1
|
Wang J, Luo Y, Jia C, Yang J, Wang B, Zheng J, Jing Y, Chen W, Yang W, Zhu G, Qin M, Li S. Successful use of defibrotide to treat allogeneic hematopoietic stem cell transplantation associated thrombotic microangiopathy in pediatric patients: report from Chinese single center. Bone Marrow Transplant 2024; 59:1483-1485. [PMID: 39043925 DOI: 10.1038/s41409-024-02368-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024]
Affiliation(s)
- Jiayu Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yanhui Luo
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chenguang Jia
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jun Yang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Bin Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jie Zheng
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yuanfang Jing
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Chen
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Yang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Guanghua Zhu
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Maoquan Qin
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Sidan Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Tsakiris DA, Gavriilaki E, Chanou I, Meyer SC. Hemostasis and complement in allogeneic hematopoietic stem cell transplantation: clinical significance of two interactive systems. Bone Marrow Transplant 2024; 59:1349-1359. [PMID: 39004655 PMCID: PMC11452340 DOI: 10.1038/s41409-024-02362-8] [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: 03/16/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024]
Abstract
Hematopoietic stem cell transplantation (HCT) represents a curative treatment option for certain malignant and nonmalignant hematological diseases. Conditioning regimens before HCT, the development of graft-versus-host disease (GVHD) in the allogeneic setting, and delayed immune reconstitution contribute to early and late complications by inducing tissue damage or humoral alterations. Hemostasis and/or the complement system are biological regulatory defense systems involving humoral and cellular reactions and are variably involved in these complications after allogeneic HCT. The hemostasis and complement systems have multiple interactions, which have been described both under physiological and pathological conditions. They share common tissue targets, such as the endothelium, which suggests interactions in the pathogenesis of several serious complications in the early or late phase after HCT. Complications in which both systems interfere with each other and thus contribute to disease pathogenesis include transplant-associated thrombotic microangiopathy (HSCT-TMA), sinusoidal obstruction syndrome/veno-occlusive disease (SOS/VOD), and GVHD. Here, we review the current knowledge on changes in hemostasis and complement after allogeneic HCT and how these changes may define clinical impact.
Collapse
Affiliation(s)
| | - Eleni Gavriilaki
- Second Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Chanou
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Thessaloniki, Greece
| | - Sara C Meyer
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Cheng G, Smith MA, Phelan R, Brazauskas R, Strom J, Ahn KW, Hamilton B, Peterson A, Savani B, Schoemans H, Schoettler M, Sorror M, Higham C, Kharbanda S, Dvorak CC, Zinter MS. Epidemiology of Diffuse Alveolar Hemorrhage in Pediatric Allogeneic Hematopoietic Cell Transplantation Recipients. Transplant Cell Ther 2024; 30:1017.e1-1017.e12. [PMID: 39089527 DOI: 10.1016/j.jtct.2024.07.022] [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: 06/06/2024] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/04/2024]
Abstract
Diffuse alveolar hemorrhage (DAH) is a life-threatening pulmonary toxicity that can arise after hematopoietic cell transplantation (HCT). Risk factors and outcomes are not well understood owing to a sparsity of cases spread across multiple centers. The objectives of this epidemiologic study were to characterize the incidence, outcomes, transplantation-related risk factors and comorbid critical care diagnoses associated with post-HCT DAH. Retrospective analysis was performed in a multicenter cohort of 6995 patients age ≤21 years who underwent allogeneic HCT between 2008 and 2014 identified through the Center for International Blood and Marrow Transplant Research registry and cross-matched with the Virtual Pediatric Systems database to obtain critical care characteristics. A multivariable Cox proportional hazard model was used to determine risk factors for DAH. Logistic regression models were used to determine critical care diagnoses associated with DAH. Survival outcomes were analyzed using both a landmark approach and Cox regression, with DAH as a time-varying covariate. DAH occurred in 81 patients at a median of 54 days post-HCT (interquartile range, 23 to 160 days), with a 1-year post-transplantation cumulative incidence probability of 1.0% (95% confidence interval [CI], .81% to 1.3%) and was noted in 7.6% of all pediatric intensive care unit patients. Risk factors included receipt of transplantation for nonmalignant hematologic disease (reference: malignant hematologic disease; hazard ratio [HR], 1.98; 95% CI, 1.22 to 3.22; P = .006), use of a calcineurin inhibitor (CNI) plus mycophenolate mofetil (MMF) as graft-versus-host disease (GVHD) prophylaxis (referent: CNI plus methotrexate; HR, 1.89; 95% CI, 1.07 to 3.34; P = .029), and grade III-IV acute GVHD (HR, 2.67; 95% CI, 1.53-4.66; P < .001). Critical care admitted patients with DAH had significantly higher rates of systemic hypertension, pulmonary hypertension, pericardial disease, renal failure, and bacterial/viral/fungal infections (P < .05) than those without DAH. From the time of DAH, median survival was 2.2 months, and 1-year overall survival was 26% (95% CI, 17% to 36%). Among all HCT recipients, the development of DAH when considered was associated with a 7-fold increase in unadjusted all-cause post-HCT mortality (HR, 6.96; 95% CI, 5.42 to 8.94; P < .001). In a landmark analysis of patients alive at 2 months post-HCT, patients who developed DAH had a 1-year overall survival of 33% (95% CI, 18% to 49%), compared to 82% (95% CI, 81% to 83%) for patients without DAH (P < .001). Although DAH is rare, it is associated with high mortality in the post-HCT setting. Our data suggest that clinicians should have a heightened index of suspicion of DAH in patients with pulmonary symptoms in the context of nonmalignant hematologic indication for HCT, use of CNI + MMF as GVHD prophylaxis, and severe acute GVHD. Further investigations and validation of modifiable risk factors are warranted given poor outcomes.
Collapse
Affiliation(s)
- Geoffrey Cheng
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of California, San Francisco, California.
| | - Michael A Smith
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, California
| | - Rachel Phelan
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ruta Brazauskas
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Joelle Strom
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kwang Woo Ahn
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Andrew Peterson
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Bipin Savani
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | - Christine Higham
- Department of Pediatrics, Division of Allergy, Immunology, and BMT, University of California, San Francisco, California
| | - Sandhya Kharbanda
- Department of Pediatrics, Division of Allergy, Immunology, and BMT, University of California, San Francisco, California
| | - Christopher C Dvorak
- Department of Pediatrics, Division of Allergy, Immunology, and BMT, University of California, San Francisco, California
| | - Matt S Zinter
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, California; Department of Pediatrics, Division of Allergy, Immunology, and BMT, University of California, San Francisco, California
| |
Collapse
|
4
|
Liu C, Liu M, Liu X, Li B, Gao L, Wu S, Ji Q, Zhang Z, Zhang S, Xiao P, Lu J, Li J, Hu S. The efficacy and safety of third-party umbilical blood/umbilical cord mesenchymal stem cell assisted related haploid hematopoietic stem cell transplantation in pediatric patients with acute leukemia: an observational study. Ther Adv Hematol 2024; 15:20406207241277549. [PMID: 39372558 PMCID: PMC11452895 DOI: 10.1177/20406207241277549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/25/2024] [Indexed: 10/08/2024] Open
Abstract
Background There is limited data on third-party umbilical cord blood (UCB) or mesenchymal stem cell (MSC) transplantation-assisted haploidentical hematopoietic stem cell transplantation (haplo-HSCT) in pediatric patients. Objective To evaluate the efficacy and safety of UCB and MSC transplantation-assisted haplo-HSCT in pediatric patients with acute leukemia (AL). Design Observational study. Methods Clinical data of 152 children with AL undergoing haplo-HSCT at the Children's Hospital of Soochow University between January 2020 and June 2022 were collected. The patients were divided into the haplo-HSCT + UCB group (n = 76), haplo-HSCT + MSC group (n = 31), and haplo-HSCT group (n = 45). Hematopoietic reconstruction time, complications within 30 days after transplantation, and survival and recurrence at 3 years after transplantation were compared among the groups. Results Multivariate analysis revealed that haplo-HSCT with MSC and human leukocyte antigen (HLA) matching ⩾6/10 were independent factors reducing engraftment syndrome (ES) incidence. There were no significant differences among the groups in the hematopoietic reconstruction time or incidence of complications within 30 days after transplantation (p > 0.05). Overall survival, relapse-free survival, cumulative incidence of relapse, cumulative incidence of hematological relapse, and 3-year transplant-related mortality were not significantly different (p > 0.05). The incidence of adverse reactions in the haplo-HSCT + UCB group was 97.3% within 4 h after UCB infusion, with a particularly high occurrence rate of 94.7% for hypertension. No transfusion-related adverse reactions occurred after the transfusion of umbilical cord MSC in the haplo-HSCT + MSC group. Conclusion MSC-assisted haplo-HSCT can reduce ES incidence after transplantation in pediatric patients with AL. UCB infusion is associated with a high incidence of reversible hypertension. However, no adverse reactions were observed in umbilical cord MSC transfusion.
Collapse
Affiliation(s)
- Chang Liu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Minyuan Liu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Xin Liu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Bohan Li
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Li Gao
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Shuiyan Wu
- Department of Hematological Intensive Care Unit, Children’s Hospital of Soochow University, Suzhou, China
| | - Qi Ji
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Zhiqi Zhang
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Senlin Zhang
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Peifang Xiao
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Jun Lu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Jie Li
- Department of Hematology, Children’s Hospital of Soochow University, No.92 Zhongnan Street, Industrial Park, Suzhou 215003, China
| | - Shaoyan Hu
- Department of Hematology, Children’s Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, China
| |
Collapse
|
5
|
Schoettler ML, Ofori J, Bryson E, Spencer K, Qayed M, Stenger E, Bidgoli A, Chonat S, Westbrook A, Williams KM. Real-World Application of Recently Proposed ASTCT/CIBMTR/EBMT/APBMT Consensus Risk Stratification for Transplantation-Associated Thrombotic Microangiopathy in Children. Transplant Cell Ther 2024; 30:929.e1-929.e6. [PMID: 38936547 DOI: 10.1016/j.jtct.2024.06.017] [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: 05/28/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Consensus diagnostic and risk stratification of transplantation-associated thrombotic microangiopathy (TA-TMA) was recently achieved from international transplantation groups. Although the proposed diagnostic criteria have been applied to multiple pediatric cohorts, there are scant data applying the novel risk stratification approach in children with TA-TMA. In this retrospective cohort study, all children undergoing an allogeneic HCT or autologous HCT for neuroblastoma were prospectively screened for TA-TMA, diagnosed, and risk-stratified using the Jodele criteria from August 2019 to October 2023. Our institutional practice during the study period was treat all Jodele intermediate-risk (IR) and high-risk (HR) patients with eculizumab. Harmonization risk stratification criteria were applied retrospectively. All survival analyses were calculated from the day of TA-TMA diagnosis. To identify which specific harmonization high-risk features were the most important predictors for nonrelapse mortality (NRM), full and reduced logistical regression models were tested. The lowest Bayes information criterion and optimal Mallows CP statistic were used to identify the best subset. The analysis was performed with SAS 9.4 (SAS Institute, Cary, NC). Fifty-two children were diagnosed with TA-TMA during the study period, at a median of 37.5 days post-HCT (range, 3 to 735 days). Using Jodele risk stratification, 11 (21%) were SR, 21 (40%) were IR, and 20 (39%) were HR. Forty (77%) were treated with eculizumab. There were no statistically significant differences in NRM among Jodele risk groups, although overall survival (OS) differed significantly. Using the harmonized stratification, 49 children (94%) were stratified as HR and 3 as standard risk (SR), there were no statistically significant differences in NRM or OS between groups. Eight children (15.4%) were classified as SR using Jodele risk stratification but restratified as HR using the harmonization criteria. One child (12.5%) died in the setting of severe GVHD, and the remaining 7 were alive at the last follow-up. In a best subset model, lactate dehydrogenase (LDH) level >2 times the upper limit of normal (ULN) (odds ratio [OR], 6.52, 95% confidence interval [CI], .96 to 44.3; P = .05), grade II-IV acute graft-versus-host disease (GVHD) at the time of TA-TMA diagnosis (OR, 15.4; 95% CI, 2.14 to 110.68; P = .01), and organ dysfunction at the time of TA-TMA (OR, 21.5; 95% CI, 2.96 to 156.37; P = .002) were significantly associated with NRM; elevated sC5b-9, urine protein/creatinine ratio, and viral infections were not significantly associated with NRM. Using these best-fit criteria, 14 patients were classified as SR and 38 were classified as HR, NRM was significantly higher, and OS was significantly lower. In this cohort of children with TA-TMA, retrospective application of the harmonization criteria resulted in more patients stratified as HR compared to use of the previously described Jodele criteria. The intention of the harmonization criteria was to identify those at greatest risk of poor outcomes; while all harmonization SR patients survived, this risk stratification was very sensitive. Previous criticisms of harmonization risk stratification include limited access to sC5b-9 testing. These data suggest that organ dysfuncion, acute GVHD, and LDH >2 times ULN are the most important predictors of NRM in this cohort, allowing risk stratification even in the absence of available sC5b-9 testing. Additional studies are needed to validate these findings.
Collapse
Affiliation(s)
- Michelle L Schoettler
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia.
| | - Joel Ofori
- Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Elyse Bryson
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Kathleen Spencer
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Muna Qayed
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Elizabeth Stenger
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Alan Bidgoli
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Satheesh Chonat
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| | - Adrianna Westbrook
- Pediatric Biostatistics Core, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Kirsten M Williams
- Aflac Blood and Cancer Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia
| |
Collapse
|
6
|
Davitt M, Offenbacher R, Lee MA, Loeb DM, Manwani D, Mitchell W, Weiser DA. Atypical hemolytic uremic syndrome during induction chemotherapy in neuroblastoma, a rare phenomenon or common congenital predisposition? Pediatr Blood Cancer 2024; 71:e31175. [PMID: 38961591 DOI: 10.1002/pbc.31175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024]
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a complement-mediated thrombotic microangiopathy sometimes associated with germline variants in genes of the complement system. Clinical findings of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury arise due to aberrant complement protein activation in the circulation. A 13-month-old boy with metastatic neuroblastoma (NB) developed aHUS during his first cycle of induction chemotherapy with germline testing revealing a complement factor H (CFH) gene mutation, currently classified as a variant of uncertain significance (VUS). Now he is in disease remission after successful complement blockade therapy, thus highlighting a unique presentation of aHUS in a patient with newly diagnosed NB.
Collapse
Affiliation(s)
- Meghan Davitt
- Lisa Dean Mosely Foundation Institute for Cancer and Blood Disorders, Nemours Children's Hospital, Wilmington, Delaware, USA
| | - Rachel Offenbacher
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Michelle A Lee
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David M Loeb
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Deepa Manwani
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | - William Mitchell
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Daniel A Weiser
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
7
|
Benavent N, Cañete A, Argilés B, Juan-Ribelles A, Bonanad S, Oto J, Medina P. Delving into the clinical impact of NETs in pediatric cancer. Pediatr Res 2024:10.1038/s41390-024-03437-4. [PMID: 39095576 DOI: 10.1038/s41390-024-03437-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024]
Abstract
Pediatric cancer, a complex and heterogeneous group of diseases, continues to challenge medical research and treatment strategies. Despite advances in precision medicine and immunotherapy, certain aggressive subtypes of pediatric cancer are resistant to conventional therapies, requiring further exploration of potential therapeutic targets. Neutrophil extracellular traps (NETs), net-like structures released by neutrophils, have emerged as a potential player in the pediatric cancer landscape. However, our understanding of their role in pediatric oncology remains limited. This systematic review examines the current state of the NETs literature in pediatric cancer, focusing on the most frequent subtypes. The review reveals the scarcity of research in this area, highlighting the need for further investigation. The few studies available suggest that NETs may influence infection risk, treatment resistance and prognosis in certain pediatric malignancies. Although the field is still in its infancy, it holds great promise for advancing our understanding of pediatric cancer biology and potential therapeutic pathways. IMPACT: This review identifies a significant gap in research on neutrophil extracellular traps (NETs) in pediatric cancer. It provides a summary of existing studies and their promising findings and potential, as well as a comprehensive overview of current research on NETs in certain tumor types. It also emphasizes the lack of specific studies in pediatric cancer. The review encourages the prioritization of NET research in pediatric oncology, with the aim of improving prognosis and developing new treatments through increased understanding and targeted studies.
