1
|
Mlakar V, Dupanloup I, Gloor Y, Ansari M. Haplotype Inference Using Long-Read Nanopore Sequencing: Application to GSTA1 Promoter. Mol Biotechnol 2025; 67:2512-2519. [PMID: 38886308 PMCID: PMC12055866 DOI: 10.1007/s12033-024-01213-7] [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: 03/26/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Recovering true haplotypes can have important clinical consequences. The laboratory process is difficult and is, therefore, most often done through inference. In this paper, we show that when using the Oxford nanopore sequencing technology, we could recover the true haplotypes of the GSTA1 promoter region. Eight LCL cell lines with potentially ambiguous haplotypes were used to characterize the efficacy of Oxford nanopore sequencing to phase the correct GSTA1 promoter haplotypes. The results were compared to Sanger sequencing and inferred haplotypes in the 1000 genomes project. The average read length was 813 bp out of a total PCR length of 1336 bp. The best coverage of sequencing was in the middle of the PCR product and decreased to 50% at the PCR ends. SNPs separated by less than 200 bp showed > 90% of correct haplotypes, while at the distance of 1089 bp, this proportion still exceeded 58%. The number of cycles influences the generation of hybrid haplotypes but not extension or annealing time. The results demonstrate that this long sequencing reads methodology, can accurately determine the haplotypes without the need for inference. The technology proved to be robust but the success of phasing nonetheless depends on the distances and frequencies of SNPs.
Collapse
Affiliation(s)
- Vid Mlakar
- CANSEARCH Research Laboratory, Geneva University Medical School, Rue Michel Servet 1, 1211, Geneva, Switzerland.
| | - Isabelle Dupanloup
- CANSEARCH Research Laboratory, Geneva University Medical School, Rue Michel Servet 1, 1211, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yvonne Gloor
- CANSEARCH Research Laboratory, Geneva University Medical School, Rue Michel Servet 1, 1211, Geneva, Switzerland
| | - Marc Ansari
- CANSEARCH Research Laboratory, Geneva University Medical School, Rue Michel Servet 1, 1211, Geneva, Switzerland
- Onco-Hematology Unit, Pediatric Department, Geneva University Hospital, Rue Willy-Donzé 6, 1205, Geneva, Switzerland
| |
Collapse
|
2
|
Seydoux C, Passweg JR, Heim D, Halter J, Rentsch KM, Medinger M. Busulfan Once versus Four Times daily: Impact on Pharmacokinetics, Organ Toxicities and Survival After Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2025:S2666-6367(25)01158-3. [PMID: 40373977 DOI: 10.1016/j.jtct.2025.04.019] [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: 12/31/2024] [Revised: 04/01/2025] [Accepted: 04/29/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Therapeutic drug monitoring of busulfan (Bu) used as conditioning for allogeneic stem cell transplantation (allo-HSCT) is recommended as pharmacokinetics (PK) display variability. Since 2019, we give Bu 1x/day (Bu-Q24) instead of 4x/day (Bu-Q6) for practical convenience, despite limited studies evaluating the best way of application. OBJECTIVES Our aim was to analyze the correlation between Bu administration (Bu-Q6 versus Bu-Q24), Bu-PK and clinical outcome in 256 adult patients receiving Bu as conditioning regimen for allo-HSCT. STUDY DESIGN This is a retrospective study of adult patients receiving a myeloablative chemotherapeutic regimen containing Bu to treat hematological malignancies. The population was separated into two groups according to Bu administration, namely Bu-Q6 and Bu-Q24. RESULTS A total of 133 patients received Bu-Q6 and 123 Bu-Q24. Bu-Q6 patients were more commonly treated with cyclophosphamide and Bu-Q24 with fludarabin. Bu-Q6 showed lower cumulative area-under-the-curve values than Bu-Q24 (63.78mg*h/L in Bu-Q6 and 70.12mg*h/L in Bu-Q24, p=0.06). Only 44% of the patients fell within the 1st AUC FDA target range in Bu-Q6 versus 62% in Bu-Q24 (p<0.01). Overall, Bu-Q24 appeared to be superior to Bu-Q6 for most outcomes, showing lower incidence of toxicity grade≥II (78% vs. 90%, p=0.02), with less uro-renal (14% in Bu-Q24 vs. 26% in Bu-Q6; p=0.02), pulmonary (2% vs. 8%, p=0.05) and gastro-intestinal toxicities (10% vs. 17%, p<0.01). Patient receiving Bu-Q24 had fewer infections (51% vs. 65%; p=0.04), particularly bacterial (33% vs. 47%, p=0.03) and fungal infections (10% vs. 20%; p=0.03). At 2 years, Bu-Q24 tended to have lower treatment-related mortality (5% vs. 10%, p=0.13), relapse rate (37% vs. 42%, p=0.55) and incidence of acute and chronic graft-versus-host-disease (24% vs. 28%; 32% vs. 36%, respectively). The overall survival was 81% (95% confidence interval (CI) 74%-89%) in Bu-Q24 and 69% (95% CI 62%-77%, p=0.03) in Bu-Q6, without impact on the relapse rate or incidence of acute and chronic graft-versus-host-disease. The only benefit of Bu-Q6 was mucositis grade≥III, with an incidence of 36% versus 60% in Bu-Q24 (p<0.01). At 2 years, patients with a cumulative AUC < 59.11 mg*h/L had the lowest TRM, without impact on the OS. Bu clearance was largely influenced by BMI and age > 60 years. CONCLUSION Bu administered once a day shows benefit both in the short and long term compared to Bu administered four times a day, but data are heterogeneus, Bu-Q24 being more commonly associated with use of fludarabine, Bu-Q6 with use of cyclophosphamide in this study.
Collapse
Affiliation(s)
- Claire Seydoux
- Division of Hematology, University Hospital of Basel, Basel, Switzerland.
| | - Jakob R Passweg
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Dominik Heim
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Joerg Halter
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Katharina M Rentsch
- Department of Laboratory Medicine, University Hospital of Basel, Basel, Switzerland
| | - Michael Medinger
- Division of Hematology, University Hospital of Basel, Basel, Switzerland.
