1
|
Nirmal G, Kharya G, Shankar R, Singh S, Paul S, Choudhary M, Chadha V, Iskandarov K, Begali S, Bakane A, Chatterjee G. A Comparative Analysis of Low Dose Grafalon ® Versus Thymoglobuline ® as Serotherapy in Hematopoietic Stem Cell Transplant in Pediatric and Young Adult Population. Pediatr Hematol Oncol 2024; 41:557-571. [PMID: 39310983 DOI: 10.1080/08880018.2024.2398523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 11/05/2024]
Abstract
Anti-thymocyte globulin (ATG) forms an essential component of conditioning in hematopoietic stem cell transplantation (HSCT). Due to the shift of donor preference to alternate donors, reliance on rabbit-ATG (rATG) has increased. Two different forms of rATG (Thymoglobuline® and Grafalon®) are available for clinical use but data to support the use of one over the other is sparse. We retrospectively analyzed data of 144 patients who underwent allogenic-HSCT for benign hematological conditions at our center, from August 2019 to August 2023. Of these, 87 received Grafalon® and 57 received Thymoglobuline®. The majority (77.7%) underwent HSCT for hemoglobinopathies and all received pre-transplant immunosuppression. Engraftment kinetics was similar in 2 cohorts. Six patients had primary graft failure (PGF). There was no difference in the incidence of PGF stratified by serotherapy. Overall survival(OS) for the cohort was 74.9%. Kaplan-Meier estimate of OSand EFSwas significantly better in Grafalon® group than Thymoglobuline® (84.4 ± 0.04% vs 64.1% ±0.065%) (p-value= 0.04%) and (84.4 ± 0.04% and 61.2%±0.065% (p-value = 0.01)). Extensive chronic GVHD was (14%) higher in Thymoglobuline® group and (2.3%) in Grafalon®. Immune reconstitution at day + 100 was not statistically different between the two groups. On univariate analysis, Thymoglobuline® serotherapy (OR (95% CI) =4.665 (1.2-18.04))was associated with increased risk of acute grade III-IV GvHD. In our study, Grafalon® tended to have better OS, decreased incidence of acute grade III-IV GvHD, and extensive cGVHD. There was no difference in engraftment kinetics, PGF, and immune reconstitution between 2 cohorts of serotherapy.
Collapse
Affiliation(s)
- Garima Nirmal
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| | - Gaurav Kharya
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| | - Ravi Shankar
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| | - Saksham Singh
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| | - Subhasish Paul
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| | - Mohit Choudhary
- Department of Transfusion Medicine, Indraprsatha Apollo Hospital, Delhi, India
| | - Vaibhav Chadha
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| | - Kamol Iskandarov
- Department of Pediatric Hematology Oncology & BMT, National Children's Medical Centre, Tashkent, Uzbekistan
| | - Sayitov Begali
- Department of Pediatric Hematology Oncology & BMT, National Children's Medical Centre, Tashkent, Uzbekistan
| | - Atish Bakane
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| | - Goutomi Chatterjee
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, Delhi, India
| |
Collapse
|
2
|
Schlegel LS, Werbrouck C, Boettcher M, Schlegel P. Universal CAR 2.0 to overcome current limitations in CAR therapy. Front Immunol 2024; 15:1383894. [PMID: 38962014 PMCID: PMC11219820 DOI: 10.3389/fimmu.2024.1383894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has effectively complemented the treatment of advanced relapsed and refractory hematological cancers. The remarkable achievements of CD19- and BCMA-CAR T therapies have raised high expectations within the fields of hematology and oncology. These groundbreaking successes are propelling a collective aspiration to extend the reach of CAR therapies beyond B-lineage malignancies. Advanced CAR technologies have created a momentum to surmount the limitations of conventional CAR concepts. Most importantly, innovations that enable combinatorial targeting to address target antigen heterogeneity, using versatile adapter CAR concepts in conjunction with recent transformative next-generation CAR design, offer the promise to overcome both the bottleneck associated with CAR manufacturing and patient-individualized treatment regimens. In this comprehensive review, we delineate the fundamental prerequisites, navigate through pivotal challenges, and elucidate strategic approaches, all aimed at paving the way for the future establishment of multitargeted immunotherapies using universal CAR technologies.
Collapse
Affiliation(s)
- Lara Sophie Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Coralie Werbrouck
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Pediatric Hematology and Oncology, Westmead Children’s Hospital, Sydney, NSW, Australia
| |
Collapse
|
3
|
Davison GM, Opie JJ, Davids SFG, Mohammed R, Novitzky N. Early recovery of natural killer cells post T-cell depleted allogeneic stem cell transplantation using alemtuzumab "in the bag". Transpl Immunol 2024; 84:102045. [PMID: 38641148 DOI: 10.1016/j.trim.2024.102045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/03/2024] [Accepted: 04/14/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Allogeneic stem cell transplantation (SCT) is a critical therapy for haematological malignancy but may lead to acute and chronic graft versus host disease (GvHD). T-cell depletion with alemtuzumab, either in vivo or ex vivo, reduces the incidence of GvHD but is a risk factor for disease relapse and poor immune reconstitution. Natural killer (NK) cells are the first lymphocytes to recover. Classical NK cells make up >90% of the normal circulating population and can directly kill neoplastic or virally infected cells while the regulatory subset makes up <10%, secretes cytokines and is not cytotoxic. The recovery and balance of these subsets post SCT remains controversial, with most studies analysing patients who received unmanipulated grafts and in vivo immunosuppression. OBJECTIVE The aim was to assess the early recovery of NK cells in 18 consecutive patients receiving ex vivo T-cell depleted SCT and to compare the results to 25 individuals receiving haploidentical non-T cell depleted grafts. METHODS All patients received myeloablative conditioning. After stem cell collection, the stem cells of the T cell depleted group were treated "in the bag" with alemtuzumab (CAMPATH 1H) at a concentration of 1mg/108 mononuclear cells and thereafter immediately infused. For those receiving non-T cell depleted grafts, GvHD prophylaxis was with post infusion therapeutic doses of cyclophosphamide. Blood samples were collected at days 21, 28 and 90. Complete blood counts were performed on an automated analyser while lymphocyte and NK subsets were examined using multiparameter flowcytometry. NK cells were defined as lymphocytes which were CD3-/CD56+. The classical subset was recognised as CD56dim/CD16+ while the regulatory population as CD56bright/CD16-. The results for both transplant types were compared at all time points using SPSS v8 statistical software. RESULTS The recovery of lymphocytes was slow in both groups. Those receiving non-T cell depleted grafts had significantly higher T cell counts at day 21 and 28 when compared to the T cell depleted group (P < 0.05). In contrast, NK cells in the ex vivo T-cell depleted patients recovered rapidly and by day 21 was no different to normal (p > 0.05), while the non-T cell depleted group had significantly decreased numbers (p < 0.001), only recovering at day 90. Both groups had abnormal NK cell subset ratios with significantly elevated percentages of regulatory cells (p < 0.05). However, significant differences were observed between the two groups with those receiving T cell depleted grafts having lower percentages of regulatory cells as well as higher numbers of classical NK cells at day 21 and 28 (p < 0.01). CONCLUSION This study of ex vivo T-cell depleted SCT's demonstrates that NK cells recover quicker when compared to those receiving unfractionated grafts. These results may have implications for GvHD and the GvL effect which warrants further study.
