proven in healthy pediatric allogeneic hematopoietic cell donors.
Collapse
Affiliation(s)
- Elena Kurnikova
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Pavel Trakhtman
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Dmitry Pershin
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Mariya Ilyushina
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Rimma Khismatullina
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Michael Maschan
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Galina Novichkova
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| | - Alexey Maschan
- Dmitri Rogachev National Research Centre for Pediatric Hematology, Oncology and Immunology, Ministry of Health of Russian Federation, Moscow, Russia
| |
Collapse
|
9
|
Zubicaray J, Galvez E, Sebastian E, Molina B, González-Vicent M, Castillo A, Ramírez M, Madero L, Díaz MA, Sevilla J. Plerixafor-based mobilization in pediatric healthy donors with unfavorable donor/recipient body weight ratio resulted in a better CD34 + collection yield: A retrospective analysis. J Clin Apher 2020; 36:78-86. [PMID: 33079424 DOI: 10.1002/jca.21844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/12/2020] [Accepted: 09/15/2020] [Indexed: 01/04/2023]
Abstract
INTRODUCTION In order to propose risk-adapted mobilization algorithms, several authors have tried to look for predictive factors of the CD34+ yield in healthy pediatric donors. Donor recipient body weight ratio (D/R ratio) was identified as one of the main variables related with the success to achieve the target cell dose for transplantation. According to this variable we modified the mobilization schedule. MATERIAL AND METHODS We report the results of 46 mobilizations and apheresis procedures performed in our center with unfavorable D/R ratio. Mobilization was attempted by the standard regime of G-CSF (10 mcg/kg/24 hours) in 28 cases (60.9%), with high dose G-CSF (10 mcg/kg/12 hours) in 9 cases (19.6%), and with plerixafor and G-CSF single dose regime in 9 cases (19.6%). RESULTS CD34+ cell quantification before apheresis is closely related to CD34+ yield, being the only factor related to collected CD34+ cells (beta .71; P < .0001). The mobilization efficiency was higher in plerixafor group compared to the other two schedules (P < .0001). By using plerixafor for mobilization, we achieved the target CD34+ cell dose of ≥2 × 106 /kg per recipient body weight in all cases with unfavorable D/R ratio. It was observed that 17.4% of cases that not reached the established target cell dose were located in the standard or high-dose mobilization regimes. This difference is even greater for optimal collections (≥5 × 106 /kg), since of the 54.3% cases that did not reach this goal none was mobilized by plerixafor. CONCLUSION Tailoring the mobilization regime we can reach the target cell dose, even in those cases with the worst D/R ratio.
Collapse
Affiliation(s)
- Josune Zubicaray
- Hematology y Hemotherapy Unit, Hematología y Oncología Pediátricas, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Eva Galvez
- Hematology y Hemotherapy Unit, Hematología y Oncología Pediátricas, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Elena Sebastian
- Hematology y Hemotherapy Unit, Hematología y Oncología Pediátricas, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Blanca Molina
- Hematopoietic Transplant Unit, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Marta González-Vicent
- Hematopoietic Transplant Unit, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Ana Castillo
- Laboratorio Hematología y Oncología, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Manuel Ramírez
- Laboratorio Hematología y Oncología, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Luis Madero
- Hematology y Hemotherapy Unit, Hematología y Oncología Pediátricas, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Hematopoietic Transplant Unit, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Laboratorio Hematología y Oncología, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Miguel Angel Díaz
- Hematopoietic Transplant Unit, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Julian Sevilla
- Hematology y Hemotherapy Unit, Hematología y Oncología Pediátricas, Fundación para la Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| |
Collapse
|
10
|
Hölig K, Schmidt H, Hütter G, Kramer M, Teipel R, Heidrich K, Zimmer K, Heidenreich F, Blechschmidt M, Torosian T, Ordemann R, Kroschinsky F, Rücker-Braun E, Gopsca L, Wagner-Drouet EM, Oelschlaegel U, Schmidt AH, Bornhäuser M, Ehninger G, Schetelig J. Salvage treatment with plerixafor in poor mobilizing allogeneic stem cell donors: results of a prospective phase II-trial. Bone Marrow Transplant 2020; 56:635-645. [PMID: 33028987 PMCID: PMC8589660 DOI: 10.1038/s41409-020-01053-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 08/14/2020] [Accepted: 09/02/2020] [Indexed: 11/09/2022]
Abstract
We conducted a prospective clinical trial to investigate the safety and efficacy of plerixafor (P) in allogeneic peripheral blood stem cells (PBSC) donors with poor mobilization response to standard-dose granulocyte colony-stimulating factor (G-CSF), defined by <2 × 106 CD34 + cells/kg recipient body-weight (CD34+/kg RBW) after 1st apheresis. A single dose of 240 µg/kg P was injected subcutaneously at 10 p.m. on the day of the 1st apheresis. Thirty-seven allogeneic PBSC donors underwent study treatment. The median CD34+ count in peripheral blood was 15/µl on Day 1 after G-CSF alone, versus 44/µl on Day 2 after G-CSF plus P (p < 0.001). The median yield of CD34+ cells was 1.1 × 108 on Day 1 and 2.8 × 108 on Day 2. In contrast to a median yield of only 1.31 × 106 CD CD34+/kg RBW on Day 1, triggering study inclusion, a median of 3.74 × 106 CD CD34+/kg RBW were collected with G-CSF plus P on Day 2. Of 37 donors, 21 reached the target cell count of >4.5 × 106 CD34+/kg RBW (57%, 95%CI 40-73%). No donor experienced a severe adverse event requiring treatment. In conclusion, P might be considered on a case-by-case basis for healthy allogeneic donors with very poor stem cell mobilization success after G-CSF.