| |
Collapse
|
3
|
Olivo LB, Corrêa GG, Dias BB, Corrêa JARA, Schweinberger BM, do Carmo RL, Daudt LE, Costa TD, de Araujo BV. Validation of an ultra-high performance liquid chromatography/UV method to quantify busulfan in plasma: application to therapeutic drug monitoring. EINSTEIN-SAO PAULO 2025; 23:eAO0964. [PMID: 40105571 PMCID: PMC11975062 DOI: 10.31744/einstein_journal/2025ao0964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/03/2024] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Olivo et al. validated an in-house precise UHPLC/UV method for quantifying busulfan in human plasma for therapeutic monitoring. The method shows linearity (0.5-10 μg/mL) with a lower limit of quantification of 0.5 μg/mL, demonstrating accuracy and precision. It effectively supported therapeutic drug monitoring in a Brazilian public hospital by providing rapid and reliable results. ■ We validated the UHPLC/UV method for accurate busulfan quantification in plasma. ■ Inaccuracy and imprecision were below 15%, ensuring reliable therapeutic drug monitoring results. ■ This enables effective pharmacokinetic studies with rapid turnaround times in patient samples. OBJECTIVE This study aimed to validate a sensitive, accurate, and precise bioanalytical ultra-high-performance liquid chromatography coupled with ultraviolet (UHPLC/UV) method for the determination of busulfan in human plasma using 1,6-bis-(methanesulfonyloxy) hexane as an internal standard for therapeutic drug monitoring. METHODS Plasma samples were deproteinized with acetonitrile (1:2, v/v) and, after derivatization with sodium diethyl dithiocarbamate, submitted to liquid-liquid extraction with ethyl acetate and evaporation at 50ºC under a nitrogen stream. Analyses were performed on a Shimadzu® system using a C18 column and isocratic elution with methanol/water (70:30, v/v) at a flow rate of 0.4mL min-1 and detection at 277nm. RESULTS The retention times of busulfan and the IS were approximately 2.87 and 6.35 min, respectively. The plasma calibration curves were linear in the concentration range of 0.5-10 µg mL-1 with a coefficient of determination greater than 0.99. The lower limit of quantification was 0.5 µg mL-1. The inaccuracies and imprecisions of this method are less than 15%. The applicability of this method to pharmacokinetic studies was confirmed using patient samples obtained after 4 h of 3.2-5.4 mg kg-1 busulfan intermittent infusion. CONCLUSION This method demonstrated the feasibility of quantifying samples within the target concentration range and quickly releasing results to allow for busulfan therapeutic monitoring.
Collapse
Affiliation(s)
- Laura Ben Olivo
- Pharmaceutical Sciences Graduate ProgramFaculdade de FarmáciaUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Gabriel Giron Corrêa
- Hematology and Flow Cytometry UnitHospital de Clínicas de Porto AlegrePorto AlegreRSBrazil Hematology and Flow Cytometry Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
- Graduate Program in Medicine: Medical SciencesUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Bruna Bernar Dias
- Pharmaceutical Sciences Graduate ProgramFaculdade de FarmáciaUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Janaína Aparecida Risczik Arruda Corrêa
- Biochemistry UnitHospital de Clínicas de Porto AlegrePorto AlegreRSBrazil Biochemistry Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Bruna Martins Schweinberger
- Biochemistry UnitHospital de Clínicas de Porto AlegrePorto AlegreRSBrazil Biochemistry Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Raiza Lima do Carmo
- Graduate Program in Medicine: Medical SciencesUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Liane Esteves Daudt
- Hematology and Pediatric Bone Marrow Transplantation ServiceHospital de Clínicas de Porto AlegrePorto AlegreRSBrazil Hematology and Pediatric Bone Marrow Transplantation Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Teresa Dalla Costa
- Pharmaceutical Sciences Graduate ProgramFaculdade de FarmáciaUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Bibiana Verlindo de Araujo
- Pharmaceutical Sciences Graduate ProgramFaculdade de FarmáciaUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Graduate Program in Medicine: Medical SciencesUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
4
|
Ettoury S, Louati S, Saad I, Bentayebi K, Zarrik O, Bourkadi JE, Belyamani L, Daali Y, Eljaoudi R. Association of GST polymorphism with adverse drug reactions: an analysis across multiple drug categories. Expert Opin Drug Metab Toxicol 2025; 21:191-201. [PMID: 39501971 DOI: 10.1080/17425255.2024.2426616] [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: 05/24/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024]
Abstract
INTRODUCTION Adverse drug reactions (ADRs) pose a significant challenge in clinical practice, impacting patient safety and treatment outcomes. Genetic variations in drug-metabolizing enzymes, particularly glutathione S-transferases (GSTs), have been implicated in modulating individual susceptibility to ADRs. AREAS COVERED This overview aims to explore the association between GSTs genetic polymorphisms and ADRs across diverse drug categories documented in current literature. Here we cover antiepileptic, immunosuppressive, chemotherapeutic agents, analgesics, antivirals, and antibiotics. EXPERT OPINION According to the existing literature, the association between genetic polymorphisms in GST theta (GSTT1), GST mu (GSTM1), and GST pi (GSTP1) and adverse drug reaction occurrence has been frequently reported. However, the strength of these associations varies considerably among studies, with some showing inconsistent or contradictory results, underscoring the need for further investigations.
Collapse
Affiliation(s)
- Soukaina Ettoury
- Medical Biotechnology Laboratory, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Sara Louati
- Medical Biotechnology Laboratory, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Ibtissam Saad
- Medical Biotechnology Laboratory, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Kaoutar Bentayebi
- Medical Biotechnology Laboratory, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Oumaima Zarrik
- Medical Biotechnology Laboratory, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Jamal Eddine Bourkadi
- Respiratory Department, Moulay Youssef University Hospital, University of Mohammed V, Rabat, Morocco
| | - Lahcen Belyamani
- Mohammed V Military Teaching Hospital/Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Youssef Daali
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Geneva, Switzerland
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Rachid Eljaoudi
- Medical Biotechnology Laboratory, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| |
Collapse
|
5
|
Indrawijaya YYA, Artarini AA, Hamijoyo L, Iwo MI. GSTA1 gene polymorphisms are associated with cyclophosphamide effectiveness in lupus nephritis patients: A case-control study in Indonesia. NARRA J 2024; 4:e1144. [PMID: 39816074 PMCID: PMC11731938 DOI: 10.52225/narra.v4i3.1144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/14/2024] [Indexed: 01/18/2025]
Abstract
Glutathione-S-transferase alpha-1 (GSTA1) is an enzyme with high conjugation activity against aldophosphamide, a metabolite of cyclophosphamide and promoter polymorphisms in GSTA1 may influence the cyclophosphamide effectiveness. The aim of this study was to evaluate the effectiveness and side effects of cyclophosphamide in lupus nephritis patients, using GSTA1 variants as predictors. A case-control study was conducted at Hasan Sadikin Hospital, Bandung, Indonesia, involving 100 lupus nephritis patients from February 2023 to January 2024. The PCR-Sanger sequencing was used to genotype five selected single nucleotide polymorphisms (SNPs) in the GSTA1 promoter: - 52 A > G, -69 T > C, -513 A > G, -567 G > T, and -631 G > T. The endpoint was assessed after six doses of cyclophosphamide by evaluating renal function, disease activity and side effects. Results indicated that six doses of intravenous cyclophosphamide treatment improved renal function and disease activity in the patients, as evidenced by significant changes in serum creatinine (0.79 vs 0.69 mg/dL), dipstick proteinuria (3.00 vs 1.50), creatinine clearance (98.50 vs 109.50 mL/min), and Modified Systemic Lupus Erythematosus Disease Activity Index 2000 (M-SLEDAI-2 K) score (8.61 vs 6.95). The AG genotype at - 513 A > G was associated with reduced cyclophosphamide effectiveness (odds ratio (OR): 0.19; 95%CI: 0.19-0.60; p = 0.019). The GT genotype at -631 G > T independently increased the progression of anemia (OR: 2.41; 95%CI: 0.26-22.12; p = 0.040). This study highlights that the presence of GSTA1 variants affected cyclophosphamide effectiveness in lupus nephritis patients, with heterozygous polymorphisms at -513 (AA to AG) and -631 (TT to GT) predicting reduced effectiveness of cyclophosphamide by enhancing GSTA1 promoter activity, while anemia further exacerbated lupus nephritis disease severity. GSTA1 polymorphism was not associated with the presence of alopecia, amenorrhea, gastrointestinal disorders, and leukopenia during cyclophosphamide therapy.