Collapse
Affiliation(s)
- Glenda M Davison
- Division of Haematology, Department of Pathology, University of Cape Town, Cape Town, South Africa; SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa.
| | - Jessica J Opie
- Division of Haematology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| | - Saarah F G Davids
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, South Africa
| | - Rygana Mohammed
- Division of Haematology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nicolas Novitzky
- Division of Haematology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
4
|
Avenoso D, Mehra V, Slonim LB, de Farias M, Alshehri H, Bouziana S, Krishnamurthy P, Kulasekararaj A, Dazzi F, Wood H, Kenyon M, Leung YT, Anteh S, Shah MN, Hannah G, Serpenti F, Gameil A, Bourlon C, Dragoi OD, Pagliuca A, Potter V. Myeloablative Dose of Busulfan and Fludarabine Combined with In Vivo T Cell Depletion Is Safe and Effective Conditioning for Acute Myeloid Leukemia and Myelodysplastic Syndrome Patients. Transplant Cell Ther 2023; 29:698.e1-698.e6. [PMID: 37579918 DOI: 10.1016/j.jtct.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/27/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative strategy for acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). The prediction of transplantation-related mortality (TRM) using the Hematopoietic Cell Transplantation Comorbidity Index (HCT-CI) score and an arbitrary upper age limit of 55 years for administering myeloablative conditioning (MAC) are common strategies to ensure a safe procedure. The use of reduced-toxicity conditioning regimens is an additional approach to providing safe and effective myeloablation. Herein we report the outcome of AML and MDS patients conditioned with fludarabine and a myeloablative dose of busulfan (FB4) stratified by age and HCT-CI score. The primary objective was overall survival (OS) for patients age ≥55 years. Secondary objectives were total OS, TRM, graft-versus-host disease (GVHD), and GVHD, relapse-free survival (GRFS). The 2 year OS was 72% in patients age <55 and 51% in patients age ≥55. In patients age ≥55 with an HCT-CI <2, the estimated 2 year OS was 64%, with median OS not reached. In those with HCT-CI ≥2, the 2-year OS was 43%, with a median OS of 14 months. The total cumulative incidence of relapse was 30% regardless of age or HCT-CI score. FB4 conditioning regimen offers a high rate of prolonged remission with a relapse rate similar to that reported in previous studies. These positive outcomes suggest that this conditioning platform can be offered to patients age ≥55 years in the absence of comorbidities, and that age should not be the sole determinant of conditioning intensity.
Collapse
Affiliation(s)
- Daniele Avenoso
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom.
| | - Varun Mehra
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Liron Barnea Slonim
- King's College Hospital NHS Foundation Trust, Department of Histopathology, Denmark Hill, London, United Kingdom
| | - Madson de Farias
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Hassan Alshehri
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Styliani Bouziana
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Pramila Krishnamurthy
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Austin Kulasekararaj
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Francesco Dazzi
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Henry Wood
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Michelle Kenyon
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Ye Ting Leung
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Sandra Anteh
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Mili Naresh Shah
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Guy Hannah
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Fabio Serpenti
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Amna Gameil
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Christianne Bourlon
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Oana Diana Dragoi
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Antonio Pagliuca
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| | - Victoria Potter
- King's College Hospital NHS Foundation Trust, Department of Haematological Medicine, Denmark Hill, London, United Kingdom
| |
Collapse
|
5
|
Impact of in vivo T-cell depletion in patients with myelodysplastic syndromes undergoing allogeneic hematopoietic stem cell transplant: a registry study from the Chronic Malignancies Working Party of the EBMT. Bone Marrow Transplant 2022; 57:768-774. [DOI: 10.1038/s41409-022-01620-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/18/2021] [Accepted: 02/14/2022] [Indexed: 11/08/2022]
|
6
|
Tvedt THA, Vo AK, Bruserud Ø, Reikvam H. Cytokine Release Syndrome in the Immunotherapy of Hematological Malignancies: The Biology behind and Possible Clinical Consequences. J Clin Med 2021; 10:jcm10215190. [PMID: 34768710 PMCID: PMC8585070 DOI: 10.3390/jcm10215190] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cytokine release syndrome (CRS) is an acute systemic inflammatory syndrome characterized by fever and multiple organ dysfunction associated with (i) chimeric antigen receptor (CAR)-T cell therapy, (ii) therapeutic antibodies, and (iii) haploidentical allogeneic stem cell transplantation (haplo-allo-HSCT). Severe CRS can be life-threatening in some cases and requires prompt management of those toxicities and is still a great challenge for physicians. The pathophysiology of CRS is still not fully understood, which also applies to the identifications of predictive biomarkers that can forecast these features in advance. However, a broad range of cytokines are involved in the dynamics of CRS. Treatment approaches include both broad spectrum of immunosuppressant, such as corticosteroids, as well as more specific inhibition of cytokine release. In the present manuscript we will try to review an update regarding pathophysiology, etiology, diagnostics, and therapeutic options for this serious complication.
Collapse
Affiliation(s)
| | - Anh Khoi Vo
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
- Clinic for Medicine, Haukeland University Hospital, 5020 Bergen, Norway
| | - Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway; (A.K.V.); (Ø.B.)
- Clinic for Medicine, Haukeland University Hospital, 5020 Bergen, Norway
- Correspondence:
| |
Collapse
|
7
|
Russell AL, Prince C, Lundgren TS, Knight KA, Denning G, Alexander JS, Zoine JT, Spencer HT, Chandrakasan S, Doering CB. Non-genotoxic conditioning facilitates hematopoietic stem cell gene therapy for hemophilia A using bioengineered factor VIII. Mol Ther Methods Clin Dev 2021; 21:710-727. [PMID: 34141826 PMCID: PMC8181577 DOI: 10.1016/j.omtm.2021.04.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/29/2021] [Indexed: 01/09/2023]
Abstract
Hematopoietic stem and progenitor cell (HSPC) lentiviral gene therapy is a promising strategy toward a lifelong cure for hemophilia A (HA). The primary risks associated with this approach center on the requirement for pre-transplantation conditioning necessary to make space for, and provide immune suppression against, stem cells and blood coagulation factor VIII, respectively. Traditional conditioning agents utilize genotoxic mechanisms of action, such as DNA alkylation, that increase risk of sterility, infection, and developing secondary malignancies. In the current study, we describe a non-genotoxic conditioning protocol using an immunotoxin targeting CD117 (c-kit) to achieve endogenous hematopoietic stem cell depletion and a cocktail of monoclonal antibodies to provide transient immune suppression against the transgene product in a murine HA gene therapy model. This strategy provides high-level engraftment of hematopoietic stem cells genetically modified ex vivo using recombinant lentiviral vector (LV) encoding a bioengineered high-expression factor VIII variant, termed ET3. Factor VIII procoagulant activity levels were durably elevated into the normal range and phenotypic correction achieved. Furthermore, no immunological rejection or development of anti-ET3 immunity was observed. These preclinical data support clinical translation of non-genotoxic antibody-based conditioning in HSPC LV gene therapy for HA.
