101
|
López-Corral L, Caballero-Velázquez T, López-Godino O, Rosiñol L, Pérez-Vicente S, Fernandez-Avilés F, Krsnik I, Morillo D, Heras I, Morgades M, Rifon JJ, Sampol A, Iniesta F, Ocio EM, Martin J, Rovira M, Cabero M, Castilla-Llorente C, Ribera JM, Torres-Juan M, Moraleda JM, Martinez C, Vázquez A, Gutierrez G, Caballero D, San Miguel JF, Mateos MV, Pérez-Simón JA. Response to Novel Drugs before and after Allogeneic Stem Cell Transplantation in Patients with Relapsed Multiple Myeloma. Biol Blood Marrow Transplant 2019; 25:1703-1712. [PMID: 31054983 DOI: 10.1016/j.bbmt.2019.04.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/21/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022]
Abstract
Multiple myeloma (MM) remains as an incurable disease and, although allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative approach, most patients ultimately relapse, and their treatment remains challenging. Because allo-HSCT can modify not only the biology of the disease, but also the immune system and the microenvironment, it can potentially enhance the response to rescue therapies. Information on the efficacy and safety of novel drugs in patients relapsing after allo-HSCT is lacking, however. The objectives of this study were to evaluate the efficacy and toxicity of rescue therapies in patients with MM who relapsed after allo-HSCT, as well as to compare their efficacy before and after allo-HSCT. This retrospective multicenter study included 126 consecutive patients with MM who underwent allo-HSCT between 2000 and 2013 at 8 Spanish centers. All patients engrafted. The incidence of grade II-IV acute graft-versus-host disease (GVHD) was 47%, and nonrelapse mortality within the first 100 days post-transplantation was 13%. After a median follow-up of 92 months, overall survival (OS) was 51% at 2 years and 43% at 5 years. The median progression-free survival after allo-HSCT was 7 months, whereas the median OS after relapse was 33 months. Patients relapsing in the first 6 months after transplantation had a dismal prognosis compared with those who relapsed later (median OS, 11 months versus 120 months; P < .001). The absence of chronic GVHD was associated with reduced OS after relapse (hazard ratio, 3.44; P < .001). Most patients responded to rescue therapies, including proteasome inhibitors (PIs; 62%) and immunomodulatory drugs (IMiDs; 77%), with a good toxicity profile. An in-depth evaluation, including the type and intensity of PI- and IMiD-based combinations used before and after allo-HSCT, showed that the overall response rate and duration of response after allo-HSCT were similar to those seen in the pretransplantation period. Patients with MM who relapse after allo-HSCT should be considered candidates for therapy with new drugs, which can achieve similar response rates with similar durability as seen in the pretransplantation period. This pattern does not follow the usual course of the disease outside the transplantation setting, where response rates and time to progression decreases with each consecutive line of treatment.
Collapse
Affiliation(s)
- Lucia López-Corral
- Hematology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, Centro de Investigación del Cáncer-IBMCC, Spain.
| | - Teresa Caballero-Velázquez
- Hematology Department, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/CIBERON/Universidad de Sevilla, Spain
| | - Oriana López-Godino
- Hematology Department, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, IMIB, Universidad de Murcia, Spain
| | - Laura Rosiñol
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Sabina Pérez-Vicente
- Hematology Department, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/CIBERON/Universidad de Sevilla, Spain
| | | | - Isabel Krsnik
- Hematology Department, Instituto de Investigación Puerta de Hierro Majadahonda, Madrid, Spain
| | - Daniel Morillo
- Hematology Department, Instituto de Investigación Puerta de Hierro Majadahonda, Madrid, Spain
| | - Inmaculada Heras
- Hematology Department, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, IMIB, Universidad de Murcia, Spain
| | - Mireia Morgades
- Department of Hematology, ICO Badalona-Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Jose J Rifon
- Hematology Service, Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | - Antonia Sampol
- Hematology Department, Hospital Son Espases, Palma de Mallorca, Spain
| | - Francisca Iniesta
- Hematology Department, University Hospital of Virgen de la Arrixaca, Murcia, Spain
| | - Enrique-María Ocio
- Hematology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, Centro de Investigación del Cáncer-IBMCC, Spain
| | - Jesús Martin
- Hematology Department, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/CIBERON/Universidad de Sevilla, Spain
| | - Montserrat Rovira
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Martín Cabero
- Hematology Department, Instituto de Investigación Puerta de Hierro Majadahonda, Madrid, Spain
| | | | - Josep-María Ribera
- Department of Hematology, ICO Badalona-Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Marta Torres-Juan
- Hematology Department, Hospital Son Espases, Palma de Mallorca, Spain
| | - Jose María Moraleda
- Hematology Department, University Hospital of Virgen de la Arrixaca, Murcia, Spain
| | - Carmen Martinez
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Alejandro Vázquez
- Hematology Department, Instituto de Investigación Puerta de Hierro Majadahonda, Madrid, Spain
| | - Gonzalo Gutierrez
- Department of Hematology, Amyloidosis and Myeloma Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - Dolores Caballero
- Hematology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, Centro de Investigación del Cáncer-IBMCC, Spain
| | - Jesús F San Miguel
- Hematology Service, Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | - María-Victoria Mateos
- Hematology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, Centro de Investigación del Cáncer-IBMCC, Spain
| | - Jose Antonio Pérez-Simón
- Hematology Department, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/CIBERON/Universidad de Sevilla, Spain
| |
Collapse
|
102
|
Pretreatment CD34 +/CD38 - Cell Burden as Prognostic Factor in Myelodysplastic Syndrome Patients Receiving Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1560-1566. [PMID: 30928626 DOI: 10.1016/j.bbmt.2019.03.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 12/16/2022]
Abstract
Myelodysplastic syndrome (MDS) is a highly heterogeneous clonal hematopoietic disorder. Allogeneic hematopoietic stem cell transplantation (HSCT) remains the only curative treatment and is of particular interest in patients at high risk for progression to acute myeloid leukemia (AML). In MDS, CD34+/CD38- cells possess MDS stem cell potential, and secondary AML (sAML) clones originate from the MDS disease stage. However, the prognostic impact of the pretreatment stem cell population burden in MDS remains unknown. We retrospectively analyzed the prognostic impact of the pretreatment CD34+/CD38- cell burden in 124 MDS patients who received allogeneic HSCT at our institution. A high pretreatment bone marrow CD34+/CD38- cell burden (≥1%) was associated with worse genetic risk and a higher incidence of blast excess. Patients with a high CD34+/CD38- cell burden had a significantly higher cumulative incidence of MDS relapse, a higher cumulative incidence of secondary AML, and a trend for shorter overall survival after allogeneic HSCT. In multivariable analyses this prognostic impact was shown to be independent of other clinical and cytogenetic risk factors in MDS. Patients suffering MDS relapse or progression to AML also had a higher pre-treatment CD34+/CD38- cell burden as a continuous variable. The observed prognostic impact is likely mediated by MDS stem cells within the CD34+/CD38- cell population initiating MDS relapse or progression to AML. New therapeutic strategies targeting MDS stem cells might improve outcomes.
Collapse
|
103
|
Ali S, Dunmore HM, Karres D, Hay JL, Salmonsson T, Gisselbrecht C, Sarac SB, Bjerrum OW, Hovgaard D, Barbachano Y, Nagercoil N, Pignatti F. The EMA Review of Mylotarg (Gemtuzumab Ozogamicin) for the Treatment of Acute Myeloid Leukemia. Oncologist 2019; 24:e171-e179. [PMID: 30898889 DOI: 10.1634/theoncologist.2019-0025] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/20/2019] [Indexed: 11/17/2022] Open
Abstract
On February 22, 2018, the Committee for Medicinal Products for Human Use (CHMP) adopted a positive opinion, recommending the granting of a marketing authorization for the medicinal product gemtuzumab ozogamicin (Mylotarg; Pfizer, New York City, NY), intended for the treatment of acute myeloid leukemia. Mylotarg was designated as an orphan medicinal product on October 18, 2000. The applicant for this medicinal product was Pfizer Limited (marketing authorization now held by Pfizer Europe MA EEIG).The demonstrated benefit with Mylotarg is improvement in event-free survival. This has been shown in the pivotal ALFA-0701 (MF-3) study. In addition, an individual patient data meta-analysis from five randomized controlled trials (3,325 patients) showed that the addition of Mylotarg significantly reduced the risk of relapse (odds ratio [OR] 0.81; 95% CI: 0.73-0.90; p = .0001), and improved overall survival at 5 years (OR 0.90; 95% CI: 0.82-0.98; p = .01) [Lancet Oncol 2014;15:986-996]. The most common (>30%) side effects of Mylotarg when used together with daunorubicin and cytarabine are hemorrhage and infection.The full indication is as follows: "Mylotarg is indicated for combination therapy with daunorubicin (DNR) and cytarabine (AraC) for the treatment of patients age 15 years and above with previously untreated, de novo CD33-positive acute myeloid leukemia (AML), except acute promyelocytic leukemia (APL)."The objective of this article is to summarize the scientific review done by the CHMP of the application leading to regulatory approval in the European Union. The full scientific assessment report and product information, including the Summary of Product Characteristics, are available on the European Medicines Agency website (www.ema.europa.eu). IMPLICATIONS FOR PRACTICE: This article reflects the scientific assessment of Mylotarg (gemtuzumab ozogamicin; Pfizer, New York City, NY) use for the treatment of acute myeloid leukemia based on important contributions from the rapporteur and co-rapporteur assessment teams, Committee for Medicinal Products for Human Use members, and additional experts following the application for a marketing authorization from the company. It's a unique opportunity to look at the data from a regulatory point of view and the importance of assessing the benefit-risk.
Collapse
Affiliation(s)
- Sahra Ali
- European Medicines Agency, London, United Kingdom
| | - Helen-Marie Dunmore
- Medicines and Healthcare Products Regulatory Agency Licensing, London, United Kingdom
| | | | - Justin L Hay
- Medicines and Healthcare Products Regulatory Agency Licensing, London, United Kingdom
| | | | | | | | | | | | - Yolanda Barbachano
- Medicines and Healthcare Products Regulatory Agency Licensing, London, United Kingdom
| | | | | |
Collapse
|
104
|
Potdar RR, Gupta S, Giebel S, Savani BN, Varadi G, Nagler A, Blamek S. Current Status and Perspectives of Irradiation-Based Conditioning Regimens for Patients with Acute Leukemia Undergoing Hematopoietic Stem Cell Transplantation. Clin Hematol Int 2019; 1:19-27. [PMID: 34595407 PMCID: PMC8432382 DOI: 10.2991/chi.d.190218.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 02/18/2019] [Indexed: 11/01/2022] Open
Abstract
Acute myeloid leukemia and acute lymphoblastic leukemia are the most common indications for allogeneic hematopoietic stem cell transplantation. Total body irradiation (TBI) is an important part of conditioning regimens. TBI-based regimens offer advantages in sanctuary sites but are associated with significant risks of early and late side effects, including pulmonary toxicity, growth retardation, and second malignancy. TBI is also associated with technical problems, such as dose heterogeneity. With evolving techniques in radiation oncology, it is possible to focus the dose to the entire skeleton while sparing the rest of the body. This technique is called total marrow irradiation (TMI). TMI is able to deliver the same or higher doses to bone marrow while reducing toxicity. With the success of TMI, we are moving toward ultra-personalized conditioning. We review the clinical role of the irradiation-based regimens currently in clinical use, emphasizing on their strengths and limitations. Novel technologies with targeted irradiation accompanied by the modern imaging techniques and increased knowledge of the disease process can help us achieve our goal of maximum response with minimum toxicity.
Collapse
Affiliation(s)
- Rashmika R Potdar
- Division of Hematology and Oncology, Department of Internal Medicine, Einstein Medical Center, Philadelphia, PA, USA
| | - Sorab Gupta
- Division of Hematology and Oncology, Department of Internal Medicine, Einstein Medical Center, Philadelphia, PA, USA
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie Institute-Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center & Veterans Affairs Medical Center, Nashville, TN, USA
| | - Gabor Varadi
- Division of Hematology and Oncology, Department of Internal Medicine, Einstein Medical Center, Philadelphia, PA, USA
| | - Arnon Nagler
- Hematology and Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Slawomir Blamek
- Department of Radiotherapy, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| |
Collapse
|
105
|
Sun YQ, Huang XJ, Xu LP, Zhang XH, Yan CH, Liu KY, Wang Y. [Impact of mycophenolate mofetil prophylaxis duration on acute graft-versus-host disease after haploidentical stem cell transplantation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 39:286-291. [PMID: 29779322 PMCID: PMC7342124 DOI: 10.3760/cma.j.issn.0253-2727.2018.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Objective: To investigate the impact of mycophenolate mofetil (MMF) prophylaxis duration on acute graft-versus-host disease (aGVHD) after haploidentical stem cell transplantation (haplo-HSCT) using 'Beijing Protocol'. Methods: Adult patients (≥14 years) received haplo-HSCT in Peking University Institute of Hematology from Sep, 2016 to Mar, 2017 were retrospectively reviewed if they fulfilled the criterias: ①diagnosed with hematological maligancies; ②standard-risk status at haplo-HSCT. A total of 237 patients [including 102 patients with long MMF duration (defined as started on day -9 with 100 mg/d, adjusted to 500 mg/d from day +30 and discontinued on day +45 to +60 or occurrence of CMV/EBV reactivation or late-onset hemorrhagic cytitis), and 135 patients with short MMF duration (defined as started on day -9 with 500 mg/d and discontinued on the day achieved neutrophil engraftment)] were reviewed. The incidence of aGVHD, virus infection and overall survival (OS) were compared between the two groups. Results: The median durations of MMF prophylaxis of long and short duration groups were 27(7-71) and 15(9-24) days, respectively after haplo-HSCT. There were no differences of baseline characteristics (including sex, patient age, disease, mismatched HLA loci, donor-recipient relation, donor-recipient sex and donor age) between the two groups. The incidences of the grade Ⅱ-Ⅳ and Ⅲ/Ⅳ aGVHD in long and short duration groups were 31.1% versus 17.6% (P=0.018) and 7.4% verus 7.8% (P=0.900), respectively. The duration of MMF prophylaxis was not found to be associated with gradeⅡ-Ⅳ aGVHD by the multivariate analysis. There were no significant differences in terms of CMV viremia, EBV viremia, hemorrhagic cytitis and OS between the two groups. Conclusion: Prophylaxis with short duration MMF in the setting of 'Beijing protocol' haplo-SCT was not associated with increased acute GVHD with no impact on OS, which indicated that short duration MMF might be a feasible GVHD prophylaxis regimen.
