1
|
Keller MD, Hanley PJ, Chi YY, Aguayo-Hiraldo P, Dvorak CC, Verneris MR, Kohn DB, Pai SY, Dávila Saldaña BJ, Hanisch B, Quigg TC, Adams RH, Dahlberg A, Chandrakasan S, Hasan H, Malvar J, Jensen-Wachspress MA, Lazarski CA, Sani G, Idso JM, Lang H, Chansky P, McCann CD, Tanna J, Abraham AA, Webb JL, Shibli A, Keating AK, Satwani P, Muranski P, Hall E, Eckrich MJ, Shereck E, Miller H, Mamcarz E, Agarwal R, De Oliveira SN, Vander Lugt MT, Ebens CL, Aquino VM, Bednarski JJ, Chu J, Parikh S, Whangbo J, Lionakis M, Zambidis ET, Gourdine E, Bollard CM, Pulsipher MA. Antiviral cellular therapy for enhancing T-cell reconstitution before or after hematopoietic stem cell transplantation (ACES): a two-arm, open label phase II interventional trial of pediatric patients with risk factor assessment. Nat Commun 2024; 15:3258. [PMID: 38637498 PMCID: PMC11026387 DOI: 10.1038/s41467-024-47057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Viral infections remain a major risk in immunocompromised pediatric patients, and virus-specific T cell (VST) therapy has been successful for treatment of refractory viral infections in prior studies. We performed a phase II multicenter study (NCT03475212) for the treatment of pediatric patients with inborn errors of immunity and/or post allogeneic hematopoietic stem cell transplant with refractory viral infections using partially-HLA matched VSTs targeting cytomegalovirus, Epstein-Barr virus, or adenovirus. Primary endpoints were feasibility, safety, and clinical responses (>1 log reduction in viremia at 28 days). Secondary endpoints were reconstitution of antiviral immunity and persistence of the infused VSTs. Suitable VST products were identified for 75 of 77 clinical queries. Clinical responses were achieved in 29 of 47 (62%) of patients post-HSCT including 73% of patients evaluable at 1-month post-infusion, meeting the primary efficacy endpoint (>52%). Secondary graft rejection occurred in one child following VST infusion as described in a companion article. Corticosteroids, graft-versus-host disease, transplant-associated thrombotic microangiopathy, and eculizumab treatment correlated with poor response, while uptrending absolute lymphocyte and CD8 T cell counts correlated with good response. This study highlights key clinical factors that impact response to VSTs and demonstrates the feasibility and efficacy of this therapy in pediatric HSCT.
Collapse
Affiliation(s)
- Michael D Keller
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
- Division of Allergy and Immunology, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University School of Medicine, Washington, DC, USA
| | - Patrick J Hanley
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University School of Medicine, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Yueh-Yun Chi
- Department of Pediatrics and Preventative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paibel Aguayo-Hiraldo
- Cancer and blood disease institute, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Christopher C Dvorak
- Division of Pediatric Allergy, Immunology, and BMT, University of California San Francisco, San Francisco, CA, USA
| | - Michael R Verneris
- Department of Pediatrics and Division of Child's Cancer and Blood Disorders, Children's Hospital Colorado and University of Colorado, Denver, CO, USA
| | - Donald B Kohn
- Department of Microbiology, Immunology & Molecular Genetics and Department of Pediatrics David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Division of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sung-Yun Pai
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Blachy J Dávila Saldaña
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Benjamin Hanisch
- Division of Pediatric Infectious Diseases, Children's National Hospital, Washington, DC, USA
| | - Troy C Quigg
- Pediatric Blood & Bone Marrow Transplant and Cellular Therapy, Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Roberta H Adams
- Center for Cancer and Blood Disorders, Phoenix Children's/Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Ann Dahlberg
- Clinical Research Division, Fred Hutch Cancer Center/Seattle Children's Hospital/University of Washington, Seattle, WA, USA
| | | | - Hasibul Hasan
- Cancer and blood disease institute, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Jemily Malvar
- Cancer and blood disease institute, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | | | - Christopher A Lazarski
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Gelina Sani
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - John M Idso
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Haili Lang
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Pamela Chansky
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Chase D McCann
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Jay Tanna
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Allistair A Abraham
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University School of Medicine, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Jennifer L Webb
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
- Division of Hematology, Children's National Hospital, Washington, DC, USA
| | - Abeer Shibli
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
| | - Amy K Keating
- Pediatric Stem Cell Transplant, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
| | - Prakash Satwani
- Division of Pediatric Hematology/Oncology and Stem Cell Transplantation, Columbia University Medical Center, New York, NY, USA
| | - Pawel Muranski
- Division of Pediatric Hematology/Oncology and Stem Cell Transplantation, Columbia University Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Erin Hall
- Division of Pediatric Hematology/Oncology/Bone Marrow Transplant, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Michael J Eckrich
- Pediatric Transplant and Cellular Therapy, Levine Children's Hospital, Wake Forest School of Medicine, Charlotte, NC, USA
| | - Evan Shereck
- Division of Hematology and Oncology, Oregon Health & Science Univ, Portland, OR, USA
| | - Holly Miller
- Center for Cancer and Blood Disorders, Phoenix Children's/Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Ewelina Mamcarz
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rajni Agarwal
- Division of Pediatric Hematology/Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University, Palo Alto, CA, USA
| | - Satiro N De Oliveira
- Division of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mark T Vander Lugt
- Division of Pediatric Hematology/Oncology/BMT, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, MI, USA
| | - Christen L Ebens
- Division of Pediatric Blood and Marrow Transplant & Cellular Therapy, University of Minnesota MHealth Fairview Masonic Children's Hospital, Minneapolis, MI, USA
| | - Victor M Aquino
- Division of Pediatric Hematology/Oncology, University of Texas, Southwestern Medical Center Dallas, Dallas, TX, USA
| | - Jeffrey J Bednarski
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - Julia Chu
- Division of Pediatric Allergy, Immunology, and BMT, University of California San Francisco, San Francisco, CA, USA
| | - Suhag Parikh
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Jennifer Whangbo
- Cancer and Blood Disorders Center, Dana Farber Institute and Boston Children's Hospital, Boston, MA, USA
| | - Michail Lionakis
- Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Elias T Zambidis
- Pediatric Blood and Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth Gourdine
- Cancer and blood disease institute, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Catherine M Bollard
- Center for Cancer & Immunology Research, Children's National Hospital, Washington, DC, USA
- GW Cancer Center, George Washington University School of Medicine, Washington, DC, USA
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Michael A Pulsipher
- Division of Pediatric Hematology/Oncology, Intermountain Primary Children's Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
2
|
De Simone P, Battistella S, Lai Q, Ducci J, D'Arcangelo F, Marchetti P, Russo FP, Burra P. Immunosuppression for older liver transplant recipients. Transplant Rev (Orlando) 2024; 38:100817. [PMID: 38128152 DOI: 10.1016/j.trre.2023.100817] [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: 11/16/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Older liver transplant recipients have a lower risk of acute rejection than younger patients (9% for patients aged ≥65 years versus 23% for those aged 18-34 years) and are more vulnerable to immunosuppression-related complications. The number of liver transplant recipients ≥65 years has risen to 22% in Europe and the US, but limited information is available on the optimal immunosuppressive regimen for these patients. In this review, we discuss the appropriate management of immunosuppressive agents in older adults to minimize adverse events while avoiding acute rejection. The way the body processes drugs greatly depends on age. In the case of calcineurin inhibitor drugs, aging reduces hepatic metabolism, leading to changes in their pharmacokinetics. Corticosteroids also show decreased clearance as the patient ages. In severe cases of hypoalbuminemia, dose adjustment of mycophenolate acid derivatives may be necessary. However, the pharmacokinetic profiles of the mammalian target of rapamycin inhibitors, basiliximab, and rabbit anti-thymocyte globulin remain unaffected by age. Furthermore, age-related frailty may impact drug metabolism and require tailored interventions and closer follow-up. Although there is limited research, elderly liver transplant recipients require less immunosuppression with double or triple-agent regimens, lower exposure to calcineurin inhibitors, and a shorter course of corticosteroids. The usage of mammalian target of rapamycin inhibitors in older transplant populations has not been specifically investigated, and thus their usage should align with indications for younger patient groups.
Collapse
Affiliation(s)
- Paolo De Simone
- Liver Transplant Program, University of Pisa Medical School Hospital, Pisa, Italy; Department of Surgical, Medical, Biochemical Pathology and Intensive Care, University of Pisa, Italy.
| | - Sara Battistella
- Gastroenterology, Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, Padua University Hospital, Padua, Italy
| | - Quirino Lai
- General Surgery and Organ Transplantation Unit, La Sapienza University of Rome, Italy
| | - Juri Ducci
- Liver Transplant Program, University of Pisa Medical School Hospital, Pisa, Italy
| | - Francesca D'Arcangelo
- Gastroenterology, Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, Padua University Hospital, Padua, Italy
| | - Piero Marchetti
- Diabetology Unit, University of Pisa Medical School Hospital, Pisa, Italy
| | - Francesco Paolo Russo
- Gastroenterology, Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, Padua University Hospital, Padua, Italy
| | - Patrizia Burra
- Gastroenterology, Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, Padua University Hospital, Padua, Italy
| |
Collapse
|
3
|
Takahashi T, Teramoto M, Matsumoto K, Jaber MM, Tamaki H, Ikegame K, Yoshihara S, Kaida K. Population Pharmacokinetics of Total Rabbit Anti-thymocyte Globulin in Non-obese Adult Patients Undergoing Hematopoietic Cell Transplantation for Hematologic Malignancy. Clin Pharmacokinet 2023; 62:1081-1091. [PMID: 37284975 DOI: 10.1007/s40262-023-01252-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2023] [Indexed: 06/08/2023]
Abstract
BACKGROUND AND OBJECTIVES Rabbit anti-thymocyte globulin (rATG), a therapeutic polyclonal antibody against human T cells, is commonly used in conditioning therapy prior to allogeneic hematopoietic cell transplantation (HCT). Previous studies successfully developed an individualized rATG dosing regimen based on "active" rATG population PK (popPK) analysis, while "total" rATG can be a more logistically favorable alternative for early HCT outcomes. We conducted a novel popPK analysis of total rATG. METHODS Total rATG concentration was measured in adult human-leukocyte antigen (HLA) mismatched HCT patients who received a low-dose rATG regimen (total 2.5-3 mg/kg) within 3 days prior to HCT. PopPK modeling and simulation was performed using nonlinear mixed effect modeling approach. RESULTS A total of 504 rATG concentrations were available from 105 non-obese patients with hematologic malignancy (median age 47 years) treated in Japan. The majority had acute leukemia or malignant lymphoma (94%). Total rATG PK was described by a two-compartment linear model. Influential covariate relations include ideal body weight [positively on both clearance (CL) and central volume of distribution], baseline serum albumin (negatively on CL), CD4+ T cell dose (positively on CL), and baseline serum IgG (positively on CL). Simulated covariate effects predicted that early total rATG exposures were affected by ideal body weight. CONCLUSIONS This novel popPK model described the PK of total rATG in the adult HCT patients who received a low-dose rATG conditioning regimen. This model can be used for model-informed precision dosing in the settings with minimal baseline rATG targets (T cells), and early clinical outcomes are of interest.
Collapse
Affiliation(s)
- Takuto Takahashi
- Pediatric Stem Cell Transplantation, Boston Children's Hospital/Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, USA.
| | - Masahiro Teramoto
- Department of Hematology, Hyogo Medical University Hospital, Nishinomiya, Hyogo, Japan
| | - Kana Matsumoto
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Kyoto, Japan
| | - Mutaz M Jaber
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, USA
| | - Hiroya Tamaki
- Department of Hematology, Hyogo Medical University Hospital, Nishinomiya, Hyogo, Japan
| | - Kazuhiro Ikegame
- Department of Hematology, Hyogo Medical University Hospital, Nishinomiya, Hyogo, Japan
| | - Satoshi Yoshihara
- Department of Hematology, Hyogo Medical University Hospital, Nishinomiya, Hyogo, Japan
| | - Katsuji Kaida
- Department of Hematology, Hyogo Medical University Hospital, Nishinomiya, Hyogo, Japan
| |
Collapse
|
4
|
Muckenhuber M, Mucha J, Mengrelis K, How C, Reindl-Schwaighofer R, Heinzel A, Kainz V, Worel N, Berlakovich G, Edinger M, Oberbauer R, Wekerle T. Optimum timing of antithymocyte globulin in relation to adoptive regulatory T cell therapy. Am J Transplant 2023; 23:84-92. [PMID: 36695625 DOI: 10.1016/j.ajt.2022.09.002] [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: 06/19/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 01/13/2023]
Abstract
Reducing the recipient's T cell repertoire is considered to increase the efficacy of regulatory T cell (Treg) therapy. This necessitates timing the administration of antithymocyte globulin (ATG) early enough before adoptive cell therapy (ACT) so that residual serum ATG does not deplete the transferred Tregs. The optimum time point in this regard has not been defined. Herein, we report the effects of residual serum ATG on the viability of an in vitro expanded Treg cell product used in a clinical trial of ACT in kidney transplant recipients (NCT03867617). Patients received ATG monotherapy (either 6 or 3 mg/kg body weight) without concomitant immunosuppression 2 to 3 weeks before transplantation and Treg transfer. An anti-ATG immunoglobulin G (IgG) immune response was elicited in all patients within 14 days. In turn, the elimination of total and Treg-specific ATG was accelerated substantially over control patients receiving the same dose of ATG with concomitant immunosuppression. However, ATG serum concentrations of <1 μg/mL, which had previously been reported as subtherapeutic threshold, triggered apoptosis of Tregs in vitro. Therefore, ATG levels need to decline to lower levels than those previously thought for efficacious Treg transfer. In 5 of 6 patients, such low levels of serum ATG considered safe for Treg transfer were reached within 2 weeks after ATG administration.