Collapse
Affiliation(s)
- Athena L. Russell
- Graduate Program in Genetics and Molecular Biology, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Chengyu Prince
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Taran S. Lundgren
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Kristopher A. Knight
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | | | - Jordan S. Alexander
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jaquelyn T. Zoine
- Graduate Program in Cancer Biology, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - H. Trent Spencer
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Expression Therapeutics, LLC, Tucker, GA 30084, USA
| | - Shanmuganathan Chandrakasan
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christopher B. Doering
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Expression Therapeutics, LLC, Tucker, GA 30084, USA
| |
Collapse
|
8
|
Gorfinkel L, Demsky C, Pashankar F, Kupfer G, Shah NC. Bone marrow transplant using fludarabine-based reduced intensity conditioning regimen with in vivo T cell depletion in patients with Fanconi anemia. Pediatr Transplant 2021; 25:e14009. [PMID: 33755277 DOI: 10.1111/petr.14009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/15/2020] [Accepted: 02/16/2021] [Indexed: 11/28/2022]
Abstract
FA is the most common cause of inherited BMF syndromes. The only cure for BMF in FA remains HSCT. Due to DNA instability in FA, RIC has been used to decrease immediate and late complications of HSCT. Most FA conditioning regimens in mismatched and unrelated donor transplants rely on TBI, which increases the risk of secondary malignancies. Most of the non-TBI conditioning regimens use an ex vivo T-cell depletion approach, but this is not feasible at all pediatric stem cell transplant programs. To evaluate the success of HSCT in patients with FA using non-TBI conditioning regimens with in vivo T-cell depletion approach. HSCT using non-TBI based conditioning was performed on two siblings with FA. The first sibling underwent matched unrelated donor transplant with a BM graft using fludarabine, alemtuzumab, busulfan, and cyclophosphamide conditioning and cyclosporine and mycophenolate as GVHD prophylaxis. The second sibling underwent MSD transplant with UCB and BM grafts using similar approach, but without busulfan and mycophenolate. Both siblings had engraftment without signs of acute or chronic GVHD. Acute post-transplant complications included brief viral reactivations. At last follow-up, both siblings continued to have full immune reconstitution with stable chimerism. Conditioning regimens without radiation and inclusion of alemtuzumab can lead to successful engraftment without development of GVHD and reduce risk of developing secondary neoplasms, even with unrelated donor transplants.
Collapse
Affiliation(s)
- Lev Gorfinkel
- Yale New Haven Children's Hospital, New Haven, Connecticut, USA
| | - Carolyn Demsky
- Yale New Haven Children's Hospital, New Haven, Connecticut, USA
| | - Farzana Pashankar
- Yale New Haven Children's Hospital, New Haven, Connecticut, USA.,Yale School of Medicine, New Haven, Connecticut, USA
| | - Gary Kupfer
- Yale New Haven Children's Hospital, New Haven, Connecticut, USA.,Yale School of Medicine, New Haven, Connecticut, USA
| | - Niketa C Shah
- Yale New Haven Children's Hospital, New Haven, Connecticut, USA.,Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
9
|
Brown M, Abasov R, Salerno D, Shore TB, Gergis U, Mayer S, Phillips A, Hsu J, Kodiyanplakkal RPL, Pasciolla M, van Besien K. Impact of alemtuzumab dosing and low-dose total body irradiation on cytomegalovirus infection in allogeneic hematopoietic stem cell transplantation. Leuk Lymphoma 2020; 61:3024-3026. [PMID: 32654572 DOI: 10.1080/10428194.2020.1791855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Maxwell Brown
- Department of Pharmacy, NewYork-Presbyterian/Weill Cornell Medical Center
| | - Rza Abasov
- Department of Pharmacy, NewYork-Presbyterian/Weill Cornell Medical Center
| | - David Salerno
- Department of Pharmacy, NewYork-Presbyterian/Weill Cornell Medical Center
| | - Tsiporah B Shore
- Division of Hematology and Oncology, Department of Medicine, NewYork-Presbyterian, Weill Cornell Medical Center
| | - Usama Gergis
- Division of Hematology and Oncology, Department of Medicine, NewYork-Presbyterian, Weill Cornell Medical Center
| | - Sebastian Mayer
- Division of Hematology and Oncology, Department of Medicine, NewYork-Presbyterian, Weill Cornell Medical Center
| | - Adrienne Phillips
- Division of Hematology and Oncology, Department of Medicine, NewYork-Presbyterian, Weill Cornell Medical Center
| | - Jingmei Hsu
- Division of Hematology and Oncology, Department of Medicine, NewYork-Presbyterian, Weill Cornell Medical Center
| | | | - Michelle Pasciolla
- Department of Pharmacy, NewYork-Presbyterian/Weill Cornell Medical Center
| | - Koen van Besien
- Division of Hematology and Oncology, Department of Medicine, NewYork-Presbyterian, Weill Cornell Medical Center
| |
Collapse
|
10
|
Woelfinger P, Epp K, Schaefer L, Kriege D, Theobald M, Bopp T, Wagner-Drouet EM. CD52-negative T cells predict acute graft-versus-host disease after an alemtuzumab-based conditioning regimen. Br J Haematol 2020; 191:253-262. [PMID: 32410220 DOI: 10.1111/bjh.16706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/09/2020] [Indexed: 11/28/2022]
Abstract
Allogeneic haematopoietic stem cell transplantation (HSCT) after a reduced-intensity conditioning (RIC) regimen with fludarabine, melphalan and alemtuzmab is an effective therapy for haematological malignancies. Alemtuzumab, a monoclonal antibody against CD52, a glycosylphosphatidylinositol-anchor-bound surface protein on lymphocytes, depletes T cells to prevent graft-versus-host disease (GVHD). Despite this, acute and chronic GVHD (a/cGVHD) remain life-threatening complications after HSCT. The aim of the present study was to identify parameters to predict GVHD. In 69 patients after HSCT, T-cell subsets were functionally analysed. Reconstitution of CD52neg T cells and CD52neg regulatory T cells (Tregs) correlated with onset, severity and clinical course of aGVHD. Patients with aGVHD showed significantly lower levels of CD52pos T cells compared to patients with cGVHD or without GVHD (P < 0·001). Analysis of T-cell reconstitution revealed a percentage of <40% of CD52pos CD4pos T cells or CD52pos Tregs at day +50 as a risk factor for the development of aGVHD. In contrast, CD52neg Tregs showed significant decreased levels of glycoprotein A repetitions predominant (GARP; P < 0·001), glucocorticoid-induced TNFR-related protein (GITR; P < 0·001), chemokine receptor (CXCR3; P = 0·023), C-C chemokine receptor type 5 (CCR5; P = 0·004), but increased levels of immunoglobulin-like transcript 3 (ILT3; P = 0·001), as well as a reduced suppressive capacity. We conclude that reconstitution of CD52neg T cells and CD52neg Tregs is a risk factor for development of aGVHD.