Collapse
Affiliation(s)
- Y Q Sun
- The Institute of Hematology, People's Hospital of Peking University, Beijing 100044, China
| | | | | | | | | | | | | |
Collapse
|
106
|
Jentzsch M, Bill M, Grimm J, Schulz J, Beinicke S, Häntschel J, Goldmann K, Pönisch W, Franke GN, Vucinic V, Cross M, Behre G, Lange T, Niederwieser D, Schwind S. Prognostic Impact of Blood MN1 Copy Numbers Before Allogeneic Stem Cell Transplantation in Patients With Acute Myeloid Leukemia. Hemasphere 2019; 3:e167. [PMID: 31723806 PMCID: PMC6745933 DOI: 10.1097/hs9.0000000000000167] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/27/2018] [Indexed: 01/02/2023] Open
Abstract
High expression of the leukemia-associated gene meningioma-1 (MN1) is frequently found at diagnosis of acute myeloid leukemia (AML) and associates with adverse outcomes. The presence of measurable residual disease (MRD) in complete remission (CR) indicates high risk of relapse and worse outcome in AML patients. However, the prognostic impact of MN1 expression levels as MRD marker has not been evaluated. Digital droplet polymerase chain reaction (ddPCR) is a novel technique allowing sensitive and specific absolute gene expression quantification. We retrospectively analyzed 124 AML patients who received allogeneic hematopoietic stem cell transplantation (HSCT) in CR or CR with incomplete peripheral recovery. Absolute MN1 copy numbers in peripheral blood were assessed prior to HSCT (median 7; range 0-29 days) using ddPCR. High pre-HSCT MN1/Abelson murine leukemia viral oncogene homolog 1 gene (ABL1) copy numbers associated with a higher cumulative incidence of relapse after HSCT and-in relapsing patients-shorter time to relapse. In multivariable analysis, high pre-HSCT MN1/ABL1 copy numbers remained an independent prognosticator for relapse after HSCT. Patients with the highest pre-HSCT MN1/ABL1 copy numbers also had the highest risk of relapse. MN1 copy number assessment also added prognostic information to nucleophosmin 1 gene (NPM1) mutation- and brain and acute leukemia, cytoplasmic (BAALC) and Wilm's tumor gene 1 (WT1) expression-based MRD evaluation. Our study demonstrates the feasibility of the novel ddPCR technique for MN1/ABL1 copy number assessment as a marker for MRD. Evaluation of MN1/ABL1 copy numbers allows the identification of patients at high risk of relapse, independently of other diagnostic risk factors and MRD markers.
Collapse
Affiliation(s)
- Madlen Jentzsch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Marius Bill
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Juliane Grimm
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Julia Schulz
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Stefanie Beinicke
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Janine Häntschel
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Karoline Goldmann
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Wolfram Pönisch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | | | - Vladan Vucinic
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Michael Cross
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Gerhard Behre
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Thoralf Lange
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | | | - Sebastian Schwind
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
107
|
Kennedy VE, Muffly LS. Assessment of older adult candidates for allogeneic hematopoietic cell transplantation: updates and remaining questions. Expert Rev Hematol 2019; 12:99-106. [DOI: 10.1080/17474086.2019.1568236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Vanessa E. Kennedy
- Department of Internal Medicine, Division of Hospital Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lori S. Muffly
- Department of Internal Medicine, Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
108
|
Monaco F, Scott BL, Chauncey TR, Petersen FB, Storer BE, Baron F, Flowers ME, Deeg HJ, Maloney DG, Storb R, Sandmaier BM. Total body irradiation dose escalation decreases risk of progression and graft rejection after hematopoietic cell transplantation for myelodysplastic syndromes or myeloproliferative neoplasms. Haematologica 2019; 104:1221-1229. [PMID: 30630975 PMCID: PMC6545836 DOI: 10.3324/haematol.2018.199398] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/02/2019] [Indexed: 11/09/2022] Open
Abstract
A non-myeloablative regimen of fludarabine and 200 cGy total body irradiation combined with post-grafting immunosuppression with mycophenolate mofetil and a calcineurin inhibitor facilitates allogeneic hematopoietic cell transplantation from HLA-matched related or unrelated donors in older patients and/or those with comorbidities. However, outcomes of prior studies have been disappointing in patients with myelodysplastic syndromes or myeloproliferative neoplasms due to high incidences of progression or graft failure (together termed hematopoietic cell transplantation-failure). We hypothesized that escalating the total body irradiation dose may improve the outcomes and subsequently performed a phase II total body irradiation dose-escalation trial. Patients with median age 66 years were enrolled in two arms to receive non-myeloablative conditioning followed by hematopoietic cell transplantation with total body irradiation dose escalation for excessive hematopoietic cell transplantation-failure: Arm A: myeloproliferative neoplasm/myelodysplastic syndrome low risk (n=36); and Arm B: myelodysplastic syndrome high-risk/chronic myelomonocytic leukemia (n=41). Total body irradiation dose levels were: Level-1 (300 cGy), Level-2 (400 cGy), or Level-3 (450 cGy). Patients received intravenous fludarabine 30 mg/m2 for three days. Total body irradiation was administered on day 0 followed by infusion of peripheral blood stem cells from HLA-matched related (n=30) or unrelated (n=47) donors. Post-grafting immunosuppression with mycophenolate mofetil and cyclosporine was administered. The primary end point was day 200 hematopoietic cell transplant failure, with the objective of reducing the incidence to <20%. The primary end point was reached on Arm A at dose Level-1 (300 cGy total body irradiation) with a cumulative incidence of day 200 hematopoietic cell transplant failure of 11%, and on Arm B at dose Level-3 (450 cGy) with a cumulative incidence of day 200 hematopoietic cell transplant failure of 9%. Increasing the total body irradiation dose leads to a higher success rate with non-myeloablative conditioning by reducing relapse and rejection. Further studies are necessary to decrease non-relapse mortality, especially among patients with high-risk disease. Trial registered under clinicaltrials.gov identifier: NCT00397813.
Collapse
Affiliation(s)
| | - Bart L Scott
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | - Thomas R Chauncey
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA.,VA Puget Sound Health Care System, Seattle, WA, USA
| | | | - Barry E Storer
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | | | - Mary E Flowers
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | - H Joachim Deeg
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | - David G Maloney
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | - Rainer Storb
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA .,University of Washington, Seattle, WA, USA
| |
Collapse
|
109
|
Munshi PN, Rowley SD, Korngold R. Hematopoietic Stem Cell Transplantation for Malignant Diseases. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00083-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
110
|
Guru Murthy GS, Hari PN, Szabo A, Pasquini M, Narra R, Khan M, Abedin S, Chhabra S, Dhakal B, D'Souza A, Drobyski WR, Rizzo JD, Runaas L, Shah NN, Shaw B, Saber W, Fenske T, Hamadani M. Outcomes of Reduced-Intensity Conditioning Allogeneic Hematopoietic Cell Transplantation Performed in the Inpatient versus Outpatient Setting. Biol Blood Marrow Transplant 2018; 25:827-833. [PMID: 30572109 DOI: 10.1016/j.bbmt.2018.12.069] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) with reduced-intensity conditioning (RIC) is commonly performed as an inpatient procedure. The feasibility and outcomes of RIC allo-HCT in the outpatient setting is not known. We performed a single-center retrospective cohort study of patients aged ≥ 18years with hematologic malignancies who underwent RIC allo-HCT either in the inpatient or outpatient setting. Donor types included HLA-matched sibling and well-matched unrelated donors. The objectives were to compare the survival, complications, charges, and incidences of relapse, nonrelapse mortality (NRM), and acute and chronic graft-versus-host disease (GVHD) between the 2 groups. Between 2014 and 2017, 151 eligible patients were included, with 116 undergoing RIC allo-HCT in the inpatient setting and 35 patients undergoing RIC allo-HCT in the outpatient setting. Baseline characteristics were comparable between the 2 groups except for a higher proportion of patients with myeloma in the outpatient cohort (inpatient 15.5% versus outpatient 37.1%). The cumulative incidence of grades II to IV acute GVHD (inpatient 25.2% versus outpatient 25.7%), grades III to IV acute GVHD (inpatient 10.4% versus outpatient 8.5%), chronic GVHD (inpatient 38.3% versus outpatient 51.6%), NRM at 1 year (inpatient 10.8% versus outpatient 3.2%), and relapse (inpatient 24.8% versus outpatient 33.2%) did not significantly differ between the 2 cohorts. One-year progression-free survival (inpatient 64.4% versus outpatient 63.6%, P = .39) and overall survival (inpatient 73.8% versus outpatient 82.8%, P = .93) were also not significantly different between the 2 groups. The proportion of patients who developed neutropenic fever (inpatient 25.8% versus outpatient 8.5%, P = .03) and mucositis (inpatient 50.8% versus outpatient 8.5%, P < .001) and who required total parenteral nutrition (inpatient 20.6% versus outpatient 5.7%, P = .04) were more frequent in the inpatient cohort. About 51.5% of the outpatient cohort never required hospital admission in the first 100days. Outpatient HCT resulted in significantly lower charges than inpatient HCT in the first 100days (median charges: inpatient $339,621 versus outpatient $247,334; P < .001). On multivariate analysis the site of the HCT (outpatient versus inpatient) was not a significant predictor of either overall or progression-free survival. Outpatient RIC allo-HCT is feasible and safe with daily outpatient evaluation and aggressive supportive care resulting in outcomes comparable with those who received the transplant in the inpatient setting.
Collapse
Affiliation(s)
- Guru Subramanian Guru Murthy
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Parameswaran N Hari
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aniko Szabo
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Marcelo Pasquini
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ravi Narra
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Muhammad Khan
- Division of Hospital Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sameem Abedin
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Saurabh Chhabra
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Binod Dhakal
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anita D'Souza
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - William R Drobyski
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J Douglas Rizzo
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lyndsey Runaas
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nirav N Shah
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Bronwen Shaw
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Wael Saber
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Timothy Fenske
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
111
|
Craddock CF. We do still transplant CML, don't we? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:177-184. [PMID: 30504307 PMCID: PMC6246013 DOI: 10.1182/asheducation-2018.1.177] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The remarkable clinical activity of tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML) has transformed patient outcome. Consequently, allogeneic stem cell transplantation (allo-SCT) is no longer the only treatment modality with the ability to deliver long-term survival. In contrast to the central position it held in the treatment algorithm 20 years ago, allografting is now largely reserved for patients with either chronic-phase disease resistant to TKI therapy or advanced-phase disease. Over the same period, progress in transplant technology, principally the introduction of reduced intensity conditioning regimens coupled with increased donor availability, has extended transplant options in patients with CML whose outcome can be predicted to be poor if they are treated with TKIs alone. Consequently, transplantation is still a vitally important, potentially curative therapeutic modality in selected patients with either chronic- or advanced-phase CML. The major causes of transplant failure in patients allografted for CML are transplant toxicity and disease relapse. A greater understanding of the distinct contributions made by various factors such as patient fitness, patient-donor HLA disparity, conditioning regimen intensity, and transplant toxicity increasingly permits personalized transplant decision making. At the same time, advances in the design of conditioning regimens coupled with the use of adjunctive posttransplant cellular and pharmacologic therapies provide opportunities for reducing the risk of disease relapse. The role of SCT in the management of CML will grow in the future because of an increase in disease prevalence and because of continued improvements in transplant outcome.