Collapse
Affiliation(s)
- Moritz Muckenhuber
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Jasmin Mucha
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Konstantinos Mengrelis
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Christopher How
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Heinzel
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Verena Kainz
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Nina Worel
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Gabriela Berlakovich
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Matthias Edinger
- Leibniz Institute of Immunotherapy, Regensburg, Germany; Department of Internal Medicine 3 (Hematology and Oncology), University Hospital Regensburg, Regensburg, Germany
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
5
|
Waller EK. Donor NK cells facilitate thymopoiesis in allo-BMT. Blood 2022; 140:2307-2308. [PMID: 36454592 DOI: 10.1182/blood.2022017856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
6
|
Wang H, Zhao Y, Fang S, Wang L, Peng B, Yang J, Wang N, Du J, Li F, Jin X, Luan S, Wu X, Dou L, Liu D. Optimal Active Anti-Thymocyte Globulin Exposure Associated with Minimum Risk of Virus Reactivation and Comparable Acute Graft-Versus-Host Disease Under Adult Myeloablative Haploidentical Peripheral Blood Stem Cell Transplantation. Transplant Cell Ther 2022; 28:332.e1-332.e10. [PMID: 35314377 DOI: 10.1016/j.jtct.2022.03.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 11/15/2022]
Abstract
Anti-thymocyte globulin (ATG) is often included in the conditioning regimen to prevent graft-versus-host disease (GVHD) in allogeneic hematopoietic cell transplantation (allo-HCT). However, the risk of virus reactivation increases significantly. We conducted a single-center prospective study to identify the optimal ATG exposure that ensures engraftment, effectively prevents acute GVHD, and reduces the risk of virus reactivation without increasing relapse of malignant diseases in haploidentical peripheral blood stem cell transplantation (haplo-PBSCT). From September 2018 to June 2020, 106 patients (median age, 32 years) with malignant hematological diseases who received haplo-PBSCT for the first time were enrolled. All patients received 10 mg/kg rabbit ATG (thymoglobulin) divided for 4 days (days -5 to -2). Pre-transplant, post-transplant, and total areas under the concentration-time curve (AUCs) of active ATG were calculated. Total AUC of active ATG was shown to be the best predictor for virus reactivation and acute GVHD of grades II to IV or grades III and IV. The optimal total AUC range of active ATG was 100 to 148.5 UE/mL/day. The median time was 14 versus 13 days (P = .184) for myeloid engraftment and 13 versus 13 days (P = .263) for platelet engraftment in the optimal and non-optimal AUC groups, respectively. The optimal AUC group showed a lower cumulative incidence of cytomegalovirus (CMV) reactivation and persistent CMV viremia than the non-optimal AUC group: 60.6% (95% confidence interval [CI], 48.3%-73.1%) versus 77.1% (95% CI, 64.5%-87.7%; P = .016) and 31.5% (95% CI, 21.2%-45.3%) versus 56.3% (95% CI, 42.9%-70.4%; P = .007), respectively. The cumulative incidence of persistent Epstein-Barr virus (EBV) viremia in the optimal AUC group was significantly lower than the non-optimal total AUC group: 33.1% (95% CI, 22.5%-46.8%) versus 52.6% (95% CI, 39.3%-67.2%; P = .048). However, there was no difference in EBV reactivation (P = .752). Similar outcomes were observed for grade II to IV and grade III and IV acute GVHD between the two groups: 48.6% (95% CI, 36.8%-62.0%) versus 37.0% (95% CI, 24.8%-52.5%; P = .113) and 10.4% (95% CI, 4.8%-21.7%) versus 4.2% (95% CI, 1.0%-15.6%; P = .234, respectively. Relapse, non-relapse mortality, and disease-free survival demonstrated no significant differences between the two groups. But, overall survival at 2 years tended to increase in the optimal AUC group: 75.7% (95% CI, 62.4%-84.8%) versus 57.8% (95% CI, 42.4%-70.4%; P = .061). These data support an optimal active ATG exposure of 110 to 148.5 UE/mL/day in haplo-PBSCT. Individualized dosing of ATG in allo-HCT might reduce the risk of virus reactivation and effectively prevent acute GVHD simultaneously.
Collapse
Affiliation(s)
- HaiTao Wang
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Chinese PLA General Hospital, Beijing, China
| | | | - Shu Fang
- Chinese PLA General Hospital, Beijing, China; School of Medicine, Nankai University, Tianjin, China
| | - LiLi Wang
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Bo Peng
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Nan Wang
- Chinese PLA General Hospital, Beijing, China
| | - JiShan Du
- Chinese PLA General Hospital, Beijing, China
| | - Fei Li
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - XiangShu Jin
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - SongHua Luan
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - XiaoXiong Wu
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - LiPing Dou
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Chinese PLA General Hospital, Beijing, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - DaiHong Liu
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
7
|
van der Stoep MYEC, Oostenbrink LVE, Bredius RGM, Moes DJAR, Guchelaar HJ, Zwaveling J, Lankester AC. Therapeutic Drug Monitoring of Conditioning Agents in Pediatric Allogeneic Stem Cell Transplantation; Where do We Stand? Front Pharmacol 2022; 13:826004. [PMID: 35330826 PMCID: PMC8940165 DOI: 10.3389/fphar.2022.826004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is an established curative treatment that has significantly improved clinical outcome of pediatric patients with malignant and non-malignant disorders. This is partly because of the use of safer and more effective combinations of chemo- and serotherapy prior to HSCT. Still, complications due to the toxicity of these conditioning regimens remains a major cause of transplant-related mortality (TRM). One of the most difficult challenges to further improve HSCT outcome is reducing toxicity while maintaining efficacy. The use of personalized dosing of the various components of the conditioning regimen by means of therapeutic drug monitoring (TDM) has been the topic of interest in the last decade. TDM could play an important role, especially in children who tend to show greater pharmacokinetic variability. However, TDM should only be performed when it has clear added value to improve clinical outcome or reduce toxicity. In this review, we provide an overview of the available evidence for the relationship between pharmacokinetic parameters and clinical outcome or toxicities of the most commonly used conditioning agents in pediatric HSCT.
Collapse
Affiliation(s)
- M. Y. Eileen C. van der Stoep
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: M. Y. Eileen C. van der Stoep,
| | - Lisa V. E. Oostenbrink
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Robbert G. M. Bredius
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Dirk Jan A. R. Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Juliette Zwaveling
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Arjan C. Lankester
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
8
|
Algaze CA, Margetson TD, Sutherland SM, Kwiatkowski DM, Maeda K, Navaratnam M, Samreth SP, Price EP, Zook NB, Yang JK, Hollander SA. Impact of a clinical pathway on acute kidney injury in patients undergoing heart transplant. Pediatr Transplant 2022; 26:e14166. [PMID: 34727417 DOI: 10.1111/petr.14166] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND To evaluate the impact of a clinical pathway on the incidence and severity of acute kidney injury in patients undergoing heart transplant. METHODS This was a 2.5-year retrospective evaluation using 3 years of historical controls within a cardiac intensive care unit in an academic children's hospital. Patients undergoing heart transplant between May 27, 2014, and April 5, 2017 (pre-pathway) and May 1, 2017, and November 30, 2019 (pathway) were included. The clinical pathway focused on supporting renal perfusion through hemodynamic management, avoiding or delaying nephrotoxic medications, and providing pharmacoprophylaxis against AKI. RESULTS There were 57 consecutive patients included. There was an unadjusted 20% reduction in incidence of any acute kidney injury (p = .05) and a 17% reduction in Stage 2/3 acute kidney injury (p = .09). In multivariable adjusted analysis, avoidance of Stage 2/3 acute kidney injury was independently associated with the clinical pathway era (AOR -1.3 [95% CI -2.5 to -0.2]; p = .03), achieving a central venous pressure of or less than 12 mmHg (AOR -1.3 [95% CI -2.4 to -0.2]; p = .03) and mean arterial pressure above 60 mmHg (AOR -1.6 [95% CI -3.1 to -0.01]; p = .05) in the first 48 h post-transplant, and older age at transplant (AOR - 0.2 [95% CI -0.2 to -0.06]; p = .002). CONCLUSIONS This report describes a renal protection clinical pathway associated with a reduction in perioperative acute kidney injury in patients undergoing heart transplant and highlights the importance of normalizing perioperative central venous pressure and mean arterial blood pressure to support optimal renal perfusion.
Collapse
Affiliation(s)
- Claudia A Algaze
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA.,Center for Pediatric and Maternal Value, Stanford University School of Medicine, Palo Alto, California, USA
| | - Tristan D Margetson
- Department of Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Scott M Sutherland
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - David M Kwiatkowski
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Katsuhide Maeda
- Department of Surgery, Stanford University School of Medicine, Palo Alto, California, USA
| | - Manchula Navaratnam
- Department of Anesthesia, Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, California, USA
| | - Sarah P Samreth
- Center for Pediatric and Maternal Value, Stanford University School of Medicine, Palo Alto, California, USA
| | - Elizabeth P Price
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Nina B Zook
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Jeffrey K Yang
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Seth A Hollander
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
9
|
Long-term survival with mixed chimerism in patients with AML and MDS transplanted after conditioning with targeted busulfan, fludarabine, and thymoglobulin. Bone Marrow Transplant 2021; 57:198-206. [PMID: 34741096 DOI: 10.1038/s41409-021-01518-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 11/08/2022]
Abstract
We evaluated long-term outcome in 40 patients with MDS or AML, transplanted from related or unrelated donors following conditioning with targeted busulfan (Bu, over 4 days), fludarabine (Flu, 120 [n = 23] or 250 [n = 17] mg/m2) and thymoglobulin (THY). Compared to 95 patients conditioned with Bu/Cyclophosphamide (Cy) without THY, BuFluTHY-conditioned patients had lower rates of chronic graft-vs.-host disease (GVHD). Adjusted hazard ratios (HR) for BuFlu(120)THY and BuFlu(250)THY-conditioned patients were 1.60 (95% confidence interval (CI) 0.66-3.86) and 1.87 (0.68-5.11), respectively, for relapse; 0.77 (0.30-1.99) and 1.32 (0.54-3.23) for non-relapse mortality; 0.81 (0.42-1.57) and 1.38 (0.72-2.57) for overall mortality; and 0.78 (0.30-2.05) and 1.62 (0.63-4.41) for relapse or death (failure for relapse-free survival). At one year, 45% of BuFlu(120 or 250)THY-conditioned patients had mixed CD3+ chimerism compared to 0% with BuCy (p < 0.0001). None of 7 patients with long-term mixed chimerism had chronic GVHD; two relapsed, five remained stable mixed chimeras. THY is effective in reducing chronic GVHD, and long-term mixed T-cell chimerism can be compatible with relapse-free survival. However, Thy may also be associated with an increased risk of relapse and, dose-dependent, with non-relapse mortality.