Collapse
Affiliation(s)
- Kristina Hölig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | | | - Gero Hütter
- Cellex Collection Center GmbH, Dresden, Germany
| | - Michael Kramer
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Raphael Teipel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Katharina Heidrich
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Kristin Zimmer
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Falk Heidenreich
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany.,DKMS gemeinnützige GmbH, Clinical Trials Unit, Dresden, Germany
| | - Matthias Blechschmidt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | | | | | - Frank Kroschinsky
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Elke Rücker-Braun
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Laszlo Gopsca
- National Institute of Hematology and Infectious Diseases, Department of Hematology and Stem Cell Transplantation, Budapest, Hungary
| | - Eva Maria Wagner-Drouet
- Medizinische Klinik und Poliklinik III, Hämatologie, Internistische Onkologie, Pneumologie, Universitätsmedizin Mainz, Mainz, Germany
| | - Uta Oelschlaegel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | | | - Martin Bornhäuser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany.,Center for Regenerative Therapies, Dresden, Germany
| | - Gerhard Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Johannes Schetelig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, TU, Dresden, Germany. .,DKMS gemeinnützige GmbH, Clinical Trials Unit, Dresden, Germany.
| |
Collapse
|
11
|
Quatrini L, Tumino N, Moretta F, Besi F, Vacca P, Moretta L. Helper Innate Lymphoid Cells in Allogenic Hematopoietic Stem Cell Transplantation and Graft Versus Host Disease. Front Immunol 2020; 11:582098. [PMID: 33101308 PMCID: PMC7554507 DOI: 10.3389/fimmu.2020.582098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Helper Innate Lymphoid Cells (hILCs), including ILC1s, ILC2s, and ILC3s, are mainly localized at the mucosal barriers where they play an important role in tissue regeneration and homeostasis through the secretion of specific sets of cytokines. The recent identification of a circulating ILC precursor able to generate all ILC mature subsets in physiological conditions, suggests that “ILC-poiesis” may be important in the context of hematopoietic stem cell transplantation (HSCT). Indeed, in HSCT the conditioning regimen (chemotherapy and radiotherapy) and Graft vs Host Disease (GvHD) may cause severe damages to mucosal tissues. Therefore, it is conceivable that rapid reconstitution of the hILC compartment may be beneficial in HSCT, by promoting mucosal tissue repair/regeneration and providing protection from opportunistic infections. In this review, we will summarize the evidence for a role of hILCs in allogenic HSCT for the treatment of hematological malignancies in all its steps, from the preparative regimen to the immune reconstitution in the recipient. The protective properties of hILCs at the mucosal barrier interfaces make them an attractive target to exploit in future cellular therapies aimed at improving allogenic HSCT outcome.
Collapse
Affiliation(s)
- Linda Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicola Tumino
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesca Moretta
- Department of Laboratory Medicine, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Francesca Besi
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Paola Vacca
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
12
|
Pre-transplant myeloid and immune suppression, upfront plerixafor mobilization and post-transplant cyclophosphamide: novel strategy for haploidentical transplant in sickle cell disease. Bone Marrow Transplant 2020; 56:492-504. [PMID: 32929175 DOI: 10.1038/s41409-020-01054-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/30/2020] [Accepted: 09/02/2020] [Indexed: 11/09/2022]
Abstract
Allogenic hematopoietic stem cell transplant is the only curative option for symptomatic sickle cell disease (SCD). HLA haploidentical related donor transplants are associated with high graft failure rates. We conceptualized a novel protocol (APOLLO protocol) using pre-transplant immune and myelosuppression (PTIS) using fludarabine, cyclophosphamide, and dexamethasone followed by augmented John Hopkins protocol by adding thiotepa to conditioning. Twenty-five consecutive patients suffering from symptomatic SCD were enrolled into the study. We added upfront plerixafor to granulocyte colony stimulating factor (GCSF) for mobilization of healthy donors. Graft versus host disease (GvHD) prophylaxis was done using post-transplant cyclophosphamide, sirolimus, and mycophenolate mofetil. Graft failure was not seen in any of our patients. Five patients developed acute grade II/IV GvHD (4 classical acute, 1 late onset), 3 had limited chronic GvHD. Out of 25 evaluable patients, 22 are alive and disease free, making an overall survival (OS) and disease-free survival (DFS) of 88% with a median follow up of 485 days (range 198-802). T-cell-replete haploidentical transplant with PTIS, augmented John Hopkins conditioning and plerixafor based mobilization is a safe and effective way of treating patients suffering from SCD with minimal or no risk of graft failure and acceptable GvHD rates.