Collapse
Affiliation(s)
- Nuria Benavent
- Clinical and Translational Research in Cancer, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain.
| | - Adela Cañete
- Clinical and Translational Research in Cancer, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
- Pediatric Oncology and hematology Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Bienvenida Argilés
- Pediatric Oncology and hematology Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Antonio Juan-Ribelles
- Clinical and Translational Research in Cancer, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
- Pediatric Oncology and hematology Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Santiago Bonanad
- Thrombosis and Haemostasis Unit, Hematology Service, La Fe University and Polytechnic Hospital, Valencia, Spain
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Julia Oto
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| | - Pilar Medina
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, Valencia, Spain
| |
Collapse
|
8
|
Kafa K, Hoell JI. Transplant-associated thrombotic microangiopathy in pediatrics: incidence, risk factors, therapeutic options, and outcome based on data from a single center. Front Oncol 2024; 14:1399696. [PMID: 39050576 PMCID: PMC11266128 DOI: 10.3389/fonc.2024.1399696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Background Transplant-associated thrombotic microangiopathy (TA-TMA) is a critical complication of hematopoietic stem cell transplantation. Awareness about TA-TMA has increased in recent years, resulting in the implementation of TA-TMA screening in most centers. Methods Retrospective analysis of children who underwent autologous or allogeneic hematopoietic stem cell transplantation at our center between January 2018 and December 2022 was conducted to evaluate the incidence, clinical features, and outcomes of TA-TMA following the administration of different therapeutic options. Results A total of 45 patients comprised the study cohort, of whom 10 developed TA-TMA with a cumulative incidence of 22% by 100 days after transplantation. Patients with and without TA-TMA in our cohort displayed an overall survival of 80% and 88%, respectively (p = 0.48), and a non-relapse mortality of 0% and 5.7%, respectively (p = 0.12), at 1 year after transplantation. Risk factors for TA-TMA development included allogeneic transplantation and total body irradiation-based conditioning regime. Among the 10 patients with TA-TMA, 7 did not meet the high-risk criteria described by Jodele and colleagues. Of these seven patients, two responded to calcineurin-inhibitor withdrawal without further therapy and five developed multiorgan dysfunction syndrome and were treated with anti-inflammatory steroids (prednisone), and all responded to therapy. The three patients with high-risk TA-TMA were treated with complement blockade or prednisone, and all responded to therapy. Conclusion TA-TMA is a multifactorial complication with high morbidity rates. Patients with high-risk TA-TMA may benefit from complement blockade using eculizumab. No consensus has been reached regarding therapy for patients who do not meet high-risk criteria. Our analysis showed that these patients may respond to anti-inflammatory treatment with prednisone.
Collapse
Affiliation(s)
- Kinan Kafa
- Department of Pediatric Hematology and Oncology, University Hospital Halle (Saale), Halle, Germany
| | | |
Collapse
|
9
|
Yamada S, Sakai K, Kubo M, Okumura H, Asakura H, Miyamoto T, Matsumoto M. Excessive cleavage of von Willebrand factor multimers by ADAMTS13 may predict the progression of transplant-associated thrombotic microangiopathy. Res Pract Thromb Haemost 2024; 8:102517. [PMID: 39247211 PMCID: PMC11378204 DOI: 10.1016/j.rpth.2024.102517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 09/10/2024] Open
Abstract
Background Transplant-associated thrombotic microangiopathy (TA-TMA) is a fatal complication of hematopoietic stem cell transplantation and is characterized by severe thrombocytopenia, hemolytic anemia, and organ dysfunction. In response to several possible triggers, dynamic multimetric change in von Willebrand factor (VWF) may contribute to inducing microthrombi in circulation in TA-TMA. Objectives By performing VWF multimer analysis and measuring VWF-degradation product (DP), we unraveled the relationship between multimeric changes in circulating VWF and the pathogenesis of TA-TMA. Methods This study analyzed 135 plasma samples from 14 patients who underwent allogeneic hematopoietic stem cell transplantation at a single institute. VWF-associated markers, namely VWF:antigen (VWF:Ag), VWF-DP/VWF:Ag ratio, VWF:ristocetin cofactor activity, VWF:ristocetin cofactor activity/VWF:Ag ratio, and ADAMTS13 activity, were analyzed in these samples collected every 7 days. Results There were 2 patients with definite thrombotic microangiopathy (TMA) and 6 patients who presented with probable TMA that did not progress to definite TMA. Each plasma sample was classified into 3 groups: definite TMA, probable TMA, and non-TMA. VWF multimer analysis showed the absence of high-molecular-weight VWF multimers in probable TMA, whereas the appearance of unusually large VWF multimers was observed in definite TMA. The median value of the VWF-DP/VWF:Ag ratio in probable TMA was elevated to 4.17, suggesting that excessive cleavage of VWF multimers by VWF cleaving enzyme, ADAMTS13, resulted in the loss of high-molecular-weight VWF multimers. Conclusion During the transition from probable to definite TMA, drastic VWF multimer changes imply a switch from bleeding to thrombotic tendencies. Extensive VWF-DP and VWF multimer analyses provided novel insights.
Collapse
Affiliation(s)
- Shinya Yamada
- Department of Hematology, Kanazawa University, Kanazawa City, Ishikawa, Japan
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara City, Nara, Japan
- Department of Hematology, Toyama Prefectural Central Hospital, Toyama City, Toyama, Japan
| | - Kazuya Sakai
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara City, Nara, Japan
| | - Masayuki Kubo
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara City, Nara, Japan
- Department of Hematology, Nara Medical University, Kashihara City, Nara, Japan
| | - Hirokazu Okumura
- Department of Hematology, Toyama Prefectural Central Hospital, Toyama City, Toyama, Japan
| | - Hidesaku Asakura
- Department of Hematology, Kanazawa University, Kanazawa City, Ishikawa, Japan
| | - Toshihiro Miyamoto
- Department of Hematology, Kanazawa University, Kanazawa City, Ishikawa, Japan
| | - Masanori Matsumoto
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara City, Nara, Japan
- Department of Hematology, Nara Medical University, Kashihara City, Nara, Japan
| |
Collapse
|
10
|
Lee BJ, Arter Z, Doh J, Griffin SP, Vittayawacharin P, Atallah S, Shieh KR, Tran M, Jodele S, Kongtim P, Ciurea SO. Eculizumab for Shiga-toxin-induced hemolytic uremic syndrome in adults with neurological involvement. EJHAEM 2024; 5:548-553. [PMID: 38895057 PMCID: PMC11182416 DOI: 10.1002/jha2.902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 06/21/2024]
Abstract
The role of eculizumab in treating Shiga-toxin-producing Escherichia coli (STEC) hemolytic uremic syndrome (HUS) patients with neurological involvement remains unclear. We describe two distinctly different STEC-HUS patients with neurologic involvement successfully managed with eculizumab, and perform a literature review of all published cases. Both patients had complete resolution of neurological symptoms after initiation of eculizumab. Eighty patients with STEC-HUS treated with eculizumab were identified in the literature, 68.7% had complete resolution of neurological symptoms. Based on our experience and literature review, three prevailing themes were noted: 1) Early eculizumab administration optimized neurological outcomes, 2) Symptom resolution may not be immediate, neurological symptoms may initially worsen before improvement, and 3) Plasma exchange yielded no benefit. Early administration of eculizumab may reverse neurotoxicity in patients with STEC-HUS.
Collapse
Affiliation(s)
- Benjamin J. Lee
- Department of PharmacyUniversity of California Irvine HealthOrangeCaliforniaUSA
- Department of Clinical Pharmacy PracticeSchool of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Zhaohui Arter
- Department of MedicineDivision of Hematology‐OncologyChao Family Comprehensive Cancer CenterUniversity of California Irvine HealthOrangeCaliforniaUSA
| | - Jean Doh
- Department of PharmacyUniversity of California Irvine HealthOrangeCaliforniaUSA
| | - Shawn P. Griffin
- Department of PharmacyUniversity of California Irvine HealthOrangeCaliforniaUSA
- Department of Clinical Pharmacy PracticeSchool of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Pongthep Vittayawacharin
- Department of MedicineDivision of Hematology‐OncologyChao Family Comprehensive Cancer CenterUniversity of California Irvine HealthOrangeCaliforniaUSA
- Division of HematologyDepartment of MedicineFaculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Steven Atallah
- Department of PharmacyUniversity of California Irvine HealthOrangeCaliforniaUSA
- Department of Clinical Pharmacy PracticeSchool of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kevin R. Shieh
- Department of MedicineDivision of Hematology‐OncologyChao Family Comprehensive Cancer CenterUniversity of California Irvine HealthOrangeCaliforniaUSA
| | - Minh‐Ha Tran
- Department of Pathology and Laboratory MedicineDivision of Transfusion MedicineUniversity of California Irvine HealthOrangeCaliforniaUSA
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and ImmuneDeficiencyCancer and Blood Disease InstituteCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Piyanuch Kongtim
- Department of MedicineDivision of Hematology‐OncologyChao Family Comprehensive Cancer CenterUniversity of California Irvine HealthOrangeCaliforniaUSA
| | - Stefan O. Ciurea
- Department of MedicineDivision of Hematology‐OncologyChao Family Comprehensive Cancer CenterUniversity of California Irvine HealthOrangeCaliforniaUSA
| |
Collapse
|
11
|
Benítez Carabante MI, Bueno D, Alonso García L, López Torija I, Marsal J, Fernandez Navarro JM, Uria Oficialdegui ML, Panesso M, Molina B, Beléndez Bieler C, Palomo P, Pérez Martínez A, Diaz-de-Heredia C. Use of Eculizumab in Pediatric Patients with High-Risk Transplantation-Associated Thrombotic Microangiopathy: Outcomes and Risk Factors Associated with Response and Survival. A Retrospective Study on Behalf of the Spanish Group for Hematopoietic Transplantation and Cellular Therapy (GETH-TC). Transplant Cell Ther 2024; 30:601.e1-601.e13. [PMID: 38521410 DOI: 10.1016/j.jtct.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
Transplantation-associated thrombotic microangiopathy (TA-TMA) is associated with high morbidity and mortality. Although survival has improved significantly with the introduction of eculizumab, the need for improvement remains, especially in high-risk patients. This study aimed to describe the results obtained with eculizumab in a pediatric cohort with the attempt to define which risk factors could determine the response to treatment. We designed a national multicenter retrospective study of children treated with eculizumab for high-risk TA-TMA. The study cohort comprised 29 patients who had undergone a first (n = 28) or second (n = 1) allogeneic hematopoietic stem cell transplantation (HSCT) for malignant (n = 17) or nonmalignant (n = 12) disease. The median time from HSCT to TA-TMA diagnosis was 154 days (interquartile range [IQR], 103 to 263 days). Eleven patients (38%) who were initially diagnosed with low- to intermediate-risk TA-TMA progressed to high-risk TA-TMA (hrTA-TMA), within a median time of 4 days (IQR, 1 to 33 days). SC5b-9 was increased in 90% of 20 patients in whom it was measured. Renal (n = 12), pulmonary (n = 1), and intestinal (n = 1) biopsy confirmed the diagnosis in 12 of 14 patients (85%). Seventeen patients (58%) had extrarenal involvement with serositis (n = 13; 44,8%), pulmonary (n = 12; 41,4%), gastrointestinal (n = 8; 27.6%), cardiovascular (n = 7; 24.1%), or central nervous system (CNS) (n = 2; 6.9%) involvement. The median time from hrTA-TMA diagnosis to the initiation of eculizumab was 7 days (IQR, 1 to 8 days). Overall, 19 patients (65.5%) responded to eculizumab, of whom 17 (58.6%) achieved a complete response and 2 (6.9%) achieved a partial response. The remaining 10 patients (34.5%) did not show any of response. The overall response rate to eculizumab for TA-TMA was 27.59% (95% confidence interval [CI], 14.87% to 47.66%) at 1 month, 55.17% (95% CI, 38.43% to 73.48%) at 3 months, and 62.07% (95% CI, 45.10% to 79.13%) at 6 months after eculizumab initiation. In multivariate analysis, the pulmonary involvement decreased the probability of response (hazard ratio [HR], .18; P = .0298). The 1-year overall survival (OS) was 55.2% (95% CI, 35.6% to 71.0%) for the whole cohort and 83.3% (95% CI, 56.7% to 94.3%) for patients who responded to eculizumab. Pulmonary involvement (HR, 14.93; P = .0043) and CNS involvement (HR, 8.63; P = .0497) were associated with a statistically significant decrease in survival. We found that patients diagnosed with hrTA-TMA with pulmonary involvement had a poor response to eculizumab, and that patients with pulmonary and CNS involvement had significantly decreased survival. Given these results, we hypothesize that providing eculizumab therapy at an early stage of the disease before organ damage is established might significantly improve the response and, consequently, survival.
Collapse
Affiliation(s)
- María Isabel Benítez Carabante
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d´Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - David Bueno
- Department of Pediatric Hematology and Oncology, Hospital Infantil Universitario La Paz, idiPAZ Research Institute, Madrid, Spain
| | - Laura Alonso García
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d´Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Iván López Torija
- Department of Pediatric Hematology and Oncology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Julia Marsal
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain
| | | | - María Luz Uria Oficialdegui
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d´Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Melissa Panesso
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d´Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Blanca Molina
- Department of Pediatric Hematology and Oncology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Cristina Beléndez Bieler
- Department of Pediatric Hematology and Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pilar Palomo
- Department of Pediatric Hematology and Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Antonio Pérez Martínez
- Department of Pediatric Hematology and Oncology, Hospital Infantil Universitario La Paz, idiPAZ Research Institute, Madrid, Spain
| | - Cristina Diaz-de-Heredia
- Division of Pediatric Hematology and Oncology, Hospital Universitari Vall d´Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.
| |
Collapse
|
12
|
Tazoe K, Harada N, Makuuchi Y, Kuno M, Takakuwa T, Okamura H, Hirose A, Nakamae M, Nishimoto M, Nakashima Y, Koh H, Hino M, Nakamae H. Systemic inflammatory autoimmune disease before allogeneic hematopoietic stem cell transplantation is a risk factor for death in patients with myelodysplastic syndrome or chronic myelomonocytic leukemia. Ann Hematol 2024; 103:2059-2072. [PMID: 38662207 DOI: 10.1007/s00277-024-05772-2] [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: 07/24/2023] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Myelodysplastic syndrome (MDS) is well known to be complicated by systemic inflammatory autoimmune disease (SIADs). However, it remains unclear how the prognosis after allogenic hematopoietic stem cell transplantation (allo-HSCT) in patients with MDS is impacted by SIADs that occur before allo-HSCT. Therefore, we hypothesized that SIADs before allo-HSCT may be a risk factor for negative outcomes after allo-HSCT in patients with MDS. We conducted a single-center, retrospective, observational study of sixty-nine patients with MDS or chronic myelomonocytic leukemia who underwent their first allo-HCT. Fourteen of the patients had SIADs before allo-HSCT. In multivariate analysis, the presence of SIADs before allo-HSCT was an independent risk factor for overall survival (HR, 3.36, 95% confidence interval: 1.34-8.42, p = 0.009). Endothelial dysfunction syndrome was identified in five of 14 patients with SIADs who required immunosuppressive therapy or intensive chemotherapy, and notably, all patients with uncontrollable SIADs at allo-HSCT developed serious endothelial dysfunction syndrome and died in the early phase after allo-HSCT. The development of SIADs in the context of MDS is thought to reflect the degree of dysfunction of hematopoietic cells in MDS and suggests a higher risk of disease progression. In addition, MDS patients with SIADs before allo-HSCT are considered to be at higher risk of endothelial dysfunction syndrome because of preexisting vascular endothelial dysfunction due to SIADs. In conclusion, SIADs before allo-HSCT constitute an independent risk factor for death in MDS patients undergoing allo-HSCT.