Collapse
|
10
|
Azzopardi N, Longuet H, Ternant D, Thibault G, Gouilleux-Gruart V, Lebranchu Y, Büchler M, Gatault P, Paintaud G. Relationship Between Antithymocyte Globulin Concentrations and Lymphocyte Sub-Populations in Kidney Transplant Patients. Clin Pharmacokinet 2021; 61:111-122. [PMID: 34292526 DOI: 10.1007/s40262-021-01053-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Rabbit antithymocyte globulins (rATGs) are polyclonal antibodies used to prevent acute cellular rejection in kidney transplantation. Their dosing remains largely empirical and the question of an individualized dose is still unresolved. METHODS Data from a prospective study in 17 kidney transplant patients were used to develop a model describing the dose-concentration-response relationship of rATG with T-lymphocyte subpopulation counts over time. The model was validated using an independent cohort of kidney transplant patients treated by rATG in the same center. RESULTS Pharmacokinetics of rATG was described using a two-compartment model integrating a third compartment and a target-mediated elimination for active rATG. The kinetics of CD3+, CD4+, CD8+, and CD3-CD56+ cell counts over time were described by a pharmacokinetic-pharmacodynamic model with transit compartments, integrating both CD3-CD56+-independent and CD3-CD56+-dependent rATG-mediated lymphocyte depletion, and a positive feedback. Elimination of rATG was influenced by age and body surface area, while its distribution was also influenced by body surface area. CD3+ proliferation rate decreased with age and CD3-CD56+-mediated elimination was influenced by the V158F-FCGR3A polymorphism. Binary efficacy and tolerance endpoints were defined as a CD3+ count < 20 mm-3 for at least 7 days and a CD4+ count > 200 mm-3 at 1 year, respectively. Simulations showed that increasing or decreasing the standard 6-mg/kg dose will impact both tolerance and efficacy, while a dose decrease may be beneficial in elderly patients. CONCLUSIONS Our results can be used to design prospective clinical trials testing dose individualization based on patients' characteristics. CLINICAL TRIAL REGISTRATION Eudract No. 2009-012673-35.
Collapse
Affiliation(s)
| | - Hélène Longuet
- Department of Nephrology and Clinical Immunology, CHRU de Tours, Tours, France
| | - David Ternant
- University of Tours, EA4245 T2I, Tours, France. .,Department of Medical Pharmacology, CHRU de Tours, 37044, Tours, France.
| | - Gilles Thibault
- University of Tours, EA7501 GICC, Tours, France.,Laboratory of Immunology, CHRU de Tours, Tours, France
| | - Valérie Gouilleux-Gruart
- University of Tours, EA7501 GICC, Tours, France.,Laboratory of Immunology, CHRU de Tours, Tours, France
| | | | - Matthias Büchler
- Department of Nephrology and Clinical Immunology, CHRU de Tours, Tours, France.,University of Tours, EA4245 T2I, Tours, France
| | - Philippe Gatault
- Department of Nephrology and Clinical Immunology, CHRU de Tours, Tours, France.,University of Tours, EA4245 T2I, Tours, France
| | - Gilles Paintaud
- University of Tours, EA4245 T2I, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, 37044, Tours, France
| |
Collapse
|
11
|
Marfil-Garza BA, Hefler J, Bermudez De Leon M, Pawlick R, Dadheech N, Shapiro AMJ. Progress in Translational Regulatory T Cell Therapies for Type 1 Diabetes and Islet Transplantation. Endocr Rev 2021; 42:198-218. [PMID: 33247733 DOI: 10.1210/endrev/bnaa028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Regulatory T cells (Tregs) have become highly relevant in the pathophysiology and treatment of autoimmune diseases, such as type 1 diabetes (T1D). As these cells are known to be defective in T1D, recent efforts have explored ex vivo and in vivo Treg expansion and enhancement as a means for restoring self-tolerance in this disease. Given their capacity to also modulate alloimmune responses, studies using Treg-based therapies have recently been undertaken in transplantation. Islet transplantation provides a unique opportunity to study the critical immunological crossroads between auto- and alloimmunity. This procedure has advanced greatly in recent years, and reports of complete abrogation of severe hypoglycemia and long-term insulin independence have become increasingly reported. It is clear that cellular transplantation has the potential to be a true cure in T1D, provided the remaining barriers of cell supply and abrogated need for immune suppression can be overcome. However, the role that Tregs play in islet transplantation remains to be defined. Herein, we synthesize the progress and current state of Treg-based therapies in T1D and islet transplantation. We provide an extensive, but concise, background to understand the physiology and function of these cells and discuss the clinical evidence supporting potency and potential Treg-based therapies in the context of T1D and islet transplantation. Finally, we discuss some areas of opportunity and potential research avenues to guide effective future clinical application. This review provides a basic framework of knowledge for clinicians and researchers involved in the care of patients with T1D and islet transplantation.
Collapse
Affiliation(s)
| | - Joshua Hefler
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Mario Bermudez De Leon
- Department of Molecular Biology, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey, Nuevo Leon, Mexico
| | - Rena Pawlick
- Department of Surgery, University of Alberta, Edmonton, Canada
| | | | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, Canada.,Clinical Islet Transplant Program, University of Alberta, Edmonton, Canada
| |
Collapse
|
12
|
Keogh SJ, Dalle JH, Admiraal R, Pulsipher MA. Serotherapy as Graft-Versus-Host Disease Prophylaxis in Haematopoietic Stem Cell Transplantation for Acute Lymphoblastic Leukaemia. Front Pediatr 2021; 9:805189. [PMID: 35071142 PMCID: PMC8771860 DOI: 10.3389/fped.2021.805189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/01/2021] [Indexed: 12/26/2022] Open
Abstract
Serotherapy comprising agents such as anti-thymocyte globulin, anti-T-lymphocyte globulin, and the anti-CD52 monoclonal antibody alemtuzumab is used widely to reduce the incidence of graft-versus-host disease (GvHD) after paediatric haematopoietic stem cell transplantation (HSCT). The outcome of transplants using matched unrelated donors now approaches that of matched sibling donors. This is likely due to better disease control in recipients, the use of donors more closely human-leukocyte antigen (HLA)-matched to recipients, and more effective graft-versus-host disease (GvHD) prophylaxis. The price paid for reduced GvHD is slower immune reconstitution of T cells and thus more infections. This has led to studies looking to optimise the amount of serotherapy used. The balance between prevention of GvHD on one side and prevention of infections and relapse on the other side is quite delicate. Serotherapy is given with chemotherapy-/radiotherapy-based conditioning prior to HSCT. Due to their long half-lives, agents used for serotherapy may be detectable in patients well after graft infusion. This exposes the graft-infused T cells to a lympholytic effect, impacting T-cell recovery. As such, excessive serotherapy dosing may lead to no GvHD but a higher incidence of infections and relapse of leukaemia, while under-dosing may result in a higher chance of serious GvHD as immunity recovers more quickly. Individualised dosing is being developed through studies including retrospective analyses of serotherapy exposure, population pharmacokinetic modelling, therapeutic drug monitoring in certain centres, and the development of dosing models reliant on factors including the patient's peripheral blood lymphocyte count. Early results of "optimal" dosing strategies for serotherapy and conditioning chemotherapy show promise of improved overall survival.
Collapse
Affiliation(s)
- Steven J Keogh
- Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Jean-Hugues Dalle
- Hôpital Robert Debré, GHU AP-HP. Nord Université de Paris, Paris, France
| | - Rick Admiraal
- Princess Maxima Center for Pediatric Oncology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Michael A Pulsipher
- Children's Hospital Los Angeles, Cancer and Blood Disease Institute, Los Angeles, CA, United States
| |
Collapse
|
13
|
Antirelapse effect of pretransplant exposure to rabbit antithymocyte globulin. Blood Adv 2020; 3:1394-1405. [PMID: 31043372 DOI: 10.1182/bloodadvances.2018030247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/24/2019] [Indexed: 01/15/2023] Open
Abstract
It remains unknown why rabbit antithymocyte globulin (ATG; Thymoglobulin) has not affected relapse after hematopoietic cell transplantation (HCT) in randomized studies. We hypothesized that high pre-HCT ATG area under the curve (AUC) would be associated with a low incidence of relapse, whereas high post-HCT AUC would be associated with a high incidence of relapse. We measured serum levels of ATG capable of binding to mononuclear cells (MNCs), lymphocytes, T cells, CD4 T cells, or CD33 cells. We estimated pre- and post-HCT AUCs in 152 adult recipients of myeloablative conditioning and blood stem cells. High pre-HCT AUCs of MNC- and CD33 cell-binding ATG were associated with a low incidence of relapse and high relapse-free survival (RFS). There was a trend toward an association of high post-HCT AUC of lymphocyte-binding ATG with a high incidence of relapse and low RFS. High pre-HCT AUCs were also associated with faster engraftment and had no impact on graft-versus-host disease (GVHD) or fatal infections. High post-HCT AUCs were associated with a low risk of GVHD, seemed associated with an increased risk of fatal infections, and had no impact on engraftment. In conclusion, pre-HCT AUC seems to have a positive, whereas post-HCT AUC seems to have a negative, impact on relapse.
Collapse
|
14
|
Oostenbrink LVE, Jol-van der Zijde CM, Jansen-Hoogendijk AM, Pool ES, van Halteren AGS, Moes DJAR, Bredius RGM, Mohseny AB, Smiers FJW, van Tol MJD, Schilham MW, Lankester AC. Proceeding of the European Group for Blood and Marrow Transplantation (EBMT) congress on sickle cell disease, 16-17 may 2019, Regensburg, Germany: What is the impact of antithymocyte globulin pharmacokinetics on haploidentical hematopoietic stem cell transplantation? Hematol Oncol Stem Cell Ther 2020; 13:61-65. [PMID: 32202242 DOI: 10.1016/j.hemonc.2019.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/11/2019] [Indexed: 01/18/2023] Open
Abstract
Antithymocyte globulin (ATG) is a widely accepted part of the conditioning regimen applied in the setting of hematopoietic stem cell transplantation (HSCT) to prevent graft rejection and graft-versus-host disease. Although weight-based dosing of ATG has been introduced to optimize ATG dosing, substantial variance in clearance of active ATG, the actual lymphocyte binding component, remains a challenge. Therefore, further research regarding ATG pharmacokinetics and pharmacodynamics in different HSCT settings and in patients with different types of underlying diseases is required.
Collapse
Affiliation(s)
- Lisa V E Oostenbrink
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands.
| | | | | | - Emma S Pool
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Dirk Jan A R Moes
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Robbert G M Bredius
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Alex B Mohseny
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Frans J W Smiers
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten J D van Tol
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Marco W Schilham
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Arjan C Lankester
- Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
15
|
Kako S, Kanda Y, Onizuka M, Aotsuka N, Usuki K, Tachibana T, Kobayashi T, Kato J, Yano S, Shimizu H, Shono K, Tanaka M, Tsukamoto S, Mori T, Yamazaki E, Najima Y, Hangaishi A, Hoshino T, Watanabe R, Matsumoto K, Okamoto S. Allogeneic hematopoietic stem cell transplantation for aplastic anemia with pre-transplant conditioning using fludarabine, reduced-dose cyclophosphamide, and low-dose thymoglobulin: A KSGCT prospective study. Am J Hematol 2020; 95:251-257. [PMID: 31804748 DOI: 10.1002/ajh.25693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 11/10/2022]
Abstract
The optimal pre-transplant conditioning for aplastic anemia (AA) remains unclear. We performed a prospective study on allogeneic transplantation from a related or unrelated donor for adult patients with AA. We assessed whether reduced-dose cyclophosphamide (CY) could decrease toxicity while maintaining engraftment, and low-dose thymoglobulin could safely prevent graft-vs-host disease (GVHD). The pre-transplant conditioning regimen consisted of fludarabine 120 mg/m2 , CY 100 mg/kg, and thymoglobulin 2.5 mg/kg with or without 2 Gy of total body irradiation. Twenty-seven patients with a median age of 36 years were analyzed. Sixteen patients received graft from related donors. The stem cell source was bone marrow in 26 patients. All of the patients but one, who died early, achieved neutrophil engraftment at a median of 19 days. Mixed chimerism was observed in six and five patients at days 30 and 90, respectively. Only one patient experienced secondary engraftment failure with complete donor-type chimerism. None of the patients developed severe acute GVHD. The cumulative incidence of chronic GVHD was 37.7% at 1 year. The overall survival rate was 96.3% at 1 year and 3 years. A high EB virus-DNA load was detected in one patient at days 60. No one developed EBV-lymphoproliferative disorder within a year. The results suggest that the conditioning regimen in this study was safe and effective. However, relatively high incidence of chronic GVHD needs further improvement.