Collapse
Affiliation(s)
- Pascal Woelfinger
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Katharina Epp
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Lukas Schaefer
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Diana Kriege
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Eva-Maria Wagner-Drouet
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
11
|
Chander V, Gangenahalli G. Emerging strategies for enhancing the homing of hematopoietic stem cells to the bone marrow after transplantation. Exp Cell Res 2020; 390:111954. [PMID: 32156602 DOI: 10.1016/j.yexcr.2020.111954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/26/2022]
Abstract
Bone marrow failure is the primary cause of death after nuclear accidents or intentional exposure to high or low doses of ionizing radiation. Hematopoietic stem cell transplantation is the most potent treatment procedure for patients suffering from several hematopoietic malignancies arising after radiation injuries. Successful hematopoietic recovery after transplantation depends on efficient homing and subsequent engraftment of hematopoietic stem cells in specific niches within the bone marrow. It is a rapid and coordinated process in which circulating cells actively enter the bone marrow through the process known as transvascular migration, which involves the tightly regulated relay of events that finally leads to homing of cells in the bone marrow. Various adhesion molecules, chemokines, glycoproteins, integrins, present both on the surface of stem cells and sinusoidal endothelium plays a critical role in transvascular migration. But despite having an in-depth knowledge of homing and engraftment and the key events that regulate it, we are still not completely able to avoid graft failures and post-transplant mortalities. This deems it necessary to design a flawless plan for successful transplantation. Here, in this review, we will discuss the current clinical methods used to overcome graft failures and their flaws. We will also discuss, what are the new approaches developed in the past 10-12 years to selectively deliver the hematopoietic stem cells in the bone marrow by adopting proper targeting strategies that can help revolutionize the field of regenerative and translational medicine.
Collapse
Affiliation(s)
- Vikas Chander
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences, Delhi, 110054, India
| | - Gurudutta Gangenahalli
- Division of Stem Cell & Gene Therapy Research, Institute of Nuclear Medicine & Allied Sciences, Delhi, 110054, India.
| |
Collapse
|
12
|
Admiraal R, Jol-van der Zijde CM, Furtado Silva JM, Knibbe CAJ, Lankester AC, Boelens JJ, Hale G, Etuk A, Wilson M, Adams S, Veys P, van Kesteren C, Bredius RGM. Population Pharmacokinetics of Alemtuzumab (Campath) in Pediatric Hematopoietic Cell Transplantation: Towards Individualized Dosing to Improve Outcome. Clin Pharmacokinet 2019; 58:1609-1620. [PMID: 31131436 PMCID: PMC6885503 DOI: 10.1007/s40262-019-00782-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Alemtuzumab (Campath®) is used to prevent graft-versus-host disease and graft failure following pediatric allogeneic hematopoietic cell transplantation. The main toxicity includes delayed immune reconstitution, subsequent viral reactivations, and leukemia relapse. Exposure to alemtuzumab is highly variable upon empirical milligram/kilogram dosing. METHODS A population pharmacokinetic (PK) model for alemtuzumab was developed based on a total of 1146 concentration samples from 206 patients, aged 0.2-19 years, receiving a cumulative intravenous dose of 0.2-1.5 mg/kg, and treated between 2003 and 2015 in two centers. RESULTS Alemtuzumab PK were best described using a two-compartment model with a parallel saturable and linear elimination pathway. The linear clearance pathway, central volume of distribution, and intercompartmental distribution increased with body weight. Blood lymphocyte counts, a potential substrate for alemtuzumab, did not impact clearance. CONCLUSION The current practice with uniform milligram/kilogram doses leads to highly variable exposures in children due to the non-linear relationship between body weight and alemtuzumab PK. This model may be used for individualized dosing of alemtuzumab.
Collapse
Affiliation(s)
- Rick Admiraal
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
| | - Cornelia M Jol-van der Zijde
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Catherijne A J Knibbe
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Arjan C Lankester
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap Jan Boelens
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
- Stem Cell Transplant and Cellular Therapies, Memorial Sloane Kettering Cancer Center, New York, NY, USA
| | | | - Aniekan Etuk
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Melanie Wilson
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Stuart Adams
- Department of Haematology, Camelia Botnar Laboratories, Great Ormond Street Hospital, London, UK
| | - Paul Veys
- Bone Marrow Transplantation Department, Great Ormond Street Hospital, London, UK
| | - Charlotte van Kesteren
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
- Pediatric Blood and Marrow Transplantation Program, Prinses Maxima Center, Utrecht, The Netherlands
| | - Robbert G M Bredius
- Division of Stem Cell Transplantation, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
13
|
McIlroy G, Nikolousis E, Abou-Zeid N, Shankara P, Kishore B, Kaparou M, Lovell R, Elmoamly S, Davies D, Horgan C, Shenouda A, Kanellopoulos A. Mixed T-cell chimerism at 3 months followed by donor lymphocyte infusion is independently associated with favorable outcomes in alemtuzumab-based reduced-intensity allogeneic hematopoietic stem cell transplantation. Leuk Lymphoma 2019; 61:202-205. [DOI: 10.1080/10428194.2019.1650177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Graham McIlroy
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Emmanouil Nikolousis
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Nervana Abou-Zeid
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Paneesha Shankara
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Bhuvan Kishore
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Maria Kaparou
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Richard Lovell
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Shereef Elmoamly
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
- Internal Medicine and Clinical Hematology Department, Kasr Alainy Faculty of Medicine, Cairo University, Giza, Egypt
| | - David Davies
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Claire Horgan
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Amir Shenouda
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| | - Alexandros Kanellopoulos
- Department of Haematology, University Hospitals Birmingham, Birmingham, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
14
|
Okano T, Imai K, Tsujita Y, Mitsuiki N, Yoshida K, Kamae C, Honma K, Mitsui-Sekinaka K, Sekinaka Y, Kato T, Hanabusa K, Endo E, Takashima T, Hiroki H, Yeh TW, Tanaka K, Nagahori M, Tsuge I, Bando Y, Iwasaki F, Shikama Y, Inoue M, Kimoto T, Moriguchi N, Yuza Y, Kaneko T, Suzuki K, Matsubara T, Maruo Y, Kunitsu T, Waragai T, Sano H, Hashimoto Y, Tasaki K, Suzuki O, Shirakawa T, Kato M, Uchiyama T, Ishimura M, Tauchi T, Yagasaki H, Jou ST, Yu HH, Kanegane H, Kracker S, Durandy A, Kojima D, Muramatsu H, Wada T, Inoue Y, Takada H, Kojima S, Ogawa S, Ohara O, Nonoyama S, Morio T. Hematopoietic stem cell transplantation for progressive combined immunodeficiency and lymphoproliferation in patients with activated phosphatidylinositol-3-OH kinase δ syndrome type 1. J Allergy Clin Immunol 2019; 143:266-275. [DOI: 10.1016/j.jaci.2018.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 03/20/2018] [Accepted: 04/09/2018] [Indexed: 11/15/2022]
|
15
|
Hematopoietic stem cell transplantation for adult sickle cell disease in the era of universal donor availibility. Bone Marrow Transplant 2018; 53:1390-1400. [DOI: 10.1038/s41409-018-0193-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/13/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
|
16
|
Kruizinga MD, van Tol MJ, Bekker V, Netelenbos T, Smiers FJ, Bresters D, Jansen-Hoogendijk AM, van Ostaijen-ten Dam MM, Kollen WJ, Zwaginga JJ, Lankester AC, Bredius RG. Risk Factors, Treatment, and Immune Dysregulation in Autoimmune Cytopenia after Allogeneic Hematopoietic Stem Cell Transplantation in Pediatric Patients. Biol Blood Marrow Transplant 2018; 24:772-778. [DOI: 10.1016/j.bbmt.2017.12.782] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/09/2017] [Indexed: 10/18/2022]
|
17
|
Neumann T, Schneidewind L, Thiele T, Pink D, Schulze M, Schmidt C, Krüger W. No indication of increased infection rates using low-dose alemtuzumab instead of anti-thymocyte globulin as graft-versus-host disease prophylaxis before allogeneic stem cell transplantation. Transpl Infect Dis 2017; 20. [PMID: 29178248 DOI: 10.1111/tid.12822] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/20/2017] [Accepted: 08/25/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alemtuzumab as part of the conditioning protocol is effective in reducing graft-versus-host disease (GvHD), but may be associated with increased infection rates, especially when using high doses (ie, 100 mg). METHODS We performed a retrospective, single-center, case-control study analyzing the rates of neutropenic fever, cytomegalovirus (CMV) reactivation, Epstein-Barr virus (EBV) reactivation, clinical manifest toxoplasmosis, and clinical manifest human herpesvirus-6 (HHV6) infection using low-dose alemtuzumab in comparison with anti-thymocyte globulin (ATG) as GvHD prophylaxis before allogeneic stem cell transplantation. Forty-four patients transplanted from unrelated donors between 2001 and 2012 were matched by age, diagnosis, and conditioning regimen and treated either with alemtuzumab 10 mg at day -2 (respectively, 20 mg in case of mismatch transplantation) or ATG. ATG Fresenius (10 mg/kg for 3 days) or Thymoglobulin (2 mg/kg for 3 days) were used. RESULTS Rates of CMV reactivation, EBV reactivation, and clinical manifest HHV6 infection or toxoplasmosis did not differ significantly between both groups until 2 years after transplantation. No case of post-transplant lymphoproliferative disorder was observed. Also, rates of neutropenic fever during inpatient treatment after transplantation did not differ significantly in both groups. CONCLUSION We saw no indication of increased infections rates when using low-dose alemtuzumab as GvHD prophylaxis before allogeneic stem cell transplantation in this retrospective analysis.