Collapse
Affiliation(s)
- Kumiyo Tazoe
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
- Department of Hematology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Naonori Harada
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.
- Department of Hematology, Fuchu Hospital, Osaka, Japan.
| | - Yosuke Makuuchi
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masatomo Kuno
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Teruhito Takakuwa
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Okamura
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Asao Hirose
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mika Nakamae
- Department of Clinical Laboratory, Osaka Metropolitan University Hospital, Osaka, Japan
- Department of Laboratory Medicine and Medical Informatics, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mitsutaka Nishimoto
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yasuhiro Nakashima
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hideo Koh
- Department of Preventive Medicine and Environmental Health, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masayuki Hino
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
13
|
Hambrick HR, Fei L, Pavia K, Kaplan J, Mizuno T, Tang P, Schuler E, Benoit S, Girdwood ST. Cystatin C Outperforms Creatinine in Predicting Cefepime Clearance in Pediatric Stem Cell Transplant Recipients. Transplant Cell Ther 2024; 30:614.e1-614.e11. [PMID: 38522579 PMCID: PMC11155626 DOI: 10.1016/j.jtct.2024.03.021] [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: 12/19/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Pediatric hematopoietic stem cell transplant (HSCT) patients are at risk of developing both sepsis and altered kidney function. Cefepime is used for empiric coverage post-HSCT and requires dose adjustment based on kidney function. Since cefepime's antimicrobial efficacy is determined by the time free concentrations exceed bacterial minimum inhibitory concentration (MIC), it is important to assess kidney function accurately to ensure adequate concentrations. Serum creatinine (SCr) is routinely used to estimate glomerular filtration rate (eGFR) but varies with muscle mass, which can be significantly lower in HSCT patients, making SCr an inaccurate kidney function biomarker. Cystatin C (CysC) eGFR is independent of muscle mass, though steroid use increases CysC. Objectives of this study were to describe how eGFR impacts cefepime pharmacokinetic/pharmacodynamic (PK/PD) target attainment in pediatric HSCT patients, to investigate which method of estimating GFR (SCr, CysC, combined) best predicts cefepime clearance, and to explore additional predictors of cefepime clearance. Patients admitted to the pediatric HSCT unit who received ≥2 cefepime doses were prospectively enrolled. We measured total cefepime peak/trough concentrations between the second and fourth cefepime doses and measured SCr and CysC if not already obtained clinically within 24h of cefepime samples. eGFRs were calculated with Chronic Kidney Disease in Children U25 equations. Bayesian estimates of cefepime clearance were determined with a pediatric cefepime PK model and PK software MwPharm++. Simple linear regression was used to compare cefepime clearance normalized to body surface area (BSA) to BSA-normalized SCr-, CysC-, and SCr-/CysC-eGFRs, while multiple linear regression was used to account for additional predictors of cefepime clearance. For target attainment, we assessed the percentage of time free cefepime concentrations exceeded 1x MIC (%fT>1x MIC) and 4x MIC (%fT>4x MIC) using a susceptibility breakpoint of 8 mg/L for Pseudomonas aeruginosa. We enrolled 53 patients (ages 1 to 30 years, median 8.9 years). SCr- and CysC-eGFRs were lower in patients who attained 100% fT>1xMIC compared to those who did not attain this target: 115 versus 156 mL/min/1.73m2 (p = .01) for SCr-eGFR and 73.5 versus 107 mL/min/1.73m2 (p < .001) for CysC-eGFR. SCr-eGFR was weakly positively correlated with cefepime clearance (adjusted [a]r2= 0.14), while CysC-eGFR and SCr-/CysC-eGFR had stronger positive correlations (ar2 = 0.30 CysC, ar2 = 0.28 combo. There was a weak, significant linear association between increasing CysC-eGFR and decreased %fT>1xMIC (ar2 = 0.32) and %fT>4xMIC (ar2 = 0.14). No patients with a CysC-eGFR >120 mL/min/1.73 m2 achieved 100% fT>1xMIC or 50% fT>4x MIC. In multiple regression models, underlying diagnosis of hemoglobinopathy (in all models) and being pretransplant (in SCr and combined models) were associated with increased cefepime clearance, while concomitant use of calcineurin inhibitors was associated with decreased cefepime clearance in all models. Overall, the combo-eGFR model with timing pretransplant, hemoglobinopathy, and use of calcineurin inhibitors had the best performance (ar2 = 0.63). CysC-based eGFRs (CysC alone and combined) predicted cefepime clearance better than SCr-eGFR, even after considering steroid use. Increasing CysC eGFR correlated with decreased probability of PD target attainment, raising concerns for underdosing at high eGFRs. CysC should be included when estimating kidney function to provide adequate dosing of cefepime in pediatric HSCT patients.
Collapse
Affiliation(s)
- H Rhodes Hambrick
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| | - Lin Fei
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kathryn Pavia
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jennifer Kaplan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Tomoyuki Mizuno
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Peter Tang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Erin Schuler
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Stefanie Benoit
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sonya Tang Girdwood
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Translational and Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
14
|
Hill W, Sotlar K, Hautmann A, Kolb HJ, Ullmann J, Hausmann A, Schmidt M, Tischer J, Pham TT, Rank A, Hoechstetter MA. Late transplant-associated thrombotic microangiopathy verified in bone marrow biopsy specimens is associated with chronic GVHD and viral infections. Eur J Haematol 2024; 112:819-831. [PMID: 38243840 DOI: 10.1111/ejh.14174] [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: 10/07/2023] [Revised: 01/04/2024] [Accepted: 01/07/2024] [Indexed: 01/22/2024]
Abstract
OBJECTIVES To describe late transplant-associated thrombotic microangiopathy (TA-TMA) as chronic endothelial complication in bone marrow (BM) after allogeneic hematopoietic stem cell transplantation (HSCT). METHODS BM specimens along with conventional diagnostic parameters were assessed in 14 single-institutional patients with late TA-TMA (more than 100 days after HCST), including 11 late with history of early TA-TMA, 10 with early TA-TMA (within 100 days), and 12 non TA-TMA patients. Three non-HSCT patients served as control. The time points of BM biopsy were +1086, +798, +396, and +363 days after HSCT, respectively. RESULTS Late TA-TMA patients showed an increase of CD34+ and von Willebrand Factor (VWF)+ microvascular endothelial cells with atypical VWF+ conglomerates forming thickened VWF+ plaque sinus in the BM compared to patients without late TA-TMA and non-HSCT. Severe chronic (p = .002), steroid-refractory GVHD (p = .007) and reactivation of HHV6 (p = .002), EBV (p = .003), and adenovirus (p = .005) were pronounced in late TA-TMA. Overall and relapse-free survival were shorter in late TA-TMA than in patients without late TA-TMA (5-year OS and RFS: 78.6% vs. 90.2%, 71.4% vs. 86.4%, respectively). CONCLUSION Chronic allo-immune microangiopathy in BM associated with chronic, steroid-refractory GVHD and/or viral infections are key findings of late, high-risk TA-TMA, which deserves clinical attention.
Collapse
Affiliation(s)
- Wolfgang Hill
- Department of Internal Medicine III, University Hospital Munich, University of Munich, Munich, Germany
| | - Karl Sotlar
- University Institute of Pathology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Anke Hautmann
- Private Practice and Day Clinic for Hematology and Oncology, Regensburg, Germany
| | - Hans-Jochem Kolb
- Department of Internal Medicine III, University Hospital Munich, University of Munich, Munich, Germany
| | - Johanna Ullmann
- Department of Internal Medicine III, University Hospital Munich, University of Munich, Munich, Germany
| | - Andreas Hausmann
- Department of Internal Medicine I, Academic Teaching Hospital, München Klinik Schwabing, Munich, Germany
| | - Michael Schmidt
- Munich Cancer Registry, Institute for Medical Information Processing, Biometry, and Epidemiology, University of Munich, Munich, Germany
| | - Johanna Tischer
- Department of Internal Medicine III, University Hospital Munich, University of Munich, Munich, Germany
| | - Thu-Trang Pham
- Department of Internal Medicine I, Academic Teaching Hospital, München Klinik Schwabing, Munich, Germany
| | - Andreas Rank
- Department of Hematology and Oncology, University Hospital Augsburg, University of Augsburg, Augsburg, Germany
| | - Manuela A Hoechstetter
- Department of Internal Medicine I, Academic Teaching Hospital, München Klinik Schwabing, Munich, Germany
| |
Collapse
|
15
|
Nusrat S, Davis H, MacDougall K, George JN, Nakamura R, Borogovac A. Thrombotic Microangiopathy After Hematopoietic Stem Cell and Solid Organ Transplantation: A Review for Intensive Care Physicians. J Intensive Care Med 2024; 39:406-419. [PMID: 37990516 DOI: 10.1177/08850666231200193] [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] [Indexed: 11/23/2023]
Abstract
Intensive care physicians may assume the primary care of patients with transplant-associated thrombotic microangiopathy (TA-TMA), an uncommon but potentially critical complication of hematopoietic stem cell transplants (HSCTs) and solid organ transplants. TA-TMA can have a dramatic presentation with multiple organ dysfunction syndrome (MODS) associated with high morbidity and mortality. The typical presenting clinical features are hemolytic anemia, thrombocytopenia, refractory hypertension, proteinuria and worsening renal failure. Intestinal involvement, with abdominal pain, nausea and vomiting, gastrointestinal bleeding, and ascites are also common. Cardiopulmonary involvement may develop from various causes including pulmonary arteriolar hypertension, pleural and pericardial effusions, and diffuse alveolar hemorrhage. Due to other often concurrent complications after HSCT, early diagnosis and effective management of TA-TMA may be challenging. Close collaboration between ICU and transplant physicians, along with other relevant specialists, is needed to best manage these patients. There are currently no approved therapies for the treatment of TA-TMA. Plasma exchange and rituximab are not recommended unless circulating factor H (CFH) antibodies or thrombotic thrombocytopenic purpura (TTP; ADAMTS activity < 10%) are diagnosed or highly suspected. The role of the complement pathway activation in the pathophysiology of TA-TMA has led to the successful use of targeted complement inhibitors, such as eculizumab. However, the relatively larger studies using eculizumab have been mostly conducted in the pediatric population with limited data on the adult population. This review is focused on the role of intensive care physicians to emphasize the clinical approach to patients with suspected TA-TMA and to discuss diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Sanober Nusrat
- Department of Medicine, Division of Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hugh Davis
- Division of Pulmonary and Critical Care Medicine, City of Hope, Duarte, CA, USA
| | - Kira MacDougall
- Department of Medicine, Division of Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - James N George
- Department of Medicine, Division of Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ryotaro Nakamura
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Azra Borogovac
- Department of Hematology and Hematopoietic Cell Transplantation, Lennar Foundation Cancer Center, City of Hope, Irvine, CA, USA
| |
Collapse
|
16
|
Marco DN, Molina M, Guio AM, Julian J, Fortuna V, Fabregat-Zaragoza VL, Salas MQ, Monge-Escartín I, Riu-Viladoms G, Carcelero E, Roma JR, Llobet N, Arcarons J, Suárez-Lledó M, Rosiñol L, Fernández-Avilés F, Rovira M, Brunet M, Martínez C. Effects of CYP3A5 Genotype on Tacrolimus Pharmacokinetics and Graft-versus-Host Disease Incidence in Allogeneic Hematopoietic Stem Cell Transplantation. Pharmaceuticals (Basel) 2024; 17:553. [PMID: 38794124 PMCID: PMC11124388 DOI: 10.3390/ph17050553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Tacrolimus (Tac) is pivotal in preventing acute graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (alloHSCT). It has been reported that genetic factors, including CYP3A5*3 and CYP3A4*22 polymorphisms, have an impact on Tac metabolism, dose requirement, and response to Tac. There is limited information regarding this topic in alloHSCT. The CYP3A5 genotype and a low Tac trough concentration/dose ratio (Tac C0/D ratio) can be used to identify fast metabolizers and predict the required Tac dose to achieve target concentrations earlier. We examined 62 Caucasian alloHSCT recipients with a fast metabolizer phenotype (C0/dose ratio ≤ 1.5 ng/mL/mg), assessing CYP3A5 genotypes and acute GVHD incidence. Forty-nine patients (79%) were poor metabolizers (2 copies of the variant *3 allele) and 13 (21%) were CYP3A5 expressers (CYP3A5*1/*1 or CYP3A5*1/*3 genotypes). CYP3A5 expressers had lower C0 at 48 h (3.7 vs. 6.2 ng/mL, p = 0.03) and at 7 days (8.6 vs. 11.4 ng/mL, p = 0.04) after Tac initiation, tended to take longer to reach Tac therapeutic range (11.8 vs. 8.9 days, p = 0.16), and had higher incidence of both global (92.3% vs. 38.8%, p < 0.001) and grade II-IV acute GVHD (61.5% vs. 24.5%, p = 0.008). These results support the adoption of preemptive pharmacogenetic testing to better predict individual Tac initial dose, helping to achieve the therapeutic range and reducing the risk of acute GVHD earlier.
Collapse
Affiliation(s)
- Daniel N. Marco
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Mònica Molina
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Ana-María Guio
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Judit Julian
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, IDIBAPS, CIBERehd, Hospital Clínic, 08036 Barcelona, Spain; (J.J.); (V.F.); (M.B.)
| | - Virginia Fortuna
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, IDIBAPS, CIBERehd, Hospital Clínic, 08036 Barcelona, Spain; (J.J.); (V.F.); (M.B.)
| | | | - María-Queralt Salas
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Inés Monge-Escartín
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, 08036 Barcelona, Spain; (I.M.-E.); (G.R.-V.); (E.C.); (J.R.R.)
| | - Gisela Riu-Viladoms
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, 08036 Barcelona, Spain; (I.M.-E.); (G.R.-V.); (E.C.); (J.R.R.)
| | - Esther Carcelero
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, 08036 Barcelona, Spain; (I.M.-E.); (G.R.-V.); (E.C.); (J.R.R.)
| | - Joan Ramón Roma
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, 08036 Barcelona, Spain; (I.M.-E.); (G.R.-V.); (E.C.); (J.R.R.)
| | - Noemí Llobet
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Jordi Arcarons
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - María Suárez-Lledó
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Laura Rosiñol
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Francesc Fernández-Avilés
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Montserrat Rovira
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| | - Mercè Brunet
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, IDIBAPS, CIBERehd, Hospital Clínic, 08036 Barcelona, Spain; (J.J.); (V.F.); (M.B.)
| | - Carmen Martínez
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Cancer and Hematological Diseases, Instituto de Investigación Biomédica August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (D.N.M.); (M.M.); (A.-M.G.); (M.-Q.S.); (N.L.); (J.A.); (M.S.-L.); (L.R.); (F.F.-A.); (M.R.)
| |
Collapse
|
17
|
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
|
18
|
Takahashi T, Watkins B, Bratrude B, Neuberg D, Hebert K, Betz K, Yu A, Choi SW, Davis J, Duncan C, Giller R, Grimley M, Harris AC, Jacobsohn D, Lalefar N, Farhadfar N, Pulsipher MA, Shenoy S, Petrovic A, Schultz KR, Yanik GA, Blazar BR, Horan JT, Langston A, Kean LS, Qayed M. The Adverse Event Landscape of Stem Cell Transplant: Evidence for aGVHD Driving Early Transplant Associated Toxicities. Transplant Cell Ther 2024:S2666-6367(24)00317-8. [PMID: 38583802 DOI: 10.1016/j.jtct.2024.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/09/2024]
Abstract
Although unrelated-donor (URD) hematopoietic cell transplantation (HCT) is associated with many toxicities, a detailed analysis of adverse events, as defined by the Common Terminology Criteria for Adverse Events (CTCAE), has not previously been curated. This represents a major unmet need, especially as it relates to assessing the safety of novel agents. We analyzed a detailed AE database from the "ABA2" randomized, double-blind, placebo-controlled clinical trial of abatacept for acute graft-versus-host disease (aGVHD) prevention, for which the FDA mandated a detailed AE assessment through Day +180, and weekly neutrophil and platelet counts through Day +100. These were analyzed for their relationship to key transplant outcomes, with a major focus on the impact of aGVHD on the development/severity of AEs. A total of 2102 AEs and 1816 neutrophil/platelet counts were analyzed from 142 8/8-HLA-matched URD HCT recipients on ABA2 (placebo cohort, n = 69, abatacept cohort, n = 73). This analysis resulted in 2 major observations. (1) Among graft source, conditioning intensity, age, and Grade 2 to 4 aGVHD, only aGVHD impacted Grade 3 to 5 AE acquisition after the first month post-transplant. (2) The development of Grade 3 to 4 aGVHD was associated with thrombocytopenia. We have created a detailed resource for the transplant community by which to contextualize clinical toxicities after transplant. It has identified aGVHD as a major driver of post-HCT Grade 3 to 5 AEs, and underscored a link between aGVHD and thrombocytopenia. This establishes a critical safety framework upon which the impact of novel post-transplant aGVHD therapeutics should be evaluated. This trial was registered at www.clinicaltrials.gov (#NCT01743131).