Collapse
Affiliation(s)
- Yen YA. Indrawijaya
- Doctoral Program, School of Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
- Department of Pharmacy, Faculty of Medicine and Health Sciences, Universitas Islam Negeri Maulana Malik Ibrahim, Malang, Indonesia
| | - Aluicia A. Artarini
- Laboratory of Pharmaceutical Biotechnology, School of Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
| | - Laniyati Hamijoyo
- Department of Internal Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Immunology Study Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Maria I. Iwo
- Department of Pharmacology-Clinical Pharmacy, School of Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
| |
Collapse
|
6
|
Domingos V, Nezvalova-Henriksen K, Dadkhah A, Moreno-Martinez ME, Ben Hassine K, Pires V, Kröger N, Bauters T, Hassan M, Duncan N, Kalwak K, Ansari M, Langebrake C, Admiraal R. A practical guide to therapeutic drug monitoring in busulfan: recommendations from the Pharmacist Committee of the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant 2024; 59:1641-1653. [PMID: 39271948 DOI: 10.1038/s41409-024-02413-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/01/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Busulfan (Bu) is an important component of many conditioning regimens for allogeneic hematopoietic cell transplantation. The therapeutic window of Bu is well characterized, with strong associations between Bu exposure and the clinical outcome in adults (strongest evidence in myelo-ablative setting) and children (all settings). We provide an overview of the literature on Bu as well as a step-by-step guide to the implementation of Bu therapeutic drug monitoring (TDM). The guide covers the clinical, pharmacological, laboratory and administrative aspects of the procedure. Through this document, we aim to support centers in implementing TDM for Bu to further enhance the success rates of HCT and improve patient outcomes. The Pharmacist Committee of the European Society for Blood and Marrow Transplantation (EBMT) encourages all centers to perform TDM for Bu in the aforementioned indications.
Collapse
Affiliation(s)
- Vera Domingos
- Department of Pharmacy, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | | | - Adrin Dadkhah
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hospital Pharmacy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria-Estela Moreno-Martinez
- Pharmacy Department, Hospital de la Santa Creu I Sant Pau, IIB Sant Pau, Barcelona, Spain
- School of Health Sciences Blanquerna, University Ramon Lull, Barcelona, Spain
| | - Khalil Ben Hassine
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
| | - Vera Pires
- Department of Pharmacy, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tiene Bauters
- Department of Pharmacy, Ghent University Hospital, Ghent, Belgium
| | - Moustapha Hassan
- Experimental Cancer Medicine, Division of Biomolecular and Cellular Medicine (BCM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nick Duncan
- Pharmacy department, Queen Elizabeth Hospital, Birmingham, UK
| | - Krzysztof Kalwak
- Department of Pediatric Hematology, Oncology and BMT, Wroclaw Medical University, Wroclaw, Poland
| | - Marc Ansari
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
| | - Claudia Langebrake
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hospital Pharmacy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rick Admiraal
- Department of Stem Cell Transplantation, Princess Maxima Centre for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
7
|
Castelli S, Thorwarth A, van Schewick C, Wendt A, Astrahantseff K, Szymansky A, Lodrini M, Veldhoen S, Gratopp A, Mall MA, Eggert A, Deubzer HE. Management of Busulfan-Induced Lung Injury in Pediatric Patients with High-Risk Neuroblastoma. J Clin Med 2024; 13:5995. [PMID: 39408056 PMCID: PMC11477708 DOI: 10.3390/jcm13195995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Integrating the cytotoxic drug busulfan into a high-dose chemotherapy regimen prior to autologous hematopoietic stem cell rescue in patients with high-risk neuroblastoma has improved the survival of children battling this deadly disease. Busulfan-induced toxicities can, however, be severe. Here, we describe the diagnosis and successful treatment of acute pulmonary injury by total-body-weight-adjusted busulfan therapy in two children with high-risk neuroblastoma. Case series: Patient 1 developed life-threatening biphasic acute respiratory failure on days +60 and +100 after busulfan therapy, requiring intubation and invasive mechanical ventilation. Despite intensive anti-inflammatory and immunomodulatory therapy, including systemic corticosteroids, topical inhalation regimens, azithromycin, nintedanib and extracorporal photopheresis, patient 1 required extended intensive care measures and non-invasive respiratory support for a total of 20 months. High-resolution computed tomography showed diffuse intra-alveolar and interstitial patterns. Patient 2 developed partial respiratory failure with insufficient oxygen saturation and dyspnea on day +52 after busulfan therapy. Symptoms were resolved after 6 months of systemic corticosteroids, topical inhalation regimens and azithromycin. High-resolution computed tomography showed atypical pneumonic changes with ground-glass opacities. While both patients fully recovered without evidence of pulmonary fibrosis, cancer therapy had to be paused and then modified until full recovery from busulfan-induced lung injury. Conclusions: Busulfan-induced lung injury requires prompt diagnosis and intervention. Symptoms and signs are nonspecific and difficult to differentiate from other causes. Therapeutic busulfan drug level monitoring and the identification of patients at risk for drug overdosing through promoter polymorphisms in the glutathione S-transferase alpha 1 gene encoding the main enzyme in busulfan metabolism are expected to reduce the risk of busulfan-induced toxicities.