Collapse
Affiliation(s)
- Thomas Neumann
- Department of Haematology, Oncology, Transplantation, University Medical Centre, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Laila Schneidewind
- Department of Haematology, Oncology, Transplantation, University Medical Centre, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Thomas Thiele
- Institute of Immunology and Transfusion Medicine, University Medical Centre, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Daniel Pink
- Department of Haematology, Oncology, HELIOS Klinikum Bad Saarow, Bad Saarow, Germany
| | - Meike Schulze
- Department of Internal Medicine, Wolgast Hospital, Wolgast, Germany
| | - Christian Schmidt
- Department of Haematology, Oncology, Transplantation, University Medical Centre, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - William Krüger
- Department of Haematology, Oncology, Transplantation, University Medical Centre, Ernst-Moritz-Arndt University, Greifswald, Germany
| |
Collapse
|
18
|
Ali R, Ramdial J, Algaze S, Beitinjaneh A. The Role of Anti-Thymocyte Globulin or Alemtuzumab-Based Serotherapy in the Prophylaxis and Management of Graft-Versus-Host Disease. Biomedicines 2017; 5:biomedicines5040067. [PMID: 29186076 PMCID: PMC5744091 DOI: 10.3390/biomedicines5040067] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/01/2017] [Accepted: 11/20/2017] [Indexed: 12/02/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplant is an established treatment modality for hematologic and non-hematologic diseases. However, it is associated with acute and long-term sequelae which can translate into mortality. Graft-versus-host disease (GVHD) remains a glaring obstacle, especially with the advent of reduced-intensity conditioning. Serotherapy capitalizes on antibodies which target T cells and other immune cells to mitigate this effect. This article focuses on the utility of two such agents: anti-thymocyte globulin (ATG) and alemtuzumab. ATG has demonstrated benefit in prophylaxis against GVHD, especially in the chronic presentation. However, there is limited impact of ATG on overall survival and it has little utility in the treatment context. There may be an initial improvement, particularly in skin manifestations, but no substantial benefit has been elicited. Alemtuzumab has shown benefit in both prophylaxis and treatment of GVHD, but at the consequence of a more profound immunosuppressive phase, mandating aggressive viral prophylaxis. There remains heterogeneity in the doses and regimens of the agents, with no standardized protocol in place. Furthermore, it seems that once steroid-refractory GVHD has been established, there is little that can be offered to offset the ultimately dismal outcome. Here we present a systematic overview of ATG- or alemtuzumab-based serotherapy in the prophylaxis and management of GVHD.
Collapse
Affiliation(s)
- Robert Ali
- Hematology/Medical Oncology Fellow, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Jeremy Ramdial
- Hematology/Medical Oncology Fellow, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Sandra Algaze
- Internal Medicine Residency Program, University of Miami/Miller School of Medicine, Miami, FL 33136, USA.
| | - Amer Beitinjaneh
- Associate Professor of Medicine, Stem Cell Transplant and Cellular Therapy Program, Sylvester Comprehensive Cancer Center, University of Miami/Miller School of Medicine; Miami, FL 33136, USA.
| |
Collapse
|
19
|
Hilal T, Slone S, Peterson S, Bodine C, Gul Z. Cytomegalovirus reactivation is associated with a lower rate of early relapse in myeloid malignancies independent of in-vivo T cell depletion strategy. Leuk Res 2017; 57:37-44. [PMID: 28279876 DOI: 10.1016/j.leukres.2017.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/22/2017] [Accepted: 02/26/2017] [Indexed: 11/19/2022]
Abstract
The association between cytomegalovirus (CMV) reactivation and relapse risk has not been evaluated in relation to T cell depletion strategies. We evaluated 93 patients who underwent allogeneic hematopoietic stem cell transplantation (HSCT) and analyzed the association between T cell depletion strategies with the cumulative incidence of relapse and CMV reactivation. A total of 33% of patients who received ATG vs. 34% who received alemtuzumab developed CMV reactivation. The cumulative incidence of relapse was 3% at 1year and 20% at 3 years in patients with CMV reactivation vs. 30% at 1year and 38% at 3 years in patients without CMV reactivation (p=0.02). When analyzed separately, this effect persisted in the myeloid, but not the lymphoid group. There was a numerical trend towards increased non-relapse mortality (NRM) in patients with CMV reactivation, especially in the myeloid group. The choice of T cell depleting agent and the rate of CMV reactivation were not associated with different overall survival (OS) rates. These results suggest that the choice of T cell depletion strategy may have similar effects on rates of CMV reactivation, disease relapse, and survival. Further studies examining these variables in patients not exposed to in-vivo T cell depleting agents may be of interest.