Collapse
|
13
|
Unrelated donor vs HLA-haploidentical α/β T-cell- and B-cell-depleted HSCT in children with acute leukemia. Blood 2018; 132:2594-2607. [PMID: 30348653 DOI: 10.1182/blood-2018-07-861575] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/03/2018] [Indexed: 12/13/2022] Open
Abstract
Traditionally, hematopoietic stem cell transplantation (HSCT) from both HLA-matched related and unrelated donors (UD) has been used for treating children with acute leukemia (AL) in need of an allograft. Recently, HLA-haploidentical HSCT after αβ T-cell/B-cell depletion (αβhaplo-HSCT) was shown to be effective in single-center studies. Here, we report the first multicenter retrospective analysis of 127 matched UD (MUD), 118 mismatched UD (MMUD), and 98 αβhaplo-HSCT recipients, transplanted between 2010 and 2015, in 13 Italian centers. All these AL children were transplanted in morphological remission after a myeloablative conditioning regimen. Graft failure occurred in 2% each of UD-HSCT and αβhaplo-HSCT groups. In MUD vs MMUD-HSCT recipients, the cumulative incidence of grade II to IV and grade III to IV acute graft-versus-host disease (GVHD) was 35% vs 44% and 6% vs 18%, respectively, compared with 16% and 0% in αβhaplo-HSCT recipients (P < .001). Children treated with αβhaplo-HSCT also had a significantly lower incidence of overall and extensive chronic GVHD (P < .01). Eight (6%) MUD, 32 (28%) MMUD, and 9 (9%) αβhaplo-HSCT patients died of transplant-related complications. With a median follow-up of 3.3 years, the 5-year probability of leukemia-free survival in the 3 groups was 67%, 55%, and 62%, respectively. In the 3 groups, chronic GVHD-free/relapse-free (GRFS) probability of survival was 61%, 34%, and 58%, respectively (P < .001). When compared with patients given MMUD-HSCT, αβhaplo-HSCT recipients had a lower cumulative incidence of nonrelapse mortality and a better GRFS (P < .001). These data indicate that αβhaplo-HSCT is a suitable therapeutic option for children with AL in need of transplantation, especially when an allele-matched UD is not available.
Collapse
|
14
|
Foulds GA, Vadakekolathu J, Abdel-Fatah TMA, Nagarajan D, Reeder S, Johnson C, Hood S, Moseley PM, Chan SYT, Pockley AG, Rutella S, McArdle SEB. Immune-Phenotyping and Transcriptomic Profiling of Peripheral Blood Mononuclear Cells From Patients With Breast Cancer: Identification of a 3 Gene Signature Which Predicts Relapse of Triple Negative Breast Cancer. Front Immunol 2018; 9:2028. [PMID: 30254632 PMCID: PMC6141692 DOI: 10.3389/fimmu.2018.02028] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/17/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Interactions between the immune system and tumors are highly reciprocal in nature, leading to speculation that tumor recurrence or therapeutic resistance could be influenced or predicted by immune events that manifest locally, but can be detected systemically. Methods: Multi-parameter flow cytometry was used to examine the percentage and phenotype of natural killer (NK) cells, myeloid-derived suppressor cells (MDSCs), monocyte subsets and regulatory T (Treg) cells in the peripheral blood of of 85 patients with breast cancer (50 of whom were assessed before and after one cycle of anthracycline-based chemotherapy), and 23 controls. Transcriptomic profiles of peripheral blood mononuclear cells (PBMCs) in 23 patients were generated using a NanoString gene profiling platform. Results: An increased percentage of immunosuppressive cells such as granulocytic MDSCs, intermediate CD14++CD16+ monocytes and CD127negCD25highFoxP3+ Treg cells was observed in patients with breast cancer, especially patients with stage 3 and 4 disease, regardless of ER status. Following neoadjuvant chemotherapy, B cell numbers decreased significantly, whereas monocyte numbers increased. Although chemotherapy had no effect on the percentage of Treg, MDSC and NK cells, the expression of inhibitory receptors CD85j, LIAR and NKG2A and activating receptors NKp30 and NKp44 on NK cells increased, concomitant with a decreased expression of NKp46 and DNAM-1 activating receptors. Transcriptomic profiling revealed a distinct group of 3 patients in the triple negative breast cancer (TNBC) cohort who expressed high levels of mRNA encoding genes predominantly involved in inflammation. The analysis of a large transcriptomic dataset derived from the tumors of patients with TNBC revealed that the expression of CD163, CXCR4, THBS1 predicted relapse-free survival. Conclusions: The peripheral blood immunome of patients with breast cancer is influenced by the presence and stage of cancer, but not by molecular subtypes. Furthermore, immune profiling coupled with transcriptomic analyses of peripheral blood cells may identify patients with TNBC that are at risk of relapse after chemotherapy.