Collapse
Affiliation(s)
- Charles F Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| |
Collapse
|
112
|
Grimm J, Bill M, Jentzsch M, Beinicke S, Häntschel J, Goldmann K, Schulz J, Cross M, Franke G, Behre G, Vucinic V, Pönisch W, Lange T, Niederwieser D, Schwind S. Clinical impact of clonal hematopoiesis in acute myeloid leukemia patients receiving allogeneic transplantation. Bone Marrow Transplant 2018; 54:1189-1197. [DOI: 10.1038/s41409-018-0413-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/21/2022]
|
113
|
Le Bourgeois A, Labopin M, Leclerc M, de Latour RP, Bourhis JH, Ceballos P, Orvain C, Wallet HL, Bilger K, Blaise D, Rubio MT, Guillaume T, Mohty M, Chevallier P, on behalf of Société Francophone de Greffe de Moelle et de Thérapie Cellulaire. Clofarabine/busulfan-based reduced intensity conditioning regimens provides very good survivals in acute myeloid leukemia patients in complete remission at transplant: a retrospective study on behalf of the SFGM-TC. Oncotarget 2018; 9:36603-36612. [PMID: 30564300 PMCID: PMC6290956 DOI: 10.18632/oncotarget.26391] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/01/2018] [Indexed: 11/25/2022] Open
Abstract
Background Clofarabine has been proved to have higher anti-leukemic myeloid activity compared to fludarabine, a drug extensively used as part of reduced intensity conditioning (RIC) for allogeneic stem cell transplantation (allo-SCT). Results Eighty-four patients were included. The majority of patients had acute myeloid leukemia (AML, n = 63). Sixty-one patients were in complete remission (AML n = 55). With a median follow up of 31 months (range: 5.7-74.1), 2-year overall (OS) and disease-free (DFS) survivals, relapse incidence (RI), non-relapse mortality (NRM) and graft-versus-host disease (GVHD)/relapse free survival (GRFS) were 64.5% (53.8-75.2); 57.2% (46.2-68.2); 27.7% (18.2-37.9); 15.1% (8.2-23.9) and 43.6% (32.5-54.7), respectively. Considering AML in remission, 2-year OS, DFS, RI, NRM and GRFS were 74.2% (62-86.5); 66.8% (53.6-79.9); 23.4% (12.7-36); 9.8% (3.5-19.9) and 50.9% (36.9-64.9), respectively. Two-year outcomes were similar between CloB2A1 and CloB2A2 sub-groups. In multivariate analysis, active disease at transplant was the only factor adversely impacting 2 years outcomes. Conclusions CloB2A2/A1 RIC regimen provides very good results for AML patients allografted in CR and could be retained as a new RIC platform for these patients. Materials and Methods This was a retrospective study including all patients who received a clofarabine/busulfan based RIC allo-SCT for myeloid malignancies and reported within the SFGM-TC registry. RIC regimen consisted of clofarabine 30 mg/m2/day 4 to 5 days (Clo), busulfan 3.2 mg/kg/day 2 days (B2) and 2.5 mg/kg/day of rabbit anti-thymocyte globulin 1 or 2 days (A1 or A2). The primary objective of the study was to report the main outcomes of the whole cohort at 2 years.
Collapse
Affiliation(s)
| | - Myriam Labopin
- Department of Hematology, Hôpital Saint Antoine, Paris, France
| | - Mathieu Leclerc
- Department of Hematology, Hôpital Henri Mondor, Créteil, France
| | | | | | - Patrice Ceballos
- Department of Hematology, CHU de Montpellier, Montpellier, France
| | | | | | - Karin Bilger
- Department of Hematology, CHU Strasbourg, Strasbourg, France
| | - Didier Blaise
- Department of Hematology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, Marseille, France
| | | | | | - Mohamad Mohty
- Department of Hematology, Hôpital Saint Antoine, Paris, France
| | | | - on behalf of Société Francophone de Greffe de Moelle et de Thérapie Cellulaire
- Department of Hematology, CHU Hôtel Dieu, Nantes, France
- Department of Hematology, Hôpital Saint Antoine, Paris, France
- Department of Hematology, Hôpital Henri Mondor, Créteil, France
- Department of Hematology, Hôpital Saint Louis, Université Paris 7, Denis Diderot, Paris, France
- Department of Hematology, Hôpital Gustave Roussy, Paris, France
- Department of Hematology, CHU de Montpellier, Montpellier, France
- Department of Hematology, CHU d’Angers, Angers, France
- Department of Hematology, Centre Hospitalier Lyon Sud, Lyon, France
- Department of Hematology, CHU Strasbourg, Strasbourg, France
- Department of Hematology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, Marseille, France
- Department of Hematology, CHU Nancy, Nancy, France
| |
Collapse
|
114
|
Villa NY, McFadden G. Virotherapy as Potential Adjunct Therapy for Graft-Vs-Host Disease. CURRENT PATHOBIOLOGY REPORTS 2018; 6:247-263. [PMID: 30595970 PMCID: PMC6290699 DOI: 10.1007/s40139-018-0186-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This review discusses the pathophysiology, risk factors, and the advances in the prevention or treatment of graft-vs-host disease (GvHD) by exploiting adjunct virotherapy. In addition, nonviral adjunct therapeutic options for the prevention of GvHD in the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT) are discussed. The role of oncolytic viruses to treat different HSCT-eligible hematological cancers is also considered and correlated with the issue of GvHD in the context of allo-HSCT. RECENT FINDINGS Emerging therapies focused on the prevention or treatment of GvHD include the use of regulatory T cells (Tregs), mesenchymal stem cells (MSCs), microbiome manipulation, B cell inhibitors, among others. Our lab and others have reported that an oncolytic DNA virus from the Poxviridae family, called myxoma virus (MYXV), not only exhibits oncolytic activity against various hematologic malignancies like multiple myeloma (MM) or acute myeloid leukemia (AML) but also, in addition, ex vivo MYXV treatment of human allogeneic-bone marrow transplants (allo-BMT), or allo-peripheral blood mononuclear cell (allo-PBMC) transplants can abrogate GvHD in xenografted mice without impairing graft-vs-tumor (GvT) effects against residual cancer. To date, this is the first and the only oncolytic virus with a dual potential of mediating oncolysis against a residual cancer target and also inhibiting or preventing GvHD following allo-HSCT. SUMMARY This review discusses how oncolytic virotherapy can be applied as a potential adjunct therapy for the potential treatment of GvHD. In addition, we highlight major emerging nonviral therapies currently studied for the treatment or prevention of GvHD. We also review the emerging oncolytic virotherapies against different hematological cancers currently eligible for allo-HSCT and highlight the potential role of the oncolytic virus MYXV to decrease GvHD while maintaining or enhancing the positive benefits of GvT.
Collapse
Affiliation(s)
- Nancy Y. Villa
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287 USA
| | - Grant McFadden
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85287 USA
| |
Collapse
|
115
|
Suen WCW, Lee WYW, Leung KT, Pan XH, Li G. Natural Killer Cell-Based Cancer Immunotherapy: A Review on 10 Years Completed Clinical Trials. Cancer Invest 2018; 36:431-457. [PMID: 30325244 DOI: 10.1080/07357907.2018.1515315] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
NK cell cancer immunotherapy is an emerging anti-tumour therapeutic strategy that explores NK cell stimulation. In this review, we address strategies developed to circumvent limitations to clinical application of NK cell-based therapies, and comprehensively review the design and results of clinical trials conducted in the past 10 years (2008-2018) to test their therapeutic potential. NK cell-based immunotherapy of solid cancers remains controversial, but merit further detailed investigation.
Collapse
Affiliation(s)
- Wade Chun-Wai Suen
- a Department of Orthopaedics and Traumatology, Faculty of Medicine , The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin , Hong Kong.,b Department of Orthopaedics and Traumatology , Bao-An People's Hospital , Shenzhen , PR China.,c Department of Haematology , University of Cambridge , Cambridge , UK
| | - Wayne Yuk-Wai Lee
- a Department of Orthopaedics and Traumatology, Faculty of Medicine , The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin , Hong Kong.,d Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences , The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin , Hong Kong
| | - Kam-Tong Leung
- e Department of Paediatrics, Faculty of Medicine , The Chinese University of Hong Kong , Shatin , Hong Kong
| | - Xiao-Hua Pan
- b Department of Orthopaedics and Traumatology , Bao-An People's Hospital , Shenzhen , PR China
| | - Gang Li
- a Department of Orthopaedics and Traumatology, Faculty of Medicine , The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin , Hong Kong.,d Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences , The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin , Hong Kong.,f The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System , The Chinese University of Hong Kong Shenzhen Research Institute , Shenzhen , PR China
| |
Collapse
|
116
|
Sheth V, Nachmias B, Grisariu S, Avni B, Or R, Shapira M. Augmented myeloablative conditioning with thiotepa in acute myeloid leukemia - improved outcomes with similar toxicity. Leuk Lymphoma 2018; 60:726-733. [PMID: 30277100 DOI: 10.1080/10428194.2018.1510495] [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] [Indexed: 10/28/2022]
Abstract
Myeloablative doses of busulfan (Bu) with fludarabine (Flu) have reduced toxicity, however, limited by an increased relapse rate. We aimed to improve outcome of Flu-Bu regimen by augmentation with thiotepa (TT) (10 mg/kg). Eighty-nine patients with AML, 44 patients conditioned with Flu-Bu (group 1), and 45 patients augmented with TT (Flu-Bu-TT, group 2), were retrospectively analyzed. Primary objectives were toxicity and outcomes. Major toxicities were comparable: mucositis (p = 1.0), sepsis (p = .7), severe venocclusive disease of liver (VOD) (p = 1.0), and non-relapse mortality (NRM) (22 vs. 22%, p = .7). Five-year disease-free survival was significantly better in group 2 compared to group 1 (62 vs. 38%, p = .02). Five-year overall survival (OS) showed trend toward benefit in group 2 (62 vs. 42%, p = .06). Lower relapse rate in group 2 (14 vs. 46%, p = .005) contributed to better outcomes. Augmented regimen has better disease-free survival (DFS) (mainly due to reduced relapse rate) and similar toxicities as compared to Flu-Bu. Key points Assessing the addition of TT to myeloablative conditioning (Flu, Bu) in patients undergoing allogeneic stem cell transplant for acute myeloid leukemia with regard to relapse rate, disease-free survival and toxicity. Addition of thiotepa improves disease-free survival and shows trend toward benefit in overall survival, by reducing relapses without additional toxicity.
Collapse
Affiliation(s)
- Vipul Sheth
- a Bone Marrow Transplantation and Cancer Immunotherapy Department , Hadassah - Ein Kerem , Jerusalem , Israel
| | - Boaz Nachmias
- a Bone Marrow Transplantation and Cancer Immunotherapy Department , Hadassah - Ein Kerem , Jerusalem , Israel
| | - Sigal Grisariu
- a Bone Marrow Transplantation and Cancer Immunotherapy Department , Hadassah - Ein Kerem , Jerusalem , Israel
| | - Batia Avni
- a Bone Marrow Transplantation and Cancer Immunotherapy Department , Hadassah - Ein Kerem , Jerusalem , Israel
| | - Reuven Or
- a Bone Marrow Transplantation and Cancer Immunotherapy Department , Hadassah - Ein Kerem , Jerusalem , Israel
| | - Michael Shapira
- a Bone Marrow Transplantation and Cancer Immunotherapy Department , Hadassah - Ein Kerem , Jerusalem , Israel
| |
Collapse
|
117
|
Saraceni F, Beohou E, Labopin M, Arcese W, Bonifazi F, Stepensky P, Aljurf M, Bruno B, Pioltelli P, Passweg J, Sociè G, Santarone S, Yakoub-Agha I, Lanza F, Savani BN, Mohty M, Nagler A. Thiotepa, busulfan and fludarabine compared to busulfan and cyclophosphamide as conditioning regimen for allogeneic stem cell transplant from matched siblings and unrelated donors for acute myeloid leukemia. Am J Hematol 2018; 93:1211-1219. [PMID: 30033639 DOI: 10.1002/ajh.25225] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 11/09/2022]
Abstract
Busulfan plus cyclophosphamide (BuCy) is the traditional conditioning regimen for allogeneic stem cell transplant (allo-SCT) for young, fit patients with acute myeloid leukemia (AML). The thiotepa-busulfan-fludarabine (TBF) protocol has recently demonstrated promising outcome in cord blood and haploidentical SCT; however, there is limited evidence about this regimen in transplant from matched siblings (MSD) and unrelated donors (UD). We retrospectively compared outcomes of 2523 patients aged 18-50 with AML in remission, undergoing transplant from MSD or UD prepared with either TBF or BuCy conditioning. A 1:3 pair-matched analysis was performed: 146 patients receiving TBF were compared with 438 patients receiving BuCy. Relapse risk was significantly lower in the TBF when compared with BuCy group (HR 0.6, P = .02), while NRM did not differ. No significant difference was observed in LFS and OS between the two regimens. TBF was associated with a trend towards higher risk of grades III-IV aGVHD (HR 1.8, P = .06) and inferior cGVHD (HR 0.7, P = .04) when compared with BuCy. In patients undergoing transplant in first remission, the advantage for TBF in terms of relapse was more evident (HR 0.4, P = .02), leading to a trend for better LFS in favor of TBF (HR 0.7, P = .10), while OS did not differ between the two cohorts. In conclusion, TBF represents a valid myeloablative conditioning regimen providing significantly lower relapse and similar survival when compared with BuCy. Patients in first remission appear to gain the most from this protocol, as in this subgroup a tendency for better LFS was observed when compared with BuCy.