Collapse
Affiliation(s)
- Shinichi Kako
- Division of HematologyJichi Medical University Saitama Medical Center Saitama Japan
| | - Yoshinobu Kanda
- Division of HematologyJichi Medical University Saitama Medical Center Saitama Japan
| | - Makoto Onizuka
- Department of Hematology and OncologyTokai University School of Medicine Kanagawa Japan
| | - Nobuyuki Aotsuka
- Division of Hematology‐OncologyJapanese Red Cross Society Narita Hospital Chiba Japan
| | - Kensuke Usuki
- Department of HematologyNTT Medical Center Tokyo Tokyo Japan
| | - Takayoshi Tachibana
- Department of HematologyYokohama City University Medical Center Yokohama Japan
| | - Takeshi Kobayashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases CenterKomagome Hospital Tokyo Japan
| | - Jun Kato
- Division of Hematology, Department of MedicineKeio University School of Medicine Tokyo Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal MedicineThe Jikei University School of Medicine Tokyo Japan
| | - Hiroaki Shimizu
- Department of Medicine and Clinical ScienceGunma University Gunma Japan
| | - Katsuhiro Shono
- Department of HematologyChiba Aoba Municipal Hospital Chiba Japan
| | | | | | - Takehiko Mori
- Division of Hematology, Department of MedicineKeio University School of Medicine Tokyo Japan
| | - Etsuko Yamazaki
- Department of Hematology and Clinical ImmunologyYokohama City University School of Medicine Kanagawa Japan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases CenterKomagome Hospital Tokyo Japan
| | - Akira Hangaishi
- Department of HematologyNTT Medical Center Tokyo Tokyo Japan
| | - Takumi Hoshino
- Leukemia Research CenterSaiseikai Maebashi Hospital Gunma Japan
| | - Reiko Watanabe
- Department of Hematology, Saitama Medical CenterSaitama Medical University Saitama Japan
| | - Kenji Matsumoto
- Department of HematologyKanagawa Cancer Center Kanagawa Japan
- Department of Hematology and Clinical ImmunologyYokohama City University School of Medicine Kanagawa Japan
| | - Shinichiro Okamoto
- Division of Hematology, Department of MedicineKeio University School of Medicine Tokyo Japan
| | | |
Collapse
|
16
|
Amrani ME, Admiraal R, Willaert L, Ebskamp-van Raaij LJC, Lacna AM, Hack CE, Huitema ADR, Nierkens S, van Maarseveen EM. Quantification of T Cell Binding Polyclonal Rabbit Anti-thymocyte Globulin in Human Plasma with Liquid Chromatography Tandem-Mass Spectrometry. AAPS J 2020; 22:43. [PMID: 32030538 PMCID: PMC7005072 DOI: 10.1208/s12248-020-0419-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/07/2020] [Indexed: 11/30/2022] Open
Abstract
The addition of rabbit anti-human thymocyte globulin (ATG) to the conditioning regimen prior to allogeneic hematopoietic cell transplantation has significantly reduced the risk of graft-versus-host disease (GvHD) and graft failure. However, ATG has a small therapeutic window. Overexposure of ATG post-HCT hampers T cell immune reconstitution and has been associated with increased relapse rates and viral reactivations, whereas underexposure has been associated with an increased incidence of GvHD, both of which lead to increased mortality. Therapeutic drug monitoring of T cell binding ATG plasma levels provides a means to optimize dosing for patients at high risk for graft failure to ensure timely T cell immune reconstitution and subsequently increase survival chances. This manuscript describes the first liquid chromatography tandem-mass spectrometry (LC-MS/MS) method to quantify the pharmacologically active fraction of polyclonal ATG in plasma. This was achieved through immunoaffinity purification of active ATG from plasma with Jurkat T cells. After the binding and washing, samples were eluted, denatured, and trypsin-digested. Signature peptides originating from the IgG constant chain were measured with LC-MS/MS. Critical method parameters were optimized, and the method was successfully validated following European Medicines Agency (EMA) guidelines. The method covered the therapeutic range of ATG and was validated at a lower limit of quantification (LLOQ) of 1 AU/mL with an overall CV and bias of 11.8% and - 2.5%, respectively. In conclusion, we developed a LC-MS/MS-based method to quantify active polyclonal rabbit ATG in human plasma. We suggest that this novel assay can be used to monitor and optimize dosing of ATG in clinical practice.
Collapse
Affiliation(s)
- Mohsin El Amrani
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Room no. D.00.318A, Internal post no. D.00.204, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Rick Admiraal
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Lobke Willaert
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Room no. D.00.318A, Internal post no. D.00.204, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Lysette J C Ebskamp-van Raaij
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Amelia M Lacna
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - C Erik Hack
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Alwin D R Huitema
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Room no. D.00.318A, Internal post no. D.00.204, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Erik M van Maarseveen
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, Room no. D.00.318A, Internal post no. D.00.204, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Irure J, Sango C, San Segundo D, Fernández-Fresnedo G, Ruiz JC, Benito-Hernández A, Asensio E, López-Hoyos M, Rodrigo E. Late Plasma Cell Depletion After Thymoglobulin Induction in Kidney Transplant Recipients. EXP CLIN TRANSPLANT 2019; 17:732-738. [DOI: 10.6002/ect.2018.0261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Aversa F, Pierini A, Ruggeri L, Martelli MF, Velardi A. The Evolution of T Cell Depleted Haploidentical Transplantation. Front Immunol 2019; 10:2769. [PMID: 31827475 PMCID: PMC6890606 DOI: 10.3389/fimmu.2019.02769] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022] Open
Abstract
Work on bone marrow transplantation from haploidentical donor has been proceeding for over 20 years all over the world and new transplant procedures have been developed. To control both graft rejection and graft vs. host disease, some centers have preferred to enhance the intensity of the conditioning regimens and the post-transplant immune suppression in the absence of graft manipulation; others have concentrated on manipulating the graft in the absence of any additional post-transplant immune suppressive agent. Due to the current high engraftment rates, the low incidence of graft-vs.-host disease and regimen related mortality, transplantation from haploidentical donors have been progressively offered even to elderly patients. Overall, survivals compare favorably with reports on transplants from unrelated donors. Further improvements will come with successful implementation of strategies to enhance post-transplant immune reconstitution and to prevent leukemia relapse.
Collapse
Affiliation(s)
- Franco Aversa
- Hematology and Bone Marrow Transplantation Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonio Pierini
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Loredana Ruggeri
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Massimo Fabrizio Martelli
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
19
|
Zhang P, Tey SK. Adoptive T Cell Therapy Following Haploidentical Hematopoietic Stem Cell Transplantation. Front Immunol 2019; 10:1854. [PMID: 31447852 PMCID: PMC6691120 DOI: 10.3389/fimmu.2019.01854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/23/2019] [Indexed: 12/24/2022] Open
Abstract
Delayed immune reconstitution and the consequently high rates of leukemia relapse and infectious complications are the main limitations of haploidentical hematopoietic stem cell transplantation. Donor T cell addback can accelerate immune reconstitution but the therapeutic window between graft-vs.-host disease and protective immunity is very narrow in the haploidentical transplant setting. Hence, strategies to improve the safety and efficacy of adoptive T cell transfer are particularly relevant in this setting. Adoptive T cell transfer strategies in haploidentical transplantation include the use of antigen-specific T cells, allodepletion and alloanergy induction, immune modulation by the co-infusion of regulatory cell populations, and the use of safety switch gene-modified T cells. Whilst common principles apply, there are features that are unique to haploidentical transplantation, where HLA-mismatching directly impacts on immune reconstitution, and shared vs. non-shared HLA-allele can be an important consideration in antigen-specific T cell therapy. This review will also present an update on safety switch gene-modified T cells, which can be conditionally deleted in the event of severe graft- vs.-host disease or other adverse events. Herpes Virus Simplex Thymidine Kinase (HSVtk) and inducible caspase-9 (iCasp9) are safety switches that have undergone multicenter studies in haploidentical transplantation with encouraging results. These gene-modified cells, which are trackable long-term, have also provided important insights on the fate of adoptively transferred T cells. In this review, we will discuss the biology of post-transplant T cell immune reconstitution and the impact of HLA-mismatching, and the different cellular therapy strategies that can help accelerate T cell immune reconstitution after haploidentical transplantation.
Collapse
Affiliation(s)
- Ping Zhang
- Clinical Translational Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Siok-Keen Tey
- Clinical Translational Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia.,Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
20
|
Effect of plasmapheresis on ATG (Thymoglobulin) clearance prior to adoptive T cell transfer. Bone Marrow Transplant 2019; 54:2110-2116. [PMID: 30890771 DOI: 10.1038/s41409-019-0505-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/06/2019] [Accepted: 03/02/2019] [Indexed: 01/01/2023]
Abstract
The effect of anti-thymocyte globulin (ATG) on the outcome of hematopoietic stem cell transplantation (SCT) is dependent on formulation, dose and exposure. However, ATG levels are not routinely measured and therapeutic levels are not well defined. In ex vivo T cell-deplete SCT, the potential effect of residual ATG has important implications on the timing of adoptive T cell transfer. Here we measured active rabbit ATG concentration using a flow cytometry-based method that can be implemented in any laboratory. Three adult patients received 6 mg/kg Thymoglobulin over 4 days, leading to peak plasma active ATG concentration of 20.8 ± 1.4 µg/mL, suggesting volume of distribution of 16-19 L. The half-life of active ATG was 6.1 ± 0.7 days and plasmapheresis at Day 25 ± 1 post-transplant reduced mean plasma concentration from 1.25 to 0.61 µg/mL. Total ATG and active ATG do not have a constant relationship because of differences in volumes of distribution and half-lives. Thymoglobulin can mediate antibody-dependent cell-mediated cytotoxicity (ADCC) in vitro at concentrations as low as 0.03 µg/mL, with a log-linear relationship between ATG concentration and ADCC. Plasmapheresis can remove ATG but likely has modest biological impact when performed 4 weeks after 6 mg/kg ATG.
Collapse
|
21
|
Oostenbrink LVE, Jol-van der Zijde CM, Kielsen K, Jansen-Hoogendijk AM, Ifversen M, Müller KG, Lankester AC, van Halteren AGS, Bredius RGM, Schilham MW, van Tol MJD. Differential Elimination of Anti-Thymocyte Globulin of Fresenius and Genzyme Impacts T-Cell Reconstitution After Hematopoietic Stem Cell Transplantation. Front Immunol 2019; 10:315. [PMID: 30894854 PMCID: PMC6414431 DOI: 10.3389/fimmu.2019.00315] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/06/2019] [Indexed: 01/11/2023] Open
Abstract
Anti-thymocyte globulin (ATG) is a lymphocyte depleting agent applied in hematopoietic stem cell transplantation (HSCT) to prevent rejection and Graft-vs.-Host Disease (GvHD). In this study, we compared two rabbit ATG products, ATG-Genzyme (ATG-GENZ), and ATG-Fresenius (ATG-FRES), with respect to dosing, clearance of the active lymphocyte binding component, post-HSCT immune reconstitution and clinical outcome. Fifty-eigth pediatric acute leukemia patients (n = 42 ATG-GENZ, n = 16 ATG-FRES), who received a non-depleted bone marrow or peripheral blood stem cell graft from an unrelated donor were included. ATG-GENZ was given at a dosage of 6-10 mg/kg; ATG-FRES at 45-60 mg/kg. The active component of ATG from both products was cleared at different rates. Within the ATG-FRES dose range no differences were found in clearance of active ATG or T-cell re-appearance. However, the high dosage of ATG-GENZ (10 mg/kg), in contrast to the low dosage (6-8 mg/kg), correlated with prolonged persistence of active ATG and delayed T-cell reconstitution. Occurrence of serious acute GvHD (grade III-IV) was highest in the ATG-GENZ-low dosage group. These results imply that dosing of ATG-GENZ is more critical than dosing of ATG-FRES due to the difference in clearance of active ATG. This should be taken into account when designing clinical protocols.