Collapse
Affiliation(s)
- Sveva Castelli
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Anne Thorwarth
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Claudia van Schewick
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Anke Wendt
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Annabell Szymansky
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Marco Lodrini
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Simon Veldhoen
- Department of Pediatric Radiology, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Alexander Gratopp
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Lung Research (DLZ), Associated Partner Site Berlin, 89337 Berlin, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, 89337 Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| | - Hedwig E. Deubzer
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of Charité and Max-Delbrück-Center of Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany
- Max-Delbrück Center of Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| |
Collapse
|
8
|
Ben Hassine K, Seydoux C, Khier S, Daali Y, Medinger M, Halter J, Heim D, Chalandon Y, Schanz U, Nair G, Cantoni N, Passweg JR, Satyanarayana Uppugunduri CR, Ansari M. Pharmacokinetic Modeling and Simulation with Pharmacogenetic Insights Support the Relevance of Therapeutic Drug Monitoring for Myeloablative Busulfan Dosing in Adult HSCT. Transplant Cell Ther 2024; 30:332.e1-332.e15. [PMID: 38081414 DOI: 10.1016/j.jtct.2023.12.003] [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: 09/17/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 01/01/2024]
Abstract
Therapeutic drug monitoring (TDM) of busulfan (Bu) is well-established in pediatric hematopoietic stem cell transplantation (HSCT), but its use in adults is limited due to a lack of clear recommendations and scarcity of evidence regarding its utility. GSTA1 promoter variants are reported to affect Bu clearance in both adults and pediatric patients. This study aimed to evaluate the value of preemptive genotyping GSTA1 and body composition (obesity) in individualizing Bu dosing in adults, through pharmacokinetic (PK) modeling and simulations. A population pharmacokinetic (PopPK) model was developed and validated with data from 60 adults who underwent HSCT. Simulations assessed different dosing scenarios based on body size metrics and GSTA1 genotypes. Due to the limited number of obese patients in the cohort, the effect of obesity on Bu pharmacokinetics (PK) was evaluated in silico using a physiologically-based pharmacokinetic (PBPK) model and relevant virtual populations from Simcyp software. Patients with at least 1 GSTA1*B haplotype had 17% lower clearance on average. PopPK simulations indicated that adjusting doses based on genotype increased the probability of achieving the target exposure (3.7 to 5.5 mg.h/L) from 53% to 60 % in GSTA1*A homozygous patients, and from 50% to 61% in *B carriers. Still, Approximately 40% of patients would not achieve this therapeutic window without TDM. A 2-sample optimal design was validated for routine model-based Bu first dose AUC0-∞ estimation, and the model was implemented in the Tucuxi user-friendly TDM software. PBPK simulations confirmed body surface area-based doses of 29 to 31 mg/m2/6h as the most appropriate, regardless of obesity status. This study emphasizes the importance of individualized Bu dosing strategies in adults to achieve therapeutic targets. Preemptive genotyping alone may not have a significant clinical impact, and routine TDM may be necessary for optimal transplantation outcomes.
Collapse
Affiliation(s)
- Khalil Ben Hassine
- Department of Pediatrics, Gynecology and Obstetrics, Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claire Seydoux
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Sonia Khier
- Pharmacokinetic and Modeling Department, School of Pharmacy, Montpellier University, Montpellier, France; Probabilities and Statistics Department, Institut Montpelliérain Alexander Grothendieck (IMAG), CNRS, UMR 5149, Inria, Montpellier University, Montpellier, France
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, University Hospital of Geneva, Geneva, Switzerland; Faculty of Medicine & Sciences, University of Geneva, Geneva, Switzerland
| | - Michael Medinger
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Joerg Halter
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Dominik Heim
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Yves Chalandon
- Division of Hematology, Bone Marrow Transplant Unit, University Hospital of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Urs Schanz
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
| | - Gayathri Nair
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
| | - Nathan Cantoni
- Division of Oncology, Hematology and Transfusion Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Jakob R Passweg
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Chakradhara Rao Satyanarayana Uppugunduri
- Department of Pediatrics, Gynecology and Obstetrics, Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marc Ansari
- Department of Pediatrics, Gynecology and Obstetrics, Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland.
| |
Collapse
|
9
|
Allard J, Bucher S, Ferron PJ, Launay Y, Fromenty B. Busulfan induces steatosis in HepaRG cells but not in primary human hepatocytes: Possible explanations and implication for the prediction of drug-induced liver injury. Fundam Clin Pharmacol 2024; 38:152-167. [PMID: 37665028 DOI: 10.1111/fcp.12951] [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: 01/23/2023] [Revised: 07/27/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND The antineoplastic drug busulfan can induce different hepatic lesions including cholestasis and sinusoidal obstruction syndrome. However, hepatic steatosis has never been reported in patients. OBJECTIVES This study aimed to determine whether busulfan could induce steatosis in primary human hepatocytes (PHH) and differentiated HepaRG cells. METHODS Neutral lipids were determined in PHH and HepaRG cells. Mechanistic investigations were performed in HepaRG cells by measuring metabolic fluxes linked to lipid homeostasis, reduced glutathione (GSH) levels, and expression of genes involved in lipid metabolism and endoplasmic reticulum (ER) stress. Analysis of two previous transcriptomic datasets was carried out. RESULTS Busulfan induced lipid accumulation in HepaRG cells but not in six different batches of PHH. In HepaRG cells, busulfan impaired VLDL secretion, increased fatty acid uptake, and induced ER stress. Transcriptomic data analysis and decreased GSH levels suggested that busulfan-induced steatosis might be linked to the high expression of glutathione S-transferase (GST) isoenzyme A1, which is responsible for the formation of the hepatotoxic sulfonium cation conjugate. In keeping with this, the GST inhibitor ethacrynic acid and the chemical chaperone tauroursodeoxycholic acid alleviated busulfan-induced lipid accumulation in HepaRG cells supporting the role of the sulfonium cation conjugate and ER stress in steatosis. CONCLUSION While the HepaRG cell line is an invaluable tool for pharmacotoxicological studies, it might not be always an appropriate model to predict and mechanistically investigate drug-induced liver injury. Hence, we recommend carrying out toxicological investigations in both HepaRG cells and PHH to avoid drawing wrong conclusions on the potential hepatotoxicity of drugs and other xenobiotics.