Collapse
Affiliation(s)
- Talal Hilal
- Department of Internal Medicine, University of Kentucky, Lexington, KY, United States; Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, United States.
| | - Stacey Slone
- Biostatistics Shared Resource Facility, University of Kentucky Markey Cancer Center, Lexington, KY, United States
| | - Shawn Peterson
- Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Charles Bodine
- Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Zartash Gul
- Division of Hematology and Blood and Marrow Transplant, University of Kentucky Markey Cancer Center, Lexington, KY, United States; Division of Hematology, University of Cincinnati Cancer Center, Cincinnati, OH, United States
| |
Collapse
|
20
|
Pharmacokinetics, Pharmacodynamics, and Pharmacogenomics of Immunosuppressants in Allogeneic Hematopoietic Cell Transplantation: Part II. Clin Pharmacokinet 2016; 55:551-93. [PMID: 26620047 DOI: 10.1007/s40262-015-0340-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Part I of this article included a pertinent review of allogeneic hematopoietic cell transplantation (alloHCT), the role of postgraft immunosuppression in alloHCT, and the pharmacokinetics, pharmacodynamics, and pharmacogenomics of the calcineurin inhibitors and methotrexate. In this article (Part II), we review the pharmacokinetics, pharmacodynamics, and pharmacogenomics of mycophenolic acid (MPA), sirolimus, and the antithymocyte globulins (ATG). We then discuss target concentration intervention (TCI) of these postgraft immunosuppressants in alloHCT patients, with a focus on current evidence for TCI and on how TCI may improve clinical management in these patients. Currently, TCI using trough concentrations is conducted for sirolimus in alloHCT patients. Several studies demonstrate that MPA plasma exposure is associated with clinical outcomes, with an increasing number of alloHCT patients needing TCI of MPA. Compared with MPA, there are fewer pharmacokinetic/dynamic studies of rabbit ATG and horse ATG in alloHCT patients. Future pharmacokinetic/dynamic research of postgraft immunosuppressants should include '-omics'-based tools: pharmacogenomics may be used to gain an improved understanding of the covariates influencing pharmacokinetics as well as proteomics and metabolomics as novel methods to elucidate pharmacodynamic responses.
Collapse
|
21
|
Neumann T, Schneidewind L, Thiele T, Schulze M, Klenner AF, Busemann C, Pink D, Greinacher A, Dölken G, Krüger WH. Reduced platelet transfusions and earlier platelet engraftment using alemtuzumab-based conditioning regimen in allogeneic stem cell transplantation. J Cancer Res Clin Oncol 2016; 142:1091-7. [PMID: 26779644 DOI: 10.1007/s00432-016-2114-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/04/2016] [Indexed: 11/24/2022]
Abstract
PURPOSE In patients undergoing allogeneic stem cell transplantation, conditioning regimens containing alemtuzumab instead of anti-thymocyte globulin (ATG) may result in an earlier platelet engraftment and a reduced number of platelet transfusions. METHODS We performed a retrospective, single-center, case-control study analyzing time to engraftment and transfusion needs using alemtuzumab in comparison with ATG as part of conditioning protocol. RESULTS Median values for time to platelet engraftment, number of transfused platelet concentrates and number of transfused red cell concentrates were 12 versus 19.5 days (p < 0.001), 2 versus 14 (p < 0.001) and 6 versus 14.5 (p = 0.003) in the alemtuzumab and ATG group. Time to leukocyte engraftment did not differ with median 15 days in both groups. Patients in the ATG group showed a significant higher decrease in platelet count during conditioning (68 vs. 29 %, p = 0.001), leading to significant lower median platelet counts at the day of stem cell infusion (38 vs. 95.5 Gpt/l, p = 0.008), and higher values for median C-reactive protein after first antibody infusion (69.0 vs. 43.6 mg/l, p = 0.001) compared with alemtuzumab group. Test for significance was done by using Wilcoxon rank-sum test. Subgroup analysis considering the type of ATG used (Thymoglobulin vs. ATG Fresenius) revealed that differences between alemtuzumab and ATG group were more due to effects of ATG Fresenius than Thymoglobulin. CONCLUSIONS The use of alemtuzumab in comparison with ATG as part of the conditioning regimen may be an approach to reduce the number of transfused platelet and red cell concentrates after allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Thomas Neumann
- Department of Hematology, Oncology, Transplantation, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany.
| | - Laila Schneidewind
- Department of Urology, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Thomas Thiele
- Institute of Immunology and Transfusion Medicine, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Meike Schulze
- Department of Hematology, Oncology, Transplantation, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Anne F Klenner
- Department of Hematology, Oncology, Transplantation, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Christoph Busemann
- Department of Hematology, Oncology, Transplantation, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Daniel Pink
- Department of Hematology, Oncology, HELIOS Klinikum Bad Saarow, Bad Saarow, Germany
| | - Andreas Greinacher
- Institute of Immunology and Transfusion Medicine, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Gottfried Dölken
- Department of Hematology, Oncology, Transplantation, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - William H Krüger
- Department of Hematology, Oncology, Transplantation, University Medical Center, Ernst-Moritz-Arndt University, Greifswald, Germany
| |
Collapse
|
22
|
Patel K, Parmar S, Shah S, Shore T, Gergis U, Mayer S, van Besien K. Comparison of Subcutaneous versus Intravenous Alemtuzumab for Graft-versus-Host Disease Prophylaxis with Fludarabine/Melphalan-Based Conditioning in Matched Unrelated Donor Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2015; 22:456-61. [PMID: 26524732 PMCID: PMC7128235 DOI: 10.1016/j.bbmt.2015.10.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/23/2015] [Indexed: 11/27/2022]
Abstract
The objective of this study was to compare infusion-related reactions and outcomes of using subcutaneous (subQ) alemtuzumab versus intravenous (i.v.) alemtuzumab as graft-versus-host disease (GVHD) prophylaxis for matched unrelated donor stem cell transplantations. Outcomes include incidence of cytomegalovirus (CMV)/Epstein-Barr (EBV) viremia, development of CMV disease or post-transplantation lymphoproliferative disorder, fatal infections, acute and chronic GVHD, time to engraftment, relapse rate, and survival. We conducted a retrospective study of all adult matched unrelated donor stem cell transplantations patients who received fludarabine/melphalan with subQ or i.v. alemtuzumab in combination with tacrolimus as part of their conditioning for unrelated donor transplantation at New York-Presbyterian/Weill Cornell Medical Center from January 1, 2012 to March 21, 2014. Alemtuzumab was administered at a total cumulative dose of 100 mg (divided over days −7 to −3). Forty-six patients received an unrelated donor stem cell transplantation with fludarabine/melphalan and either subQ (n = 26) or i.v. (n = 20) alemtuzumab in combination with tacrolimus. Within the evaluable population, 130 subQ and 100 i.v. alemtuzumab doses were administered. For the primary outcome, ≥grade 2 infusion-related reactions occurred in 11 (8%) versus 25 (25%) infusions in the subQ and i.v. cohorts, respectively (P = .001). Overall, 12 injections (9%) in the subQ arm versus 26 infusions (26%) in the i.v. arm experienced an infusion-related reaction of any grade (P = .001). There were no significant differences between the subQ and i.v. arms in rates of reactivation of CMV/EBV, development of CMV disease or post-transplantation lymphoproliferative disorder, fatal infections, acute and chronic GVHD, relapse, or survival. Subcutaneous administration of alemtuzumab for GVHD prophylaxis was associated with fewer infusion-related reactions compared with i.v. administration in the SCT setting. Incidences of acute and chronic GVHD were similar between both arms. There was also no difference in reactivation of CMV/EBV viremia, development of CMV disease or post-transplantation lymphoproliferative disorder, fatal infections, relapse, or survival. We conducted a retrospective comparison of intravenous versus subcutaneous administration of alemtuzumab for graft-versus-host disease prophylaxis in unrelated donor transplant recipients. We observed a significant reduction in grade ≥ 2 infusion-related reactions after subcutaneous injections. The incidences of acute and chronic graft-versus-host disease were similar. There was no difference in incidence of viral reactivation or fatal bacterial infections. There was no difference in 30-day mortality or 1-year overall survival between the 2 arms.