Collapse
Affiliation(s)
| | | | - Katrine Kielsen
- Institute for Inflammation Research, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Marianne Ifversen
- Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Klaus G Müller
- Institute for Inflammation Research, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Department of Paediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Arjan C Lankester
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Robbert G M Bredius
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Marco W Schilham
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Maarten J D van Tol
- Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
22
|
Kako S, Gomyo A, Akahoshi Y, Harada N, Kameda K, Ugai T, Wada H, Ishihara Y, Kawamura K, Sakamoto K, Sato M, Terasako-Saito K, Kimura SI, Kikuchi M, Nakasone H, Kanda J, Kanda Y. Haploidentical transplantation using low-dose alemtuzumab: Comparison with haploidentical transplantation using low-dose thymoglobulin. Eur J Haematol 2019; 102:256-264. [PMID: 30578673 DOI: 10.1111/ejh.13204] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To establish the optimal strategy for haploidentical hematopoietic stem cell transplantation (HSCT). METHODS We performed a prospective study on haploidentical HSCT using low-dose alemtuzumab. Alemtuzumab was added at 0.25 mg/kg for 2 days. The primary outcome measure was the survival rate with the engraftment of donor cells and without grade III-IV acute graft-vs-host disease (GVHD) at 60 days after transplantation. RESULTS Fourteen adult patients with advanced hematological disease were enrolled. The primary outcome measure was achieved in 86% of the patients. Six patients experienced relapse/progression. Non-relapse death was observed in three patients, and all of them had a history of previous allogeneic HSCT. Overall survival and progression-free survival rates at 1 year were 51% and 43%, respectively. Four patients were suspected to have herpes simplex virus infection and three had aseptic meningitis under the use of acyclovir at 200 mg. There were no deaths due to viral infection. Compared to those who underwent haploidentical HSCT using thymoglobulin, patients with alemtuzumab showed a slower recovery of CD8+ T-cells and lower incidences of GVHD and EB virus reactivation. CONCLUSIONS Haploidentical HSCT using low-dose alemtuzumab can be performed safely. We need to overcome the high relapse/progression rate in non-remission patients.
Collapse
Affiliation(s)
- Shinichi Kako
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Ayumi Gomyo
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yu Akahoshi
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Naonori Harada
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Kazuaki Kameda
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Tomotaka Ugai
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Hidenori Wada
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yuko Ishihara
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Koji Kawamura
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Kana Sakamoto
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Miki Sato
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Kiriko Terasako-Saito
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Shun-Ichi Kimura
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Misato Kikuchi
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Hideki Nakasone
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Junya Kanda
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| |
Collapse
|
23
|
Admiraal R, Boelens JJ. Pharmacotherapy in Pediatric Hematopoietic Cell Transplantation. Handb Exp Pharmacol 2019; 261:471-489. [PMID: 31375921 DOI: 10.1007/164_2019_247] [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] [Indexed: 12/23/2022]
Abstract
Hematopoietic cell transplantation (HCT) is a curative treatment option for both malignant and nonmalignant diseases. Success of the procedure mainly depends on disease control and treatment-related complications. Pharmacotherapy plays a major role in HCT and significantly impacts the outcomes. Main drug use within HCT includes conditioning, GvHD prophylaxis, and prevention/treatment of infections.Increasing evidence suggests individualized dosing in (pediatric) HCT may improve outcome. Dose individualization may result in a better predictable drug treatment in terms of safety and efficacy, including timely immune reconstitution after HCT and optimal tumor or disease control, which may result in improved survival chances.
Collapse
Affiliation(s)
- R Admiraal
- Blood and Marrow Transplantation Program, Prinses Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J J Boelens
- Stem Cell Transplantation and Cellular Therapies Pediatrics, New York, NY, USA. .,Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
24
|
Jamani K, Dabas R, Kangarloo SB, Prokopishyn NL, Luider J, Dharmani-Khan P, Khan FM, Daly A, Storek J. Rabbit Antithymocyte Globulin Serum Levels: Factors Impacting the Levels and Clinical Outcomes Impacted by the Levels. Biol Blood Marrow Transplant 2018; 25:639-647. [PMID: 30572108 DOI: 10.1016/j.bbmt.2018.12.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
Abstract
Antithymocyte globulin (ATG) levels and clearance vary significantly among patients receiving the same weight-based dose of ATG. To date, ATG area under the curve (AUC), its determinants, and its impact on clinical outcomes have been examined in pediatric hematopoietic cell transplant (HCT) and adult nonmyeloablative HCT. Here we set out to examine ATG AUC in 219 uniformly treated adults undergoing myeloablative allogeneic HCT at our institution. Sera were collected for the determination of pre- or post-HCT ATG AUC. The lowest quintiles of pre- and post-HCT AUC were associated with inferior chronic graft-versus-host disease (GVHD) and relapse-free survival (cGRFS) and a higher risk of acute GVHD, respectively. The highest pre- or post-HCT ATG AUC quintiles were not associated with risk of death, nonrelapse mortality, or relapse. Factors most strongly associated with AUC were day -2 recipient absolute lymphocyte count, body mass index (BMI), and graft lymphocyte content. To achieve ideal pre-HCT AUC (avoiding low AUC to maximize cGRFS) in this HCT setting, ATG dosing will need to take into consideration recipient weight, BMI, and blood and graft lymphocyte counts. Further studies are required to develop a modern ATG dosing schema and to demonstrate that adjusting ATG dose to target a particular AUC is feasible and leads to improved outcomes.
Collapse
Affiliation(s)
- Kareem Jamani
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Rosy Dabas
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Shahbal B Kangarloo
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Nicole L Prokopishyn
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Joanne Luider
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Poonam Dharmani-Khan
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Faisal M Khan
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Andrew Daly
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| | - Jan Storek
- Alberta Blood and Marrow Transplant Program, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
25
|
Kumar A, Reljic T, Hamadani M, Mohty M, Kharfan-Dabaja MA. Antithymocyte globulin for graft-versus-host disease prophylaxis: an updated systematic review and meta-analysis. Bone Marrow Transplant 2018; 54:1094-1106. [PMID: 30446739 DOI: 10.1038/s41409-018-0393-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 12/11/2022]
Abstract
Graft-versus-host disease (GVHD) remains a limiting factor for successful allogeneic hematopoietic cell transplantation (allo-HCT). Conflicting data exist on the benefit of ATG on post-transplant survival. We performed a systematic review of randomized controlled trials (RCTs) to assess benefits and harms of thymoglobulin and Fresenius (re-branded as Grafalon) ATG formulations in patients undergoing allo-HCT for a variety of hematologic malignancies and bone marrow failure syndromes. A comprehensive search of MEDLINE, EMBASE, and Cochrane Library was performed. Data on methodological quality, benefits, and harms were extracted for each trial and pooled under a random-effects model. Eight RCTs (1134 patients) met the inclusion criteria. Methodological quality ranged from moderate to very low. Pooled results showed no difference in overall survival (OS) with the use of ATG (hazard ratio (HR) = 0.97; 95% confidence interval (CI) = 0.74-1.28; P = 0.83). ATG reduced grade II/III acute GVHD (risk ratio (RR) = 0.61; 95% CI = 0.48-0.77; P < 0.0001), grade III/IV acute GVHD (RR = 0.52; 95% CI = 0.34-0.81; P = 0.004), and chronic GVHD (RR = 0.52; 95% CI = 0.40-0.69; P < 0.00001) without an increase in non-relapse mortality (NRM) (RR = 0.91; 95% CI = 0.74-1.13; P = 0.40). Future studies with better methodological quality are needed to provide conclusive answers related to optimal dosing and timing of ATG for prevention of GVHD.
Collapse
Affiliation(s)
- Ambuj Kumar
- Program for Comparative Effectiveness Research, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| | - Tea Reljic
- Program for Comparative Effectiveness Research, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Mehdi Hamadani
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mohamad Mohty
- Department of Haematology, Saint Antoine Hospital, University Pierre & Marie Curie and Inserm UMRs938, Paris, France
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology, Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
26
|
Ducloux D, Bamoulid J, Daguindau E, Rebibou JM, Courivaud C, Saas P. Antithymocytes globulins: Time to revisit its use in kidney transplantation? Int Rev Immunol 2018; 37:183-191. [DOI: 10.1080/08830185.2018.1455194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- D. Ducloux
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, Interactions hôte-greffon-tumeur, Federation hospitalo-universitaire INCREASE, LabEX LipSTIC, Besançon, France
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - J. Bamoulid
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, Interactions hôte-greffon-tumeur, Federation hospitalo-universitaire INCREASE, LabEX LipSTIC, Besançon, France
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - E. Daguindau
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, Interactions hôte-greffon-tumeur, Federation hospitalo-universitaire INCREASE, LabEX LipSTIC, Besançon, France
- Department of Hematology, CHU Besançon, Besançon, France
| | - J. M. Rebibou
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, Interactions hôte-greffon-tumeur, Federation hospitalo-universitaire INCREASE, LabEX LipSTIC, Besançon, France
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Dijon, Dijon, France
| | - C. Courivaud
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, Interactions hôte-greffon-tumeur, Federation hospitalo-universitaire INCREASE, LabEX LipSTIC, Besançon, France
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - P. Saas
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, Interactions hôte-greffon-tumeur, Federation hospitalo-universitaire INCREASE, LabEX LipSTIC, Besançon, France
- INSERM, CHU Besançon, Besançon, France
| |
Collapse
|
27
|
Fuchs E. Haploidentical Hematopoietic Cell Transplantation. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00106-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
28
|
Tandra A, Covut F, Cooper B, Creger R, Brister L, McQuigg B, Caimi P, Malek E, Tomlinson B, Lazarus HM, Otegbeye F, Kolk M, de Lima M, Metheny L. Low dose anti-thymocyte globulin reduces chronic graft-versus-host disease incidence rates after matched unrelated donor transplantation. Leuk Lymphoma 2017; 59:1644-1651. [PMID: 29199482 DOI: 10.1080/10428194.2017.1390234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Anti-thymocyte globulin (ATG) is often added to hematopoietic stem cell transplant conditioning regimens to prevent graft rejection and reduce graft-versus-host disease (GVHD). Doses used in retrospective and prospective clinical trials have ranged from 2.5 to 20 mg/kg with rates of grade II-IV acute GVHD and chronic GVHD up to 40 and 60%, respectively. We retrospectively compared outcomes in recipients of matched unrelated donor (MUD) grafts given low dose rabbit ATG IV 3 mg/kg (n = 52) versus recipients of matched related donor (MRD) grafts (n = 48) without ATG. One year cumulative incidence of chronic GVHD was 25.2% in the MUD group versus 33.3% in the MRD group (p = .5). One-year cumulative incidence of extensive chronic GVHD was 9.6% in the MUD group versus 26.6% in the MRD group (p = .042). Our analysis supports the use of low dose ATG in MUD transplantation as an effective therapy to prevent chronic GVHD.
Collapse
Affiliation(s)
- Anand Tandra
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Fahrettin Covut
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Brenda Cooper
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Richard Creger
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Lauren Brister
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Bernadette McQuigg
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Paolo Caimi
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Ehsan Malek
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Ben Tomlinson
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Hillard M Lazarus
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Folashade Otegbeye
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Merle Kolk
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Marcos de Lima
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| | - Leland Metheny
- a Stem Cell Transplant Program , University Hospitals Cleveland Medical Center and Case Western Reserve University , Cleveland , OH , USA
| |
Collapse
|
29
|
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: 2.3] [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
|
30
|
The Role of Low-dose Anti-thymocyte Globulin as Standard Prophylaxis in Mismatched and Matched Unrelated Hematopoietic Peripheral Stem Cell Transplantation for Hematologic Malignancies. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:658-666. [DOI: 10.1016/j.clml.2017.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/04/2017] [Accepted: 06/08/2017] [Indexed: 01/22/2023]
|
31
|
Busca A, Aversa F. In-vivo or ex-vivo T cell depletion or both to prevent graft-versus-host disease after hematopoietic stem cell transplantation. Expert Opin Biol Ther 2017; 17:1401-1415. [PMID: 28846051 DOI: 10.1080/14712598.2017.1369949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Hematopoietic stem cell transplantation (HSCT) represents a widely accepted therapeutic strategy for the treatment of hematologic disorders which are otherwise considered incurable. Alloreactive T cells infused with the stem cell inoculum may generate graft-versus-host disease (GVHD) representing one the most relevant obstacles to the successful outcome of patients receiving allogeneic HSCT. Areas covered: In this review, the authors provide an overview of the most recent approaches of T-cell depletion (TCD) including ex-vivo αβ+ TCD and in-vivo TCD with anti-thymocyte globulin (ATG). Expert opinion: Ex vivo depletion of donor T-cells prevents both acute and chronic GVHD without the need for any additional posttransplant immunological prophylaxis either in haploidentical HSCT and HLA matched transplants. Three prospective trials evaluating the efficacy of ATG in matched unrelated donor transplant recipients demonstrated that ATG reduces the incidence of both acute and chronic GVHD without a significant increase of relapse rate, and similar results have been reported in the setting of blood stem cell grafts from matched sibling donors.