Collapse
Affiliation(s)
- Takuto Takahashi
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Benjamin Watkins
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Emory University, Atlanta, Georgia
| | - Brandi Bratrude
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Donna Neuberg
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kyle Hebert
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kayla Betz
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alison Yu
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jeffrey Davis
- BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christine Duncan
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Roger Giller
- Center for Cancer and Blood Disorders, Children Hospital of Colorado, University of Colorado, Aurora, Colorado
| | - Michael Grimley
- University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Andrew C Harris
- Memorial Sloan Kettering Cancer Center, New York City, New York
| | - David Jacobsohn
- Children's National Health System, Washington, District of Columbia
| | - Nahal Lalefar
- University of California San Francisco, UCSF Benioff Children's Hospital Oakland, Oakland, California
| | | | - Michael A Pulsipher
- Spencer Fox Eccles School of Medicine at the University of Utah, Intermountain Primary Children's Hospital, Salt Lake City, Utah
| | - Shalini Shenoy
- Washington University School of Medicine, St Louis, Missouri
| | - Aleksandra Petrovic
- Seattle Children's Hospital and Fred Hutch Cancer Center, Seattle, Washington
| | - Kirk R Schultz
- BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - John T Horan
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amelia Langston
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Leslie S Kean
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, and Emory University, Atlanta, Georgia
| |
Collapse
|
19
|
Masuda Y, Yamazaki S, Honda A, Masamoto Y, Kurokawa M. Isolated massive pleural effusion as a manifestation of chronic graft versus host disease successfully treated with corticosteroid. Ann Hematol 2024; 103:1403-1407. [PMID: 38285080 PMCID: PMC10940441 DOI: 10.1007/s00277-024-05643-w] [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: 12/02/2023] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Isolated pleural effusion is a rare manifestation of chronic graft versus host disease (cGVHD) after hematopoietic stem cell transplantation (HSCT). We herein report a 58-year-old woman presenting with massive pleural effusion approximately 1 year after allogeneic HSCT, who was successfully treated with corticosteroid. She had discontinued tacrolimus approximately 1 month before she presented with pleural effusion, which was attributed to cGVHD after a thorough exclusion process. This case illustrates a unique manifestation of atypical cGVHD and highlights the need for prompt therapy initiation.
Collapse
Affiliation(s)
- Yasutaka Masuda
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Sho Yamazaki
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Akira Honda
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Yosuke Masamoto
- Department of Cell Therapy and Transplantation Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
- Department of Cell Therapy and Transplantation Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
20
|
Jodele S, Dandoy CE, Aguayo-Hiraldo P, Lane A, Teusink-Cross A, Sabulski A, Mizuno K, Laskin BL, Freedman J, Davies SM. A prospective multi-institutional study of eculizumab to treat high-risk stem cell transplantation-associated TMA. Blood 2024; 143:1112-1123. [PMID: 37946262 PMCID: PMC10972707 DOI: 10.1182/blood.2023022526] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT High-risk, complement mediated, untreated transplant-associated thrombotic microangiopathy (hrTMA) has dismal outcomes due to multi-organ dysfunction syndrome (MODS). The complement C5 blocker eculizumab shows promising results in hrTMA, but has not been prospectively studied in hematopoietic stem cell transplant (HCT) recipients. We performed the first multi-institutional prospective study in children and young adults to evaluate eculizumab as an early targeted intervention for hrTMA/MODS. We hypothesized that eculizumab would more than double survival in HCT recipients with hrTMA, compared to our prior study of prospectively screened, untreated hrTMAs serving as historical controls. HrTMA features (elevated terminal complement (sC5b-9) and proteinuria measured by random urine protein/creatinine ratio (≥1mg/mg)) were required for inclusion. The primary endpoint was survival at 6 six-months from hrTMA diagnosis. Secondary endpoints were cumulative incidence of MODS 6 months after hrTMA diagnosis and 1-year posttransplant survival. Eculizumab dosing included intensive loading, induction, and maintenance phases for up to 24 weeks of therapy. All 21 evaluated study subjects had MODS. Primary and secondary study endpoints were met by demonstrating survival of 71% (P < .0001) 6 months after hrTMA diagnosis and 62% 1 year after transplant. Of fifteen survivors, 11 (73%) fully recovered organ function and are well. Our study demonstrates significant improvement in survival and recovery of organ function in hrTMA using an intensified eculizumab dosing and real time biomarker monitoring. This study serves as a benchmark for planning future studies that should focus on preventative measures or targeted therapy to be initiated prior to organ injury. This trial was registered at www.clinicaltrials.gov as #NCT03518203.
Collapse
Affiliation(s)
- Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Christopher E. Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Paibel Aguayo-Hiraldo
- Division of Bone Marrow Transplantation, Children’s Hospital of Los Angeles, Los Angeles, CA
- Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Ashley Teusink-Cross
- Department of Pharmacy, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Anthony Sabulski
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kana Mizuno
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Benjamin L. Laskin
- Division of Nephrology, Children’s Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Jason Freedman
- Division of Oncology, Children’s Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Stella M. Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Disease Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
21
|
Schoettler ML, French K, Harris A, Bryson E, Deeb L, Hudson Z, Obordo J, Chandrakasan S, Parikh S, Watkins B, Stenger E, Qayed M, Chonat S, Westbrook A, Switchenko J, Williams KM. D-dimer and sinusoidal obstructive syndrome-novel poor prognostic features of thrombotic microangiopathy in children after hematopoietic cellular therapy in a single institution prospective cohort study. Am J Hematol 2024; 99:370-379. [PMID: 38164997 DOI: 10.1002/ajh.27186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/03/2023] [Indexed: 01/03/2024]
Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA) is a common, severe complication of allogeneic hematopoietic cellular therapy (HCT). Even when treated in many studies, morbidity and mortality rates are high. This prospective single-institution cohort study serially enrolled all allogeneic HCT recipients from August 2019-August 2022. Patients were universally screened for TA-TMA and intermediate and high-risk patients were immediately treated with eculizumab. Sub-distribution cox-proportional hazards models were used to identify sub-distribution hazard ratios (sHR) for multi-organ dysfunction (MOD) and non-relapse-related mortality (NRM). Of 136 patients, 36 (26%) were diagnosed with TA-TMA and 21/36 (58%) developed MOD, significantly more than those without TA-TMA, (p < .0001). Of those with TA-TMA, 18 (50%) had high-risk TA-TMA (HR-TA-TMA), 11 (31%) had intermediate-risk TA-TMA (IR-TA-TMA), and 8 (22%) had standard risk (SR-TA-TMA). Twenty-six were treated with eculizumab (1/8 SR, 7/11 IR, and 18/18 HR). Elevated D-dimer predicted the development of MOD (sHR 7.6, 95% confidence interval [CI] 1.8-32.3). Children with concurrent sinusoidal obstructive syndrome (SOS) and TA-TMA had an excess risk of MOD of 34% and data supported a biologic interaction. The adjusted NRM risk was significantly higher in the TA-TMA patients (sHR 10.54, 95% CI 3.8-29.2, p < .0001), despite prompt treatment with eculizumab. Significant RF for NRM in TA-TMA patients included SOS (HR 2.89, 95% 1.07-7.80) and elevated D-dimer (HR 3.82, 95% CI 1.14-12.84). An unrelated donor source and random urine protein to creatine ratio ≥2 mg/mg were significantly associated with no response to eculizumab (odds ratio 15, 95% CI 2.0-113.6 and OR 6.5, 95% CI 1.1-38.6 respectively). TA-TMA was independently associated with NRM despite early diagnosis and treatment with eculizumab in this large pediatric transplant cohort. Prognostic implications of D-dimer in TA-TMA merit further investigation as this is a readily accessible biomarker. Concurrent SOS is an exclusion criterion of many ongoing clinical trials, but these data highlight these patients could benefit from novel therapeutic approaches. Multi-institutional clinical trials are needed to understand the impact of TA-TMA-targeted therapies.
Collapse
Affiliation(s)
- Michelle L Schoettler
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Kaley French
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Anora Harris
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Elyse Bryson
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Laura Deeb
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Zuri Hudson
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Jeremy Obordo
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Shanmuganathan Chandrakasan
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Suhag Parikh
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Benjamin Watkins
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Elizabeth Stenger
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Satheesh Chonat
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | - Adrianna Westbrook
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| | | | - Kirsten M Williams
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Pediatric Hematopoietic Cellular Therapy, Atlanta, Georgia, USA
| |
Collapse
|
22
|
Azimi S, Hajifathali A, Parkhideh S, Salamzadeh J, Rahmati-Kamel B, Dastan F, Mehdizadeh M, Abiyarghamsari M, Tavakoli-Ardakani M. Efficacy and Safety of a DOAC Compared to Unfractionated Heparin and A Low Molecular Weight Heparin in the Prevention of Thromboembolism in Hematopoietic Stem Cell Transplant Recipients. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2024; 23:e143213. [PMID: 39005732 PMCID: PMC11246640 DOI: 10.5812/ijpr-143213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 07/16/2024]
Abstract
Background Hematopoietic stem cell transplantation (HSCT) patients are at risk of thromboembolic events, making thromboprophylaxis crucial. Objectives This study aimed to compare apixaban, a direct factor Xa inhibitor (DOAC), with dalteparin and unfractionated heparin for thromboprophylaxis in HSCT recipients. The safety outcome included the assessment of hemorrhagic events. Methods In this open-label randomized clinical trial, 182 HSCT recipients were divided into three groups: Apixaban (n = 61, 2.5 mg two times a day), dalteparin (n = 59, 5000 IU daily), and unfractionated heparin (n = 62, 5000 IU twice daily). These anticoagulant regimens were administered after central vein catheterization and during hospitalization. The primary clinical outcome was the risk of thrombosis, and the secondary outcome was the rate of bleeding. Relevant laboratory results were analyzed using appropriate statistical tests. Results Among the 61 patients in the apixaban group, six experienced thrombosis (9.83%), with four (6.65%) of them on anticoagulants. In the dalteparin group, three patients (5%) developed thrombosis, two of whom (3.38%) were on anticoagulants. In the heparin group, all four thrombosis cases (6.4%) occurred in patients on anticoagulants (P = 0.543 overall and P = 0.776 in anticoagulant users). Only two cases of bleeding were reported (1.09% overall), one in the dalteparin group (1.69%) and the other in the apixaban group (1.63%). Conclusions Apixaban, dalteparin, and heparin demonstrated similar effectiveness in preventing thromboembolism in HSCT recipients. Furthermore, the comparison of bleeding rates across the study groups did not reveal significant differences. Larger studies with higher event rates may yield more precise conclusions.
Collapse
Affiliation(s)
- Saeed Azimi
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayeh Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamshid Salamzadeh
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Farzaneh Dastan
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Chronic Respiratory Disease Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Mahshid Mehdizadeh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdiye Abiyarghamsari
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Internal Medicines, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maria Tavakoli-Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Sadowska-Klasa A, Dukat-Mazurek A, Zielińska H, Dębska-Zielkowska J, Piekarska A, Moszkowska G, Mensah-Glanowska P, Zaucha JM. Incidence and Role of Recipient-Specific Antibodies in Allogeneic Hematopoietic Cell Transplantation from Mismatched Related Donors. Transplant Cell Ther 2024; 30:99.e1-99.e10. [PMID: 37875214 DOI: 10.1016/j.jtct.2023.10.015] [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: 09/01/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 10/26/2023]
Abstract
High titer of donor-specific antibodies (DSAs) increases the risk of graft rejection after mismatched related hematopoietic cell transplantation (HCT). There are no data regarding the incidence of anti-HLA recipient-specific antibodies (RSAs) and their role after transplantation. Here we aimed to identify the incidence of RSAs in a mismatched related hematopoietic cell donor population and their possible impact on immune-mediated complications, such as acute graft-versus-host disease (aGVHD), and complications resulting from endothelial injury, such as transplantation-associated thrombotic microangiopathy (TA-TMA) and veno-occlusive disease (VOD). We prospectively analyzed the incidence of anti-HLA antibodies in 28 mismatched related pairs of recipients and their donors who underwent HCT at our center between 2020 and 2022. In positive samples screened for anti-HLA class I and/or II antibodies, the specificity of the HLA antibodies was analyzed. All recipients had a hematologic malignancy and received a myeloablative conditioning regimen and immunosuppression consisting of post-transplantation cyclophosphamide, tacrolimus, and mycophenolate mofetil. Patients were tested for TA-TMA and aGVHD development during routine post-transplantation visits up to 100 days post-transplantation. We used modified Jodele criteria for TA-TMA diagnosis, and based aGVHD grading on the MAGIC criteria. VOD was assessed using the European Society for Blood and Marrow Transplantation. Anti-HLA antibodies were detected in 12 donors (43%) and in 9 recipients (32%). There were no significant differences between donors and recipients according to age (median, 42 years [range, 17 to 69 years] versus 39 years [range, 8 to 68 years]), sex, or pregnancy history. No transfusion history was noted in the donor group (P < .05). RSA antibodies were present more often than DSAs and were detected in 9 out of 12 (75%) anti-HLA-positive donors and in only 2 out of 9 (22%) recipients, respectively (P < .05). During the follow-up, 11 patients (39%) developed aGVHD, including grade I-II in 9 (32%) and grade III-IV in 2 (7%). Twelve patients (43%) met the criteria for TA-TMA, and only 1 patient (3.5%) was diagnosed with VOD by day 100 post-HCT. RSAs were detected significantly more often in the TA-TMA group; among 12 patients diagnosed with TA-TMA, 7 (58%) had RSAs (P < .05). We did not find a correlation between RSAs and aGVHD. The patient with VOD did not have an RSA-positive donor. There was no difference in membrane attack complex (MAC) concentration in the RSA-positive group on day 30 and day 60 post-HCT; however, there was a trend toward higher MAC concentration in the RSA-positive group on day 100 (median, 912 ng/mL [range, 788 to 1120 ng/mL] versus 616 ng/mL [range, 352 to 1244 ng/mL]; P = .055). Patients with RSA suffered more often from platelet and red blood cell decreases or transfusion refractoriness, and increased lactate dehydrogenase activity was observed in all RSA-positive cases. The donor immune status and the presence of RSA may be associated with higher rates of TA-TMA in mismatched HCT recipients. Antibody-mediated complement activation might be an additional factor influencing TA-TMA occurrence.