Collapse
Affiliation(s)
- Francesco Saraceni
- Hematology and Stem Cell Transplant, Romagna Transplant Network; Ravenna Italy
| | - Eric Beohou
- EBMT Paris Study Office, Saint Antoine Hospital; Paris France
| | - Myriam Labopin
- EBMT Paris Study Office, Saint Antoine Hospital; Paris France
| | - William Arcese
- Tor Vergata University of Rome, Stem Cell Transplant Unit, Policlinico Universitario Tor Vergata; Rome Italy
| | - Francesca Bonifazi
- Bologna University, S. Orsola-Malpighi Hospital, Institute of Hematology L and A Seràgnoli; Bologna Italy
| | - Polina Stepensky
- Department of Bone Marrow Transplantation; Hadassah University Hospital; Jerusalem Israel
| | - Mahmoud Aljurf
- King Faisal Specialist Hospital and Research Centre, Oncology (Section of Adult Haematolgy/BMT); Riyadh Saudi Arabia
| | - Benedetto Bruno
- S.S.C.V.D Trapianto di Cellule Staminali, A.O.U Citta della Salute e della Scienza di Torino; Torino Italy
| | - Pietro Pioltelli
- Ospedale San Gerardo, Clinica Ematologica dell'Universita Milano-Bicocca; Monza Italy
| | - Jakob Passweg
- Department of Hematology; University Hospital; Basel Switzerland
| | - Gerard Sociè
- Department of Hematology; BMT, Hospital St. Louis; Paris France
| | - Stella Santarone
- Ospedale Civile, Dipartimento di Ematologia, Medicina Trasfusionale e Biotecnologie; Pescara Italy
| | | | - Francesco Lanza
- Hematology and Stem Cell Transplant, Romagna Transplant Network; Ravenna Italy
| | | | - Mohamad Mohty
- Department of Haematology; Saint Antoine Hospital; Paris France
| | - Arnon Nagler
- EBMT Paris Study Office, Saint Antoine Hospital; Paris France
- Department of Bone Marrow Transplantation; Chaim Sheba Medical Center; Tel-Hashomer Israel
| | | |
Collapse
|
118
|
Chhabra S, Liu Y, Hemmer MT, Costa L, Pidala JA, Couriel DR, Alousi AM, Majhail NS, Stuart RK, Kim D, Ringden O, Urbano-Ispizua A, Saad A, Savani BN, Cooper B, Marks DI, Socie G, Schouten HC, Schoemans H, Abdel-Azim H, Yared J, Cahn JY, Wagner J, Antin JH, Verdonck LF, Lehmann L, Aljurf MD, MacMillan ML, Litzow MR, Solh MM, Qayed M, Hematti P, Kamble RT, Vij R, Hayashi RJ, Gale RP, Martino R, Seo S, Hashmi SK, Nishihori T, Teshima T, Gergis U, Inamoto Y, Spellman SR, Arora M, Hamilton BK. Comparative Analysis of Calcineurin Inhibitor-Based Methotrexate and Mycophenolate Mofetil-Containing Regimens for Prevention of Graft-versus-Host Disease after Reduced-Intensity Conditioning Allogeneic Transplantation. Biol Blood Marrow Transplant 2018; 25:73-85. [PMID: 30153491 DOI: 10.1016/j.bbmt.2018.08.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/09/2018] [Indexed: 01/11/2023]
Abstract
The combination of a calcineurin inhibitor (CNI) such as tacrolimus (TAC) or cyclosporine (CYSP) with methotrexate (MTX) or with mycophenolate mofetil (MMF) has been commonly used for graft-versus-host disease (GVHD) prophylaxis after reduced-intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (alloHCT), but there are limited data comparing efficacy of the 2 regimens. We evaluated 1564 adult patients who underwent RIC alloHCT for acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML), and myelodysplastic syndrome (MDS) from 2000 to 2013 using HLA-identical sibling (matched related donor [MRD]) or unrelated donor (URD) peripheral blood graft and received CYSP or TAC with MTX or MMF for GVHD prophylaxis. Primary outcomes of the study were acute and chronic GVHD and overall survival (OS). The study divided the patient population into 4 cohorts based on regimen: MMF-TAC, MMF-CYSP, MTX-TAC, and MTX-CYSP. In the URD group, MMF-CYSP was associated with increased risk of grade II to IV acute GVHD (relative risk [RR], 1.78; P < .001) and grade III to IV acute GVHD (RR, 1.93; P = .006) compared with MTX-TAC. In the URD group, use of MMF-TAC (versus MTX-TAC) lead to higher nonrelapse mortality. (hazard ratio, 1.48; P = .008). In either group, no there was no difference in chronic GVHD, disease-free survival, and OS among the GVHD prophylaxis regimens. For RIC alloHCT using MRD, there are no differences in outcomes based on GVHD prophylaxis. However, with URD RIC alloHCT, MMF-CYSP was inferior to MTX-based regimens for acute GVHD prevention, but all the regimens were equivalent in terms of chronic GVHD and OS. Prospective studies, targeting URD recipients are needed to confirm these results.
Collapse
Affiliation(s)
- Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.
| | - Ying Liu
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Institute for Health and Society, Department of Biostatistics, 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
| | - Luciano Costa
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joseph A Pidala
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Daniel R Couriel
- Division of Hematology and Hematologic Malignancies, Utah Blood and Marrow Transplant Program, Salt Lake City, Utah
| | - Amin M Alousi
- Department of Stem Cell Transplantation, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Navneet S Majhail
- Blood & Marrow Transplant Program, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | - Robert K Stuart
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Dennis Kim
- Department of Medical Oncology & Hematology, Princess Margaret Cancer Center, Toronto, Canada
| | - Olle Ringden
- Division of Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alvaro Urbano-Ispizua
- Department of Hematology, IDIBAPS, Institute of Research Josep Carreras, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Ayman Saad
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brenda Cooper
- Department of Medicine-Hematology and Oncology, University Hospitals Case Medical Center, Cleveland, Ohio
| | - David I Marks
- Adult Bone Marrow Transplant, University Hospitals Bristol NHS Trust, Bristol, United Kingdom
| | - Gerard Socie
- Department of Hematology, Hôpital Saint Louis, Paris, France
| | - Harry C Schouten
- Department of Hematology, Academische Ziekenhuis, Maastricht, the Netherlands
| | - Helene Schoemans
- Department of Hematology, 26-Department of Pediatrics, University Hospital of Leuven, Leuven, Belgium
| | - Hisham Abdel-Azim
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Jean Yared
- Blood & Marrow Transplantation Program, Division of Hematology/Oncology, Department of Medicine, Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland
| | - Jean-Yves Cahn
- Department of Hematology, CHU Grenoble Alpes, Grenoble, France
| | - John Wagner
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Joseph H Antin
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Leo F Verdonck
- Departmentt of Hematology/Oncology, Isala Clinic, Zwolle, the Netherlands
| | - Leslie Lehmann
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mahmoud D Aljurf
- Department of Oncology, King Faisal Specialist Hospital Center & Research, Riyadh, Saudi Arabia
| | - Margaret L MacMillan
- University of Minnesota Blood and Marrow Transplant Program, Minneapolis, Minnesota
| | - Mark R Litzow
- Division of Hematology and Transplant Center, Mayo Clinic Rochester, Rochester, Minnesota
| | - Melhem M Solh
- Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, Georgia
| | - Muna Qayed
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - 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
| | - Ravi Vij
- Division of Hematology and Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Robert J Hayashi
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Robert P Gale
- Hematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Rodrigo Martino
- Divison of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Sachiko Seo
- Department of Hematology & Oncology, National Cancer Research Center East, Chiba, Japan
| | - Shahrukh K Hashmi
- Department of Oncology, King Faisal Specialist Hospital Center & Research, Riyadh, Saudi Arabia; Division of Hematology and Transplant Center, Mayo Clinic Rochester, Rochester, Minnesota
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Takanori Teshima
- Department of Hematology Kyushu University Hospital, Sapporo, Japan
| | - Usama Gergis
- Hematolgic Malignancies & Bone Marrow Transplant, Department of Medical Oncology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - Yoshihiro Inamoto
- Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be The Match, Minneapolis, Minnesota
| | - Mukta Arora
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, Minnesota.
| | - Betty K Hamilton
- Blood & Marrow Transplant Program, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| |
Collapse
|
119
|
Inman CF, Eldershaw SA, Croudace JE, Davies NJ, Sharma-Oates A, Rai T, Pearce H, Sirovica M, Chan YLT, Verma K, Zuo J, Nagra S, Kinsella F, Nunnick J, Amel-Kashipaz R, Craddock C, Malladi R, Moss P. Unique features and clinical importance of acute alloreactive immune responses. JCI Insight 2018; 3:97219. [PMID: 29769441 PMCID: PMC6012511 DOI: 10.1172/jci.insight.97219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 04/19/2018] [Indexed: 01/22/2023] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) can cure some patients with hematopoietic malignancy, but this relies on the development of a donor T cell alloreactive immune response. T cell activity in the first 2 weeks after allo-SCT is crucial in determining outcome, despite the clinical effects of the early alloreactive immune response often not appearing until later. However, the effect of the allogeneic environment on T cells is difficult to study at this time point due to the effects of profound lymphopenia. We approached this problem by comparing T cells at week 2 after allograft to T cells from autograft patients. Allograft T cells were present in small numbers but displayed intense proliferation with spontaneous cytokine production. Oligoclonal expansions at week 2 came to represent a substantial fraction of the established T cell pool and were recruited into tissues affected by graft-versus-host disease. Transcriptional analysis uncovered a range of potential targets for immune manipulation, including OX40L, TWEAK, and CD70. These findings reveal that recognition of alloantigen drives naive T cells toward a unique phenotype. Moreover, they demonstrate that early clonal T cell responses are recruited to sites of subsequent tissue damage and provide a range of targets for potential therapeutic immunomodulation. Alloreactive response T cells at 2 weeks after allo-SCT displayed intense proliferation with spontaneous cytokine production, and were recruited into tissues affected by GvHD.
Collapse
Affiliation(s)
- Charlotte F Inman
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Suzy A Eldershaw
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Joanne E Croudace
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Nathaniel J Davies
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Archana Sharma-Oates
- Centre for Computational Biology, Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Tanuja Rai
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Mirjana Sirovica
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Y L Tracey Chan
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Kriti Verma
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Sandeep Nagra
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Francesca Kinsella
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and
| | - Jane Nunnick
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Rasoul Amel-Kashipaz
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Charles Craddock
- Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Ram Malladi
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and.,Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, and.,Birmingham Health Partners, Department of Haematology, Queen Elizabeth Hospital, Birmingham, United Kingdom
| |
Collapse
|
120
|
How and when I do allogeneic transplant in CLL. Blood 2018; 132:31-39. [PMID: 29752258 DOI: 10.1182/blood-2018-01-785998] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/02/2018] [Indexed: 12/11/2022] Open
Abstract
Allogenic stem cell transplantation (allo-SCT) has been considered the treatment of choice for high-risk patients with chronic lymphocytic leukemia (CLL) and the only approach offered with curative intent in this disease. The availability novel agents, including the B-cell receptor inhibitors ibrutinib, acalabrutinib, and idelalisib, as well as venetoclax, which targets the BCL2 pathway, and the success of these agents in treating high-risk disease patients have made it more difficult to assess who should be considered for allo-SCT and when in the treatment course. In this review, I will discuss the different treatment options available for the treatment of high-risk CLL and how allo-SCT fits into the treatment algorithm in the era of novel agents.
Collapse
|
121
|
Peccatori J, Bregni M. Allogeneic Non-Myeloablative Peripheral-Blood Stem Cell Transplant in Solid Tumors. TUMORI JOURNAL 2018; 1:S32-3. [PMID: 12658902 DOI: 10.1177/03008916020016s111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Jacopo Peccatori
- Division of Hematology and Bone Marrow Transplant Unit, San Raffaele Institute, Milan, Italy
| | | |
Collapse
|
122
|
McCune JS, Storer B, Thomas S, McKiernan J, Gupta R, Sandmaier BM. Inosine Monophosphate Dehydrogenase Pharmacogenetics in Hematopoietic Cell Transplantation Patients. Biol Blood Marrow Transplant 2018; 24:1802-1807. [PMID: 29656138 DOI: 10.1016/j.bbmt.2018.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/03/2018] [Indexed: 01/18/2023]
Abstract
We evaluated inosine monophosphate dehydrogenase (IMPDH) 1 and IMPDH2 pharmacogenetics in 247 recipient-donor pairs after nonmyeloablative hematopoietic cell transplant (HCT). Patients were conditioned with total body irradiation + fludarabine and received grafts from related or unrelated donors (10% HLA mismatch), with postgraft immunosuppression of mycophenolate mofetil (MMF) with a calcineurin inhibitor. Recipient and donor IMPDH genotypes (rs11706052, rs2278294, rs2278293) were not associated with day 28 T cell chimerism, acute graft-versus-host disease (GVHD), disease relapse, cytomegalovirus reactivation, nonrelapse mortality, or overall survival. Recipient IMPDH1 rs2278293 genotype was associated with a lower incidence of chronic GVHD (hazard ratio, .72; P = .008) in nonmyeloablative HCT recipients. Additional studies are needed to confirm these results with the goal of identifying predictive biomarkers to MMF that lower GVHD.