Collapse
Affiliation(s)
- Alessandro Busca
- a SSD Trapianto di Cellule Staminali , AOU Citta' della Salute e della Scienza , Torino , Italy
| | - Franco Aversa
- b Hematology and BMT Unit , University of Parma , Parma , Italy
| |
Collapse
|
32
|
A New Enzyme-Linked Immunosorbent Assay for a Total Anti-T Lymphocyte Globulin Determination: Development, Analytical Validation, and Clinical Applications. Ther Drug Monit 2017; 39:282-289. [PMID: 28399040 DOI: 10.1097/ftd.0000000000000407] [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 Anti-T lymphocyte globulin (ATLG) modulates the alloreactivity of T lymphocytes, reducing the risk of immunological posttransplant complications, in particular rejection and graft-versus-host disease, after allogeneic hematopoietic stem cell transplantation (HSCT). We developed and validated a new enzyme-linked immunosorbent assay (ELISA) method to measure serum levels of total ATLG and evaluate the pharmacokinetics (PK) of the drug in children with β-Thalassemia, receiving allogeneic HSCT. METHODS Diluted serum samples were incubated with Goat-anti-Rabbit IgG antibody coated on a microtiter plate and then, with Goat-anti-Human IgG labeled with horseradish peroxidase. After incubation and washings, substrate solution was added and absorbance was read at 492 nm. ATLG concentrations in samples were determined by interpolation from a standard curve (range: 200-0.095 ng/mL), prepared by diluting a known amount of ATLG in phosphate-buffered saline (PBS). Low, medium, and high-quality control concentrations were 1.56, 6.25, and 25 ng/mL, respectively. This method was developed and validated within the acceptance criteria in compliance with the Guidelines for a biological method validation: the sensitivity of the method was 0.095 ng/mL. We analyzed serum samples from 14 children with β-Thalassemia who received ATLG (Grafalon) at a dose of 10 mg/kg administered as intravenous (IV) infusion on days -5, -4, and -3 before HSCT (day 0). Blood sampling for PK evaluation was performed on days -5, -4, and -3 before and after drug infusion; and then from day -2 to +56. RESULTS The median total ATLG levels pre-IVand post-IV were 0 and 118 mcg/mL on day -5; 85.9 and 199.2 mcg/mL on day -4; 153 and 270.9 mcg/mL on day -3, respectively. The median PK values of CL was 0.0029 (range: 0.0028-0.0057) L·kg·d, Vd was 0.088 (range: 0.025-0.448) L/kg and t1/2 was 20.2 (range: 5.8-50.2) days. CONCLUSIONS These data suggest that given the marked interindividual variability of total ATLG disposition, the development of a validated ELISA method and the possibility to measure PK parameters in paediatric populations are essential steps to optimize drug therapeutic regimens.
Collapse
|
33
|
Admiraal R, Nierkens S, de Witte MA, Petersen EJ, Fleurke GJ, Verrest L, Belitser SV, Bredius RGM, Raymakers RAP, Knibbe CAJ, Minnema MC, van Kesteren C, Kuball J, Boelens JJ. Association between anti-thymocyte globulin exposure and survival outcomes in adult unrelated haemopoietic cell transplantation: a multicentre, retrospective, pharmacodynamic cohort analysis. LANCET HAEMATOLOGY 2017; 4:e183-e191. [PMID: 28330607 DOI: 10.1016/s2352-3026(17)30029-7] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/30/2017] [Accepted: 02/06/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND Anti-thymocyte globulin (ATG) is used to prevent graft-versus-host disease (GvHD) after allogeneic haemopoietic cell transplantation (HCT). However, ATG can also cause delayed immune reconstitution of T cells, negatively affecting survival. We studied the relation between exposure to ATG and clinical outcomes in adult patients with acute leukaemia and myelodysplastic syndrome. METHODS We did a retrospective, pharmacokinetic-pharmacodynamic analysis of data from patients with acute lymphoid leukaemia, acute myeloid leukaemia, or myelodysplastic syndrome receiving their first T-cell repleted allogeneic peripheral blood stem cell HCT with ATG (thymoglobulin) as part of non-myeloablative conditioning from March 1, 2004, to June 1, 2015. Patients received a cumulative intravenous dose of 8 mg/kg divided over 4 days, starting on day -8 before HCT. Active ATG concentrations were measured using a validated bioassay and pharmacokinetic exposure measures (maximum concentration, concentration at time of infusion of the graft, time to reach a concentration of 1 arbitary unit [AU] per day/mL, area under the curve [AUC], and the AUC before and after HCT) were calculated with a validated population pharmacokinetic model. The main outcome of interest was 5-year overall survival, defined as days to death from any cause or last follow-up. Other outcomes were relapse-related mortality, non-relapse mortality, event-free survival, acute and chronic GvHD, and assessment of current and optimum dosing. We used Cox proportional hazard models and Fine-Gray competing risk models for the analyses. FINDINGS 146 patients were included. ATG exposure after HCT was shown to be the best predictor for 5-year overall survival. Optimum exposure after transplantation was determined to be 60-95 AU per day/mL. Estimated 5-year overall survival in the group who had optimum exposure (69%, 95% CI 55-86) was significantly higher than in the group who had below optimum exposure (32%, 20-51, p=0·00037; hazard ratio [HR] 2·41, 95% CI 1·15-5·06, p=0·020) and above optimum exposure (48%, 37-62, p=0·030; HR 2·11, 95% CI 1·04-4·27, p=0·038). Patients in the optimum exposure group had a greater chance of event-free survival than those in the below optimum exposure group (HR 2·54, 95% CI 1·29-5·00, p=0·007; HR for the above optimum group: 1·83, 0·97-3·47, p=0·063). Above-optimum exposure led to higher relapse-related mortality compared with optimum exposure (HR 2·66, 95% CI 1·12-6·31; p=0·027). Below optimum exposure increased non-relapse mortality compared with optimum exposure (HR 4·36, 95% CI 1·60-11·88; p=0·0040), grade 3-4 acute GvHD (3·09, 1·12-8·53; p=0·029), but not chronic GvHD (2·38, 0·93-6·08; p=0·070). Modelled dosing based on absolute lymphocyte counts led to higher optimum target attainment than did weight-based dosing. INTERPRETATION Exposure to ATG affects survival after HCT in adults, stressing the importance of optimum ATG dosing. Individualised dosing of ATG, based on lymphocyte counts rather than bodyweight, might improve survival chances after HCT. FUNDING Netherlands Organization for Health Research and Development and Queen Wilhelma Fund for Cancer Research.
Collapse
Affiliation(s)
- Rick Admiraal
- Paediatric Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands; Department of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Moniek A de Witte
- Adult Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Eefke J Petersen
- Adult Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Ger-Jan Fleurke
- Adult Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Luka Verrest
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Svetlana V Belitser
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Robbert G M Bredius
- Department of Paediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Reinier A P Raymakers
- Adult Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Catherijne A J Knibbe
- Department of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, Netherlands
| | - Monique C Minnema
- Adult Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Charlotte van Kesteren
- Paediatric Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jurgen Kuball
- Adult Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jaap J Boelens
- Paediatric Blood and Marrow Transplant Program, University Medical Centre Utrecht, Utrecht, Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands.
| |
Collapse
|
34
|
Yeung MY, Gabardi S, Sayegh MH. Use of polyclonal/monoclonal antibody therapies in transplantation. Expert Opin Biol Ther 2017; 17:339-352. [PMID: 28092486 DOI: 10.1080/14712598.2017.1283400] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION For over thirty years, antibody (mAb)-based therapies have been a standard component of transplant immunosuppression, and yet much remains to be learned in order for us to truly harness their therapeutic capabilities. Current mAbs used in transplant directly target and destroy graft-destructive immune cells, interrupt cytokine and costimulation-dependent T and B cell activation, and prevent down-stream complement activation. Areas covered: This review summarizes our current approaches to using antibody-based therapies to prevent and treat allograft rejection. It also provides examples of promising novel mAb therapies, and discusses the potential for future mAb development in transplantation. Expert opinion: The broad capability of antibodies, in parallel with our growing ability to synthetically modulate them, offers exciting opportunities to develop better biologic therapeutics. In order to do so, we must further our understanding about the basic biology underlying allograft rejection, and gain better appreciation of how characteristics of therapeutic antibodies affect their efficacy.
Collapse
Affiliation(s)
- Melissa Y Yeung
- a Transplantation Research Center, Renal Division , Brigham and Women's Hospital, Harvard Medical School , Boston , Massachusetts , United States
| | - Steven Gabardi
- a Transplantation Research Center, Renal Division , Brigham and Women's Hospital, Harvard Medical School , Boston , Massachusetts , United States
| | - Mohamed H Sayegh
- a Transplantation Research Center, Renal Division , Brigham and Women's Hospital, Harvard Medical School , Boston , Massachusetts , United States.,b Faculty of Medicine, Professor of Medicine and Immunology , American University of Beirut , Beirut , Lebanon
| |
Collapse
|
35
|
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: 2.4] [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
|
36
|
Baron F, Mohty M, Blaise D, Socié G, Labopin M, Esteve J, Ciceri F, Giebel S, Gorin NC, Savani BN, Schmid C, Nagler A. Anti-thymocyte globulin as graft-versus-host disease prevention in the setting of allogeneic peripheral blood stem cell transplantation: a review from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Haematologica 2016; 102:224-234. [PMID: 27927772 DOI: 10.3324/haematol.2016.148510] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/24/2016] [Indexed: 11/09/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is increasingly used as treatment for patients with life-threatening blood diseases. Its curative potential is largely based on immune-mediated graft-versus-leukemia effects caused by donor T cells contained in the graft. Unfortunately, donor T cells are also the cause of graft-versus-host disease. The vast majority of human leukocyte antigen-matched allogeneic hematopoietic stem cell transplants are nowadays carried out with peripheral blood stem cells as the stem cell source. In comparison with bone marrows, peripheral blood stem cells contain more hematopoietic stem/progenitor cells but also one log more T cells. Consequently, the use of peripheral blood stem cells instead of bone marrow has been associated with faster hematologic recovery and a lower risk of relapse in patients with advanced disease, but also with a higher incidence of chronic graft-versus-host disease. These observations have been the basis for several studies aimed at assessing the impact of immunoregulation with anti-thymocyte globulin on transplantation outcomes in patients given human leukocyte antigen-matched peripheral blood stem cells from related or unrelated donors. After a brief introduction on anti-thymocyte globulin, this article reviews recent studies assessing the impact of anti-thymocyte globulin on transplantation outcomes in patients given peripheral blood stem cells from human leukocyte antigen-matched related or unrelated donors as well as in recipients of grafts from human leukocyte antigen haploidentical donors.
Collapse
Affiliation(s)
| | - Mohamad Mohty
- Hopital Saint-Antoine, AP-HP, Paris, France.,Université Pierre & Marie Curie, Paris, France.,INSERM UMRs U938, Paris, France
| | - Didier Blaise
- Aix Marseille Univ, CNRS, INSERM, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Gérard Socié
- AP-HP, Hematology Transplantation, Hospital Saint-Louis, Paris, France
| | - Myriam Labopin
- Hopital Saint-Antoine, AP-HP, Paris, France.,INSERM UMRs U938, Paris, France
| | - Jordi Esteve
- Department of Hematology, Hospital Clinic, Barcelona, Spain
| | - Fabio Ciceri
- Department of Hematology, Ospedale San Raffaele, Università degli Studi, Milano, Italy
| | - Sebastian Giebel
- Maria Sklodowska-Curie Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | | | - Bipin N Savani
- Long term Transplant Clinic, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christoph Schmid
- Klinikum Augsburg, Department of Hematology and Oncology, University of Munich, Augsburg, Germany
| | - Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel.,EBMT Paris Office, Hospital Saint Antoine, Paris, France
| |
Collapse
|
37
|
Murata M, Ikegame K, Morishita Y, Ogawa H, Kaida K, Nakamae H, Ikeda T, Nishida T, Inoue M, Eto T, Kubo K, Sakura T, Mori T, Uchida N, Ashida T, Matsuhashi Y, Miyazaki Y, Ichinohe T, Atsuta Y, Teshima T. Low-dose thymoglobulin as second-line treatment for steroid-resistant acute GvHD: an analysis of the JSHCT. Bone Marrow Transplant 2016; 52:252-257. [PMID: 27869808 DOI: 10.1038/bmt.2016.247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/15/2016] [Accepted: 08/11/2016] [Indexed: 01/08/2023]
Abstract
A nationwide retrospective study for the clinical outcomes of 99 patients who had received thymoglobulin at a median total dose of 2.5 mg/kg (range, 0.5-18.5 mg/kg) as a second-line treatment for steroid-resistant acute GvHD was conducted. Of the 92 evaluable patients, improvement (complete or partial response) was observed in 55 patients (60%). Multivariate analysis demonstrated that male sex and grade III and IV acute GvHD were associated with a lower improvement rate, whereas thymoglobulin dose (<2.0, 2.0-3.9 and ⩾4.0 mg/kg) was NS. Factors associated with significantly higher nonrelapse mortality included higher patient age (⩾50 years), grade IV acute GvHD, no improvement of GvHD and higher dose of thymoglobulin (hazard ratio, 2.55; 95% confidence interval, 1.34-4.85; P=0.004 for 2.0-3.9 mg/kg group and 1.79; 0.91-3.55; P=0.093 for ⩾4.0 mg/kg group). Higher dose of thymoglobulin was associated with a higher incidence of bacterial infections, CMV antigenemia and any additional infection. Taken together, low-dose thymoglobulin at a median total dose of 2.5 mg/kg provides a comparable response rate to standard-dose thymoglobulin reported previously, and <2.0 mg/kg thymoglobulin is recommended in terms of the balance between efficacy and adverse effects.