Collapse
Affiliation(s)
- Alicja Sadowska-Klasa
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland.
| | - Anna Dukat-Mazurek
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Hanna Zielińska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Agnieszka Piekarska
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Grażyna Moszkowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Jan Maciej Zaucha
- Department of Hematology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
24
|
Jesudas R, Takemoto CM. Where have all the platelets gone? HIT, DIC, or something else? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:43-50. [PMID: 38066886 PMCID: PMC10727081 DOI: 10.1182/hematology.2023000465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Thrombocytopenia in ill children is common; accurately diagnosing the underlying etiology is challenging and essential for appropriate management. Triggers for accelerated consumption of platelets are numerous; common downstream mechanisms of clearance include platelet trapping in microvascular thrombi, phagocytosis, and platelet activation. Thrombocytopenia with microangiopathic hemolytic anemia (MAHA) is frequently due to disseminated intravascular coagulation. Thrombotic microangiopathy (TMA) is a subgroup of MAHA. Specific TMA syndromes include thrombotic thrombocytopenic purpura, complement-mediated TMA (CM-TMA), and Shiga toxin-mediated hemolytic uremic syndrome. Isolated thrombocytopenia is characteristic of immune thrombocytopenia; however, concomitant cytopenias are frequent in critically ill patients, making the diagnosis difficult. Immune thrombocytopenia with large vessel thrombosis is a feature of heparin-induced thrombocytopenia and antiphospholipid antibody syndrome. In addition, thrombocytopenia is common with macrophage activation, which is characteristic of hemophagocytic lymphohistiocytosis. While thrombocytopenia in ill patients can be driven by hypoproliferative processes such as myelosuppression and/or bone marrow failure, this review will focus on consumptive thrombocytopenia due to immune and nonimmune causes.
Collapse
|
25
|
McEwan A, Greenwood M, Ward C, Ritchie D, Szer J, Gardiner E, Colic A, Sipavicius J, Panek-Hudson Y, Kerridge I. Diagnosis and management of endothelial disorders following haematopoietic stem cell transplantation. Intern Med J 2023; 53:2162-2174. [PMID: 37528613 DOI: 10.1111/imj.16188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/09/2023] [Indexed: 08/03/2023]
Abstract
Haematopoietic stem cell transplantation is a mainstay of therapy for numerous malignant and nonmalignant diseases. Endothelial activation and dysfunction occur after stem cell transplantation, driven by various patient- and transplant-specific factors. This can manifest as one of the relatively uncommon endothelial injury syndromes, such as sinusoidal obstruction syndrome, transplant-associated thrombotic microangiopathy, idiopathic pneumonia syndrome, capillary leak syndrome, engraftment syndrome or posterior reversible encephalopathy syndrome. This review focuses on the pathogenesis, classification and diagnosis of these disorders, as well as provides guidance on risk mitigation and treatment.
Collapse
Affiliation(s)
- Ashley McEwan
- Haematology Department, Royal North Shore Hospital, Sydney, New South Wales, Australia
- Haematology Department, Liverpool Hospital, Sydney, New South Wales, Australia
- South West Sydney Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Matthew Greenwood
- Haematology Department, Royal North Shore Hospital, Sydney, New South Wales, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Christopher Ward
- Haematology Department, Royal North Shore Hospital, Sydney, New South Wales, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| | - David Ritchie
- Peter MacCallum Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Haematology Department, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- University of Melbourne, University of Melbourne, Melbourne, Victoria, Australia
| | - Jeff Szer
- Peter MacCallum Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Haematology Department, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- University of Melbourne, University of Melbourne, Melbourne, Victoria, Australia
| | - Elizabeth Gardiner
- John Curtin School of Medical Research at the Australian National University, Canberra, Australian Capital Territory, Australia
| | - Andriana Colic
- Haematology Department, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Julija Sipavicius
- Haematology Department, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Yvonne Panek-Hudson
- Peter MacCallum Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Haematology Department, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Ian Kerridge
- Haematology Department, Royal North Shore Hospital, Sydney, New South Wales, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
26
|
Koo J, Ziady AG, Reynaud D, Abdullah S, Luebbering N, Kahn S, Langenberg L, Strecker L, Lake K, Dandoy CE, Lane A, Myers KC, Sabulski A, Good S, Nalapareddy K, Solomon M, Siefert ME, Skala E, Jodele S, Davies SM. Increased Body Mass Index Augments Endothelial Injury and Clinical Outcomes after Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2023; 29:704.e1-704.e8. [PMID: 37625594 PMCID: PMC10840974 DOI: 10.1016/j.jtct.2023.08.022] [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: 06/08/2023] [Revised: 08/11/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
Higher body mass index (BMI) is characterized as a chronic inflammatory state with endothelial dysfunction. Endothelial injury after allogeneic hematopoietic stem cell transplantation (allo-HSCT) puts patients at risk for such complications as transplantation-associated thrombotic microangiopathy (TA-TMA) and acute graft-versus-host-disease (aGVHD). To evaluate the impact of increased BMI on endothelial injury after allo-HSCT in pediatric and young adult patients, we conducted a retrospective cohort study evaluating 476 consecutive allo-HSCT children and young adult recipients age 0 to 20 years. Our analysis was subdivided based on distinct age categories (<2 years and 2 to 20 years). BMI was considered as a variable but was also expressed in standard deviations from the mean adjusted for age and sex (z-score), based on established criteria from the World Health Organization (age <2 years) and the Centers for Disease Control and Prevention (age 2 to 20 years) to account for differences associated with age. Primary endpoints included the incidences of TA-TMA and aGVHD. Increased BMI z-score was associated with TA-TMA after allo-HSCT in patients age <2 years (median, 18.1; IQR, 17 to 20; P = .006) and in patients age 2 to 20 years (median, 18.7; IQR, 16 to 21.9; P = .02). Higher BMI z-score correlated with TA-TMA risk in both age groups, with a BMI z-score of .9 in the younger cohort and .7 (IQR, -.4 to 1.6; P = .04) in the older cohort. Increased BMI z-score was associated with an increased risk of TA-TMA in a multivariate analysis of the entire cohort (odds ratio [OR], 1.2; 95% confidence interval [CI], 1.05 to 1.37; P = .008). Multivariate analysis also demonstrated that patients with BMI in the 85th percentile or greater had an increased risk of developing TA-TMA compared to those with a lower BMI percentile (OR, 2.66; 95% CI, 1.62 to 4.32; P < .001). Baseline and day +7 ST2 levels were elevated in subjects with TA-TMA compared to those without TA-TMA in both age groups. Baseline sC5b-9 concentration was not correlated with BMI z-score, but sC5b-9 concentration was increased markedly by 7 days post-allo-HSCT in patients age <2 years who later developed TA-TMA compared to those who never developed TA-TMA (P = .001). The median BMI z-score was higher for patients with aGVHD compared to patients without aGVHD (.7 [range, -3.9 to 3.9] versus .2 [range, -7.8 to 5.4]; P = .03). We show that high BMI is associated with augmented risk of endothelial injury after HSCT, specifically TA-TMA. These data identify a high-risk population likely to benefit from early interventions to prevent endothelial injury and prompt treatment of established endothelial injury.
Collapse
Affiliation(s)
- Jane Koo
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio.
| | - Assem G Ziady
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Damien Reynaud
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sheyar Abdullah
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Nathan Luebbering
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Seth Kahn
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Department of Politics, Princeton University, Princeton, New Jersey
| | - Lucille Langenberg
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Lauren Strecker
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Kelly Lake
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Christopher E Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Kasiani C Myers
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Anthony Sabulski
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Samantha Good
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kodandaramireddy Nalapareddy
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael Solomon
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Matthew E Siefert
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Emily Skala
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Stella M Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
27
|
Munjal RS, Sharma J, Polishetti S, Valleru PS, Banker H, Bandhu Gupta R, Anamika F, Jain R. Beyond Immunosuppression: The Intricate Relationship Between Tacrolimus and Microangiopathy. Cureus 2023; 15:e49351. [PMID: 38146570 PMCID: PMC10749684 DOI: 10.7759/cureus.49351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2023] [Indexed: 12/27/2023] Open
Abstract
Tacrolimus, widely known as Prograf, has become the preferred immunosuppressant for preventing graft rejection in solid organ transplant recipients, particularly in steroid-sparing regimens. Its efficacy and reduced risk of acute and chronic rejection compared to cyclosporine have made it the preferred treatment option for transplant patients. However, tacrolimus has drawbacks as it is associated with adverse effects, such as renal tubular necrosis, kidney failure, hypertension, metabolic acidosis, and new-onset diabetes mellitus. Among the less common but potentially severe complications is thrombotic microangiopathy linked to tacrolimus usage. Identifying and addressing this condition early on is crucial given its severity and potential complications. Manifestations of this microangiopathy can vary, encompassing renal, neurological, cardiac, and respiratory symptoms, and, in some cases, presenting as pancreatitis, intestinal ischemia, or skin abnormalities. Although conventional management often involves plasma exchange as the primary therapeutic option, recent insights into the pathophysiology have led to newer drugs, such as eculizumab and belatacept, offering promising outcomes. In this narrative review, we delve deeper into the underlying pathophysiological mechanisms of tacrolimus-induced thrombotic microangiopathy and aim to provide clinicians with valuable recommendations for efficient and timely treatment strategies. By understanding the complexities of this condition and staying abreast of the latest advancements in therapeutic options, healthcare providers can optimize patient outcomes and ensure safer tacrolimus administration in solid organ transplant recipients.
Collapse
Affiliation(s)
| | - Jagdish Sharma
- Internal Medicine, Manipal College of Medical Sciences, Pokhara, NPL
| | | | | | - Himanshi Banker
- Medicine and Surgery, Maulana Azad Medical College, Delhi, IND
| | | | - Fnu Anamika
- Medicine, University College of Medical Sciences, Delhi, IND
| | - Rohit Jain
- Internal Medicine, Penn State Health Hershey Medical Center, Hershey, USA
| |
Collapse
|
28
|
Ma S, Bhar S, Guffey D, Kim RB, Jamil M, Amos CI, Lee SJ, Hingorani SR, Sartain SE, Li A. Prospective Clinical and Biomarker Validation of the American Society for Transplantation and Cellular Therapy Consensus Definition for Transplantation-Associated Thrombotic Microangiopathy. Transplant Cell Ther 2023; 29:685.e1-685.e7. [PMID: 37597686 PMCID: PMC11037887 DOI: 10.1016/j.jtct.2023.08.015] [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: 06/14/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023]
Abstract
Transplantation-associated thrombotic microangiography (TA-TMA) is a disorder that causes severe complications after allogeneic hematopoietic cell transplantation (allo-HCT). Diagnosing TA-TMA is challenging because of the lack of standardized criteria. In this study, we aimed to evaluate the new TA-TMA consensus definition from the American Society for Transplantation and Cellular Therapy (ASTCT) panel as part of an ongoing prospective pediatric cohort study, and also to compare the impact and outcomes of using the current definition of clinical TMA (cTMA) versus the new consensus definition. We included patients age 0 to 18 years who underwent their first allo-HCT between May 2021 and January 2023 at Texas Children's Hospital. We compared the incidence, biomarkers, and outcomes of TA-TMA applying the previous and recently proposed screening algorithms and definitions. Whereas use of the classic microangiopathic hemolytic anemia (MAHA)-based cTMA definition led to an incidence of 12.7% by day 100 post-transplantation, the ASTCT-HR definition doubled the incidence to 28.5% by day 100. In contrast to patients with a concordant diagnosis (+/+), who had significantly worse post-transplantation survival, those reclassified as TA-TMA only by the new definition (-/+) had a significantly different prognosis (100% survival at day 100) despite the lack of TMA-directed therapy. Furthermore, biomarkers of the terminal and alternative complement pathways (sC5b9 and Ba, respectively) were significantly elevated compared with non-TMA patients around day 15 in the concordant group (+/+) but not in the discordant group (-/+). The recently proposed ASTCT consensus TA-TMA diagnosis is more sensitive and allows earlier recognition of manifestation that requires closer clinical monitoring but risks overdiagnosis and overtreatment. We recommend additional prospective validation.
Collapse
Affiliation(s)
- Shengling Ma
- Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas
| | - Saleh Bhar
- Divisions of Hematology/Oncology and Critical Care Medicine and Bone Marrow Transplantation, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Danielle Guffey
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, Texas
| | - Rock Bum Kim
- Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas
| | - Mahrukh Jamil
- Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, Texas; Section of Epidemiology and Population Science, Baylor College of Medicine, Houston, Texas
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Sangeeta R Hingorani
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Sarah E Sartain
- Section of Hematology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Ang Li
- Section of Hematology-Oncology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
29
|
Notarantonio AB, D'aveni-Piney M, Pagliuca S, Ashraf Y, Galimard JE, Xhaard A, Marçais A, Suarez F, Brissot E, Feugier P, Urien S, Bouazza N, Jacquelin S, Meatchi T, Bruneval P, Frémeaux-Bacchi V, Peffault De Latour R, Hermine O, Durey-Dragon MA, Rubio MT. Systemic complement activation influences outcomes after allogeneic hematopoietic cell transplantation: A prospective French multicenter trial. Am J Hematol 2023; 98:1559-1570. [PMID: 37483161 DOI: 10.1002/ajh.27030] [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] [Received: 04/17/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/25/2023]
Abstract
Complement activation has shown a role in murine models of graft-versus-host disease (GVHD) and in endothelial complications after allogeneic hematopoietic cell transplantation (allo-HSCT). However, its impact on post-transplant outcomes has not been so far fully elucidated. Here, we conducted a prospective multicentric trial (NCT01520623) performing serial measurements of complement proteins, regulators, and CH50 activity for 12 weeks after allo-HSCT in 85 patients receiving a myeloablative conditioning (MAC) regimen for various hematological malignancies. Twenty-six out of 85 patients showed an "activated" complement profile through the classical/lectin pathway, defined as a post-transplant decline of C3/C4 and CH50 activity. Time-dependent Cox regression models demonstrated that complement activation within the first weeks after allo-HSCT was associated with increased non-relapse mortality (hazard ratio [HR]: 3.69, 95% confident interval [CI]: 1.55-8.78, p = .003) and poorer overall survival (HR: 2.72, 95% CI: 1.37-5.39, p = .004) due to increased incidence of grade II-IV acute GVHD and in particular gastrointestinal (GI) GVHD (HR: 36.8, 95% CI: 12.4-109.1, p < .001), higher incidences of thrombotic microangiopathy (HR: 8.58, 95% CI: 2.16-34.08, p = .0022), capillary leak syndrome (HR: 7.36, 95% CI: 2.51-21.66, p = .00028), post-engraftment bacterial infections (HR: 2.37, 95% CI: 1.22-4.63, p = .0108), and EBV reactivation (HR: 3.33, 95% CI: 1.31-8.45, p = .0112). Through specific immune staining, we showed the correlation of deposition of C1q, C3d, C4d, and of C5b9 components on endothelial cells in GI GVHD lesions with the histological grade of GVHD. Altogether these findings define the epidemiology and the clinical impact of complement classical/lectin pathway activation after MAC regimens and provide a rational for the use of complement inhibitory therapeutics in a post-allo-HSCT setting.