Collapse
Affiliation(s)
- Gemma A Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Tarek M A Abdel-Fatah
- Clinical Oncology Department, Nottingham University Hospitals, Nottingham, United Kingdom
| | - Divya Nagarajan
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephen Reeder
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Catherine Johnson
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Simon Hood
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Paul M Moseley
- Clinical Oncology Department, Nottingham University Hospitals, Nottingham, United Kingdom
| | - Stephen Y T Chan
- Clinical Oncology Department, Nottingham University Hospitals, Nottingham, United Kingdom
| | - A Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Sergio Rutella
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephanie E B McArdle
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
15
|
Teipel R, Oelschlägel U, Wetzko K, Schmiedgen M, Kramer M, Rücker-Braun E, Hölig K, von Bonin M, Heidrich K, Fuchs A, Ordemann R, Kroschinsky F, Bornhäuser M, Hütter G, Schmidt H, Ehninger G, Schetelig J, Heidenreich F. Differences in Cellular Composition of Peripheral Blood Stem Cell Grafts from Healthy Stem Cell Donors Mobilized with Either Granulocyte Colony-Stimulating Factor (G-CSF) Alone or G-CSF and Plerixafor. Biol Blood Marrow Transplant 2018; 24:2171-2177. [PMID: 29935214 DOI: 10.1016/j.bbmt.2018.06.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/13/2018] [Indexed: 11/29/2022]
Abstract
This study was conducted to characterize and compare peripheral blood stem cell grafts from healthy donors who underwent granulocyte colony-stimulating factor (G-CSF) mobilization and subsequently received 1 dose of plerixafor after insufficient stem cell yields were achieved at the first apheresis. Aliquots from 35 donors were collected from the first apheresis after mobilization with G-CSF alone and from the second apheresis after additional plerixafor administration. Samples were freshly analyzed for cellular subsets by 8-color flow cytometry. Leukapheresis samples mobilized with additional plerixafor showed a significant increase of total nucleated cells, including B cells, CD4+ and CD8+ T cells, and CD34+ hematopoietic stem and progenitor cells. Absolute numbers of plasmacytoid dendritic cells were also significantly increased, whereas no changes were detected for myeloid dendritic cells. Furthermore, absolute numbers of regulatory T cells increased, with naive CD45RA+ regulatory T cells showing the highest rise. Finally, strikingly higher numbers of myeloid-derived suppressor cells were detected in the plerixafor and G-CSF-mobilized graft. The mobilization of peripheral stem cells in healthy donors with G-CSF and plerixafor led to a significant difference in cellular graft composition compared with G-CSF alone. The clinical impact of the different cell composition for the graft recipient warrants further clinical investigation.
Collapse
Affiliation(s)
- Raphael Teipel
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Uta Oelschlägel
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Katrin Wetzko
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Maria Schmiedgen
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Michael Kramer
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Elke Rücker-Braun
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Kristina Hölig
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Malte von Bonin
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Katharina Heidrich
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Anke Fuchs
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Rainer Ordemann
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; Cellex Gesellschaft für Zellgewinnung mbH, Dresden, Germany
| | - Frank Kroschinsky
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Martin Bornhäuser
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany; National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Gero Hütter
- Cellex Gesellschaft für Zellgewinnung mbH, Dresden, Germany
| | | | - Gerhard Ehninger
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Cellex Gesellschaft für Zellgewinnung mbH, Dresden, Germany
| | - Johannes Schetelig
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; DKMS, Clinical Trials Unit, Dresden, Germany
| | - Falk Heidenreich
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; DKMS, Clinical Trials Unit, Dresden, Germany.
| |
Collapse
|
16
|
Jaiswal SR, Bhakuni P, Joy A, Murli N, Rajoreya A, Chakrabarti A, Chakrabarti S. Higher CD45RA + Regulatory T Cells in the Graft Improves Outcome in Younger Patients Undergoing T Cell-Replete Haploidentical Transplantation: Where Donor Age Matters. Biol Blood Marrow Transplant 2018; 24:2025-2033. [PMID: 29906568 DOI: 10.1016/j.bbmt.2018.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/05/2018] [Indexed: 01/16/2023]
Abstract
To understand the phenomenon of early alloreactivity (EA) in younger children undergoing post-transplantation cyclophosphamide (PTCy)-based haploidentical transplantation, we studied the graft composition and the immune reconstitution in 32 consecutive patients (aged 2 to 25 years) undergoing PTCy and T cell costimulation blockade based peripheral blood stem cell transplantation with emphasis on CD45RA+ subset of regulatory T cells (Tregs). All but 1 engrafted, and 14 patients experienced EA (acute graft-versus-host disease grades II to IV, n = 8; and post-transplantation hemophagocytic syndrome, n = 6) with a cumulative incidence of 43.7%; 42% developed mild chronic graft-versus-host disease. The overall survival was 70.2% with a nonrelapse mortality of 16.8% at a median of 19 months. Age < 10 years, donor age > 45 years, and poor recovery of Tregs correlated with EA. Not Tregs but higher CD45RA+ Tregs in the graft was associated with less EA (11.7% versus 32.5%, P = .0001). Higher donor age correlated with a lower CD45RA+ Tregs in the graft (P = .01). However, only higher CD45RA+ Treg percentage in the graft favorably impacted EA as well as nonrelapse mortality and overall survival. Our study demonstrates a critical role for CD45RA+ Tregs in determining EA and outcome after PTCy-based haploidentical peripheral blood stem cell transplantation, and the age-related physiologic decline in this population might be responsible for adverse impact of donor age.