Collapse
Affiliation(s)
- Jeannine S McCune
- School of Pharmacy, University of Washington, Seattle, Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Population Sciences, City of Hope, Duarte, California; Department of Hematology and HCT, City of Hope, Duarte, California.
| | - Barry Storer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sushma Thomas
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jožefa McKiernan
- Department of Population Sciences, City of Hope, Duarte, California
| | - Rohan Gupta
- Department of Hematology and HCT, City of Hope, Duarte, California
| | - Brenda M Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
123
|
Baron F, Labopin M, Ruggeri A, Cornelissen JJ, Meijer E, Sengeloev H, Niederwieser D, De Groot MR, Schouten HC, Milpied N, Blaise D, Savani BN, Gluckman E, Mohty M, Nagler A. Impact of Donor Type in Patients with AML Given Allogeneic Hematopoietic Cell Transplantation After Low-Dose TBI-Based Regimen. Clin Cancer Res 2018; 24:2794-2803. [PMID: 29555662 DOI: 10.1158/1078-0432.ccr-17-3622] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/02/2018] [Accepted: 03/13/2018] [Indexed: 11/16/2022]
Abstract
Purpose: We assessed the impact of donor type in acute myeloid leukemia (AML) patients transplanted with 2 Gy total body irradiation (TBI)-based nonmyeloablative conditioning regimen.Patients and Methods: Data from 1,715 adult patients, with AML in CR1 or CR2 were included in this retrospective survey.Results: Donors consisted either of HLA-matched sibling donors (MSD, n = 701), 10/10 HLA-matched unrelated donors (MUD, n = 611), HLA-haploidentical donors (haplo, n = 112) or single or double umbilical cord bloods (CBT, n = 291). Chronic graft-versus-host disease (GVHD) was less frequent in CBT (28%) and in haplo (30%) patients than in MSD (50%) and MUD (51%) recipients (P < 0.001). Two-year incidence of relapse was 32%, 30%, 34%, and 34% in MSD, MUD, CBT and haplo patients, respectively (P = 0.7). Two-year overall (OS) and GVHD-free relapse-free survival (GRFS) were 59% and 29% in MSD patients, 56% and 39% in CBT recipients, 53% and 23% in MUD recipients, and 43% and 37% in haplo patients, respectively. In multivariate analyses, MUD patients had lower GRFS than MSD patients beyond day 100 (HR 1.3, P = 0.001) while CBT was associated with a better GRFS than MSD beyond day 100 (HR 0.6, P = 0.002).Conclusions: In this large cohort of AML patients transplanted following low-dose TBI-based conditioning, the relapse incidence was not affected by donor type suggesting that the intensity of GVL effects might be comparable with these four transplant approaches. Furthermore, CBT was associated with better GRFS beyond day 100 than MSD while the opposite was observed for MUD. Clin Cancer Res; 24(12); 2794-803. ©2018 AACR.
Collapse
Affiliation(s)
- Frédéric Baron
- GIGA and CHU of Liege, University of Liege, Liege, Belgium.
| | - Myriam Labopin
- EBMT Paris study office/CEREST-TC, Paris, France.,Department of Haematology, Saint Antoine Hospital, Paris, France.,INSERM UMR 938, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Annalisa Ruggeri
- Department of Haematology, Saint Antoine Hospital, Paris, France.,Eurocord, Saint Louis Hospital, Paris, France.,Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's, Roma, Italy
| | - Jan J Cornelissen
- Erasmus MC Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, the Netherlands
| | - Ellen Meijer
- VU University Medical Center, Department of Hematology, Amsterdam, the Netherlands
| | - Henrik Sengeloev
- Bone Marrow Transplant Unit L 4043, National University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Dietger Niederwieser
- University Hospital Leipzig, Division of Haematology & Oncology, Leipzig, Germany
| | - Marco R De Groot
- University Medical Center Groningen (UMCG), Dept. of Hematology, Groningen, the Netherlands
| | - Harry C Schouten
- University Hospital Maastricht, Dept. Internal Med. Hematology, Maastricht, the Netherlands
| | - Noel Milpied
- CHU Bordeaux, Hôpital Haut-Leveque, Pessac, France
| | - Didier Blaise
- Programme de Transplantation & Therapie Cellulaire, Centre de Recherche en Cancérologie de Marseille, Institut Paoli Calmettes, Marseille, France
| | - Bipin N Savani
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Mohamad Mohty
- EBMT Paris study office/CEREST-TC, Paris, France.,Department of Haematology, Saint Antoine Hospital, Paris, France.,INSERM UMR 938, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Arnon Nagler
- EBMT Paris study office/CEREST-TC, Paris, France.,Division of Hematology and Bone Marrow Transplantation, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| |
Collapse
|
124
|
Mesenchymal Stromal Cell Irradiation Interferes with the Adipogenic/Osteogenic Differentiation Balance and Improves Their Hematopoietic-Supporting Ability. Biol Blood Marrow Transplant 2018; 24:443-451. [DOI: 10.1016/j.bbmt.2017.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/02/2017] [Indexed: 12/19/2022]
|
125
|
Retière C, Willem C, Guillaume T, Vié H, Gautreau-Rolland L, Scotet E, Saulquin X, Gagne K, Béné MC, Imbert BM, Clemenceau B, Peterlin P, Garnier A, Chevallier P. Impact on early outcomes and immune reconstitution of high-dose post-transplant cyclophosphamide vs anti-thymocyte globulin after reduced intensity conditioning peripheral blood stem cell allogeneic transplantation. Oncotarget 2018; 9:11451-11464. [PMID: 29545911 PMCID: PMC5837739 DOI: 10.18632/oncotarget.24328] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 11/01/2017] [Indexed: 11/29/2022] Open
Abstract
We have compared prospectively the outcome and immune reconstitution of patients receiving either post-transplant cyclophosphamide (PTCY) (n = 30) or anti-thymocyte globulin ATG (n = 15) as Graft-versus-host disease (GVHD) prophylaxis after reduced-intensity conditioning (RIC) allogeneic peripheral blood stem cell (PBSC) transplantation (allo-SCT). The outcome and immune reconstitution of patients receiving either of these two regimens were compared prospectively. This study allowed also to investigate the impact of PTCY between haplo-identical vs matched donors and of clofarabine as part of the RIC regimen. The γ/δ T-cells, α/β T-cells (CD8+ and CD4+), NK T-cells, NK cells, B-cells, Tregs and monocytes were analyzed by flow cytometry from a total of 583 samples. In the PTCY group significant delayed platelets recovery, higher CD3+ donor chimerism, higher HHV-6 and lower EBV reactivations were observed. Early survival advantage for CD4+ T-cells, Tregs and α/β T-cells was documented in the PTCY group while it was the case for α/β T-cells, NK cells and monocytes in the ATG group. Higher counts of NK and monocytes were observed at days +30 and/or day+60 in the ATG group. Both results were retained even in the case of mismatched donors. However, higher percentages of CD4+ T-cells, α/β T-cells and Tregs were observed with haplo-identical donors in the PTCY group. Finally, clofarabine was responsible for early survival advantage of NK T-cells in the PTCY group while it abrogated the early survival advantage of γ/δ T-cells in the ATG group. In conclusion, there are marked differences in the immunological effects of ATG vs PTCY as GVHD prophylaxis for RIC PBSC allo-SCT.
Collapse
Affiliation(s)
| | - Catherine Willem
- Etablissement Français du Sang, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Thierry Guillaume
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Hematology Department, CHU, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Henri Vié
- Etablissement Français du Sang, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Laetitia Gautreau-Rolland
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Emmanuel Scotet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Xavier Saulquin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Katia Gagne
- Etablissement Français du Sang, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,LabEx Transplantex, Université de Strasbourg, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Marie C Béné
- Hematology/Biology Department, CHU, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Berthe-Marie Imbert
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR1064, Université de Nantes, Nantes, France.,Service de Virologie, CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | - Beatrice Clemenceau
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| | | | | | - Patrice Chevallier
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Hematology Department, CHU, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, F-44000, France
| |
Collapse
|
126
|
Schmalbrock LK, Bonifacio L, Bill M, Jentzsch M, Schubert K, Grimm J, Cross M, Lange T, Vucinic V, Pönisch W, Behre G, Franke GN, Niederwieser D, Schwind S. Prognostic relevance of DNMT3A R882 mutations in AML patients undergoing non-myeloablative conditioning hematopoietic stem cell transplantation. Bone Marrow Transplant 2018; 53:640-643. [PMID: 29335621 DOI: 10.1038/s41409-017-0060-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/15/2017] [Accepted: 11/09/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Laura K Schmalbrock
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Lynn Bonifacio
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Marius Bill
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Madlen Jentzsch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Karoline Schubert
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Juliane Grimm
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Michael Cross
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Thoralf Lange
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Vladan Vucinic
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Wolfram Pönisch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Gerhard Behre
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | | | | | - Sebastian Schwind
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
127
|
|
128
|
Mahr B, Granofszky N, Muckenhuber M, Wekerle T. Transplantation Tolerance through Hematopoietic Chimerism: Progress and Challenges for Clinical Translation. Front Immunol 2017; 8:1762. [PMID: 29312303 PMCID: PMC5743750 DOI: 10.3389/fimmu.2017.01762] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
The perception that transplantation of hematopoietic stem cells can confer tolerance to any tissue or organ from the same donor is widely accepted but it has not yet become a treatment option in clinical routine. The reasons for this are multifaceted but can generally be classified into safety and efficacy concerns that also became evident from the results of the first clinical pilot trials. In comparison to standard immunosuppressive therapies, the infection risk associated with the cytotoxic pre-conditioning necessary to allow allogeneic bone marrow engraftment and the risk of developing graft-vs.-host disease (GVHD) constitute the most prohibitive hurdles. However, several approaches have recently been developed at the experimental level to reduce or even overcome the necessity for cytoreductive conditioning, such as costimulation blockade, pro-apoptotic drugs, or Treg therapy. But even in the absence of any hazardous pretreatment, the recipients are exposed to the risk of developing GVHD as long as non-tolerant donor T cells are present. Total lymphoid irradiation and enriching the stem cell graft with facilitating cells emerged as potential strategies to reduce this peril. On the other hand, the long-lasting survival of kidney allografts, seen with transient chimerism in some clinical series, questions the need for durable chimerism for robust tolerance. From a safety point of view, loss of chimerism would indeed be favorable as it eliminates the risk of GVHD, but also complicates the assessment of tolerance. Therefore, other biomarkers are warranted to monitor tolerance and to identify those patients who can safely be weaned off immunosuppression. In addition to these safety concerns, the limited efficacy of the current pilot trials with approximately 40-60% patients becoming tolerant remains an important issue that needs to be resolved. Overall, the road ahead to clinical routine may still be rocky but the first successful long-term patients and progress in pre-clinical research provide encouraging evidence that deliberately inducing tolerance through hematopoietic chimerism might eventually make it from dream to reality.
Collapse
Affiliation(s)
- Benedikt Mahr
- Department of Surgery, Section of Transplantation Immunology, Medical University of Vienna, Vienna, Austria
| | - Nicolas Granofszky
- Department of Surgery, Section of Transplantation Immunology, Medical University of Vienna, Vienna, Austria
| | - Moritz Muckenhuber
- Department of Surgery, Section of Transplantation Immunology, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Department of Surgery, Section of Transplantation Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
129
|
Wang Y, Niu ZY, Guo YJ, Wang LH, Lin FR, Zhang JY. IL-11 promotes the treatment efficacy of hematopoietic stem cell transplant therapy in aplastic anemia model mice through a NF-κB/microRNA-204/thrombopoietin regulatory axis. Exp Mol Med 2017; 49:e410. [PMID: 29217821 PMCID: PMC5750475 DOI: 10.1038/emm.2017.217] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/30/2017] [Accepted: 06/07/2017] [Indexed: 12/26/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation could be of therapeutic value for aplastic anemia (AA) patients, and immunosuppressants may facilitate the efficiency of the procedure. As anti-inflammatory cytokine interleukin-11 (IL-11) has a thrombopoietic effect, its use in cases of chronic bone marrow failure, such as AA, has been proposed to induce HSC function. However, the putative mechanisms that may support this process remain poorly defined. We found that decreased miR-204-5p levels were coincident with increased proliferation in mouse HSCs following exposure to IL-11 in vitro. Through inhibiting NF-кB activity, miR-204-5p repression was demonstrated to be a downstream effect of IL-11 signaling. miR-204-5p was shown to directly target thrombopoietin (TPO) via sequence-dependent 3′-UTR repression, indicating that this microRNA-dependent pathway could serve an essential role in supporting IL-11 functions in HSCs. Increased TPO expression in HSCs following IL-11 exposure could be mimicked or blocked by inhibiting or overexpressing miR-204-5p, respectively. Consistent with these in vitro findings, IL-11 promoted HSC engraftment in a mouse model of AA, an effect that was attenuated in cells overexpressing miR-204-5p. The reduction in miR-204-5p levels is an integral component of IL-11 signaling that may play an essential role in treating AA.