Collapse
Affiliation(s)
- Anne Béatrice Notarantonio
- Service d'Hématologie, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365, IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-les-Nancy, France
| | - Maud D'aveni-Piney
- Service d'Hématologie, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365, IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-les-Nancy, France
- Laboratory of Physiopathology of Hematological Disorders and Their Therapeutic Implications, INSERM U1158 Imagine Institute, Université Paris Cité, Paris, France
| | - Simona Pagliuca
- Service d'Hématologie, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365, IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-les-Nancy, France
| | - Yayha Ashraf
- Laboratoire d'Immunologie, Hôpital Européen Georges-Pompidou, Université Paris Cité and UMR S 1138, Centre de Recherche des Cordeliers, Paris, France
| | | | - Aliénor Xhaard
- BMT Unit, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University of Paris VII, Paris, France
| | - Ambroise Marçais
- Service d'Hématologie Clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Felipe Suarez
- Service d'Hématologie Clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Eolia Brissot
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Antoine, Paris, France
| | - Pierre Feugier
- Service d'Hématologie, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365, IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-les-Nancy, France
| | - Saik Urien
- Unité de Recherche Clinique, Paris Centre Necker Cochin, Hôpital Tarnier, Paris, France
| | - Naim Bouazza
- Unité de Recherche Clinique, Paris Centre Necker Cochin, Hôpital Tarnier, Paris, France
| | - Sébastien Jacquelin
- Laboratory of Physiopathology of Hematological Disorders and Their Therapeutic Implications, INSERM U1158 Imagine Institute, Université Paris Cité, Paris, France
| | - Tchao Meatchi
- Service d'Anatomopathologie, Hôpital Européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Patrick Bruneval
- Service d'Anatomopathologie, Hôpital Européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Véronique Frémeaux-Bacchi
- Laboratoire d'Immunologie, Hôpital Européen Georges-Pompidou, Université Paris Cité and UMR S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Régis Peffault De Latour
- BMT Unit, Saint-Louis Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University of Paris VII, Paris, France
| | - Olivier Hermine
- Laboratory of Physiopathology of Hematological Disorders and Their Therapeutic Implications, INSERM U1158 Imagine Institute, Université Paris Cité, Paris, France
- Service d'Hématologie Clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Marie Agnès Durey-Dragon
- Laboratoire d'Immunologie, Hôpital Européen Georges-Pompidou, Université Paris Cité and UMR S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Marie-Thérèse Rubio
- Service d'Hématologie, Hôpital Brabois, CHRU Nancy and CNRS UMR 7365, IMoPA, Biopôle de l'Université de Lorraine, Vandoeuvre-les-Nancy, France
- Laboratory of Physiopathology of Hematological Disorders and Their Therapeutic Implications, INSERM U1158 Imagine Institute, Université Paris Cité, Paris, France
| |
Collapse
|
30
|
Leimi L, Koski JR, Kilpivaara O, Vettenranta K, Lokki AI, Meri S. Rare variants in complement system genes associate with endothelial damage after pediatric allogeneic hematopoietic stem cell transplantation. Front Immunol 2023; 14:1249958. [PMID: 37771589 PMCID: PMC10525714 DOI: 10.3389/fimmu.2023.1249958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction Complement system has a postulated role in endothelial problems after hematopoietic stem cell transplantation (HSCT). In this retrospective, singlecenter study we studied genetic complement system variants in patients with documented endotheliopathy. In our previous study among pediatric patients with an allogeneic HSCT (2001-2013) at the Helsinki University Children´s Hospital, Finland, we identified a total of 19/122 (15.6%) patients with vascular complications, fulfilling the criteria of capillary leak syndrome (CLS), venoocclusive disease/sinusoidal obstruction syndrome (VOD/SOS) or thrombotic microangiopathy (TMA). Methods We performed whole exome sequencing (WES) on 109 patients having an adequate pre-transplantation DNA for the analysis to define possible variations and mutations potentially predisposing to functional abnormalities of the complement system. In our data analysis, we focused on 41 genes coding for complement components. Results 50 patients (45.9%) had one or several, nonsynonymous, rare germline variants in complement genes. 21/66 (31.8%) of the variants were in the terminal pathway. Patients with endotheliopathy had variants in different complement genes: in the terminal pathway (C6 and C9), lectin pathway (MASP1) and receptor ITGAM (CD11b, part of CR3). Four had the same rare missense variant (rs183125896; Thr279Ala) in the C9 gene. Two of these patients were diagnosed with endotheliopathy and one with capillary leak syndrome-like problems. The C9 variant Thr279Ala has no previously known disease associations and is classified by the ACMG guidelines as a variant of uncertain significance (VUS). We conducted a gene burden test with gnomAD Finnish (fin) as the reference population. Complement gene variants seen in our patient population were investigated and Total Frequency Testing (TFT) was used for execution of burden tests. The gene variants seen in our patients with endotheliopathy were all significantly (FDR < 0.05) enriched compared to gnomAD. Overall, 14/25 genes coding for components of the complement system had an increased burden of missense variants among the patients when compared to the gnomAD Finnish population (N=10 816). Discussion Injury to the vascular endothelium is relatively common after HSCT with different phenotypic appearances suggesting yet unidentified underlying mechanisms. Variants in complement components may be related to endotheliopathy and poor prognosis in these patients.
Collapse
Affiliation(s)
- Lilli Leimi
- Pediatric Research Center, Children’s Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Jessica R. Koski
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Outi Kilpivaara
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Kim Vettenranta
- Pediatric Research Center, Children’s Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - A. Inkeri Lokki
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Seppo Meri
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Kubo H, Imataki O, Fukumoto T, Kawanaka Y, Ishida T, Kubo YH, Kida JI, Uemura M, Fujita H, Kadowaki N. Potential factors for and the prognostic impact of ascites after allogeneic hematopoietic stem cell transplantation. Sci Rep 2023; 13:13005. [PMID: 37563148 PMCID: PMC10415345 DOI: 10.1038/s41598-023-39604-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
Ascites is sometimes detected after allogeneic hematopoietic stem cell transplantation (allo-HSCT); however, since limited information is currently available, its clinical meaning remains unclear. Therefore, we herein examined potential factors for and the impact of ascites on the prognosis of patients after allo-HSCT at our institutes. Fifty-eight patients developed ascites within 90 days of allo-HSCT (small in 34 (16%), moderate-large in 24 (11%)). A multivariate analysis identified veno-occlusive disease/sinusoidal obstruction syndrome (p = 0.01) and myeloablative conditioning (p = 0.01) as significant potential factors for the development of small ascites. Thrombotic microangiopathy (TMA) (p < 0.01) was a significant potential factor for moderate-large ascites. The incidence of both small and moderate-large ascites correlated with lower overall survival (p = 0.03 for small ascites and p < 0.01 for moderate-large ascites) and higher non-relapse mortality rates (p = 0.03 for small ascites and p < 0.01 for moderate-large ascites). Lower OS and higher NRM rates correlated with the incidence of both small and moderate-large ascites. Further investigation is warranted to establish whether the clinical sign of ascites improves the diagnostic quality of TMA in a large-scale study.
Collapse
Affiliation(s)
- Hiroyuki Kubo
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan
| | - Osamu Imataki
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan.
| | - Tetsuya Fukumoto
- Department of Hematology, Takamatsu Red Cross Hospital, Takamatsu, Kagawa, Japan
| | - Yui Kawanaka
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan
| | - Tomoya Ishida
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan
| | - Yukiko Hamasaki Kubo
- Department of Hematology, Kagawa Prefectural Central Hospital, Takamatsu, Kagawa, Japan
| | - Jun-Ichiro Kida
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan
| | - Makiko Uemura
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan
| | - Haruyuki Fujita
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan
| | - Norimitsu Kadowaki
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-Town, Kita-County, Kagawa, 761-0793, Japan
| |
Collapse
|
32
|
Zhang Z, Hong W, Wu Q, Tsavachidis S, Li JR, Amos CI, Cheng C, Sartain SE, Afshar-Kharghan V, Dong JF, Bhatraju P, Martin PJ, Makar RS, Bendapudi PK, Li A. Pathway-driven rare germline variants associated with transplant-associated thrombotic microangiopathy (TA-TMA). Thromb Res 2023; 225:39-46. [PMID: 36948020 PMCID: PMC10147584 DOI: 10.1016/j.thromres.2023.03.001] [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: 12/19/2022] [Revised: 02/20/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023]
Abstract
The significance of rare germline mutations in transplant-associated thrombotic microangiopathy (TA-TMA) is not well studied. We performed a genetic association study in 100 adult TA-TMA patients vs. 98 post-transplant controls after matching by race, sex, and year. We focused on 5 pathways in complement, von Willebrand factor (VWF) function and related proteins, VWF clearance, ADAMTS13 function and related proteins, and endothelial activation (3641variants in 52 genes). In the primary analysis focused on 189 functional rare variants, no differential variant enrichment was observed in any of the pathways; specifically, 29 % TA-TMA and 33 % controls had at least 1 rare complement mutation. In the secondary analysis focused on 37 rare variants predicted to be pathogenic or likely pathogenic by ClinVar, Complement Database, or REVEL in-silico prediction tool, rare variants in the VWF clearance pathway were found to be significantly associated with TA-TMA (p = 0.008). On the gene level, LRP1 was the only one with significantly increased variants in TA-TMA in both analyses (p = 0.025 and 0.015). In conclusion, we did not find a significant association between rare variants in the complement pathway and TA-TMA; however, we discovered a new signal in the VWF clearance pathway driven by the gene LRP1 among likely pathogenic variants.
Collapse
Affiliation(s)
- Zhihui Zhang
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, TX, United States of America
| | - Wei Hong
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, TX, United States of America
| | - Qian Wu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Spiridon Tsavachidis
- Section of Epidemiology and Population Science, Baylor College of Medicine, Houston, TX, United States of America
| | - Jian-Rong Li
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, TX, United States of America
| | - Christopher I Amos
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, TX, United States of America; Section of Epidemiology and Population Science, Baylor College of Medicine, Houston, TX, United States of America
| | - Chao Cheng
- Institute for Clinical & Translational Research, Baylor College of Medicine, Houston, TX, United States of America
| | - Sarah E Sartain
- Section of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States of America
| | - Vahid Afshar-Kharghan
- Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Jing-Fei Dong
- BloodWorks Northwest Research Institute, Seattle, WA, United States of America
| | - Pavan Bhatraju
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Paul J Martin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America; Division of Medical Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States of America
| | - Robert S Makar
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA, United States of America; Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Pavan K Bendapudi
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA, United States of America; Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Ang Li
- Section of Hematology-Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
33
|
Pace A, Steiner ME, Vercellotti GM, Somani A. Endothelial cell provenance: an unclear role in transplant medicine. FRONTIERS IN TRANSPLANTATION 2023; 2:1130941. [PMID: 38993867 PMCID: PMC11235371 DOI: 10.3389/frtra.2023.1130941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/11/2023] [Indexed: 07/13/2024]
Abstract
An understanding of the interplay between both donor endothelial progenitors and the recipient endothelium (in the case of hematopoietic cell transplant) and recipient endothelial provenance upon the established donor endothelium (in the case of solid organ transplant) is unknown. It is postulated that this interplay and consequences of purported dual endothelial populations may be a component of the post-transplant disease process and contribute to complications of engraftment or rejection. To address this potential confounding and often overlooked arena of vascular biology, a directed brief overview primarily focused on literature presented over the last decade is presented herein.
Collapse
Affiliation(s)
- Autumn Pace
- University of Minnesota Medical School, Minneapolis, MN, United States
| | - Marie E. Steiner
- Department of Pediatrics, Division of Hematology/Oncology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Pediatrics, Division of Critical Care Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Gregory M. Vercellotti
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Arif Somani
- Department of Pediatrics, Division of Critical Care Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
34
|
Chen RY, Li XZ, Lin Q, Tang HY, Cui NX, Jiang L, Dai XM, Chen WQ, Deng F, Hu SY, Zhu XM. Pathological evaluation of renal complications in children following allogeneic hematopoietic stem cell transplantation: a retrospective cohort study. BMC Pediatr 2023; 23:186. [PMID: 37085779 PMCID: PMC10120150 DOI: 10.1186/s12887-023-03996-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapy for hematologic malignancies and non-malignant disorders, such as aplastic anemia, fanconi anemia, and certain immune deficiencies. Post-transplantation kidney injury is a common complication and involves a wide spectrum of structural abnormalities, including glomerular (MSPGN, mesangial proliferative glomerulonephritis; FSGS, focal segmental glomerulosclerosis; MPGN, membranoproliferative glomerulonephritis; MCD, minimal change disease), vascular (TMA, thrombotic microangiopathy), and/or tubulointerstitial (TIN, tubulointerstitial nephritis; ATI, acute tubular injury). Renal biopsy is the gold-standard examination for defining multiple etiologies of kidney impairment. Although kidney injury following HSCT has been studied, little is known about the effects of allo-HSCT on renal pathology in pediatric patients. METHODS We retrospectively analyzed renal biopsy specimens from children with kidney injury after allo-HSCT and correlated results with clinical data in the last 10 years. RESULTS Among 25 children (18 males and 7 females), three patients had proteinuria indicating nephrotic syndrome (24-hour urinary total protein/weight > 50 mg/kg/d), nine patients had severely reduced estimated glomerular filtration rate (eGFR < 30 ml/min/1.73 m2) and four patients received kidney replacement therapy (KRT). The main pathologies identified from kidney biopsies were MSPGN (n = 12), FSGS (n = 12), MPGN (n = 5), TMA (n = 4), MCD (n = 3), diffuse glomerular fibrosis (DGF, n = 2), ATI and TIN, in isolation or combined with other pathologies. The median follow-up time was 16.5 (0.5 ~ 68.0) months. Three patients died of recurrent malignancy and/or severe infection, one child developed to end-stage renal disease (ESRD), six patients (24%) had elevated serum creatinine (SCr > 100µmol/l) and nine patients (36%) still had proteinuria. CONCLUSIONS This study evaluates histomorphologic findings from kidney biopsies of pediatric recipients following allo-HSCT. Detailed evaluation of renal biopsy samples is helpful to elucidate the nature of renal insult, and may potentially identify treatable disease processes.
Collapse
Affiliation(s)
- Ru-Yue Chen
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Zhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Qiang Lin
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Han-Yun Tang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ning-Xun Cui
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lu Jiang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Mei Dai
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei-Qing Chen
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fan Deng
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shao-Yan Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xue-Ming Zhu
- Department of Pathology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
35
|
Mahmoudjafari Z, Alencar MC, Alexander MD, Johnson DJ, Yeh J, Evans MD. Hematopoietic stem cell transplantation-associated thrombotic microangiopathy and the role of advanced practice providers and pharmacists. Bone Marrow Transplant 2023:10.1038/s41409-023-01951-3. [PMID: 37059738 DOI: 10.1038/s41409-023-01951-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/01/2023] [Accepted: 03/08/2023] [Indexed: 04/16/2023]
Abstract
Hematopoietic stem cell transplantation-associated thrombotic microangiopathy (HSCT-TMA) is a severe and potentially life-threatening complication. HSCT-TMA is often underdiagnosed due to multifactorial pathophysiology and a historic lack of standard diagnostic criteria. Identification of the multi-hit hypothesis and the key role of the complement system, particularly the lectin pathway of complement, has led to development of treatments targeting the underlying pathogenesis of HSCT-TMA. Additional research is ongoing to investigate the efficacy and safety of these targeted therapies in patients with HSCT-TMA. Advanced practice providers (APPs; nurse practitioners and physician assistants) and pharmacists are critical members of the multidisciplinary HSCT team and ensure management of patients throughout the continuum of care. Additionally, pharmacists and APPs can improve patient care through medication management of complex regimens; transplant education for patients, staff, and trainees; evidence-based protocol and clinical guideline development; assessment and reporting of transplant-related outcomes; and quality improvement initiatives to improve outcomes. Understanding the presentation, prognosis, pathophysiology, and treatment options for HSCT-TMA can improve each of these efforts. Collaborative practice model for monitoring and care of HSCT-TMA. Advanced practice providers and pharmacists contribute to many aspects of patient care in transplant centers, including medication management for complex regimens; transplant education for patients, staff, and trainees; evidence-based protocol and clinical guideline development; assessment and reporting of transplant-related outcomes; and quality improvement initiatives. HSCT-TMA is a severe and potentially life-threatening complication that is often underdiagnosed. The collaboration of a multidisciplinary team of advanced practice providers, pharmacists, and physicians can optimize recognition, diagnosis, management, and monitoring of patients with HSCT-TMA, thereby improving outcomes for these patients.