Collapse
Affiliation(s)
- Sarita Rani Jaiswal
- Manashi Chakrabarti Foundation, Kolkata, India; Department of Blood and Marrow Transplantation and Hematology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India.
| | - Prakash Bhakuni
- Department of Blood and Marrow Transplantation and Hematology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Aby Joy
- Department of Blood and Marrow Transplantation and Hematology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Nisha Murli
- Department of Blood and Marrow Transplantation and Hematology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | - Ashok Rajoreya
- Department of Blood and Marrow Transplantation and Hematology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| | | | - Suparno Chakrabarti
- Manashi Chakrabarti Foundation, Kolkata, India; Department of Blood and Marrow Transplantation and Hematology, Dharamshila Narayana Superspeciality Hospital and Research Centre, New Delhi, India
| |
Collapse
|
17
|
Impact of Single-Dose Plerixafor as an Adjunct to Granulocyte Colony-Stimulating Factor-Based Peripheral Blood Stem Cell Mobilization on the Graft Composition and Outcome for T Cell-Replete Haploidentical Peripheral Blood Stem Cell Transplantation with Post-Transplantation Cyclophosphamide: A Comparative Study. Biol Blood Marrow Transplant 2017; 24:542-548. [PMID: 29191663 DOI: 10.1016/j.bbmt.2017.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/12/2017] [Indexed: 11/23/2022]
Abstract
We conducted a prospective study on T and natural killer (NK) cell subset composition of graft and transplant outcomes in T cell-replete haploidentical transplantation with a single dose of subcutaneous plerixafor (Px) added to granulocyte colony-stimulating factor (G-CSF)-based mobilization in allogeneic donors to collect 10 × 106/kg CD34+ hematopoietic stem cells (HSCs) at single apheresis. Twnety-six donors received G-CSF + Px and 25 G-CSF alone for mobilization. Despite significantly lower peripheral blood (PB) CD34+ HSCs on day 4 in the G-CSF + Px group (33 [range, 6-47] cells/µL versus 81 [range, 50-168] cells/µL in the G-CSF group; P = .0001), PB CD34+ HSC count (median 136 versus 139 cells/µL) on day 5 as well as that in the graft (2.7 versus 2.3 × 106/mL, P = .1) were comparable between the 2 groups. The total nucleated cell count was higher (3.4 versus 3.1 × 108/mL, P = .05), but CD4+ T cells (2.3 versus 2.7 × 107/mL, P = .09) were lower in the G-CSF group with mobilization of regulatory T cells being similar. NK cells were skewed toward the CD56+/16- subset in both groups, varying significantly from the steady-state NK subset ratio in PB. The time to engraftment, incidences of acute and chronic graft-versus-host disease, nonrelapse mortality, and overall survival were also similar. Addition of single-dose Px to G-CSF mobilization improves CD34 recovery and does not significantly alter the T and NK cell composition of the graft, including regulatory T cells, with no adverse impact on transplant outcomes.
Collapse
|
18
|
Outcome of children with acute leukemia given HLA-haploidentical HSCT after αβ T-cell and B-cell depletion. Blood 2017; 130:677-685. [DOI: 10.1182/blood-2017-04-779769] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/28/2017] [Indexed: 12/13/2022] Open
Abstract
Key Points
Children with AL given haplo-HSCT after αβ T- and B-cell depletion are exposed to a low risk of acute and chronic GVHD and NRM. The leukemia-free, GVHD-free survival of patients given this type of allograft is comparable to that of HLA-matched donor HSCT recipients.
Collapse
|
19
|
Abstract
Allogeneic haematopoietic stem cell transplantation (HSCT) from an human leukocyte antigen (HLA)-identical donor can be curative for eligible patients with non-malignant and malignant haematological disorders. HSCT from alternative donor sources, such as HLA-mismatched haploidentical donors, is increasingly considered as a viable therapeutic option for patients lacking HLA-matched donors. Initial attempts at haploidentical HSCT were associated with vigorous bidirectional alloreactivity, leading to unacceptably high rates of graft rejection and graft-versus-host disease (GVHD). More recently, new approaches for mitigating harmful T-cell alloreactivity that mediates GVHD, while preserving the function of tumour-reactive natural killer (NK) cells and γδ T cells, have led to markedly improved clinical outcomes, and are successfully being implemented in the clinic. This article will provide an update on in vitro strategies and in vivo approaches aimed at preventing GVHD by selectively manipulating key components of the adaptive immune response, such as T-cell receptor (TCR)-αβ T cells and CD45RA-expressing naive T cells.