Collapse
Affiliation(s)
- Yan Wang
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Zhi-Yun Niu
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Yu-Jie Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Li-Hua Wang
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Feng-Ru Lin
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| | - Jing-Yu Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, China
| |
Collapse
|
130
|
Allogeneic bone marrow transplant in the absence of cytoreductive conditioning rescues mice with β-thalassemia major. Blood Adv 2017; 1:2421-2432. [PMID: 29296892 DOI: 10.1182/bloodadvances.2017009449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/24/2017] [Indexed: 02/08/2023] Open
Abstract
β-thalassemia is a group of inherited blood disorders that result in defects in β-globin chain production. Cooley anemia (CA), or β-thalassemia major, is the most severe form of the disease and occurs when an individual has mutations in both copies of the adult β-globin gene. Patients with CA fail to make adult hemoglobin, exhibit ineffective erythropoiesis, experience severe anemia, and are transfusion dependent for life. Currently, allogeneic bone marrow transplantation (BMT) is the only cure; however, few patients have suitable donors for this procedure, which has significant morbidity and mortality. In this study, a novel humanized murine model of CA is rescued from lethal anemia by allogeneic BMT in the absence of cytoreductive conditioning. A single intravenous postnatal injection of allogeneic bone marrow results in stable, mixed hematopoietic chimerism. Five months after transplantation, donor cells accounted for approximately 90% of circulating erythrocytes and up to 15% of hematopoietic stem and progenitor cells. Transplanted mice are transfusion independent, have marked improvement of hematological indices, exhibit no growth retardation or signs of graft-versus-host disease, and are fertile. This study describes a method for the consistent engraftment of allogeneic donor hematopoietic cells that rescues a humanized mouse model of CA from lethal anemia, all in the absence of toxic cytoreductive conditioning.
Collapse
|
131
|
Takeuchi A, Kato K, Akashi K, Eto M. Cyclophosphamide-induced tolerance in kidney transplantation avoids long-term immunosuppressive therapy. Int J Urol 2017; 25:112-120. [PMID: 29105189 DOI: 10.1111/iju.13474] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 08/22/2017] [Indexed: 12/28/2022]
Abstract
There has recently been remarkable progress in immunosuppressive agents, such as tacrolimus and cyclosporine. Therefore, the rate of organ establishment has improved in transplantation. However, immunosuppressive agents generally suppress the function of T cells. Thus, opportunistic infections, such as cytomegalovirus infection, are still a major problem in kidney transplantation. Induction of specific tolerance to avoid immunosuppressive drug therapy after kidney transplantation is considered as the ultimate goal of transplantation. Various factors induce tolerance that involves establishment of hematopoietic chimerism and various cell subsets. In particular, we have carried out various studies regarding the cyclophosphamide-induced tolerance system. Tolerance is induced after establishment of hematopoietic chimerism after donor bone marrow transplantation. At the clinical stage, kidney transplantation before administration of cyclophosphamide after transfusion of bone marrow to create hematopoietic chimera is considered to be one of the most successful protocols. Furthermore, recent studies have shown the involvement of multiple populations of immune cells in preserving immunological tolerance and promoting long-term renal grafts. The present review focuses on how cyclophosphamide and other immune factors induce tolerance in kidney transplantation.
Collapse
Affiliation(s)
- Ario Takeuchi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Kato
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
132
|
Jentzsch M, Bill M, Grimm J, Schulz J, Goldmann K, Beinicke S, Häntschel J, Pönisch W, Franke GN, Vucinic V, Behre G, Lange T, Niederwieser D, Schwind S. High BAALC copy numbers in peripheral blood prior to allogeneic transplantation predict early relapse in acute myeloid leukemia patients. Oncotarget 2017; 8:87944-87954. [PMID: 29152132 PMCID: PMC5675684 DOI: 10.18632/oncotarget.21322] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
High BAALC expression levels at acute myeloid leukemia diagnosis have been linked to adverse outcomes. Recent data indicate that high BAALC expression levels may also be used as marker for residual disease following acute myeloid leukemia treatment. Allogeneic hematopoietic stem cell transplantation (HSCT) offers a curative treatment for acute myeloid leukemia patients. However, disease recurrence remains a major clinical challenge and identification of high-risk patients prior to HSCT is crucial to improve outcomes. We performed absolute quantification of BAALC copy numbers in peripheral blood prior (median 7 days) to HSCT in complete remission (CR) or CR with incomplete peripheral recovery in 82 acute myeloid leukemia patients using digital droplet PCR (ddPCR) technology. An optimal cut-off of 0.14 BAALC/ABL1 copy numbers was determined and applied to define patients with high or low BAALC/ABL1 copy numbers. High pre-HSCT BAALC/ABL1 copy numbers significantly associated with higher cumulative incidence of relapse and shorter overall survival in univariable and multivariable models. Patients with high pre-HSCT BAALC/ABL1 copy numbers were more likely to experience relapse within 100 days after HSCT. Evaluation of pre-HSCT BAALC/ABL1 copy numbers in peripheral blood by ddPCR represents a feasible and rapid way to identify acute myeloid leukemia patients at high risk of early relapse after HSCT. The prognostic impact was also observed independently of other known clinical, genetic, and molecular prognosticators. In the future, prospective studies should evaluate whether acute myeloid leukemia patients with high pre-HSCT BAALC/ABL1 copy numbers benefit from additional treatment before or early intervention after HSCT.
Collapse
Affiliation(s)
- Madlen Jentzsch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Marius Bill
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Juliane Grimm
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Julia Schulz
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Karoline Goldmann
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Stefanie Beinicke
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Janine Häntschel
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Wolfram Pönisch
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | | | - Vladan Vucinic
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Gerhard Behre
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | - Thoralf Lange
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| | | | - Sebastian Schwind
- Department of Hematology and Oncology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
133
|
Muffly L, Pasquini MC, Martens M, Brazauskas R, Zhu X, Adekola K, Aljurf M, Ballen KK, Bajel A, Baron F, Battiwalla M, Beitinjaneh A, Cahn JY, Carabasi M, Chen YB, Chhabra S, Ciurea S, Copelan E, D'Souza A, Edwards J, Foran J, Freytes CO, Fung HC, Gale RP, Giralt S, Hashmi SK, Hildebrandt GC, Ho V, Jakubowski A, Lazarus H, Luskin MR, Martino R, Maziarz R, McCarthy P, Nishihori T, Olin R, Olsson RF, Pawarode A, Peres E, Rezvani AR, Rizzieri D, Savani BN, Schouten HC, Sabloff M, Seftel M, Seo S, Sorror ML, Szer J, Wirk BM, Wood WA, Artz A. Increasing use of allogeneic hematopoietic cell transplantation in patients aged 70 years and older in the United States. Blood 2017; 130:1156-1164. [PMID: 28674027 PMCID: PMC5580273 DOI: 10.1182/blood-2017-03-772368] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/19/2017] [Indexed: 12/20/2022] Open
Abstract
In this study, we evaluated trends and outcomes of allogeneic hematopoietic cell transplantation (HCT) in adults ≥70 years with hematologic malignancies across the United States. Adults ≥70 years with a hematologic malignancy undergoing first allogeneic HCT in the United States between 2000 and 2013 and reported to the Center for International Blood and Marrow Transplant Research were eligible. Transplant utilization and transplant outcomes, including overall survival (OS), progression-free survival (PFS), and transplant-related mortality (TRM) were studied. One thousand one hundred and six patients ≥70 years underwent HCT across 103 transplant centers. The number and proportion of allografts performed in this population rose markedly over the past decade, accounting for 0.1% of transplants in 2000 to 3.85% (N = 298) in 2013. Acute myeloid leukemia and myelodysplastic syndromes represented the most common disease indications. Two-year OS and PFS significantly improved over time (OS: 26% [95% confidence interval (CI), 21% to 33%] in 2000-2007 to 39% [95% CI, 35% to 42%] in 2008-2013, P < .001; PFS: 22% [16% to 28%] in 2000-2007 to 32% [95% CI, 29% to 36%] in 2008-2013, P = .003). Two-year TRM ranged from 33% to 35% and was unchanged over time (P = .54). Multivariable analysis of OS in the modern era of 2008-2013 revealed higher comorbidity by HCT comorbidity index ≥3 (hazard ratio [HR], 1.27; P = .006), umbilical cord blood graft (HR, 1.97; P = .0002), and myeloablative conditioning (HR, 1.61; P = .0002) as adverse factors. Over the past decade, utilization and survival after allogeneic transplant have increased in patients ≥70 years. Select adults ≥70 years with hematologic malignancies should be considered for transplant.
Collapse
Affiliation(s)
- Lori Muffly
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | | | - Michael Martens
- Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, WI
| | - Ruta Brazauskas
- Center for International Blood and Marrow Transplant Research and
- Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, WI
| | - Xiaochun Zhu
- Center for International Blood and Marrow Transplant Research and
| | | | - Mahmoud Aljurf
- Department of Oncology, King Faisal Specialist Hospital Center and Research, Riyadh, Saudi Arabia
| | - Karen K Ballen
- Department of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Ashish Bajel
- Royal Melbourne Hospital, Victoria, VIC, Australia
| | - Frederic Baron
- Centre Hospitalier Universitaire de Liege, Domaine Universitaire du Sart Tilman, Liege, Belgium
| | - Minoo Battiwalla
- Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Amer Beitinjaneh
- Department of Hematology and Oncology, University of Miami, Miami, FL
| | - Jean-Yves Cahn
- Department of Hematology, University Hospital, Grenoble, France
| | - Mathew Carabasi
- Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, PA
| | - Yi-Bin Chen
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Saurabh Chhabra
- Department of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Stefan Ciurea
- Department of Stem Cell Transplantation and Cellular Therapy and
- Transplant Myeloid Study Group, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Edward Copelan
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC
| | - Anita D'Souza
- Center for International Blood and Marrow Transplant Research and
| | - John Edwards
- Indiana Blood and Marrow Transplantation, Indianapolis, IN
| | | | | | - Henry C Fung
- Department of Medical Oncology, Fox Chase Cancer Center, Temple Health, Philadelphia, PA
| | - Robert Peter Gale
- Hematology Research Centre, Division of Experimental Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| | - Sergio Giralt
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Shahrukh K Hashmi
- Department of Internal Medicine, Mayo Clinic, Minneapolis, MN
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Gerhard C Hildebrandt
- Department of Internal Medicine, University of Kentucky Chandler Medical Center, Lexington, KY
| | - Vincent Ho
- Center for Hematologic Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Hillard Lazarus
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Marlise R Luskin
- Abramson Cancer Center, University of Pennsylvania Medical Center, Philadelphia, PA
| | - Rodrigo Martino
- Divison of Clinical Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Richard Maziarz
- Adult Blood and Marrow Stem Cell Transplant Program, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Philip McCarthy
- Blood and Marrow Transplant Program, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY
| | - Taiga Nishihori
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Rebecca Olin
- Department of Medicine, University of California San Francisco Medical Center, San Francisco, CA
| | - Richard F Olsson
- Division of Therapeutic Immunology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Clinical Research Sormland, Uppsala University, Uppsala, Sweden
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, Division of Hematology/Oncology, Department of Internal Medicine, The University of Michigan Medical School, Ann Arbor, MI
| | - Edward Peres
- Bone Marrow Transplant Program, Henry Ford Hospital, Detroit, MI
| | - Andrew R Rezvani
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | - David Rizzieri
- Division of Hematologic Malignancies and Cellular Therapy, Blood and Marrow Transplant Clinic, Duke University, Durham, NC
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Harry C Schouten
- Department of Hematology, Academische Ziekenhuis, Maastricht, The Netherlands
| | - Mitchell Sabloff
- Division of Hematology, Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Matthew Seftel
- Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB, Canada
| | - Sachiko Seo
- National Cancer Research Center, East Hospital, Kashiwa, Chiba, Japan
| | - Mohamed L Sorror
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Jeff Szer
- Department Clinical Haematology and Bone Marrow Transplantation, Royal Melbourne Hospital, Victoria, VIC, Australia
| | - Baldeep M Wirk
- Division of Bone Marrow Transplant, Seattle Cancer Care Alliance, Seattle, WA
| | - William A Wood
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| | - Andrew Artz
- Section of Hematology/Oncology, University of Chicago School of Medicine, Chicago, IL
| |
Collapse
|
134
|
First French experiences of total body irradiations using helical TomoTherapy ®. Cancer Radiother 2017; 21:365-372. [DOI: 10.1016/j.canrad.2017.01.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 11/15/2022]
|
135
|
Gallo S, Sangiolo D, Carnevale Schianca F, Aglietta M, Montemurro F. Treating breast cancer with cell-based approaches: an overview. Expert Opin Biol Ther 2017; 17:1255-1264. [PMID: 28728493 DOI: 10.1080/14712598.2017.1356816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Breast cancer is the most common malignancy in women. Despite there being considerable progress in the treatment of this disease, metastatic dissemination is still considered an incurable condition at the present time, causing 500,000 deaths worldwide every year. Although most of the research efforts have been focused on pharmacological approaches, over the last three decades, the use of bone marrow and peripheral blood-derived cell therapy approaches have been attempted and developed. Areas covered: This review will briefly address cell therapy for breast cancer, including autologous stem cell transplantations for overcoming the myelosuppressive effects of high-dose chemotherapy, allogeneic stem cell transplants and adoptive immunotherapy using bone-marrow derived T-cells. Expert opinion: The treatment of breast cancer using bone marrow or peripheral-blood derived cells has evolved from a supportive care approach to allow dose escalation of conventional chemotherapy to a therapeutic strategy aimed at eliciting immune cell mediated anticancer immunity. This latter principle has led to the development of adoptive immunotherapies, either with 'natural' or genetically engineered effectors, which are being intensively investigated for their great potential against several solid tumors, including breast cancer.