Collapse
Affiliation(s)
- Zahra Mahmoudjafari
- Department of Pharmacy, University of Kansas Cancer Center, Kansas City, KS, USA.
| | - Maritza C Alencar
- Oncology Service Line, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Maurice D Alexander
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, NC, USA
| | - Darren J Johnson
- Pediatric Hematology and Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Jason Yeh
- Division of Pharmacy, MD Anderson Cancer Center, Houston, TX, USA
| | - Misty D Evans
- School of Nursing, Vanderbilt University, Nashville, TN, USA
- Sarah Cannon Pediatric Hematology/Oncology & Cellular Therapy at TriStar Centennial, Nashville, TN, USA
| |
Collapse
|
36
|
A Fatal Case of Neuroblastoma Complicated by Posterior Reversible Encephalopathy with Rapidly Evolving Transplantation-Associated Thrombotic Microangiopathy. CHILDREN 2023; 10:children10030506. [PMID: 36980064 PMCID: PMC10047442 DOI: 10.3390/children10030506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023]
Abstract
Background: Transplantation-associated thrombotic microangiopathy (TA-TMA) is a severe complication of hematopoietic stem cell transplantation and is sometimes fatal. Observations: A 4-year-old, male patient with stage M neuroblastoma (NBL) who had received an allogeneic bone marrow transplantation (BMT) from his sibling five months previously presented with rapidly progressive posterior reversible encephalopathy (PRES) complicated with TA-TMA. Although the patient was transferred to the pediatric intensive care unit, he died within one week after the onset of the latest symptoms. Conclusions: This is the first description of a fatal case of NBL complicated by PRES with rapidly evolving TA-TMA after an allogenic BMT.
Collapse
|
37
|
Fakhouri F, Schwotzer N, Frémeaux-Bacchi V. How I diagnose and treat atypical hemolytic uremic syndrome. Blood 2023; 141:984-995. [PMID: 36322940 DOI: 10.1182/blood.2022017860] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/19/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Our understanding and management of atypical hemolytic uremic syndrome (aHUS) have dramatically improved in the last decade. aHUS has been established as a prototypic disease resulting from a dysregulation of the complement alternative C3 convertase. Subsequently, prospective nonrandomized studies and retrospective series have shown the efficacy of C5 blockade in the treatment of this devastating disease. C5 blockade has become the cornerstone of the treatment of aHUS. This therapeutic breakthrough has been dulled by persistent difficulties in the positive diagnosis of aHUS, and the latter remains, to date, a diagnosis by exclusion. Furthermore, the precise spectrum of complement-mediated renal thrombotic microangiopathy is still a matter of debate. Nevertheless, long-term management of aHUS is increasingly individualized and lifelong C5 blockade is no longer a paradigm that applies to all patients with this disease. The potential benefit of complement blockade in other forms of HUS, notably secondary HUS, remains uncertain.
Collapse
Affiliation(s)
- Fadi Fakhouri
- Department of Medicine, Service of Nephrology and Hypertension, Lausanne University Hospital and Université de Lausanne, Lausanne, Switzerland
| | - Nora Schwotzer
- Department of Medicine, Service of Nephrology and Hypertension, Lausanne University Hospital and Université de Lausanne, Lausanne, Switzerland
| | - Véronique Frémeaux-Bacchi
- Laboratory of Immunology, Paris University, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Paris, France
| |
Collapse
|
38
|
Schoettler ML, Carreras E, Cho B, Dandoy CE, Ho VT, Jodele S, Moissev I, Sanchez-Ortega I, Srivastava A, Atsuta Y, Carpenter P, Koreth J, Kroger N, Ljungman P, Page K, Popat U, Shaw BE, Sureda A, Soiffer R, Vasu S. Harmonizing Definitions for Diagnostic Criteria and Prognostic Assessment of Transplantation-Associated Thrombotic Microangiopathy: A Report on Behalf of the European Society for Blood and Marrow Transplantation, American Society for Transplantation and Cellular Therapy, Asia-Pacific Blood and Marrow Transplantation Group, and Center for International Blood and Marrow Transplant Research. Transplant Cell Ther 2023; 29:151-163. [PMID: 36442770 PMCID: PMC10119629 DOI: 10.1016/j.jtct.2022.11.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
Transplantation-associated thrombotic microangiopathy (TA-TMA) is an increasingly recognized complication of hematopoietic cell transplantation (HCT) associated with significant morbidity and mortality. However, TA-TMA is a clinical diagnosis, and multiple criteria have been proposed without universal application. Although some patients have a self-resolving disease, others progress to multiorgan failure and/or death. Poor prognostic features also are not uniformly accepted. The lack of harmonization of diagnostic and prognostic markers has precluded multi-institutional studies to better understand incidence and outcomes. Even current interventional trials use different criteria, making it challenging to interpret the data. To address this urgent need, the American Society for Transplantation and Cellular Therapy, Center for International Bone Marrow Transplant Research, Asia-Pacific Blood and Marrow Transplantation, and European Society for Blood and Marrow Transplantation nominated representatives for an expert panel tasked with reaching consensus on diagnostic and prognostic criteria. The panel reviewed literature, generated consensus statements regarding diagnostic and prognostic features of TA-TMA using the Delphi method, and identified future directions of investigation. Consensus was reached on 4 key concepts: (1) TA-TMA can be diagnosed using clinical and laboratory criteria or tissue biopsy of kidney or gastrointestinal tissue; however, biopsy is not required; (2) consensus diagnostic criteria are proposed using the modified Jodele criteria with additional definitions of anemia and thrombocytopenia. TA-TMA is diagnosed when ≥4 of the following 7 features occur twice within 14 days: anemia, defined as failure to achieve transfusion independence despite neutrophil engraftment; hemoglobin decline by ≥1 g/dL or new-onset transfusion dependence; thrombocytopenia, defined as failure to achieve platelet engraftment, higher-than-expected transfusion needs, refractory to platelet transfusions, or ≥50% reduction in baseline platelet count after full platelet engraftment; lactate dehydrogenase (LDH) exceeding the upper limit of normal (ULN); schistocytes; hypertension; soluble C5b-9 (sC5b-9) exceeding the ULN; and proteinuria (≥1 mg/mg random urine protein-to-creatinine ratio [rUPCR]); (3) patients with any of the following features are at increased risk of nonrelapse mortality and should be stratified as high-risk TA-TMA: elevated sC5b-9, LDH ≥2 times the ULN, rUPCR ≥1 mg/mg, multiorgan dysfunction, concurrent grade II-IV acute graft-versus-host disease (GVHD), or infection (bacterial or viral); and (4) all allogeneic and pediatric autologous HCT recipients with neuroblastoma should be screened weekly for TA-TMA during the first 100 days post-HCT. Patients diagnosed with TA-TMA should be risk-stratified, and those with high-risk disease should be offered participation in a clinical trial for TA-TMA-directed therapy if available. We propose that these criteria and risk stratification features be used in data registries, prospective studies, and clinical practice across international settings. This harmonization will facilitate the investigation of TA-TMA across populations diverse in race, ethnicity, age, disease indications, and transplantation characteristics. As these criteria are widely used, we expect continued refinement as necessary. Efforts to identify more specific diagnostic and prognostic biomarkers are a top priority of the field. Finally, an investigation of the impact of TA-TMA-directed treatment, particularly in the setting of concurrent highly morbid complications, such as steroid-refractory GVHD and infection, is critically needed.
Collapse
Affiliation(s)
- M L Schoettler
- Department Blood and Marrow Transplantation, Children's Healthcare of Atlanta, Aflac Cancer and Blood Disorders Center, Atlanta, Georgia
| | - E Carreras
- Spanish Bone Marrow Donor Registry, Josep Carreras Foundation and Leukemia Research Institute, Barcelona, Catalunya, Spain
| | - B Cho
- Department of Internal Medicine, Catholic Blood and Marrow Transplantation Center, Seoul, Korea
| | - C E Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - V T Ho
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - S Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - I Moissev
- RM Gorbacheva Research Institute, Pavlov University, Saint-Petersburg, Russian Federation
| | | | - A Srivastava
- Department of Haematology, Christian Medical College, Vellore, India
| | - Y Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan
| | - P Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - J Koreth
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - N Kroger
- Division of Hematology, Ohio State University, Columbus, Ohio
| | - P Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - K Page
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - U Popat
- Department of Stem Cell Transplantation & Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - B E Shaw
- Center for International Blood and Marrow Transplant Research, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin; Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - A Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-Hospitalet, Barcelona, Spain
| | - R Soiffer
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - S Vasu
- Division of Hematology, Ohio State University, Columbus, Ohio.
| |
Collapse
|
39
|
Fraebel J, Engelhardt BG, Kim TK. Noninfectious Pulmonary Complications after Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2023; 29:82-93. [PMID: 36427785 DOI: 10.1016/j.jtct.2022.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/31/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
Pulmonary complications after hematopoietic stem cell transplantation (HSCT) are important sources of morbidity and mortality. Improvements in infection-related complications have made noninfectious pulmonary complications an increasingly significant driver of transplantation-related mortality. Broadly, these complications can be characterized as either early or late complications, with idiopathic pneumonia syndrome and bronchiolitis obliterans syndrome the most prevalent early and late complications, respectively. Outcomes with historical treatment consisting mainly of corticosteroids are often poor, highlighting the need for a deeper understanding of these complications' underlying disease biology to guide the adoption of novel therapies that are being increasingly used in the modern era.
Collapse
Affiliation(s)
- Johnathan Fraebel
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brian G Engelhardt
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Nashville, Tennessee; Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Immunobiology, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Nashville, Tennessee; Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee.
| |
Collapse
|
40
|
Endothelial Dysfunction Syndromes after Allogeneic Stem Cell Transplantation. Cancers (Basel) 2023; 15:cancers15030680. [PMID: 36765638 PMCID: PMC9913851 DOI: 10.3390/cancers15030680] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains the only therapy with a curative potential for a variety of malignant and non-malignant diseases. The major limitation of the procedure is the significant morbidity and mortality mainly associated with the development of graft versus host disease (GVHD) as well as with a series of complications related to endothelial injury, such as sinusoidal obstruction syndrome/veno-occlusive disease (SOS/VOD), transplant-associated thrombotic microangiopathy (TA-TMA), etc. Endothelial cells (ECs) are key players in the maintenance of vascular homeostasis and during allo-HSCT are confronted by multiple challenges, such as the toxicity from conditioning, the administration of calcineurin inhibitors, the immunosuppression associated infections, and the donor alloreactivity against host tissues. The early diagnosis of endothelial dysfunction syndromes is of paramount importance for the development of effective prophylactic and therapeutic strategies. There is an urgent need for the better understanding of the pathogenetic mechanisms as well as for the identification of novel biomarkers for the early diagnosis of endothelial damage. This review summarizes the current knowledge on the biology of the endothelial dysfunction syndromes after allo-HSCT, along with the respective therapeutic approaches, and discusses the strengths and weaknesses of possible biomarkers of endothelial damage and dysfunction.
Collapse
|
41
|
Mafosfamide, a cyclophosphamide analog, causes a proinflammatory response and increased permeability on endothelial cells in vitro. Bone Marrow Transplant 2023; 58:407-413. [PMID: 36639572 DOI: 10.1038/s41409-023-01912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Post-transplantation cyclophosphamide (PTCy) has decreased GVHD incidence. Endothelial damage in allo-HCT is caused by multiple factors, including conditioning treatments and some immunosupressants, and underlies HCT-complications as GVHD. Nevertheless, the specific impact of PTCy on the endothelium remains unclear. We evaluated the effect of mafosfamide (MAF), an active Cy analog, on endothelial cells (ECs) vs. cyclosporine A (CSA), with known damaging endothelial effect. ECs were exposed to MAF and CSA to explore changes in endothelial damage markers: (i) surface VCAM-1, (ii) leukocyte adhesion on ECs, (iii) VE-cadherin expression, (iv) production of VWF, and (v) activation of intracellular signaling proteins (p38MAPK, Akt). Results obtained (expressed in folds vs. controls) indicate that both compounds increased VCAM-1 expression (3.1 ± 0.3 and 2.8 ± 0.6, respectively, p < 0.01), with higher leukocyte adhesion (5.5 ± 0.6, p < 0.05, and 2.8 ± 0.4, respectively). VE-cadherin decreased with MAF (0.8 ± 0.1, p < 0.01), whereas no effect was observed with CSA. Production of VWF augmented with CSA (1.4 ± 0.1, p < 0.01), but diminished with MAF (0.9 ± 0.1, p < 0.05). p38MAPK activation occurred with both compounds, being more intense and faster with CSA. Both drugs activated Akt, with superior MAF effect at longer exposure. Therefore, the cyclophosphamide analog MAF is not exempt from a proinflammatory effect on the endothelium, though without modifying the subendothelial characteristics.
Collapse
|
42
|
Yamaguchi M, Mizuno M, Kitamura F, Iwagaitsu S, Nobata H, Kinashi H, Banno S, Asai A, Ishimoto T, Katsuno T, Ito Y. Case report: Thrombotic microangiopathy concomitant with macrophage activation syndrome in systemic lupus erythematosus refractory to conventional treatment successfully treated with eculizumab. Front Med (Lausanne) 2023; 9:1097528. [PMID: 36698804 PMCID: PMC9868404 DOI: 10.3389/fmed.2022.1097528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Thrombotic microangiopathy (TMA) is a rare but life-threatening complication of systemic lupus erythematosus (SLE). Macrophage activation syndrome (MAS) is also a rare, life-threatening hyperinflammatory condition that is comorbid with SLE. However, the association between TMA and MAS in patients with SLE has rarely been assessed, and the difficulty of diagnosing these conditions remains prevalent. The efficacy of eculizumab has been reported for SLE patients whose conditions are complicated with TMA. However, no study has investigated the therapeutic efficacy of eculizumab for TMA concomitant with SLE-associated MAS. Herein, we report the first case of TMA concomitant with SLE-associated MAS that was initially refractory to conventional immunosuppressive therapy but showed remarkable recovery after eculizumab treatment. Furthermore, we evaluated serum syndecan-1 and hyaluronan levels, which are biomarkers of endothelial damage. We found that these levels decreased after the administration of eculizumab, suggesting that TMA was the main pathology of the patient. This case illustrates that it is important to appropriately assess the possibility of TMA during the course of SLE-associated MAS and consider the use of eculizumab as necessary.
Collapse
Affiliation(s)
- Makoto Yamaguchi
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Masashi Mizuno
- Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumiya Kitamura
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Shiho Iwagaitsu
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Hironobu Nobata
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Hiroshi Kinashi
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Shogo Banno
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Akimasa Asai
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Takuji Ishimoto
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Takayuki Katsuno
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan,Department of Nephrology and Rheumatology, Aichi Medical University Medical Center, Okazaki, Japan
| | - Yasuhiko Ito
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan,*Correspondence: Yasuhiko Ito,
| |
Collapse
|
43
|
Lazana I. Transplant-Associated Thrombotic Microangiopathy in the Context of Allogenic Hematopoietic Stem Cell Transplantation: Where We Stand. Int J Mol Sci 2023; 24:ijms24021159. [PMID: 36674666 PMCID: PMC9863862 DOI: 10.3390/ijms24021159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA) constitutes a significant contributor to the increased morbidity and mortality after allogenic hematopoietic stem cell transplantation (allo-HSCT). TA-TMA is a heterogenous disease, characterized by the triad of endothelial cell activation, complement dysregulation and microvascular hemolytic anemia, which may affect all organs. The lack of consensus diagnostic criteria, along with the common clinical features mimicking other diseases that complicate allo-HSCT, make the diagnosis of TA-TMA particularly challenging. Significant effort has been made to recognize specific risk factors predisposing to the development of TA-TMA and to identify serum biomarkers predicting the development of the disease. With regard to treatment, therapeutic plasma exchange (TPE) has been traditionally used, although with doubtful efficacy. On the other hand, the pivotal role of complement activation in the pathophysiology of TA-TMA has led to the exploration of the therapeutic potential of complement inhibitors in this setting. Eculizumab has been proposed as a first-line therapeutic agent in TA-TMA, owing to the very promising results in both pediatric and adult clinical trials. Pharmacokinetic and pharmacodynamic studies and CH50 levels are of paramount importance in the allo-HSCT setting, as a different dosing schedule (more intensive-in dose and frequency-at the beginning) seems to be required for successful outcomes. Furthermore, Narsoplimab, a MASP-2 inhibitor, recently received a Breakthrough Therapy Designation from the FDA for the treatment of TA-TMA after allo-HSCT. Finally, the decision to withdraw the CNIs, although initially advised by the Bone and Marrow Transplant Clinical Trials Network Committee, remains debatable owing to the controversial results of recent clinical trials. This review summarizes the current updates on pathophysiology, diagnosis and therapeutic approaches and emphasizes future goals and perspectives.