Collapse
|
20
|
Li Pira G, Di Cecca S, Biagini S, Girolami E, Cicchetti E, Bertaina V, Quintarelli C, Caruana I, Lucarelli B, Merli P, Pagliara D, Brescia LP, Bertaina A, Montanari M, Locatelli F. Preservation of Antigen-Specific Functions of αβ T Cells and B Cells Removed from Hematopoietic Stem Cell Transplants Suggests Their Use As an Alternative Cell Source for Advanced Manipulation and Adoptive Immunotherapy. Front Immunol 2017; 8:332. [PMID: 28386262 PMCID: PMC5362590 DOI: 10.3389/fimmu.2017.00332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/08/2017] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem cell transplantation is standard therapy for numerous hematological diseases. The use of haploidentical donors, sharing half of the HLA alleles with the recipient, has facilitated the use of this procedure as patients can rely on availability of a haploidentical donor within their family. Since HLA disparity increases the risk of graft-versus-host disease, T-cell depletion has been used to remove alloreactive lymphocytes from the graft. Selective removal of αβ T cells, which encompass the alloreactive repertoire, combined with removal of B cells to prevent EBV-related lymphoproliferative disease, proved safe and effective in clinical studies. Depleted αβ T cells and B cells are generally discarded as by-products. Considering the possible use of donor T cells for donor lymphocyte infusions or for generation of pathogen-specific T cells as mediators of graft-versus-infection effect, we tested whether cells in the discarded fractions were functionally intact. Response to alloantigens and to viral antigens comparable to that of unmanipulated cells indicated a functional integrity of αβ T cells, in spite of the manipulation used for their depletion. Furthermore, B cells proved to be efficient antigen-presenting cells, indicating that antigen uptake, processing, and presentation were fully preserved. Therefore, we propose that separated αβ T lymphocytes could be employed for obtaining pathogen-specific T cells, applying available methods for positive selection, which eventually leads to indirect allodepletion. In addition, these functional T cells could undergo additional manipulation, such as direct allodepletion or genetic modification.
Collapse
Affiliation(s)
- Giuseppina Li Pira
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Stefano Di Cecca
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Simone Biagini
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Elia Girolami
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Elisabetta Cicchetti
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Valentina Bertaina
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Concetta Quintarelli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy; Department of "Medicina Clinica e Chirurgia", University of Naples Federico II, Naples, Italy
| | - Ignazio Caruana
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Barbarella Lucarelli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Pietro Merli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Daria Pagliara
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Letizia Pomponia Brescia
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Alice Bertaina
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Mauro Montanari
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy; Department of Pediatrics, University of Pavia, Pavia, Italy
| |
Collapse
|
21
|
Selective Depletion of αβ T Cells and B Cells for Human Leukocyte Antigen–Haploidentical Hematopoietic Stem Cell Transplantation. A Three-Year Follow-Up of Procedure Efficiency. Biol Blood Marrow Transplant 2016; 22:2056-2064. [DOI: 10.1016/j.bbmt.2016.08.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/04/2016] [Indexed: 12/16/2022]
|
22
|
Emmons R, Niemiro GM, Owolabi O, De Lisio M. Acute exercise mobilizes hematopoietic stem and progenitor cells and alters the mesenchymal stromal cell secretome. J Appl Physiol (1985) 2016; 120:624-32. [DOI: 10.1152/japplphysiol.00925.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/02/2016] [Indexed: 12/20/2022] Open
Abstract
Transplantation of hematopoietic stem and progenitor cells (HSPC), collected from peripheral blood, is the primary treatment for many hematological malignancies; however, variable collection efficacy with current protocols merits further examination into factors responsible for HSPC mobilization. HSPCs primarily reside within the bone marrow and are regulated by mesenchymal stromal cells (MSC). Exercise potently and transiently mobilizes HSPCs from the bone marrow into peripheral circulation. Thus the purpose of the present study was to evaluate potential factors in the bone marrow responsible for HSPC mobilization, investigate potential sites of HSPC homing, and assess changes in bone marrow cell populations following exercise. An acute exercise bout increased circulating HSPCs at 15 min (88%, P < 0.001) that returned to baseline at 60 min. Gene expression for HSPC homing factors (CXCL12, vascular endothelial growth factor-a, and angiopoietin-1) were increased at 15 min in skeletal muscle and HSPC content was increased in the spleen 48 h postexercise (45%, P < 0.01). Acute exercise did not alter HSPCs or MSCs quantity in the bone marrow; however, proliferation of HSPCs (40%, P < 0.001), multipotent progenitors (40%, P < 0.001), short-term hematopoietic stem cells (61%, P < 0.001), long-term hematopoietic stem cells (55%, P = 0.002), and MSCs (20%, P = 0.01) increased postexercise. Acute exercise increased the content of the mobilization agent granulocyte-colony stimulating factor, as well as stem cell factor, interleukin-3, and thrombopoeitin in conditioned media collected from bone marrow stromal cells 15 min postexercise. These findings suggest that the MSC secretome is responsible for HSPC mobilization and proliferation; concurrently, HSPCs are homing to extramedullary sites following exercise.