Collapse
Affiliation(s)
- Susanna Gallo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| | - Dario Sangiolo
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | | | - Massimo Aglietta
- a Medical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy.,b Department of Oncology , University of Turin , Turin , Italy
| | - Filippo Montemurro
- c Investigative Clinical Oncology , Candiolo Cancer Institute-FPO (IRCCS) , Candiolo , Italy
| |
Collapse
|
136
|
Storb R, Sandmaier BM. Nonmyeloablative allogeneic hematopoietic cell transplantation. Haematologica 2017; 101:521-30. [PMID: 27132278 DOI: 10.3324/haematol.2015.132860] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/05/2016] [Indexed: 11/09/2022] Open
Abstract
Most hematological malignancies occur in older patients. Until recently these patients and those with comorbidities were not candidates for treatment with allogeneic hematopoietic transplantation because they were unable to tolerate the heretofore used high-dose conditioning regimens. The finding that many of the cures achieved with allogeneic hematopoietic transplantation were due to graft-versus-tumor effects led to the development of less toxic and well-tolerated reduced intensity and nonmyeloablative regimens. These regimens enabled allogeneic engraftment, thereby setting the stage for graft-versus-tumor effects. This review summarizes the encouraging early results seen with the new regimens and discusses the two hurdles that need to be overcome for achieving even greater success, disease relapse and graft-versus-host disease.
Collapse
Affiliation(s)
- Rainer Storb
- Fred Hutchinson Cancer Research Center and the University of Washington, Seattle, WA, USA
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Research Center and the University of Washington, Seattle, WA, USA
| |
Collapse
|
137
|
Mousavi SH, Abroun S, Zarrabi M, Ahmadipanah M. The effect of maternal and infant factors on cord blood yield. Pediatr Blood Cancer 2017; 64:e26381. [PMID: 27905684 DOI: 10.1002/pbc.26381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 11/05/2016] [Accepted: 11/07/2016] [Indexed: 11/11/2022]
Abstract
Umbilical cord blood (CB) can be used as an alternative hematopoietic stem cell source for transplantation in hematological malignancy and blood disorders. The success of transplantation is highly related to the levels of total nucleated cell and CD34+ cell counts. The evaluation of optimal conditions can decrease the rate of graft rejection due to low cell count and increases the quality of CB units (CBUs) in the blood bank and the success rate of engraftment. To this end, we review the maternal and infant parameters affecting the quality and quantity of CBUs.
Collapse
Affiliation(s)
- Seyed Hadi Mousavi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Abroun
- Department of Hematology, School of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Morteza Zarrabi
- Royan Stem Cell Technology Institute, Cord Blood Bank, Tehran, Iran
| | - Mona Ahmadipanah
- Royan Stem Cell Technology Institute, Cord Blood Bank, Tehran, Iran
| |
Collapse
|
138
|
Inversion 3 Cytogenetic Abnormality in an Allogeneic Hematopoietic Cell Transplant Recipient Representative of a Donor-Derived Constitutional Abnormality. Biol Blood Marrow Transplant 2017; 23:1582-1587. [PMID: 28549770 DOI: 10.1016/j.bbmt.2017.05.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/12/2017] [Indexed: 11/23/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is an important treatment for many severe hematologic disorders; however, HCT can be associated with significant complications, including organ toxicity, graft-versus-host disease, and relapse. Another serious, but rare, complication is the transmission of hematologic and nonhematologic diseases from the donor to the recipient. With older donors, the risk of an abnormality may be increased. Here we describe the transmission of an inversion 3 constitutional cytogenetic abnormality from an unrelated donor to a recipient, and review the clinical implications of the discovery of donor-derived constitutional cytogenetic abnormalities.
Collapse
|
139
|
Mussetti A, Greco R, Peccatori J, Corradini P. Post-transplant cyclophosphamide, a promising anti-graft versus host disease prophylaxis: where do we stand? Expert Rev Hematol 2017; 10:479-492. [DOI: 10.1080/17474086.2017.1318054] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Alberto Mussetti
- Division of Hematology and Bone Marrow Transplant, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Raffaella Greco
- Hematology and Bone Marrow Transplantation (BMT) Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Jacopo Peccatori
- Hematology and Bone Marrow Transplantation (BMT) Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Corradini
- Division of Hematology and Bone Marrow Transplant, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Dipartimento di Oncologia ed Emato-oncologia, Universita’ degli Studi di Milano, Milan, Italy
| |
Collapse
|
140
|
Pulsipher MA. Reduced Intensity for Myelodysplastic Syndrome: Worth the Gamble? J Clin Oncol 2017; 35:2106-2108. [PMID: 28463632 DOI: 10.1200/jco.2017.72.8048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
141
|
Mustafa Ali M, Abounader DM, Rybicki LA, Yurch MA, Starn J, Ferraro C, Winslow V, Hamilton BK, Gerds AT, Liu H, Dean R, Hill BT, Pohlman B, Andresen S, Hanna R, Kalaycio M, Bolwell BJ, Majhail NS, Sobecks RM. Comparative Effectiveness of Busulfan and Fludarabine versus Fludarabine and 400 cGy Total Body Irradiation Conditioning Regimens for Acute Myeloid Leukemia/Myelodysplastic Syndrome. Biol Blood Marrow Transplant 2017; 23:776-781. [DOI: 10.1016/j.bbmt.2017.01.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/16/2017] [Indexed: 01/10/2023]
|
142
|
Jentzsch M, Bill M, Nicolet D, Leiblein S, Schubert K, Pless M, Bergmann U, Wildenberger K, Schuhmann L, Cross M, Pönisch W, Franke GN, Vucinic V, Lange T, Behre G, Mrózek K, Bloomfield CD, Niederwieser D, Schwind S. Prognostic impact of the CD34+/CD38- cell burden in patients with acute myeloid leukemia receiving allogeneic stem cell transplantation. Am J Hematol 2017; 92:388-396. [PMID: 28133783 DOI: 10.1002/ajh.24663] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 01/09/2023]
Abstract
In acute myeloid leukemia (AML), leukemia-initiating cells exist within the CD34+/CD38- cell compartment. They are assumed to be more resistant to chemotherapy, enriched in minimal residual disease cell populations, and responsible for relapse. Here we evaluated clinical and biological associations and the prognostic impact of a high diagnostic CD34+/CD38- cell burden in 169 AML patients receiving an allogeneic stem cell transplantation in complete remission. Here, the therapeutic approach is mainly based on immunological graft-versus-leukemia effects. Percentage of bone marrow CD34+/CD38- cell burden at diagnosis was measured using flow cytometry and was highly variable (median 0.5%, range 0%-89% of all mononuclear cells). A high CD34+/CD38- cell burden at diagnosis associated with worse genetic risk and secondary AML. Patients with a high CD34+/CD38- cell burden had shorter relapse-free and overall survival which may be mediated by residual leukemia-initiating cells in the CD34+/CD38- cell population, escaping the graft-versus-leukemia effect after allogeneic transplantation. Evaluating the CD34+/CD38- cell burden at diagnosis may help to identify patients at high risk of relapse after allogeneic transplantation. Further studies to understand leukemia-initiating cell biology and develop targeting therapies to improve outcomes of AML patients are needed.
Collapse
Affiliation(s)
- Madlen Jentzsch
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Marius Bill
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center; Columbus Ohio USA
| | - Sabine Leiblein
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Karoline Schubert
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Martina Pless
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Ulrike Bergmann
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Kathrin Wildenberger
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Luba Schuhmann
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Michael Cross
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Wolfram Pönisch
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Georg-Nikolaus Franke
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Vladan Vucinic
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Thoralf Lange
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Gerhard Behre
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center; Columbus Ohio USA
| | | | - Dietger Niederwieser
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Sebastian Schwind
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| |
Collapse
|
143
|
Bone Marrow Graft-Versus-Host Disease in Major Histocompatibility Complex-Matched Murine Reduced-Intensity Allogeneic Hemopoietic Cell Transplantation. Transplantation 2017; 101:2695-2704. [PMID: 28319565 DOI: 10.1097/tp.0000000000001733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Most clinical allogeneic hemopoietic cell transplants (alloHCT) are now performed using reduced-intensity conditioning (RIC) instead of myeloablative conditioning (MAC); however, the biology underlying this treatment remains incompletely understood. METHODS We investigated a murine model of major histocompatibility complex-matched multiple minor histocompatibility antigen-mismatched alloHCT using bone marrow (BM) cells and splenocytes from B6 (H-2) donor mice transplanted into BALB.B (H-2) recipients after RIC with fludarabine of 100 mg/kg per day for 5 days, cyclophosphamide of 60 mg/kg per day for 2 days, and total body irradiation (TBI). RESULTS The lowest TBI dose capable of achieving complete donor chimerism in this mouse strain combination was 325 cGy given as a single fraction. Mice that underwent RIC had a reduced incidence and delayed onset of graft-versus-host disease (GVHD) and significantly prolonged survival compared with MAC-transplanted recipients (TBI of 850 cGy plus cyclophosphamide of 60 mg/kg per day for 2 days). Compared with syngeneic controls, RIC mice with GVHD showed evidence of BM suppression, have anemia, reduced BM cellularity, and showed profound reduction in BM B cell lymphopoiesis associated with damage to the endosteal BM niche. This was associated with an increase in BM CD8 effector T cells in RIC mice and elevated blood and BM plasma levels of T helper1 cytokines. Increasing doses of splenocytes resulted in increased incidence of GVHD in RIC mice. CONCLUSIONS We demonstrate that the BM is a major target organ of GVHD in an informative clinically relevant RIC mouse major histocompatibility complex-matched alloHCT model by a process that seems to be driven by CD8 effector T cells.
Collapse
|
144
|
Yerushalmi R, Shem‐Tov N, Danylesko I, Shouval R, Nagler A, Shimoni A. The combination of cyclosporine and mycophenolate mofetil is less effective than cyclosporine and methotrexate in the prevention of acute graft-versus host disease after stem-cell transplantation from unrelated donors. Am J Hematol 2017; 92:259-268. [PMID: 28052467 DOI: 10.1002/ajh.24631] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/15/2016] [Accepted: 12/19/2016] [Indexed: 12/24/2022]
Abstract
Acute graft-versus-host disease (GVHD) is the major treatment-related complication after stem-cell transplantation (SCT) from unrelated-donors. Several GVHD prophylaxis regimens have been explored, but no regimen has shown superiority. We analyzed transplantation outcomes in 472 consecutive unrelated-donor SCT recipients, using cyclosporine/methotrexate (MTX, n = 314) or cyclosporine/mycophenolate-mofetil (MMF, n = 158) for GVHD prophylaxis. Neutrophil engraftment was faster after MMF, days 11 and 14, respectively (P = .001). Acute GVHD grade II-IV and III-IV occurred in 47% and 28% after MMF compared to 27% and 12% after MTX, respectively (P < .001). Nonrelapse mortality (NRM) was 44% and 24%, respectively (P < .001). Death associated with GVHD occurred in 25% and 8% (P < .0001), while other NRM causes occurred in 19% and 16%, respectively (P = .39). Relapse mortality was similar. Overall survival was better after MTX, 40% and 29%, respectively (P = .006). However, this difference had only borderline significance when adjusting for differences in patient characteristics (HR, 1.3, P = .08). To minimize potential selection bias we analyzed outcomes on the basis of an intention-to-treat like analysis. During the years 2008-2009, the leading GVHD prophylaxis regimen for unrelated-donor SCT included MMF (89% of transplants). During the other periods, MTX was used predominantly (82% of transplants). The two periods were otherwise well-matched. Acute GVHD occurred more often in 2008-2009. Death associated with GVHD occurred more often, while other NRM causes occurred less often resulting in similar NRM and overall survival. In conclusion, cyclosporine/MMF is associated with faster engraftment and possibly with less organ toxicity than cyclosporine/MTX. However, it is associated with increased rates of acute GVHD and GVHD-associated deaths.
Collapse
Affiliation(s)
- Ronit Yerushalmi
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Noga Shem‐Tov
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Ivetta Danylesko
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Roni Shouval
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Arnon Nagler
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| | - Avichai Shimoni
- The Division of Hematology and Bone Marrow TransplantationTel‐Aviv UniversityChaim Sheba Medical Center, Tel‐Hashomer Israel
| |
Collapse
|
145
|
Zheng H(B, Watkins B, Tkachev V, Yu S, Tran D, Furlan S, Zeleski K, Singh K, Hamby K, Hotchkiss C, Lane J, Gumber S, Adams A, Cendales L, Kirk AD, Kaur A, Blazar BR, Larsen CP, Kean LS. The Knife's Edge of Tolerance: Inducing Stable Multilineage Mixed Chimerism but With a Significant Risk of CMV Reactivation and Disease in Rhesus Macaques. Am J Transplant 2017; 17:657-670. [PMID: 27500470 PMCID: PMC5338742 DOI: 10.1111/ajt.14006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 08/04/2016] [Indexed: 01/25/2023]
Abstract
Although stable mixed-hematopoietic chimerism induces robust immune tolerance to solid organ allografts in mice, the translation of this strategy to large animal models and to patients has been challenging. We have previously shown that in MHC-matched nonhuman primates (NHPs), a busulfan plus combined belatacept and anti-CD154-based regimen could induce long-lived myeloid chimerism, but without T cell chimerism. In that setting, donor chimerism was eventually rejected, and tolerance to skin allografts was not achieved. Here, we describe an adaptation of this strategy, with the addition of low-dose total body irradiation to our conditioning regimen. This strategy has successfully induced multilineage hematopoietic chimerism in MHC-matched transplants that was stable for as long as 24 months posttransplant, the entire length of analysis. High-level T cell chimerism was achieved and associated with significant donor-specific prolongation of skin graft acceptance. However, we also observed significant infectious toxicities, prominently including cytomegalovirus (CMV) reactivation and end-organ disease in the setting of functional defects in anti-CMV T cell immunity. These results underscore the significant benefits that multilineage chimerism-induction approaches may represent to transplant patients as well as the inherent risks, and they emphasize the precision with which a clinically successful regimen will need to be formulated and then validated in NHP models.