Collapse
Affiliation(s)
- Khilna Patel
- Department of Pharmacy, New York-Presbyterian Hospital, New York, New York.
| | - Sapna Parmar
- Department of Pharmacy, New York-Presbyterian Hospital, New York, New York
| | - Shreya Shah
- Department of Pharmacy, New York-Presbyterian Hospital, New York, New York
| | - Tsiporah Shore
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| | - Usama Gergis
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| | - Sebastian Mayer
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| | - Koen van Besien
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| |
Collapse
|
23
|
Jain A, Sahdev I. Alemtuzumab Pharmacokinetics in Hematopoietic Stem Cell Transplants for Nonmalignant Genetic Diseases. Biol Blood Marrow Transplant 2015; 21:1337. [PMID: 25963918 DOI: 10.1016/j.bbmt.2015.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/10/2015] [Indexed: 10/23/2022]
Affiliation(s)
- Akshat Jain
- Steven and Alexandra Cohen Children's Medical Center of New York, New York, New York.
| | - Indira Sahdev
- Steven and Alexandra Cohen Children's Medical Center of New York, New York, New York
| |
Collapse
|
24
|
Chao MM, Kuehl JS, Strauss G, Hanenberg H, Schindler D, Neitzel H, Niemeyer C, Baumann I, von Bernuth H, Rascon J, Nagy M, Zimmermann M, Kratz CP, Ebell W. Outcomes of mismatched and unrelated donor hematopoietic stem cell transplantation in Fanconi anemia conditioned with chemotherapy only. Ann Hematol 2015; 94:1311-8. [PMID: 25862235 DOI: 10.1007/s00277-015-2370-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/27/2015] [Indexed: 01/13/2023]
Abstract
Fanconi anemia (FA) is a genomic instability syndrome associated with bone marrow failure, myelodysplastic syndrome (MDS), and/or acute myeloid leukemia (AML) requiring hematopoietic stem cell transplantation (HSCT) to restore normal hematopoiesis. Although low-intensity fludarabine-based preparative regimens without radiation confer excellent outcomes in FA HSCTs with HLA-matched sibling donors, outcomes for FA patients with alternative donors are less encouraging, albeit improving. We present our experience with 17 FA patients who completed mismatched related or unrelated donor HSCT using a non-radiation fludarabine-based preparative regimen at Charité University Medicine Berlin. All patients engrafted; however, one patient had unstable chimerism in the setting of multi-viral infections that necessitated a stem cell boost to revert to full donor chimerism. Forty-seven percent of patients developed grade I acute graft-verus-host disease (aGVHD). No grade II-IV aGVHD or chronic graft-versus-host disease of any severity occurred. At a median follow-up of 30 months, 88 % of patients are alive with normal hematopoiesis. Two patients died of infections 4 months post-transplantation. These results demonstrate that short-term outcomes for FA patients with mismatched and unrelated donor HSCTs can be excellent using chemotherapy only conditioning. Viral reactivation, however, was a major treatment-related complication.
Collapse
Affiliation(s)
- M M Chao
- Department of Pediatric Hematology Oncology, Hannover Medical School, Carl-Neuberg Strasse 1, 30625, Hannover, Germany,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Preemptive DLI without withdrawal of immunosuppression to promote complete donor T-cell chimerism results in favorable outcomes for high-risk older recipients of alemtuzumab-containing reduced-intensity unrelated donor allogeneic transplant: a prospective phase II trial. Bone Marrow Transplant 2014; 49:616-21. [PMID: 24801098 DOI: 10.1038/bmt.2014.2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 09/23/2013] [Accepted: 09/24/2013] [Indexed: 11/08/2022]
Abstract
Although pretransplant alemtuzumab can reduce GVHD following allogeneic transplantation, it may also increase the risk of mixed donor T-cell chimerism and infections. We hypothesized that the early use of DLI without withdrawal of immunosuppressive drugs in patients with mixed T-cell chimerism would lower the risk of relapse without significantly increasing the risk of GVHD post DLI. Thirty-six patients (median age 59 years) were treated in this phase II trial using reduced-intensity conditioning including s.c. alemtuzumab (total dose 43 mg) and a PBSC graft from a matched unrelated donor (UD). DLI without withdrawal of immunosuppressive drugs was administered to all 25 patients with <50% donor T-cell chimerism on day +60. The cumulative risks of acute and chronic GVHD were 42% and 59%, respectively. Estimated probabilities of non-relapse mortality (NRM) at day 100 and 1 year were 3% and 14%, respectively. With a median follow up 2.4 years, estimated survivals at day 100, 1 and 2 years were 97%, 71% and 57%, respectively. In multivariate analysis, the occurrence of acute GVHD was associated with an increased risk of mortality, whereas the occurrence of chronic GVHD had a protective effect, associated with decreased relapse and improved disease-free survival. Low-dose alemtuzumab and preemptive DLI provides favorable transplant outcomes including low NRM in an older patient population with high-risk malignancies undergoing UD transplantation.
Collapse
|
26
|
Lane JP, Evans PTG, Nademi Z, Barge D, Jackson A, Hambleton S, Flood TJ, Cant AJ, Abinun M, Slatter MA, Gennery AR. Low-dose serotherapy improves early immune reconstitution after cord blood transplantation for primary immunodeficiencies. Biol Blood Marrow Transplant 2013; 20:243-9. [PMID: 24225641 PMCID: PMC7110834 DOI: 10.1016/j.bbmt.2013.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 11/06/2013] [Indexed: 11/27/2022]
Abstract
Cord blood transplantation (CBT) is curative for many primary immunodeficiencies (PIDs) but is associated with risks of viral infection and graft-versus-host disease (GvHD). Serotherapy reduces GvHD but potentially increases the risk of viral infection by delaying immune reconstitution. Because many PID patients have pre-existing viral infections, the optimal dose of serotherapy is unclear. We performed a retrospective analysis in 34 consecutive PID patients undergoing CBT and compared immune reconstitution, viral infection, GvHD, mortality, and long-term immune function between high-dose (n = 11) and low-dose (n = 9) serotherapy. Serotherapy dose had no effect on neutrophil engraftment. Median CD3+ engraftment occurred at 92.5 and 97 days for high- and low-dose serotherapy, respectively. The low-dose serotherapy group had higher CD3+, CD4+, and early thymic emigrant counts at 4 months compared with the high-dose group. GvHD severity and number of viral infections did not differ between serotherapy doses. Survival from the transplantation process was 90.9% for high-dose and 100% for low-dose groups. In conclusion, low-dose serotherapy enhanced T cell reconstitution and thymopoiesis during the first year after CBT with no increase in GvHD.