Collapse
Affiliation(s)
- Russell Emmons
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Grace M. Niemiro
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Olatomide Owolabi
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
23
|
Moretta F, Petronelli F, Lucarelli B, Pitisci A, Bertaina A, Locatelli F, Mingari MC, Moretta L, Montaldo E. The generation of human innate lymphoid cells is influenced by the source of hematopoietic stem cells and by the use of G-CSF. Eur J Immunol 2016; 46:1271-8. [DOI: 10.1002/eji.201546079] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/15/2015] [Accepted: 01/26/2016] [Indexed: 01/24/2023]
Affiliation(s)
- Francesca Moretta
- Department of Internal Medicine; University of Verona; Verona Italy
- Lab. Analisi chimico-cliniche e microbiologiche; Ospedale Sacro Cuore Negrar; Verona Italy
| | - Francesca Petronelli
- U.O.C. Clinical and Experimental Immunology IPRCCS; Giannina Gaslini Institute; Genova Italy
| | - Barbarella Lucarelli
- Department of Pediatric Hematology and Oncology; IRCCS Bambino Gesù Children's Hospital; Rome Italy
| | - Angela Pitisci
- Department of Pediatric Hematology and Oncology; IRCCS Bambino Gesù Children's Hospital; Rome Italy
| | - Alice Bertaina
- Department of Pediatric Hematology and Oncology; IRCCS Bambino Gesù Children's Hospital; Rome Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology; IRCCS Bambino Gesù Children's Hospital; Rome Italy
- Department of Pediatrics; University of Pavia; Pavia Italy
| | - Maria Cristina Mingari
- U.O. Immunology; IRCCS AOU San Martino-IST; Genova Italy
- Department of Experimental Medicine; University of Genova; Genova Italy
| | - Lorenzo Moretta
- Department of Immunology; IRCCS Bambino Gesù Children's Hospital; Rome Italy
| | - Elisa Montaldo
- U.O.C. Clinical and Experimental Immunology IPRCCS; Giannina Gaslini Institute; Genova Italy
| |
Collapse
|
24
|
Yang JZ, Zhang JQ, Sun LX. Mechanisms for T cell tolerance induced with granulocyte colony-stimulating factor. Mol Immunol 2015; 70:56-62. [PMID: 26703218 DOI: 10.1016/j.molimm.2015.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 12/15/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) has been widely accepted as a mediator of T cell tolerance. The immune modulatory effect of G-CSF on T cells is believed to be mediated exclusively through other effector cells, such as monocytes, tolerogenic dendritic cells (DC), and myeloid-derived suppressor cells. Recent advances confirmed the direct effects of G-CSF in inducing immune tolerance of T cells through the G-CSF-G-CSF receptor pathway and related molecular mechanisms. This review aims to summarize the findings associated with the direct and indirect mechanisms for T cell tolerance induced with G-CSF. The role of G-CSF in preventing graft-versus-host disease (GVHD) and in treating autoimmune diseases (ADs) is also discussed. It is conceivable that G-CSF and immune cell compositions, such as tolerogenic DC and CD4(+)CD25(+)Foxp3(+) T cells, modulated by G-CSF could become an integral part of the immunomodulatory therapies against GVHD and ADs in the future.
Collapse
Affiliation(s)
- Jian-Zhu Yang
- Department of Pathology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin-Qiao Zhang
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li-Xia Sun
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
25
|
Plasmacytoid dendritic cells in allogeneic hematopoietic cell transplantation: benefit or burden? Bone Marrow Transplant 2015; 51:333-43. [PMID: 26642333 DOI: 10.1038/bmt.2015.301] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/25/2015] [Accepted: 10/27/2015] [Indexed: 11/09/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and have important roles in hematopoietic engraftment, GvHD and graft-versus-leukemia responses following allogeneic hematopoietic cell transplantation (HCT). In addition, pDCs mediate antiviral immunity, particularly as they are the body's primary cellular source of type I interferon. Given their pleiotropic roles, pDCs have emerged as cells that critically impact transplant outcomes, including overall survival. In this article, we will review the pre-clinical and clinical literature, supporting the crucial roles that pDCs assume as key immune effector cells during HCT.
Collapse
|
26
|
|
27
|
Pittari G, Filippini P, Gentilcore G, Grivel JC, Rutella S. Revving up Natural Killer Cells and Cytokine-Induced Killer Cells Against Hematological Malignancies. Front Immunol 2015; 6:230. [PMID: 26029215 PMCID: PMC4429635 DOI: 10.3389/fimmu.2015.00230] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/29/2015] [Indexed: 01/29/2023] Open
Abstract
Natural killer (NK) cells belong to innate immunity and exhibit cytolytic activity against infectious pathogens and tumor cells. NK-cell function is finely tuned by receptors that transduce inhibitory or activating signals, such as killer immunoglobulin-like receptors, NK Group 2 member D (NKG2D), NKG2A/CD94, NKp46, and others, and recognize both foreign and self-antigens expressed by NK-susceptible targets. Recent insights into NK-cell developmental intermediates have translated into a more accurate definition of culture conditions for the in vitro generation and propagation of human NK cells. In this respect, interleukin (IL)-15 and IL-21 are instrumental in driving NK-cell differentiation and maturation, and hold great promise for the design of optimal NK-cell culture protocols. Cytokine-induced killer (CIK) cells possess phenotypic and functional hallmarks of both T cells and NK cells. Similar to T cells, they express CD3 and are expandable in culture, while not requiring functional priming for in vivo activity, like NK cells. CIK cells may offer some advantages over other cell therapy products, including ease of in vitro propagation and no need for exogenous administration of IL-2 for in vivo priming. NK cells and CIK cells can be expanded using a variety of clinical-grade approaches, before their infusion into patients with cancer. Herein, we discuss GMP-compliant strategies to isolate and expand human NK and CIK cells for immunotherapy purposes, focusing on clinical trials of adoptive transfer to patients with hematological malignancies.