Collapse
Affiliation(s)
- Julien Allard
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | | | - Pierre-Jean Ferron
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1317, Rennes, France
| | - Youenn Launay
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1317, Rennes, France
| | - Bernard Fromenty
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1317, Rennes, France
| |
Collapse
|
10
|
Pai AA, Mohanan E, Panetta JC, Kulkarni UP, Illangeswaran RSS, Balakrishnan B, Jayaraman A, Edison ES, Lakshmi KM, Devasia AJ, Fouzia NA, Korula A, Abraham A, George B, Srivastava A, Mathews V, Standing JF, Balasubramanian P. Treosulfan Exposure Predicts Thalassemia-Free Survival in Patients with Beta Thalassemia Major Undergoing Allogeneic Hematopoietic Cell Transplantation. Clin Pharmacol Ther 2024; 115:116-125. [PMID: 37846495 PMCID: PMC7615782 DOI: 10.1002/cpt.3078] [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: 08/07/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
A toxicity-reduced conditioning regimen with treosulfan, fludarabine, and thiotepa in patients with high-risk β-thalassemia major has significantly improved hematopoietic stem cell transplantation (HCT) outcomes. However, complications resulting from regimen-related toxicities (RRTs), mixed chimerism, and graft rejection remain a challenge. We evaluated the dose-exposure-response relationship of treosulfan and its active metabolite S, S-EBDM, in a uniform cohort of patients with β-thalassemia major to identify whether therapeutic drug monitoring (TDM) and dose adjustment of treosulfan is feasible. Plasma treosulfan/S, S-EBDM levels were measured in 77 patients using a validated liquid chromatography with tandem mass spectrometry method, and the pharmacokinetic parameters were estimated using nlmixr2. The influence of treosulfan and S, S-EBDM exposure, and GSTA1/NQO1 polymorphisms on graft rejection, RRTs, chimerism status, and 1-year overall survival (OS), and thalassemia-free survival (TFS) were assessed. We observed that treosulfan exposure was lower in patients with graft rejection than those without (1,655 vs. 2,037 mg•h/L, P = 0.07). Pharmacodynamic modeling analysis to identify therapeutic cutoff revealed that treosulfan exposure ≥1,660 mg•hour/L was significantly associated with better 1-year TFS (97% vs. 81%, P = 0.02) and a trend to better 1-year OS (90% vs. 69%, P = 0.07). Further, multivariate analysis adjusting for known pre-HCT risk factors also revealed treosulfan exposure <1,660 mg•h/L (hazard ratio (HR) = 3.23; 95% confidence interval (CI) = 1.12-9.34; P = 0.03) and GSTA1*B variant genotype (HR = 3.75; 95% CI = 1.04-13.47; P = 0.04) to be independent predictors for inferior 1-year TFS. We conclude that lower treosulfan exposure increases the risk of graft rejection and early transplant-related mortality affecting TFS. As no RRTs were observed with increasing treosulfan exposure, TDM-based dose adjustment could be feasible and beneficial.
Collapse
Affiliation(s)
- Aswin Anand Pai
- Department of Hematology, Christian Medical College, Vellore, India
- Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | | | - John C. Panetta
- Department of Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Uday P. Kulkarni
- Department of Hematology, Christian Medical College, Vellore, India
| | | | | | - Agila Jayaraman
- Department of Hematology, Christian Medical College, Vellore, India
| | - Eunice S. Edison
- Department of Hematology, Christian Medical College, Vellore, India
| | | | - Anup J. Devasia
- Department of Hematology, Christian Medical College, Vellore, India
| | | | - Anu Korula
- Department of Hematology, Christian Medical College, Vellore, India
| | - Aby Abraham
- Department of Hematology, Christian Medical College, Vellore, India
| | - Biju George
- Department of Hematology, Christian Medical College, Vellore, India
| | - Alok Srivastava
- Department of Hematology, Christian Medical College, Vellore, India
| | - Vikram Mathews
- Department of Hematology, Christian Medical College, Vellore, India
| | - Joseph F. Standing
- Infection, Immunity and Inflammation, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Pharmacy, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | | |
Collapse
|
11
|
Seydoux C, Uppugunduri CRS, Medinger M, Nava T, Halter J, Heim D, Chalandon Y, Schanz U, Nair G, Cantoni N, Passweg JR, Ansari M. Effect of pharmacokinetics and pharmacogenomics in adults with allogeneic hematopoietic cell transplantation conditioned with Busulfan. Bone Marrow Transplant 2023; 58:811-816. [PMID: 37085674 PMCID: PMC10325946 DOI: 10.1038/s41409-023-01963-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/04/2023] [Accepted: 03/17/2023] [Indexed: 04/23/2023]
Abstract
Busulfan (Bu) combined with cyclophosphamide (Cy) is commonly used as a myeloablative conditioning regimen for allogeneic hematopoietic cell transplantation (allo-HCT). There is inter-individual variability of Bu pharmacokinetics (PK) and hence in toxicity and efficacy. The introduction of therapeutic drug monitoring (TDM) of Bu has decreased toxicity of the regimen. Hepatic metabolism of Bu is mediated through Glutathione-S-Transferases (GSTs), mainly GSTA1. Patients with GSTA1*A variants are considered normal metabolizers and GSTA1*B corresponds to poor metabolism, defined by nucleotide changes at -52 or -69 locus in GSTA1 promoter region. The aim of the study was to explore the correlation between GSTA1 polymorphisms and Bu-PK in 60 adult patients receiving an allo-HCT in the BuCyBu clinical study (ClinicalTrials.gov I, ID NCT01779882) comparing the sequence BuCy to CyBu. DNA samples prior to conditioning were genotyped for candidate variants at -52 (rs3957356) and -69 (rs3957357) loci in the GSTA1 promoter. Thirty-three % of patients were GSTA1*A*A, 49% GSTA1*A*B and 18% GSTA1*B*B. In GSTA1*A*A patients, median Bu-AUC was 3.6 ± 0.7 mg*h/L, in GSTA1*A*B 4.5 ± 1.6 and in GSTA1*B*B 4.9 ± 1.4 (AUC 35% higher than GSTA1*A*A, p = 0.03), with a similar significant correlation with Bu-clearance (p = 0.04). The correlation between GSTA1 polymorphism and AUC remained significant in multivariate linear regression analysis. There was a trend for lower non-relapse mortality (NRM) in patients with low AUC. We could not demonstrate a correlation between GSTA1 polymorphisms and NRM, acute graft-versus-host disease (aGvHD) in this small cohort, but there is a trend of higher aGvHD incidence in GSTA1*B*B patients.
Collapse
Affiliation(s)
- Claire Seydoux
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland.
| | - Chakradhara Rao Satyanarayana Uppugunduri
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Michael Medinger
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Tiago Nava
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Joerg Halter
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Dominik Heim
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Yves Chalandon
- Division of Hematology, Bone Marrow Transplant Unit, University Hospital of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Urs Schanz
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
| | - Gayathri Nair
- Department of Medical Oncology and Hematology, University Hospital of Zurich, Zurich, Switzerland
| | - Nathan Cantoni
- Division of Oncology, Hematology and Transfusion Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Jakob R Passweg
- Division of Hematology, University Hospital of Basel, Basel, Switzerland and University Basel, Basel, Switzerland
| | - Marc Ansari
- Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
12
|
Combarel D, Tran J, Delahousse J, Vassal G, Paci A. Individualizing busulfan dose in specific populations and evaluating the risk of pharmacokinetic drug-drug interactions. Expert Opin Drug Metab Toxicol 2023; 19:75-90. [PMID: 36939456 DOI: 10.1080/17425255.2023.2192924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
INTRODUCTION Busulfan is an alkylating agent widely used in the conditioning of hematopoietic stem cell transplantation possessing a complex metabolism and a large interindividual and intra-individual variability, especially in children. Combined with the strong rationale of busulfan PK/PD relationships, factors altering its clearance (e.g., weight, age, and GST-A genetic polymorphism mainly) can also affect clinical outcomes. AREAS COVERED This review aims to provide an overview of the current knowledge on busulfan pharmacokinetics, its pharmacokinetics variabilities in pediatric populations, drug-drug interactions (DDI), and their consequences regarding dose individualization. This review was based on medical literature up until October 2021. EXPERT OPINION To ensure effective busulfan exposure in pediatrics, different weight-based nomograms have been established to determine busulfan dosage and provided improved results (65 - 80% of patients correctly exposed). In addition to nomograms, therapeutic drug monitoring (TDM) of busulfan measuring plasmatic concentrations to estimate busulfan pharmacokinetic parameters can be used. TDM is now widely carried out in routine practices and aims to ensure the targeting of the reported therapeutic windows by individualizing busulfan dosing based on the clearance estimations from a previous dose.