Collapse
Affiliation(s)
- M Murata
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Ikegame
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Y Morishita
- Department of Internal Medicine, Holy Spirit Hospital, Nagoya, Japan
| | - H Ogawa
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - K Kaida
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - H Nakamae
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - T Ikeda
- Division of Hematology and Stem Cell Transplantation, Shizuoka Cancer Center, Shizuoka, Japan
| | - T Nishida
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - M Inoue
- Department of Hematology/Oncology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - T Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - K Kubo
- Department of Hematology, Aomori Prefectural Central Hospital, Aomori, Japan
| | - T Sakura
- Leukemia Research Center, Saiseikai Maebashi Hospital, Maebashi, Japan
| | - T Mori
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - N Uchida
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - T Ashida
- Division of Hematology and Rheumatology, Department of Internal Medicine, Kinki University, School of Medicine, Osakasayama, Japan
| | - Y Matsuhashi
- Department of Hematology, Kawasaki Medical School, Kurashiki, Japan
| | - Y Miyazaki
- Department of Hematology, Oita Prefectural Hospital, Oita, Japan
| | - T Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Y Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan.,Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - T Teshima
- Department of Hematology, Hokkaido University Graduate School of Medical Science, Sapporo, Japan
| |
Collapse
|
38
|
Immunity to Infections after Haploidentical Hematopoietic Stem Cell Transplantation. Mediterr J Hematol Infect Dis 2016; 8:e2016057. [PMID: 27872737 PMCID: PMC5111540 DOI: 10.4084/mjhid.2016.057] [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: 08/29/2016] [Accepted: 09/21/2016] [Indexed: 02/06/2023] Open
Abstract
The advantage of using a Human Leukocyte Antigen (HLA)-mismatched related donor is that almost every patient who does not have an HLA-identical donor or who urgently needs hematopoietic stem cell transplantation (HSCT) has at least one family member with whom shares one haplotype (haploidentical) and who is promptly available as a donor. The major challenge of haplo-HSCT is intense bi-directional alloreactivity leading to high incidences of graft rejection and graft-versus-host disease (GVHD). Advances in graft processing and pharmacologic prophylaxis of GVHD have reduced these risks and have made haplo-HSCT a viable alternative for patients lacking a matched donor. Indeed, the haplo-HSCT has spread to centers worldwide even though some centers have preferred an approach based on T cell depletion of G-CSF-mobilized peripheral blood progenitor cells (PBPCs), others have focused on new strategies for GvHD prevention, such as G-CSF priming of bone marrow and robust post-transplant immune suppression or post-transplant cyclophosphamide (PTCY). Today, the graft can be a megadose of T-cell depleted PBPCs or a standard dose of unmanipulated bone marrow and/or PBPCs. Although haplo-HSCT modalities are based mainly on high intensity conditioning regimens, recently introduced reduced intensity regimens (RIC) showed promise in decreasing early transplant-related mortality (TRM), and extending the opportunity of HSCT to an elderly population with more comorbidities. Infections are still mostly responsible for toxicity and non-relapse mortality due to prolonged immunosuppression related, or not, to GVHD. Future challenges lie in determining the safest preparative conditioning regimen, minimizing GvHD and promoting rapid and more robust immune reconstitution.
Collapse
|
39
|
Excellent T-cell reconstitution and survival depend on low ATG exposure after pediatric cord blood transplantation. Blood 2016; 128:2734-2741. [PMID: 27702800 DOI: 10.1182/blood-2016-06-721936] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/21/2016] [Indexed: 12/18/2022] Open
Abstract
Successful immune reconstitution (IR) is associated with improved outcomes following pediatric cord blood transplantation (CBT). Usage and timing of anti-thymocyte globulin (ATG), introduced to the conditioning to prevent graft-versus-host disease and graft failure, negatively influences T-cell IR. We studied the relationships among ATG exposure, IR, and clinical outcomes. All pediatric patients receiving a first CBT between 2004 and 2015 at the University Medical Center Utrecht were included. ATG-exposure measures were determined with a validated pharmacokinetics model. Main outcome of interest was early CD4+ IR, defined as CD4+ T-cell counts >50 × 106/L twice within 100 days after CBT. Other outcomes of interest included event-free survival (EFS). Cox proportional-hazard and Fine-Gray competing-risk models were used. A total of 137 patients, with a median age of 7.4 years (range, 0.2-22.7), were included, of whom 82% received ATG. Area under the curve (AUC) of ATG after infusion of the cord blood transplant predicted successful CD4+ IR. Adjusted probability on CD4+ IR was reduced by 26% for every 10-point increase in AUC after CBT (hazard ratio [HR], 0.974; P < .0001). The chance of EFS was higher in patients with successful CD4+ IR (HR, 0.26; P < .0001) and lower ATG exposure after CBT (HR, 1.005; P = .0071). This study stresses the importance of early CD4+ IR after CBT, which can be achieved by reducing the exposure to ATG after CBT. Individualized dosing of ATG to reach optimal exposure or, in selected patients, omission of ATG may contribute to improved outcomes in pediatric CBT.
Collapse
|
40
|
Antithymocyte Globulin at Clinically Relevant Concentrations Kills Leukemic Blasts. Biol Blood Marrow Transplant 2016; 22:815-24. [DOI: 10.1016/j.bbmt.2016.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/04/2016] [Indexed: 01/03/2023]
|
41
|
Nishihori T, Al-Kadhimi Z, Hamadani M, Kharfan-Dabaja MA. Antithymocyte globulin in allogeneic hematopoietic cell transplantation: benefits and limitations. Immunotherapy 2016; 8:435-47. [DOI: 10.2217/imt.15.128] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Significant advances have been made in allogeneic hematopoietic cell transplantation by reducing toxicities and optimizing its efficacy. Antithymocyte globulin (ATG) is an important in vivo T-cell depletion strategy, which reduces the risk of graft-versus-host disease in HLA-matched or -mismatched donor allografting. ATG effectively targets alloreactive T cells at the expense of potentially increasing the risk of post-hematopoietic cell transplantation infections and delayed immune reconstitution. We summarize the targets, mechanisms, various preparations of ATG, the growing role of ATG in prevention of graft-versus-host disease in various transplant modalities as well as emerging data on pharmacokinetic modeling for individualized ATG dosing. Further research is needed to optimize the ATG administration while minimizing the toxicities.
Collapse
Affiliation(s)
- Taiga Nishihori
- Department of Blood & Marrow Transplantation, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, FOB-3, Tampa, FL 33612, USA
- Department of Oncologic Sciences, Morsani College of Medicine, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| | | | | | - Mohamed A Kharfan-Dabaja
- Department of Blood & Marrow Transplantation, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, FOB-3, Tampa, FL 33612, USA
- Department of Oncologic Sciences, Morsani College of Medicine, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| |
Collapse
|
42
|
Population pharmacokinetic modeling of Thymoglobulin(®) in children receiving allogeneic-hematopoietic cell transplantation: towards improved survival through individualized dosing. Clin Pharmacokinet 2015; 54:435-46. [PMID: 25466602 DOI: 10.1007/s40262-014-0214-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND AND OBJECTIVES To prevent graft-versus-host disease and rejection in hematopoietic cell transplantation (HCT), children receive Thymoglobulin(®), a polyclonal antibody acting mainly by depleting T cells. The therapeutic window is critical as over-exposure may result in delayed immune reconstitution of donor T cells. In this study, we describe the population pharmacokinetics of Thymoglobulin(®) as a first step towards an evidence-based dosing regimen of Thymoglobulin(®) in pediatric HCT. METHODS Serum active Thymoglobulin(®) concentrations were measured in all pediatric HCTs performed between 2004 and 2012 in two pediatric HCT centers in The Netherlands. Population pharmacokinetic analysis was performed using NONMEM(®) version 7.2. RESULTS A total of 3,113 concentration samples from 280 pediatric HCTs were analyzed, with age ranging from 3 months to 23 years old. The cumulative Thymoglobulin(®) dose was 10 mg/kg in 94 % of the patients given in 4 consecutive days. A model incorporating parallel linear and concentration-dependent clearance of Thymoglobulin(®) was identified. Body weight [for linear clearance (CL) and central volume of distribution] as well as lymphocyte count pre-Thymoglobulin(®) infusion (for CL) were important covariates. As such, the current dosing regimen results in higher exposure in children with a higher bodyweight and/or a lower lymphocyte count pre-Thymoglobulin(®) infusion. CONCLUSION This model can be used to develop an individual dosing regimen for Thymoglobulin(®), based on both body weight and lymphocyte counts, once the therapeutic window has been determined. This individualized regimen may contribute to a better immune reconstitution and thus outcome of allogeneic HCT.
Collapse
|
43
|
Huang W, Yu L, Cao T, Li Y, Liu Z, Li H, Bo J, Zhao Y, Jing Y, Wang S, Zhu H, Dou L, Wang Q, Gao C. The efficacy and safety of rabbit anti-thymocyte globulin vs rabbit anti-T-lymphocyte globulin in peripheral blood stem cell transplantation from unrelated donors. Leuk Lymphoma 2015; 57:355-363. [PMID: 26118935 DOI: 10.3109/10428194.2015.1045901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The comparative efficacy and safety of antithymocyte globulin (ATG) at fixed doses in patients undergoing allogeneic peripheral blood stem cell transplantation from unrelated donors (UR-PBSCT) has not been evaluated. In this study, the records of 56 patients and 54 patients who received pre-transplant ATG-Thymoglobulin (ATG-T) at a total dose of 10 mg/kg and ATG- Fresenius (ATG-F) at a total dose of 20 mg/kg, respectively, were retrospectively analyzed. ATG-F patients had a significantly lower probability of developing chronic graft-vs-host disease (cGVHD) than those treated with ATG-T (p = 0.04). ATG-F was associated with a non-significant trend towards lower relapse rates and higher survival at 3- and 5-years of follow-up compared with ATG-T. A significantly greater proportion of ATG-T patients experienced chills and high fever than ATG-F patients (p < 0.01). The current findings suggest that ATG-F may more effectively and safely prevent cGVHD without increasing relapse rates in patients undergoing UR-PBSCT.