Collapse
Affiliation(s)
- Gianfranco Pittari
- Department of Medical Oncology, National Center for Cancer Care and Research, Hamad Medical Corporation , Doha , Qatar
| | - Perla Filippini
- Deep Immunophenotyping Core, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| | - Giusy Gentilcore
- Deep Immunophenotyping Core, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| | - Jean-Charles Grivel
- Deep Immunophenotyping Core, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| | - Sergio Rutella
- Clinical Research Center, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| |
Collapse
|
28
|
Gattillo S, Marktel S, Rizzo L, Malato S, Malabarba L, Coppola M, Assanelli A, Milani R, De Freitas T, Corti C, Bellio L, Ciceri F. Plerixafor on demand in ten healthy family donors as a rescue strategy to achieve an adequate graft for stem cell transplantation. Transfusion 2015; 55:1993-2000. [PMID: 25721167 DOI: 10.1111/trf.13059] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/20/2015] [Accepted: 01/22/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND In allogeneic hematopoietic stem cell (HSC) transplantation, the collection of an appropriate number of HSCs while maintaining a high level of safety for healthy donors is fundamental. Inadequate HSC mobilization can be seen with the standard use of granulocyte-colony-stimulating (G-CSF). Plerixafor (PL) is a chemokine receptor CXC Type 4-stromal-derived factor 1 inhibitor; its HSC-mobilizing properties are synergistic with G-CSF in poor mobilizing patients. The use of PL as adjuvant or alternative to G-CSF in healthy donors has shown a good safety profile but is so far off-label. STUDY DESIGN AND METHODS We report 10 healthy HSC donors treated with PL because of insufficient response to G-CSF alone or contraindication to G-CSF. Eight donors did not mobilize enough CD34+ cells with G-CSF alone because poor mobilizers or because insufficient HSCs were harvested according to the clinical need of the patient; in two cases G-CSF administration and marrow harvest were unfeasible or contraindicated in the donor. RESULTS The use of PL for mobilization increased the number of circulating CD34+ cells by 2.8-fold and the CD34+/kg collection by 3.0-fold. Only mild adverse events were reported (bone pain or discomfort) and not univocally attributable to PL. Rate of engraftment and graft-versus-host disease were similar to those seen in recipients of grafts from G-CSF only-mobilized donors. CONCLUSION We exposed 10 allogeneic donors to mobilization with PL. PL was well tolerated in all cases and ensured procurement of an adequate graft for transplantation resulting in a normal hematopoietic engraftment.
Collapse
Affiliation(s)
- Salvatore Gattillo
- Blood Transfusion Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sarah Marktel
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Rizzo
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simona Malato
- Blood Transfusion Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Malabarba
- Blood Transfusion Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Milena Coppola
- Blood Transfusion Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Assanelli
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Milani
- Blood Transfusion Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tiago De Freitas
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Consuelo Corti
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Bellio
- Blood Transfusion Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Blood Transfusion Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
29
|
Saraceni F, Shem-Tov N, Olivieri A, Nagler A. Mobilized peripheral blood grafts include more than hematopoietic stem cells: the immunological perspective. Bone Marrow Transplant 2015; 50:886-91. [PMID: 25665044 DOI: 10.1038/bmt.2014.330] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 01/20/2023]
Abstract
Although stem cell mobilization has been performed for more than 20 years, little is known about the effects of mobilizing agents on apheresis composition and the impact of graft cell subsets on patients' outcome. With the increasing use of plerixafor and the inclusion of poor mobilizers in autologous transplant procedures, new parameters other than CD34(+) stem cell dose are emerging; plerixafor seems to mobilize more primitive CD34(+)/CD38(-) stem cells compared with G-CSF, but their correlation with stable hematopoietic engraftment is still obscure. Immune recovery is as crucial as hematopoietic reconstitution, and higher T and natural killer cells infused within the graft have been correlated with better outcome in autologous transplant; recent studies showed increased mobilization of immune effectors with plerixafor compared with G-CSF, but further data are needed to clarify the clinical impact of these findings. In the allogeneic setting, much evidence suggests that mobilized T-cell alloreactivity is tempered by G-CSF, probably with the mediation of dendritic cells, even though no clear correlation with GVL and GVHD has been found. Plerixafor is not approved in healthy donors yet; early data suggest it might mobilize a GVHD protective balance of immune effectors, but further studies are needed to define its role in allogeneic transplant.
Collapse
Affiliation(s)
- F Saraceni
- 1] Department of Hematology and Bone Marrow Transplantation, Ospedali Riuniti, Ancona, Italy [2] Department of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - N Shem-Tov
- Department of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - A Olivieri
- Department of Hematology and Bone Marrow Transplantation, Ospedali Riuniti, Ancona, Italy
| | - A Nagler
- Department of Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|