Collapse
Affiliation(s)
- David Combarel
- Service de Pharmacologie, Département de biologie et pathologie médicale, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Université Paris-Saclay, Faculté de Pharmacie, Université Paris-Saclay, Chatenay-Malabry, France
| | - Julie Tran
- Service de Pharmacologie, Département de biologie et pathologie médicale, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Julia Delahousse
- Service de Pharmacologie, Département de biologie et pathologie médicale, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Gilles Vassal
- Gustave Roussy Comprehensive Cancer Center, & University Paris-Saclay, Villejuif, France
| | - Angelo Paci
- Service de Pharmacologie, Département de biologie et pathologie médicale, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Université Paris-Saclay, Faculté de Pharmacie, Université Paris-Saclay, Chatenay-Malabry, France
| |
Collapse
|
13
|
Balakrishnan B, Kulkarni UP, Pai AA, Illangeswaran RSS, Mohanan E, Mathews V, George B, Balasubramanian P. Biomarkers for early complications post hematopoietic cell transplantation: Insights and challenges. Front Immunol 2023; 14:1100306. [PMID: 36817455 PMCID: PMC9932777 DOI: 10.3389/fimmu.2023.1100306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Hematopoietic cell transplantation is an established curative treatment option for various hematological malignant, and non-malignant diseases. However, the success of HCT is still limited by life-threatening early complications post-HCT, such as Graft Versus Host Disease (GVHD), Sinusoidal Obstruction Syndrome (SOS), and transplant-associated microangiopathy, to name a few. A decade of research in the discovery and validation of novel blood-based biomarkers aims to manage these early complications by using them for diagnosis or prognosis. Advances in this field have also led to predictive biomarkers to identify patients' likelihood of response to therapy. Although biomarkers have been extensively evaluated for different complications, these are yet to be used in routine clinical practice. This review provides a detailed summary of various biomarkers for individual early complications post-HCT, their discovery, validation, ongoing clinical trials, and their limitations. Furthermore, this review also provides insights into the biology of biomarkers and the challenge of obtaining a universal cut-off value for biomarkers.
Collapse
Affiliation(s)
- Balaji Balakrishnan
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | | | - Aswin Anand Pai
- Department of Haematology, Christian Medical College, Vellore, India
| | | | | | - Vikram Mathews
- Department of Haematology, Christian Medical College, Vellore, India
| | - Biju George
- Department of Haematology, Christian Medical College, Vellore, India
| | | |
Collapse
|
14
|
Narendra G, Choudhary S, Raju B, Verma H, Silakari O. Role of Genetic Polymorphisms in Drug-Metabolizing Enzyme-Mediated Toxicity and Pharmacokinetic Resistance to Anti-Cancer Agents: A Review on the Pharmacogenomics Aspect. Clin Pharmacokinet 2022; 61:1495-1517. [PMID: 36180817 DOI: 10.1007/s40262-022-01174-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2022] [Indexed: 01/31/2023]
Abstract
The inter-individual differences in cancer susceptibility are somehow correlated with the genetic differences that are caused by the polymorphisms. These genetic variations in drug-metabolizing enzymes/drug-inactivating enzymes may negatively or positively affect the pharmacokinetic profile of chemotherapeutic agents that eventually lead to pharmacokinetic resistance and toxicity against anti-cancer drugs. For instance, the CYP1B1*3 allele is associated with CYP1B1 overexpression and consequent resistance to a variety of taxanes and platins, while 496T>G is associated with lower levels of dihydropyrimidine dehydrogenase, which results in severe toxicities related to 5-fluorouracil. In this context, a pharmacogenomics approach can be applied to ascertain the role of the genetic make-up in a person's response to any drug. This approach collectively utilizes pharmacology and genomics to develop effective and safe medications that are devoid of resistance problems. In addition, recently reported genomics studies revealed the impact of many single nucleotide polymorphisms in tumors. These studies emphasized the importance of single nucleotide polymorphisms in drug-metabolizing enzymes on the effect of anti-tumor drugs. In this review, we discuss the pharmacogenomics aspect of polymorphisms in detail to provide an insight into the genetic manipulations in drug-metabolizing enzymes that are responsible for pharmacokinetic resistance or toxicity against well-known anti-cancer drugs. Special emphasis is placed on different deleterious single nucleotide polymorphisms and their effect on pharmacokinetic resistance. The information provided in this report may be beneficial to researchers, especially those who are working in the field of biotechnology and human genetics, in rationally manipulating the genetic information of patients with cancer who are undergoing chemotherapy to avoid the problem of pharmacokinetic resistance/toxicity associated with drug-metabolizing enzymes.
Collapse
Affiliation(s)
- Gera Narendra
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, 147002, Patiala, Punjab, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, 147002, Patiala, Punjab, India
| | - Baddipadige Raju
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, 147002, Patiala, Punjab, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, 147002, Patiala, Punjab, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, 147002, Patiala, Punjab, India.