Collapse
Affiliation(s)
- Jonathan P Lane
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Philippa T G Evans
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Zohreh Nademi
- Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Dawn Barge
- Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Anthony Jackson
- International Centre for Life, Newcastle upon Tyne, United Kingdom
| | - Sophie Hambleton
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Terry J Flood
- Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Andrew J Cant
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Mario Abinun
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Mary A Slatter
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom
| | - Andrew R Gennery
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
27
|
Sellar RS, Peggs KS. Recent progress in managing graft-versus-host disease and viral infections following allogeneic stem cell transplantation. Future Oncol 2013; 8:1549-65. [PMID: 23231517 DOI: 10.2217/fon.12.153] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite recent reductions in transplant-related mortality, post-transplant complications such as graft-versus-host disease (GvHD) remain major obstacles to the successful application of allogeneic hematopoietic transplantation. Steroid-refractory GvHD has a poor outcome. Although there are a variety of new approaches to the treatment of refractory GvHD, many have limited evidence of efficacy. Other approaches appear to be unacceptably toxic. It would be preferable to improve GvHD prophylaxis. There is good evidence that rates of GvHD can be reduced without unacceptable reduction of the graft-versus-leukemia effect or compromising overall survival. However, prophylactic measures aimed at reducing T-cell numbers or functions are associated with high rates of reactivation of latent viruses. New technologies that allow rapid generation of virus-specific T-cells show promise to reduce the frequency and severity of such reactivations and have the potential to revolutionize the approach to post-transplant infectious complications.
Collapse
Affiliation(s)
- Rob S Sellar
- UCL Cancer Institute, Department of Haematology, London, WC1E 6BT, UK
| | | |
Collapse
|
28
|
Kumar AJ, Hexner EO, Frey NV, Luger SM, Loren AW, Reshef R, Boyer J, Smith J, Stadtmauer EA, Levine BL, June CH, Porter DL, Goldstein SC. Pilot study of prophylactic ex vivo costimulated donor leukocyte infusion after reduced-intensity conditioned allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2013; 19:1094-101. [PMID: 23635453 DOI: 10.1016/j.bbmt.2013.04.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/21/2013] [Indexed: 12/30/2022]
Abstract
Donor leukocyte infusion (DLI) can induce potent graft-versus-leukemia (GVL) activity in patients with relapsed hematologic malignancies after allogeneic hematopoietic stem cell transplantation (HSCT). Unfortunately, except in patients with chronic-phase chronic myelogenous leukemia, responses to DLI have been disappointing. GVL induction is likely to be most effective in the setting of minimal residual disease. Prevention of relapse through the provision of prophylactic DLI to high-risk patients may improve the outcome of allogeneic HSCT. We previously reported that ex vivo costimulated T cell infusion of activated DLI (aDLI) as treatment for relapse is safe and has potent GVL effects. We hypothesized that prophylactic aDLI can be given safely and prevent relapse in high-risk patients after allogeneic HSCT. Eighteen patients with acute myeolgenous leukemia (n = 14), acute lymphoblastic leukemia (n = 3), or myelodysplastic syndrome (n = 1) underwent allogeneic HSCT after a reduced-intensity conditioning (RIC) regimen with alemtuzumab, fludarabine, and busulfan. Graft-versus-host-disease (GVHD) prophylaxis consisted of tacrolimus and methotrexate with a planned early and rapid taper of tacrolimus. Patients without GVHD, off immune suppression, and in remission received aDLI at a dose of 1 × 10(7) CD3(+) cells/kg (aDLI 1) at day +120, followed by a second infusion of 1 × 10(8) CD3 cells/kg (aDLI 2) at day +180. At a median follow-up of 58 months, 5 of the 18 patients (28%) were alive, and 4 patients were in remission. Eleven patients (65%) relapsed, at a median time of 191 days. Twelve of the 18 patients received at least one aDLI, and 6 of these 12 patients also received aDLI 2. Six patients did not receive any aDLI owing to early relapse (n = 2), protocol ineligibility (n = 1), or GVHD (n = 3). Only 2 of the 12 patients who received aDLI 1 developed GVHD. Two out of the 12 patients remain in remission at the time of this report. Disease recurrence was the cause of death in 10 of the 13 patients (77%) who died. Our data indicate that prophylactic ex vivo costimulated CD3/CD28 DLI is safe, feasible, and not associated with significant GVHD. Relapse remains the major cause of treatment failure after RIC HSCT even with rapid withdrawal of immune suppression and the use of prophylactic aDLI, and better strategies to prevent relapse are needed.
Collapse
Affiliation(s)
- Anita J Kumar
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
van Besien K. Allogeneic transplantation for AML and MDS: GVL versus GVHD and disease recurrence. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2013; 2013:56-62. [PMID: 24319163 DOI: 10.1182/asheducation-2013.1.56] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Allogeneic transplantation constitutes curative treatment for acute myeloid leukemia and myelodysplastic syndrome. Its therapeutic effects are to a large extent mediated by GVL effects, but partially offset by treatment-related mortality and loss of quality of life caused by acute and chronic GVHD. Although severe acute and chronic GVHD are associated with a reduction in relapse risk, they are not associated with improved survival. Recent efforts to modulate the GVL-GVH balance include novel methods of in vitro or in vivo T-cell depletion that are associated with a minimal impact on rates of disease recurrence and a dramatically decreased risk for GVHD. Donor selection algorithms may also have a significant impact on transplantation outcomes. Low-expression HLA alleles, particularly HLA-DP, should be incorporated in selection of adult unrelated donors. Evolving data suggest that KIR typing may also be important. High-resolution HLA typing and the importance of fetal-maternal interactions in umbilical cord blood transplantation are also briefly discussed. A combination of donor selection strategies and GVHD prophylaxis methods will favorably affect long-term outcomes and create an environment suitable for effective posttransplantation interventions.
Collapse
|
30
|
Krishnamurthy P, Potter VT, Barber LD, Kulasekararaj AG, Lim ZY, Pearce RM, de Lavallade H, Kenyon M, Ireland RM, Marsh JCW, Devereux S, Pagliuca A, Mufti GJ. Outcome of donor lymphocyte infusion after T cell-depleted allogeneic hematopoietic stem cell transplantation for acute myelogenous leukemia and myelodysplastic syndromes. Biol Blood Marrow Transplant 2012; 19:562-8. [PMID: 23266740 DOI: 10.1016/j.bbmt.2012.12.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Accepted: 12/14/2012] [Indexed: 11/24/2022]
Abstract
Relapse occurs in 30%-50% of recipients of T cell-depleted (TCD) reduced-intensity conditioned (RIC) hematopoietic stem cell transplantation (HSCT) for acute myelogenous leukemia (AML) and myelodysplastic syndromes (MDS). Despite limited published supportive data, donor lymphocyte infusion (DLI) is used preemptively (pDLI) to improve donor chimerism and prevent relapse, and therapeutically (tDLI) after disease recurrence. We evaluated the efficacy and toxicity of pDLI and tDLI in 113 patients after TCD (alemtuzumab, n = 99; antithymocyte globulin, n = 14) RIC HSCT for AML or MDS. Recipients of pDLI (n = 62) had an estimated 5-year overall survival (OS) of 80% and an event-free survival of 65%. More than one-half (52%; n = 32) of the patients received pDLI within 6 months post-HSCT; despite this, the 5-year incidence of graft-versus-host disease was only 31% (95% confidence interval [CI], 19%-43%). Recipients of tDLI (n = 51) had an estimated 5-year OS of 40% and a 5-year relapse/progression rate of 69% (95% CI, 54%-81%). Recipients of tDLI at >6 months post-HSCT had a significantly superior 5-year OS after tDLI compared with those treated earlier (P = .008). The cumulative incidence of graft-versus-host disease at 5 years after tDLI was 45% (95% CI, 23%-65%). We demonstrate that pDLI safely promotes durable remission after TCD RIC HSCT for AML or MDS, and that tDLI salvages patients after late relapse with greater efficacy.
Collapse
|