Collapse
Affiliation(s)
- Wenrong Huang
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China.,b Department of Hematology , Hainan Branch of Chinese PLA General Hospital , Hainan province , PR China
| | - Li Yu
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Tingting Cao
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Yanfen Li
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Zhanxiang Liu
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Honghua Li
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Jian Bo
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Yu Zhao
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Yu Jing
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Shuhong Wang
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Haiyan Zhu
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Liping Dou
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Qunshun Wang
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| | - Chunji Gao
- a Department of Hematology and BMT , Chinese PLA General Hospital , Beijing , PR China
| |
Collapse
|
44
|
Storek J, Mohty M, Boelens JJ. Rabbit Anti–T Cell Globulin in Allogeneic Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2015; 21:959-70. [DOI: 10.1016/j.bbmt.2014.11.676] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 11/14/2014] [Indexed: 12/27/2022]
|
45
|
Admiraal R, van Kesteren C, Jol-van der Zijde CM, Lankester AC, Bierings MB, Egberts TCG, van Tol MJD, Knibbe CAJ, Bredius RGM, Boelens JJ. Association between anti-thymocyte globulin exposure and CD4+ immune reconstitution in paediatric haemopoietic cell transplantation: a multicentre, retrospective pharmacodynamic cohort analysis. LANCET HAEMATOLOGY 2015; 2:e194-203. [PMID: 26688094 DOI: 10.1016/s2352-3026(15)00045-9] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/27/2015] [Accepted: 03/02/2015] [Indexed: 02/01/2023]
Abstract
BACKGROUND Anti-thymocyte globulin (ATG) was introduced into the conditioning regimen in haemopoietic cell transplantation (HCT) to prevent graft-versus-host-disease (GvHD) and graft failure. However, ATG can also cause delayed immune reconstitution of donor T cells. We studied the relation between exposure to active ATG and clinical outcomes in children. METHODS In this retrospective analysis, all patients (age 0·2-23 years) receiving their first HCT between April 1, 2004, and April 1, 2012, who received ATG (thymoglobulin) in two Dutch paediatric HCT programmes were included. The cumulative dose of ATG was chosen according to local protocols and was given intravenously over 4 days consecutively. ATG exposure measures (maximum concentration, concentration at time of HCT, clearance, days to reach a concentration below the lympholytic concentration of one arbitrary unit [AU] per mL, total area under the curve [AUC], AUC before HCT, and AUC after HCT) were calculated using a validated population pharmacokinetic model. The main outcome of interest was immune reconstitution (defined as CD4+ T cells >0·05 × 10(9) cells per L in two consecutive measurements within 100 days). Other outcomes of interest were survival, acute and chronic GvHD, and graft failure. We used Cox proportional hazard models, logistic regression models, and Fine-Gray competing risk regressions for analyses. FINDINGS 251 patients were included. The chance of successful immune reconstitution decreased as the ATG AUC after HCT increased (odds ratio 0·991, 95% CI 0·987-0·996; p<0·0001). Within the cord blood group, we noted decreased immune reconstitution above the lowest AUC quartile (≥ 20 AU × day/mL; p=0·0024), whereas in the bone marrow or peripheral blood stem cell group, decreased immune reconstitution was noted only in the highest quartile (≥ 100 AU × day/mL; p=0·0024). Successful immune reconstitution by day 100 was associated with increased overall survival (hazard ratio [HR] 0·49, 95% CI 0·29-0·81; p=0·0047) caused by reduced non-relapse mortality (0·40, 0·21-0·77; p=0·0062), and relapse-related mortality in myeloid leukaemia (0·25, 0·08-0·76; p=0·015). An AUC before transplantation of at least 40 AU × day/mL resulted in a lower incidence of acute GvHD (grade 2-4 HR 0·979, 95% CI 0·963-0·994; p=0·0081; and grade 3-4 0·975, 0·952-0·998; p=0·033), chronic GvHD (0·983, 0·968-0·998; p=0·029), and graft failure (0·981, 0·965-0·997; p=0·020) compared with an AUC of less than 40 AU × day/mL. INTERPRETATION These results stress the importance of improving the efficacy and safety of ATG in HCT by amending dosage and timing. Individualised dosing and timing of ATG to aim for optimum exposure before and after HCT could result in improved outcomes after paediatric HCT. FUNDING Dutch Organization for Scientific Research.
Collapse
Affiliation(s)
- Rick Admiraal
- Paediatric Blood and Marrow Transplant Program, University Medical Center Utrecht, Utrecht, Netherlands; U-DANCE, Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands; Department of Paediatrics, Leiden University Medical Center, Leiden, Netherlands; Department of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, Netherlands
| | - Charlotte van Kesteren
- Paediatric Blood and Marrow Transplant Program, University Medical Center Utrecht, Utrecht, Netherlands; U-DANCE, Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands; Department of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, Netherlands
| | | | - Arjan C Lankester
- Department of Paediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Marc B Bierings
- Paediatric Blood and Marrow Transplant Program, University Medical Center Utrecht, Utrecht, Netherlands
| | - Toine C G Egberts
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Maarten J D van Tol
- Department of Paediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Catherijne A J Knibbe
- Department of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, Netherlands; Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, Netherlands
| | - Robbert G M Bredius
- Department of Paediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Jaap J Boelens
- Paediatric Blood and Marrow Transplant Program, University Medical Center Utrecht, Utrecht, Netherlands; U-DANCE, Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
46
|
Preferential depletion of host over donor T cells through in vivo decay of active rabbit-anti-thymocyte globulin levels during reduced intensity conditioning. Bone Marrow Transplant 2015; 50:829-33. [PMID: 25798677 DOI: 10.1038/bmt.2015.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/19/2014] [Accepted: 01/24/2015] [Indexed: 11/09/2022]
Abstract
Inadequate T-cell chimerism following reduced-intensity conditioning transplantation may contribute to graft rejection and disease relapse. Anti-thymocyte globulin (ATG) enhances early donor T-cell chimerism, but may also deplete donor T cells, increasing risks of infection and relapse. We prospectively tested administration of rabbit ATG (rATG) ⩾14 days before the infusion of the graft, followed by in vivo decay of active rATG levels, to selectively deplete host T cells. Twenty-three patients received rATG total dose 4.5 mg/kg on days -16 and -15, fludarabine 30 mg/m(2) per day on day -7 through -3, IV busulfan 130 mg/m(2) per day on days -4 and -3 and cyclophosphamide 1500 mg/m(2) on day -2. rATG levels were therapeutic in all patients on day -14, but were sub-therapeutic (<1 μg/mL) by day 0 in 82% of patients. Median donor T-cell chimerisms on days 30 and 180 were 100% (75-100%) and 100% (90-100%), respectively. Non-relapse mortality and relapse/progression at 48 months were 17 and 30%. Cumulative incidences of acute GvHD grades II-IV and III-IV were 39 and 9%. Median follow-up is 64 months (46-79 months). Survival and disease-free survival at 48 months were 70 and 52%. These data suggest that selective depletion of host T cells using this regimen is a feasible and effective strategy.
Collapse
|
47
|
Outcomes of peripheral blood stem cell transplantation patients from HLA-mismatched unrelated donor with antithymocyte globulin (ATG)-Thymoglobulin versus ATG-Fresenius: a single-center study. Med Oncol 2015; 32:465. [PMID: 25588925 DOI: 10.1007/s12032-014-0465-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 12/16/2014] [Indexed: 10/24/2022]
Abstract
Although antithymocyte globulin (ATG) had been widely used in hematopoietic stem cell transplantation from unrelated donor due to its ability to prevent acute and chronic graft-versus-host disease (GVHD), the comparative efficacy and safety of ATG-Thymoglobulin (ATG-T) and ATG-Fresenius (ATG-F) in patients undergoing HLA-mismatched allogeneic peripheral blood stem cell transplantation from unrelated donors (UR-PBSCT) has not been evaluated. Retrospective analysis of patients who underwent HLA-mismatched UR-PBSCT between January 2003 and December 2013 and received pre-transplant ATG-T at a total dose of 10 mg/kg or ATG-F at a total dose of 20 mg/kg was performed. Patients who received ATG-T (n = 23) or ATG-F (n = 28) had similar baseline demographic, disease, and transplant characteristics. There were no significant between-groups differences in the probability of acute GVHD (P = 0.721) and chronic GVHD (P = 0.439). ATG-F was associated with nonsignificant trends toward higher disease-free survival at 3-year follow-up compared with ATG-T (45.7 ± 11.1 vs 61.3 ± 9.7 %, respectively, P = 0.07). A significantly greater proportion of ATG-T patients experienced high fever than ATG-F patients (P < 0.01) during ATG infusion. There was no difference in the rate of infection between the two treatment groups. There were less adverse effects comparing ATG-F with ATG-T. ATG-T at a total dose of 10 mg/kg and ATG-F at a total dose of 20 mg/kg had a similar clinical outcome in the setting of HLA-mismatched UR-PBSCT.
Collapse
|
48
|
Impact of serotherapy on immune reconstitution and survival outcomes after stem cell transplantations in children: thymoglobulin versus alemtuzumab. Biol Blood Marrow Transplant 2014; 21:473-82. [PMID: 25485863 DOI: 10.1016/j.bbmt.2014.11.674] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 11/13/2014] [Indexed: 01/24/2023]
Abstract
The outcome of allogeneic hematopoietic stem cell transplantation (HSCT) is strongly affected by the kinetics of reconstitution of the immune system. This study compared the effects of antithymocyte globulin (ATG) and alemtuzumab on various outcome parameters after HSCT. The study cohort consisted of 148 children, with a median age of 9.6 years (range, .4 to 19.0), who underwent HSCT for malignant and benign hematological disorders in a single HSCT unit. Conditioning included ATG (n = 110) or alemtuzumab (n = 38). Cox proportional hazard regression analysis showed that alemtuzumab significantly delayed the recovery of CD3(+) T cells and CD4(+)as well as CD8(+) T cell subsets (P ≤ .001) and natural killer (NK) cells (P = .008) compared with ATG. In both ATG- and alemtuzumab-treated patients, shorter drug exposure lead to significantly faster recovery of T cells. Alemtuzumab was associated with lower donor chimerism 3 and 6 months after transplantation and a higher risk of disease relapse (P = .001). The overall survival and event-free survival risks were significantly lower for alemtuzumab-treated patients (P = .020 and P < .001, respectively). Patients who received alemtuzumab showed a trend to lower risk of acute graft-versus-host disease, more human adenovirus, and less Epstein-Barr virus reactivations compared with patients who received ATG. These data indicate that children treated with alemtuzumab as part of the conditioning regimen have a slower T cell and NK cell reconstitution compared with those treated with ATG, which compromises the overall and event-free survival. Prolonged length of lympholytic drug exposure delayed the T cell recovery in both ATG- and alemtuzumab-treated patients. Therefore, we recommend detailed pharmacokinetic/pharmacodynamic (PK/PD) analyses in a larger cohort of patients to develop an algorithm aiming at optimization of the serotherapy containing conditioning regimen.
Collapse
|
49
|
Population pharmacokinetic/dynamic model of lymphosuppression after fludarabine administration. Cancer Chemother Pharmacol 2014; 75:67-75. [PMID: 25374408 DOI: 10.1007/s00280-014-2618-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 10/24/2014] [Indexed: 12/20/2022]
Abstract
PURPOSE Quantitative relationships between 9-β-D-arabinofuranosyl-2-fluoroadenine (F-ara-A) concentrations and lymphosuppression have not been reported, but would be useful for regimen design. A population pharmacokinetic/pharmacodynamic model was constructed in this study using data from 41 hematopoietic cell transplant (HCT) recipients conditioned with busulfan in combination with fludarabine (total dose 120 mg/m², Protocol 1519) or with fludarabine (total dose 250 mg/m²) with rabbit antithymocyte globulin (rATG, Protocol 2041). METHODS Individual pharmacokinetic parameters were fixed to post hoc Bayesian estimates, and circulating absolute lymphocyte counts (ALC) were obtained during the 3 weeks prior to graft infusion. A semi-physiological cell-kill model with three lymphocyte transit compartments was applied and aptly characterized the time course of suppression of circulating ALC by fludarabine administration. Drug- and system-specific parameters were estimated using a maximum likelihood expectation maximization algorithm, and the final model was qualified using an internal visual predictive check. RESULTS The final model successfully characterized the time course and variability in ALC. Pharmacodynamic parameters exhibited considerable between subject variability (38.9-211 %). The HCT protocol was the only covariate associated with the pharmacodynamic parameters, specifically the lymphocyte kill rate, the transit rate between lymphocyte compartments, and the baseline ALC. CONCLUSIONS This model can be used to simulate the degree of lymphosuppression for design of future fludarabine-based conditioning regimens.
Collapse
|
50
|
Impact of rabbit ATG-containing myeloablative conditioning regimens on the outcome of patients undergoing unrelated single-unit cord blood transplantation for hematological malignancies. Bone Marrow Transplant 2014; 50:45-50. [PMID: 25330221 DOI: 10.1038/bmt.2014.216] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 06/01/2014] [Accepted: 06/24/2014] [Indexed: 11/08/2022]
Abstract
This study aimed to assess the impact of antithymocyte globulin (ATG) on patient outcome in a retrospective series of 91 patients (median age: 12 years) who underwent unrelated single-unit cord blood transplantation (allo-CBT) following a myeloablative conditioning regimen. Cord blood units were HLA-matched (6/6, n=18; 21%), one-Ag mismatched (n=30, 35%) or two-Ag mismatched (n=38; 44%). In this series, the OS, nonrelapse mortality (NRM) and cumulative incidence of relapse were 47±6%, 23±4% and 48±5%, respectively. Among 46 patients who received ATG as part of the conditioning regimen, the incidence of acute and chronic GVHD was lower than that in the group of 45 patients who did not receive ATG (20% vs 43%; P=0.03). However, multivariate statistical analysis revealed that the ATG use was associated with decreased OS and EFS rates and a high incidence of NRM (hazard ratio (HR)=1.99, 95% confidence interval (CI): 1.11-3.59, P=0.02), (HR=1.83, 95% CI: 1.08-3.10, P=0.02) and (HR=2.54, 95% CI: 1.03-6.26, P=0.04), respectively. Therefore, our results do not support the use of ATG as part of a myeloablative-conditioning regimen before single-unit allo-CBT in younger patients with hematological malignancies.
Collapse
|