| |
Collapse
|
15
|
Teslenko I, Trudeau J, Luo S, Watson CJW, Chen G, Truica CI, Lazarus P. Influence of Glutathione-S-Transferase A1*B Allele on the Metabolism of the Aromatase Inhibitor, Exemestane, in Human Liver Cytosols and in Patients Treated With Exemestane. J Pharmacol Exp Ther 2022; 382:327-334. [PMID: 35793834 PMCID: PMC9426760 DOI: 10.1124/jpet.122.001232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/21/2022] [Indexed: 11/22/2022] Open
Abstract
Exemestane (EXE) is used to treat postmenopausal women diagnosed with estrogen receptor positive (ER+) breast cancer. A major mode of metabolism of EXE and its active metabolite, 17β-dihydroexemestane, is via glutathionylation by glutathione-S-transferase (GST) enzymes. The goal of the present study was to investigate the effects of genetic variation in EXE-metabolizing GST enzymes on overall EXE metabolism. Ex vivo assays examining human liver cytosols from 75 subjects revealed the GSTA1 *B*B genotype was associated with significant decreases in S-(androsta-1,4-diene-3,17-dion-6α-ylmethyl)-L-glutathione (P = 0.034) and S-(androsta-1,4-diene-17β-ol-3-on-6α-ylmethyl)-L-gutathione (P = 0.014) formation. In the plasma of 68 ER+ breast cancer patients treated with EXE, the GSTA1 *B*B genotype was associated with significant decreases in both EXE-cysteine (cys) (29%, P = 0.0056) and 17β-DHE-cys (34%, P = 0.032) as compared with patients with the GSTA1*A*A genotype, with significant decreases in EXE-cys (Ptrend = 0.0067) and 17β-DHE-cys (Ptrend = 0.028) observed in patients with increasing numbers of the GSTA1*B allele. A near-significant (Ptrend = 0.060) trend was also observed for urinary EXE-cys levels from the same patients. In contrast, plasma and urinary 17β-DHE-Gluc levels were significantly increased (36%, P = 0.00097 and 52%, P = 0.0089; respectively) in patients with the GSTA1 *B*B genotype. No significant correlations were observed between the GSTM1 null genotype and EXE metabolite levels. These data suggest that the GSTA1*B allele is associated with interindividual differences in EXE metabolism and may play a role in interindividual variability in overall response to EXE. SIGNIFICANCE STATEMENT: The present study is the first comprehensive pharmacogenomic investigation examining the role of genetic variability in GST enzymes on exemestane metabolism. The GSTA1 *B*B genotype was found to contribute to interindividual differences in the metabolism of EXE both ex vivo and in clinical samples from patients taking EXE for the treatment of ER+ breast cancer. Since GSTA1 is a major hepatic phase II metabolizing enzyme in EXE metabolism, the GSTA1*B allele may be an important biomarker for treatment outcomes and toxicities.
Collapse
Affiliation(s)
- Irina Teslenko
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (I.T., J.T., S.L., C.J.W.W., G.C., P.L.) and Penn State University, College of Medicine, Division of Hematology and Oncology, Department of Medicine, Hershey, Pennsylvania (C.I.T.)
| | - Julia Trudeau
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (I.T., J.T., S.L., C.J.W.W., G.C., P.L.) and Penn State University, College of Medicine, Division of Hematology and Oncology, Department of Medicine, Hershey, Pennsylvania (C.I.T.)
| | - Shaman Luo
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (I.T., J.T., S.L., C.J.W.W., G.C., P.L.) and Penn State University, College of Medicine, Division of Hematology and Oncology, Department of Medicine, Hershey, Pennsylvania (C.I.T.)
| | - Christy J W Watson
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (I.T., J.T., S.L., C.J.W.W., G.C., P.L.) and Penn State University, College of Medicine, Division of Hematology and Oncology, Department of Medicine, Hershey, Pennsylvania (C.I.T.)
| | - Gang Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (I.T., J.T., S.L., C.J.W.W., G.C., P.L.) and Penn State University, College of Medicine, Division of Hematology and Oncology, Department of Medicine, Hershey, Pennsylvania (C.I.T.)
| | - Cristina I Truica
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (I.T., J.T., S.L., C.J.W.W., G.C., P.L.) and Penn State University, College of Medicine, Division of Hematology and Oncology, Department of Medicine, Hershey, Pennsylvania (C.I.T.)
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (I.T., J.T., S.L., C.J.W.W., G.C., P.L.) and Penn State University, College of Medicine, Division of Hematology and Oncology, Department of Medicine, Hershey, Pennsylvania (C.I.T.)
| |
Collapse
|
16
|
Du X, Huang C, Xue L, Jiao Z, Zhu M, Li J, Lu J, Xiao P, Zhou X, Mao C, Zhu Z, Dong J, Liu X, Chen Z, Zhang S, Ding Y, Hu S, Miao L. The Correlation Between Busulfan Exposure and Clinical Outcomes in Chinese Pediatric Patients: A Population Pharmacokinetic Study. Front Pharmacol 2022; 13:905879. [PMID: 35784763 PMCID: PMC9243314 DOI: 10.3389/fphar.2022.905879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/31/2022] [Indexed: 11/21/2022] Open
Abstract
Aims: The aims of the study were to 1) establish a population pharmacokinetic (Pop-PK) model for busulfan in Chinese pediatric patients undergoing hematopoietic stem cell transplantation (HSCT) and then estimate busulfan exposure and 2) explore the association between busulfan exposure and clinical outcomes. Methods: A total of 128 patients with 467 busulfan concentrations were obtained for Pop-PK modeling using nonlinear mixed effect model (NONMEM) software. Sixty-three patients who received the 16-dose busulfan conditioning regimen were enrolled to explore the correlations between clinical outcomes and the busulfan area under the concentration–time curve (AUC) using the Cox proportional hazards regression model, Kaplan–Meier method and logistic regression. Results: The typical values for clearance (CL) and distribution volume (V) of busulfan were 7.71 L h−1 and 42.4 L, respectively. The allometric normal fat mass (NFM) and maturation function (Fmat) can be used to describe the variability in CL, and the fat-free mass (FFM) can be used to describe the variability in V. Patients with AUCs of 950–1,600 µM × min had 83.7% (95% CI: 73.3–95.5) event-free survival (EFS) compared with 55.0% (95% CI: 37.0–81.8) for patients with low or high exposure (p = 0.024). The logistic regression analysis results showed no association between transplant-related toxicities and the busulfan AUC (p > 0.05). Conclusions: The variability in busulfan CL was related to the NFM and Fmat, while busulfan V was related to the FFM. Preliminary analysis results suggested that a busulfan AUC of 950–1,600 µM × min was associated with better EFS in children receiving the 16-dose busulfan regimen.