Collapse
Affiliation(s)
- Hengqi (Betty) Zheng
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle WA
| | | | - Victor Tkachev
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle WA
| | - Shan Yu
- Tulane National Primate Research Center, New Orleans, LA
| | - Dollnovan Tran
- Tulane National Primate Research Center, New Orleans, LA
| | - Scott Furlan
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle WA
| | - Katie Zeleski
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle WA
| | | | - Kelly Hamby
- Emory University School of Medicine, Atlanta GA
| | - Charlotte Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle WA
| | - Jennifer Lane
- Washington National Primate Research Center, University of Washington, Seattle WA
| | - Sanjeev Gumber
- Emory University School of Medicine, Atlanta GA,Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | | | | | - Amitinder Kaur
- Tulane National Primate Research Center, New Orleans, LA
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | | | - Leslie S. Kean
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle WA,Department of Pediatrics, University of Washington School of Medicine, Seattle WA,Fred Hutchinson Cancer Research Center, Seattle WA,Corresponding Author: Address: 1100 Olive Way Suite 100, Seattle WA 98101, Phone: 206-884-4079
| |
Collapse
|
146
|
Green K, Pearce K, Sellar RS, Jardine L, Nicolson PLR, Nagra S, Bigley V, Jackson G, Dickinson AM, Thomson K, Mackinnon S, Craddock C, Peggs KS, Collin M. Impact of Alemtuzumab Scheduling on Graft-versus-Host Disease after Unrelated Donor Fludarabine and Melphalan Allografts. Biol Blood Marrow Transplant 2017; 23:805-812. [PMID: 28212937 PMCID: PMC6588535 DOI: 10.1016/j.bbmt.2017.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/08/2017] [Indexed: 12/11/2022]
Abstract
Alemtuzumab conditioning is highly effective at reducing the incidence of acute and chronic graft versus host disease (GVHD) in reduced intensity fludarabine and melphalan transplantation with ciclosporin monotherapy. Less frequent and lower dose scheduling may be used with sibling donors but an optimal regimen for matched unrelated donors has not been defined. In this retrospective observational study of 313 patients, the incidence and severity of GVHD was compared in patients receiving the standard 100mg regimen (20mg on day -7 to -3), 60mg (30mg day -4 and -2) or 50mg (10mg on day -7 to -3). Patients treated with 100mg, 60mg or 50mg developed acute GVHD grade I-IV with an incidence of 74%, 65% and 64%, respectively, while 36%, 32% and 41% developed chronic GHVD. An excess of severe acute grade III/IV GVHD was observed in the 50mg cohort (15% vs. 2-6%; p = 0.016). The relative risk of severe acute grade GVHD remained more than three-fold higher in the 50mg cohort, compared with 100mg, after adjustment for differences in age, gender mismatch, CMV risk and diagnosis (p = 0.030). The findings indicate that 60mg doses of alemtuzumab is comparable to 100mg but lower dosing may increase the risk of severe grade GVHD.
Collapse
Affiliation(s)
- Kile Green
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kim Pearce
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rob S Sellar
- Cancer Institute, University College London, London, United Kingdom; Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Laura Jardine
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Northern Centre for Bone Marrow Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | - Sandeep Nagra
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Venetia Bigley
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Northern Centre for Bone Marrow Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Graham Jackson
- Northern Centre for Bone Marrow Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; Northern Institute for Cancer Research, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Anne M Dickinson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kirsty Thomson
- Cancer Institute, University College London, London, United Kingdom; Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Stephen Mackinnon
- Cancer Institute, University College London, London, United Kingdom; Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Charles Craddock
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom; Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Karl S Peggs
- Cancer Institute, University College London, London, United Kingdom; Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Matthew Collin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Northern Centre for Bone Marrow Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
147
|
Scott BL, Pasquini MC, Logan BR, Wu J, Devine SM, Porter DL, Maziarz RT, Warlick ED, Fernandez HF, Alyea EP, Hamadani M, Bashey A, Giralt S, Geller NL, Leifer E, Le-Rademacher J, Mendizabal AM, Horowitz MM, Deeg HJ, Horwitz ME. Myeloablative Versus Reduced-Intensity Hematopoietic Cell Transplantation for Acute Myeloid Leukemia and Myelodysplastic Syndromes. J Clin Oncol 2017; 35:1154-1161. [PMID: 28380315 DOI: 10.1200/jco.2016.70.7091] [Citation(s) in RCA: 503] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The optimal regimen intensity before allogeneic hematopoietic cell transplantation (HCT) is unknown. We hypothesized that lower treatment-related mortality (TRM) with reduced-intensity conditioning (RIC) would result in improved overall survival (OS) compared with myeloablative conditioning (MAC). To test this hypothesis, we performed a phase III randomized trial comparing MAC with RIC in patients with acute myeloid leukemia or myelodysplastic syndromes. Patients and Methods Patients age 18 to 65 years with HCT comorbidity index ≤ 4 and < 5% marrow myeloblasts pre-HCT were randomly assigned to receive MAC (n = 135) or RIC (n = 137) followed by HCT from HLA-matched related or unrelated donors. The primary end point was OS 18 months post-random assignment based on an intent-to-treat analysis. Secondary end points included relapse-free survival (RFS) and TRM. Results Planned enrollment was 356 patients; accrual ceased at 272 because of high relapse incidence with RIC versus MAC (48.3%; 95% CI, 39.6% to 56.4% and 13.5%; 95% CI, 8.3% to 19.8%, respectively; P < .001). At 18 months, OS for patients in the RIC arm was 67.7% (95% CI, 59.1% to 74.9%) versus 77.5% (95% CI, 69.4% to 83.7%) for those in the MAC arm (difference, 9.8%; 95% CI, -0.8% to 20.3%; P = .07). TRM with RIC was 4.4% (95% CI, 1.8% to 8.9%) versus 15.8% (95% CI, 10.2% to 22.5%) with MAC ( P = .002). RFS with RIC was 47.3% (95% CI, 38.7% to 55.4%) versus 67.8% (95% CI, 59.1% to 75%) with MAC ( P < .01). Conclusion OS was higher with MAC, but this was not statistically significant. RIC resulted in lower TRM but higher relapse rates compared with MAC, with a statistically significant advantage in RFS with MAC. These data support the use of MAC as the standard of care for fit patients with acute myeloid leukemia or myelodysplastic syndromes.
Collapse
Affiliation(s)
- Bart L Scott
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Marcelo C Pasquini
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Brent R Logan
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Juan Wu
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Steven M Devine
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - David L Porter
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Richard T Maziarz
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Erica D Warlick
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Hugo F Fernandez
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Edwin P Alyea
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Mehdi Hamadani
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Asad Bashey
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Sergio Giralt
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Nancy L Geller
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Eric Leifer
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Jennifer Le-Rademacher
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Adam M Mendizabal
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Mary M Horowitz
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - H Joachim Deeg
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| | - Mitchell E Horwitz
- Bart L. Scott and H. Joachim Deeg, Fred Hutchinson Cancer Research Center, Seattle, WA; Marcelo C. Pasquini, Brent R. Logan, Mehdi Hamadani, and Mary M. Horowitz, Medical College of Wisconsin, Milwaukee, WI; Juan Wu and Adam M. Mendizabal, Emmes Corporation, Rockville, MD; Steven M. Devine, Ohio State University Comprehensive Cancer Center, Columbus, OH; David L. Porter, University of Pennsylvania, Philadelphia, PA; Richard T. Maziarz, Oregon Health and Science University, Portland, OR; Erica D. Warlick, University of Minnesota, Minneapolis; Jennifer Le-Rademacher, Mayo Clinic, Rochester, MN; Hugo F. Fernandez, Moffitt Cancer Center, Tampa, FL; Edwin P. Alyea, Dana Farber Cancer Institute, Boston, MA; Asad Bashey, Northside Hospital Cancer Institute, Atlanta, GA; Sergio Giralt, Memorial Sloan Kettering Cancer Center, New York, NY; Nancy L. Geller and Eric Leifer, National Heart, Lung, and Blood Institute, Bethesda, MD; and Mitchell E. Horwitz, Duke University, Durham, NC
| |
Collapse
|
148
|
Dose-adapted post-transplant cyclophosphamide for HLA-haploidentical transplantation in Fanconi anemia. Bone Marrow Transplant 2017; 52:570-573. [PMID: 28067886 PMCID: PMC5382060 DOI: 10.1038/bmt.2016.301] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 01/09/2023]
Abstract
We developed a haploidentical transplantation protocol with post-transplant cyclophosphamide (CY) for in vivo T-cell depletion using a novel adapted-dosing schedule (25 mg/kg on days +3 and +4) for Fanconi Anemia. With median follow-up of 3 years (range, 37 days to 6.2 years), all six patients engrafted. Two patients with multiple co-morbidities and late referrals to transplant died from sepsis (n=2) and chronic graft-versus-host disease (GVHD) (n=1). Four patients without pre-existing co-morbidities and early transplant referrals are alive with 100% donor chimerism and excellent performance status. We conclude that modulated-dosing post-transplant CY is effective in vivo T-cell depletion to promote full donor engraftment in patients with Fanconi anemia.
Collapse
|
149
|
Fürst D, Niederwieser D, Bunjes D, Wagner EM, Gramatzki M, Wulf G, Müller CR, Neuchel C, Tsamadou C, Schrezenmeier H, Mytilineos J. Increased age-associated mortality risk in HLA-mismatched hematopoietic stem cell transplantation. Haematologica 2017; 102:796-803. [PMID: 28057735 PMCID: PMC5395120 DOI: 10.3324/haematol.2016.151340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/28/2016] [Indexed: 02/06/2023] Open
Abstract
We investigated a possible interaction between age-associated risk and HLA-mismatch associated risk on prognosis in different age categories of recipients of unrelated hematopoietic stem cell transplants (HSCT) (n=3019). Patients over 55 years of age transplanted with 8/10 donors showed a mortality risk of 2.27 (CI 1.70–3.03, P<0.001) and 3.48 (CI 2.49–4.86, P<0.001) when compared to 10/10 matched patients in the same age group and to 10/10 matched patients aged 18–35 years, respectively. Compared to 10/10 matched transplantations within each age category, the Hazards Ratio for 8/10 matched transplantation was 1.14, 1.40 and 2.27 in patients aged 18–35 years, 36–55 and above 55 years. Modeling age as continuous variable showed different levels of risk attributed to age at the time of transplantation [OS: 10/10: Hazards Ratio 1.015 (per life year); 9/10: Hazards Ratio: 1.019; 8/10: Hazards Ratio 1.026]. The interaction term was significant for 8/10 transplantations (P=0.009). Findings for disease-free survival and transplant-related mortality were similar. Statistical models were stratified for diagnosis and included clinically relevant predictors except cytomegalovirus status and Karnofsky performance status. The risk conferred by age at the time of transplantation varies according to the number of HLA-mismatches and leads to a disproportional increase in risk for elderly patients, particularly with double mismatched donors. Our findings highlight the importance of HLA-matching, especially in patients over 55 years of age, as HLA-mismatches are less well tolerated in these patients. The interaction between age-associated risk and HLA-mismatches should be considered in donor selection and in the risk assessment of elderly HSCT recipients.
Collapse
Affiliation(s)
- Daniel Fürst
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | | | - Donald Bunjes
- Department of Internal Medicine III, University of Ulm, Germany
| | - Eva M Wagner
- Department of Medicine III, Johannes Gutenberg-University Mainz, Germany
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, 2nd Department of Medicine, University of Kiel, Germany
| | - Gerald Wulf
- Department of Hematology/Oncology, Georg-August-University Göttingen, Germany
| | - Carlheinz R Müller
- ZKRD-Zentrales Knochenmarkspender-Register für Deutschland (German National Bone Marrow Donor Registry), Ulm, Germany.,DRST - German Registry for Stem Cell Transplantation, Ulm, Germany
| | - Christine Neuchel
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | - Chrysanthi Tsamadou
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | - Hubert Schrezenmeier
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, Germany.,Institute of Transfusion Medicine, University of Ulm, Germany
| | - Joannis Mytilineos
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg - Hessen, Ulm, Germany .,Institute of Transfusion Medicine, University of Ulm, Germany.,DRST - German Registry for Stem Cell Transplantation, Ulm, Germany
| |
Collapse
|
150
|
Marr KA. Infections in Hematopoietic Stem Cell Transplant Recipients. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00080-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|