Collapse
Affiliation(s)
- Ioanna Lazana
- Cell and Gene Therapy Laboratory, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
- Hematology Department, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK
| |
Collapse
|
44
|
Cammett TJ, Garlo K, Millman EE, Rice K, Toste CM, Faas SJ. Exploratory Prognostic Biomarkers of Complement-Mediated Thrombotic Microangiopathy (CM-TMA) in Adults with Atypical Hemolytic Uremic Syndrome (aHUS): Analysis of a Phase III Study of Ravulizumab. Mol Diagn Ther 2023; 27:61-74. [PMID: 36329366 DOI: 10.1007/s40291-022-00620-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Clinically validated biomarkers for monitoring of patients with complement-mediated thrombotic microangiopathy (CM-TMA) including atypical hemolytic uremic syndrome (aHUS) are unavailable. Improved characterization of biomarkers in patients with aHUS may inform treatment and monitoring for patients with CM-TMA. METHODS This analysis used data collected from 55/56 (98.2 %) adult patients with aHUS enrolled in the global Phase III study of ravulizumab (NCT02949128). Baseline (pre-treatment) patient serum, plasma and urine biomarker levels were compared with the maximum observed levels in normal donors and evaluated for associations with pre-treatment plasma exchange/infusion and dialysis status. Biomarkers were also assessed for associations with key clinical measures at baseline and with changes at 26 and 52 weeks from treatment initiation via linear regression analyses. RESULTS Complement-specific urine levels (factor Ba and sC5b-9) were elevated in >85 % of patients and are significantly associated with pre-treatment kidney dysfunction. Baseline levels of other evaluated biomarkers were elevated in >70 % of patients with aHUS, except for plasma sC5b-9 and serum sVCAM-1. Lower levels of urine complement markers at baseline are significantly associated with improvements in total urine protein and estimated glomerular filtration rate at 26 and 52 weeks of treatment. Clinical assessment of complement activation by a receiver operating characteristic analysis of Ba and sC5b-9 was more sensitive and specific in urine matrix than plasma. CONCLUSION This analysis identified a set of biomarkers that may show utility in the prognosis of CM-TMA, including their potential for measuring and predicting response to anti-C5 therapy. Further studies are required to enhance patient risk stratification and improve management of these vulnerable patients. CLINICAL TRIALS REGISTRATION NCT02949128, ClinicalTrials.gov.
Collapse
Affiliation(s)
| | | | | | - Kara Rice
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | | | - Susan J Faas
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| |
Collapse
|
45
|
Agarwal N, Rotz S, Hanna R. Medical emergencies in pediatric blood & marrow transplant and cellular therapies. Front Pediatr 2023; 11:1075644. [PMID: 36824648 PMCID: PMC9941678 DOI: 10.3389/fped.2023.1075644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/16/2023] [Indexed: 02/10/2023] Open
Abstract
Hematopoietic stem cell transplant (HCT) is used for many pediatric malignant and non-malignant diseases. However, these patients are at a high risk for emergencies post-transplant, related to prior comorbidities and treatments for the underlying disease, high dose chemotherapy regimen related toxicities, prolonged myelosuppression, and opportunistic infections due to their immunocompromised state. Emergencies can be during preparative regimen and hematopoietic progenitor cell (HPC) infusion, acute post-transplant (pre-engraftment) and late during post engraftment. Infectious complications are the most common cause of morbidity and mortality in the peri-transplant period. Sinusoidal obstructive syndrome is another life-threatening emergency seen in children undergoing HCT, especially in infants. Timely recognition and administration of defibrotide with/without steroids is key to the management of this complication. Another complication seen is transplant associated thrombotic microangiopathy. It can cause multiorgan failure if left untreated and demands urgent identification and management with complement blockade agents such as eculizumab. Cytokine release syndrome and cytokine storm is an important life-threatening complication seen after cellular therapy, and needs emergent intervention with ICU supportive care and tocilizumab. Other complications in acute period include but are not limited to: seizures from busulfan or other chemotherapy agents, PRES (posterior reversible encephalopathy syndrome), diffuse alveolar hemorrhage, idiopathic pulmonary syndrome and allergic reaction to infusion of stem cells. Acute graft versus host disease (GvHD) is a major toxicity of allogeneic HCT, especially with reduced intensity conditioning, that can affect the skin, liver, upper and lower gastrointestinal tract. There has been major development in new biomarkers for early identification and grading of GvHD, which enables application of treatment modalities such as post-transplant cyclophosphamide and JAK/STAT inhibitors to prevent and treat GvHD. Myelosuppression secondary to the chemotherapy increases risk for engraftment syndrome as well as coagulopathies, thus increasing the risk for clotting and bleeding in the pediatric population. The purpose of this article is to review recent literature in these complications seen with pediatric hematopoietic cell transplant (HCT) and cellular therapies and provide a comprehensive summary of the major emergencies seen with HCT.
Collapse
Affiliation(s)
- Nikki Agarwal
- Department of Pediatric Hematology, Oncology and Bone Marrow and Blood Transplant, Cleveland Clinic, Cleveland, OH, United States
| | - Seth Rotz
- Department of Pediatric Hematology, Oncology and Bone Marrow and Blood Transplant, Cleveland Clinic, Cleveland, OH, United States
| | - Rabi Hanna
- Department of Pediatric Hematology, Oncology and Bone Marrow and Blood Transplant, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
46
|
Yang J, Xu X, Han S, Qi J, Li X, Pan T, Zhang R, Han Y. Comparison of multiple treatments in the management of transplant-related thrombotic microangiopathy: a network meta-analysis. Ann Hematol 2023; 102:31-39. [PMID: 36547721 DOI: 10.1007/s00277-022-05069-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022]
Abstract
Hematopoietic stem cell transplantation-associated thrombotic microangiopathy (TA-TMA) is a fatal post-transplant complication. It has a high mortality rate and worse prognosis, but treatment strategies remain controversial. We screened 6 out of 3453 studies on the treatment of TA-TMA. These investigations compared 5 treatment strategies with a network meta-analysis approach. The final outcome was the proportion of patients who responded to these therapies. There were significant differences in response rates for each treatment. Achieving analysis through direct and indirect evidence in the rank probabilities shows that rTM (recombinant human soluble thrombomodulin) is most likely to be rank 1 (64.98%), Eculizumab intervention rank 2 (48.66%), ISM (immunosuppression manipulation) rank 3 (32.24%), TPE (therapeutic plasma exchange) intervention rank 4 (69.56%), and supportive care intervention rank 5 (70.20%). Eculizumab and ISM have significantly higher efficacy than supportive care (odds ratio (OR): 18.04, 18.21 respectively); and TPE having lower efficacy than all other TA-TMA therapies exception to supportive care. In our study, rTM and Eculizumab may be the best choice when treating TA-TMA.
Collapse
Affiliation(s)
- Jingyi Yang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiaoyan Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Shiyu Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaqian Qi
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Xueqian Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Tingting Pan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Rui Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yue Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China. .,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China. .,Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China. .,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China.
| |
Collapse
|
47
|
Marco DN, Salas MQ, Gutiérrez-García G, Monge I, Riu G, Carcelero E, Roma JR, Llobet N, Arcarons J, Suárez-Lledó M, Martínez N, Pedraza A, Domenech A, Rosiñol L, Fernández-Avilés F, Urbano-Ispízua Á, Rovira M, Brunet M, Martínez C. Impact of Early Intrapatient Variability of Tacrolimus Concentrations on the Risk of Graft-Versus-Host Disease after Allogeneic Stem Cell Transplantation Using High-Dose Post-Transplant Cyclophosphamide. Pharmaceuticals (Basel) 2022; 15:ph15121529. [PMID: 36558980 PMCID: PMC9784628 DOI: 10.3390/ph15121529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Tacrolimus (Tac) is a pivotal immunosuppressant agent used to prevent graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (alloHSCT). Tac is characterized by a narrow therapeutic window and a high inter-patient and intra-patient pharmacokinetic variability (IPV). Although high IPV of Tac concentrations has been associated with adverse post-transplant outcomes following solid organ transplantation, the effects of Tac IPV on alloHSCT recipients have not been determined. Tac IPV was therefore retrospectively evaluated in 128 alloHSCT recipients receiving high-dose post-transplant cyclophosphamide (PTCy) and the effects of Tac IPV on the occurrence of acute GVHD (aGVHD) were analyzed. Tac IPV was calculated from pre-dose concentrations (C0) measured during the first month after Tac initiation. The cumulative rates of grades II-IV and grades III-IV aGVHD at day +100 were 22.7% and 7%, respectively. Higher Tac IPV was associated with a greater risk of developing GVHD, with patients having IPV > 50th percentile having significantly higher rates of grades II-IV (34.9% vs. 10.8%; hazard ratio [HR] 3.858, p < 0.001) and grades III-IV (12.7% vs. 1.5%; HR 9.69, p = 0.033) aGVHD than patients having IPV ≤ 50th percentile. Similarly, patients with IPV > 75th percentile had higher rates of grades II-IV (41.9% vs. 16.5%; HR 3.30, p < 0.001) and grades III-IV (16.1% vs. 4.1%; HR 4.99, p = 0.012) aGVHD than patients with IPV ≤ 75th percentile. Multivariate analyses showed that high Tac IPV (>50th percentile) was an independent risk factor for grades II-IV (HR 2.99, p = 0.018) and grades III-IV (HR 9.12, p = 0.047) aGVHD. Determination of Tac IPV soon after alloHSCT could be useful in identifying patients at greater risk of aGVHD.
Collapse
Affiliation(s)
- Daniel N. Marco
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - María Queralt Salas
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Gonzalo Gutiérrez-García
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Inés Monge
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Gisela Riu
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Esther Carcelero
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Joan Ramón Roma
- Department of Pharmacy, Pharmacy Service, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Noemí Llobet
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Jordi Arcarons
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - María Suárez-Lledó
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Nuria Martínez
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Alexandra Pedraza
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Ariadna Domenech
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Rosiñol
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Francesc Fernández-Avilés
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Álvaro Urbano-Ispízua
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Montserrat Rovira
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Mercè Brunet
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
| | - Carmen Martínez
- Hematopoietic Stem Cell Transplantation Unit, Hematology Department, Institute of Hematology and Oncology, Hospital Clínic, IDIBAPS, 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-227-54-28; Fax: +34-93-227-54-84
| |
Collapse
|
48
|
Radnay Z, Illés Á, Udvardy M, Prohászka Z, Sinkovits G, Csányi MC, Kellermayer M, Kiss A, Hársfalvi J. Von Willebrand Factor and Platelet Levels before Conditioning Chemotherapy Indicate Bone Marrow Regeneration following Autologous Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2022; 28:830.e1-830.e7. [PMID: 36058547 DOI: 10.1016/j.jtct.2022.08.028] [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: 12/07/2021] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022]
Abstract
Autologous hematopoietic stem cell transplantation (HSCT) is often complicated by hemostatic and thrombotic events associated with endothelial cell injury. Thrombotic complications are affected by a disturbed balance between platelets, circulating von Willebrand factor (VWF), and its specific protease, ADAMTS13. HSCT-associated endothelial dysfunction, impaired hemostasis, and inflammation are interrelated processes, and research on the complex interplay of conditioning regimens from engraftment to bone marrow regeneration remains intensive. This prospective observational study comparing lymphoma and multiple myeloma (MM) patients who underwent autologous HSCT explored how platelet count, VWF level, ADAMTS13 activity, and C-reactive protein (CRP) level as potential markers (1) vary in response to therapy, (2) differ between the 2 groups, and (3) correlate with the remission state at 100 days after HSCT. We correlated the quantitative changes in platelet count and levels of VWF, ADAMTS13, and CRP with one another during HSCT and in the remission state in 45 patients with lymphoma and 59 patients with MM who underwent autologous HSCT between 2010 and 2013 at the University of Debrecen. Samples were collected at the start of conditioning chemotherapy, on the day of stem cell transplantation, and at 5, 11, and 100 days following HSCT. CRP levels peaked when platelet counts dropped to a minimum, and these changes were much more pronounced in the lymphoma group. VWF level was the highest, with lower ADAMTS13 activity, at platelet engraftment in both patient groups equally. Diagnostic evidence indicative of thrombotic complications was not found. In the lymphoma group, VWF level prior to conditioning had statistically significant correlations with platelet count, CRP level, and hemoglobin concentration at the time of bone marrow regeneration (P < .001) and during the remission state (P = .034). In the MM group, platelet count before conditioning was correlated with platelet count (P < .001) and white blood cell count (P = .012) at the time of bone marrow regeneration. The statistically significant correlation of the markers at the time of bone marrow regeneration with the preconditioning VWF levels in lymphoma and with the preconditioning platelet counts in MM might indicate the clinical significance of the bone marrow niches of arterioles and megakaryocytes, respectively, where the stem cells are located and regulated. Because preconditioning VWF levels are associated with remission after HSCT in lymphoma patients, VWF should be screened before conditioning, along with the markers used in HSCT protocols, to optimize personalized treatment and reduce therapeutic risks.
Collapse
Affiliation(s)
- Zita Radnay
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Árpád Illés
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Udvardy
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Prohászka
- Research Laboratory, 3rd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - György Sinkovits
- Research Laboratory, 3rd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Mária Csilla Csányi
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Kiss
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Jolán Hársfalvi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Hungary; Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
49
|
Spałek A, Grygoruk-Wiśniowska I, Gruenpeter K, Panz-Klapuch M, Helbig G. Spectacular and Prompt Response to Extracorporeal Photopheresis for Refractory Cutaneous Chronic Graft-Versus-Host Disease after Allogeneic Hematopoietic Stem Cell Transplantation: A Case Report. Medicina (B Aires) 2022; 58:medicina58121722. [PMID: 36556924 PMCID: PMC9787900 DOI: 10.3390/medicina58121722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a serious complication after allogenic hematopoietic stem cell transplantation (allo-HSCT), negatively affecting the morbidity and mortality of recipients. Skin involvement is the most common cGVHD manifestation with a wide range of pleomorphic features, from scleroderma to ulcerations and microangiopathic changes. Despite the access to many immunosuppressive drugs, therapy for cGVHD is challenging. Systemic steroids are recommended as the first-line treatment; but, in steroid-resistant patients, extracorporeal photopheresis (ECP) remains one of the subsequent therapeutic options. Here, we present a case report of a 31-year patient suffering from advanced steroid-refractory skin and oral mucosa cGVHD who was spectacularly treated with ECP. It was the first time we observed such "overnight" resolution of the graft-versus-host disease syndrome. The present report proves the important role of ECP in the treatment of steroid-resistant cGVHD, especially when other immunosuppressive therapies have failed.
Collapse
|
50
|
Michael M, Bagga A, Sartain SE, Smith RJH. Haemolytic uraemic syndrome. Lancet 2022; 400:1722-1740. [PMID: 36272423 DOI: 10.1016/s0140-6736(22)01202-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 11/05/2022]
Abstract
Haemolytic uraemic syndrome (HUS) is a heterogeneous group of diseases that result in a common pathology, thrombotic microangiopathy, which is classically characterised by the triad of non-immune microangiopathic haemolytic anaemia, thrombocytopenia, and acute kidney injury. In this Seminar, different causes of HUS are discussed, the most common being Shiga toxin-producing Escherichia coli HUS. Identifying the underlying thrombotic microangiopathy trigger can be challenging but is imperative if patients are to receive personalised disease-specific treatment. The quintessential example is complement-mediated HUS, which once carried an extremely high mortality but is now treated with anti-complement therapies with excellent long-term outcomes. Unfortunately, the high cost of anti-complement therapies all but precludes their use in low-income countries. For many other forms of HUS, targeted therapies are yet to be identified.
Collapse
Affiliation(s)
- Mini Michael
- Division of Pediatric Nephrology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
| | - Arvind Bagga
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Sarah E Sartain
- Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Richard J H Smith
- Department of Otolaryngology, Pediatrics and Molecular Physiology & Biophysics, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|