Collapse
Affiliation(s)
- Xiaohuan Du
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Pharmacy, The Children’s Hospital of Soochow University, Suzhou, China
| | - Chenrong Huang
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Ling Xue
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Min Zhu
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Li
- Department of Hematology and Oncology, The Children’s Hospital of Soochow University, Suzhou, China
| | - Jun Lu
- Department of Hematology and Oncology, The Children’s Hospital of Soochow University, Suzhou, China
| | - Peifang Xiao
- Department of Hematology and Oncology, The Children’s Hospital of Soochow University, Suzhou, China
| | - Xuemei Zhou
- Department of Hematology and Oncology, The Children’s Hospital of Soochow University, Suzhou, China
| | - Chenmei Mao
- Department of Pharmacy, The Children’s Hospital of Soochow University, Suzhou, China
| | - Zengyan Zhu
- Department of Pharmacy, The Children’s Hospital of Soochow University, Suzhou, China
| | - Ji Dong
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoxue Liu
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiyao Chen
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shichao Zhang
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiduo Ding
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shaoyan Hu
- Department of Hematology and Oncology, The Children’s Hospital of Soochow University, Suzhou, China
- *Correspondence: Liyan Miao, ; Shaoyan Hu,
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Science, Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Liyan Miao, ; Shaoyan Hu,
| |
Collapse
|
17
|
Seydoux C, Battegay R, Halter J, Heim D, Rentsch KM, Passweg JR, Medinger M. Impact of busulfan pharmacokinetics on outcome in adult patients receiving an allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2022; 57:903-910. [PMID: 35361896 PMCID: PMC9200635 DOI: 10.1038/s41409-022-01641-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 03/02/2022] [Accepted: 03/11/2022] [Indexed: 11/09/2022]
Abstract
Busulfan (Bu) is widely used in conditioning regimens before allogeneic hematopoietic cell transplantation, with variable metabolism due to interindividual differences of pharmacokinetics (PK). The purpose of this study was to correlate pharmacokinetics and clinical outcomes. Lower-AUC, in range-AUC and higher-AUC were defined as ±25% of the targeted Bu-AUC. In 2019, we changed Bu dosing from 4×/day (Bu-4) to 1×/day (Bu-1) for ease of application. AUC-target range was reached in 46% of patients; 40% were in low-AUC and 14% in high-AUC. Among all toxicities, viral and fungal infections were significantly more frequent in high-AUC compared with low-AUC (20% vs. 8%; p = 0.01 and 37% vs. 17%; p = 0.03). Bu-1 showed lower PK values (66% vs. 36% of Bu-4 in low-AUC; p < 0.01) and higher incidence of mucositis (p = 0.02). Long-term outcomes at 2 years showed a higher non-relapse mortality (NRM) (p < 0.01) and higher relative risk of death in the high-AUC group compared to the other groups. Cumulative incidence of relapse and acute/chronic GvHD were not significantly different. The optimal cut-off in Bu-AUC associated with low NRM was 969 µmol/l*min (ROC AUC 0.67, sensitivity 0.86 and specificity 0.47) for Bu-4. In conclusion, low-AUC BU-PK seems of benefit regarding NRM and survival.
Collapse
Affiliation(s)
- Claire Seydoux
- Division of Hematology, University Hospital of Basel, Basel, Switzerland.
| | - Raphael Battegay
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Joerg Halter
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Dominik Heim
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Katharina M Rentsch
- Department of Laboratory Medicine, University Hospital of Basel, Basel, Switzerland
| | - Jakob R Passweg
- Division of Hematology, University Hospital of Basel, Basel, Switzerland
| | - Michael Medinger
- Division of Hematology, University Hospital of Basel, Basel, Switzerland.
| |
Collapse
|
18
|
Dadkhah A, Wicha SG, Kröger N, Müller A, Pfaffendorf C, Riedner M, Badbaran A, Fehse B, Langebrake C. Population Pharmacokinetics of Busulfan and Its Metabolite Sulfolane in Patients with Myelofibrosis Undergoing Hematopoietic Stem Cell Transplantation. Pharmaceutics 2022; 14:pharmaceutics14061145. [PMID: 35745718 PMCID: PMC9229330 DOI: 10.3390/pharmaceutics14061145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
For patients with myelofibrosis, allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains the only curative treatment to date. Busulfan-based conditioning regimens are commonly used, although high inter-individual variability (IIV) in busulfan drug exposure makes individual dose selection challenging. Since data regarding the IIV in patients with myelofibrosis are sparse, this study aimed to develop a population pharmacokinetic (PopPK) model of busulfan and its metabolite sulfolane in patients with myelofibrosis. The influence of patient-specific covariates on the pharmacokinetics of drug and metabolite was assessed using non-linear mixed effects modeling in NONMEM®. We obtained 523 plasma concentrations of busulfan and its metabolite sulfolane from 37 patients with myelofibrosis. The final model showed a population clearance (CL) and volume of distribution (Vd) of 0.217 L/h/kg and 0.82 L/kg for busulfan and 0.021 L/h/kg and 0.65 L/kg for its metabolite. Total body weight (TBW) and a single-nucleotide polymorphism of glutathione-S-transferase A1 (GSTA1 SNP) displayed a significant impact on volume of distribution and metabolite clearance, respectively. This is the first PopPK-model developed to describe busulfan’s pharmacokinetics in patients with myelofibrosis. Incorporating its metabolite sulfolane into the model not only allowed the characterization of the covariate relationship between GSTA1 and the clearance of the metabolite but also improved the understanding of busulfan’s metabolic pathway.
Collapse
Affiliation(s)
- Adrin Dadkhah
- Hospital Pharmacy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
- Correspondence: ; Tel.: +49-40-7410-58517
| | - Sebastian Georg Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany; (S.G.W.); (C.P.)
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (N.K.); (A.B.); (B.F.)
| | - Alexander Müller
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Christoph Pfaffendorf
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany; (S.G.W.); (C.P.)
| | - Maria Riedner
- Technology Platform Mass Spectrometry, University of Hamburg, 20146 Hamburg, Germany;
| | - Anita Badbaran
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (N.K.); (A.B.); (B.F.)
| | - Boris Fehse
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (N.K.); (A.B.); (B.F.)
| | - Claudia Langebrake
- Hospital Pharmacy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (N.K.); (A.B.); (B.F.)
| |
Collapse
|
19
|
Nijstad AL, Barnett S, Lalmohamed A, Bérénos IM, Parke E, Carruthers V, Tweddle DA, Kong J, Zwaan CM, Huitema ADR, Veal GJ. Clinical pharmacology of cytotoxic drugs in neonates and infants: Providing evidence-based dosing guidance. Eur J Cancer 2022; 164:137-154. [PMID: 34865945 PMCID: PMC8914347 DOI: 10.1016/j.ejca.2021.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/11/2021] [Accepted: 11/01/2021] [Indexed: 01/29/2023]
Abstract
Cancer in neonates and infants is a rare but challenging entity. Treatment is complicated by marked physiological changes during the first year of life, excess rates of toxicity, mortality, and late effects. Dose optimisation of chemotherapeutics may be an important step to improving outcomes. Body size-based dosing is used for most anticancer drugs used in infants. However, dose regimens are generally not evidence based, and dosing strategies are frequently inconsistent between tumour types and treatment protocols. In this review, we collate available pharmacological evidence supporting dosing regimens in infants for a wide range of cytotoxic drugs. A systematic review was conducted, and available data ranked by a level of evidence (1-5) and a grade of recommendation (A-D) provided on a consensus basis, with recommended dosing approaches indicated as appropriate. For 9 of 29 drugs (busulfan, carboplatin, cyclophosphamide, daunorubicin, etoposide, fludarabine, isotretinoin, melphalan and vincristine), grade A was scored, indicating sufficient pharmacological evidence to recommend a dosing algorithm for infants. For busulfan and carboplatin, sufficient data were available to recommend therapeutic drug monitoring in infants. For eight drugs (actinomycin D, blinatumomab, dinutuximab, doxorubicin, mercaptopurine, pegaspargase, thioguanine and topotecan), some pharmacological evidence was available to guide dosing (graded as B). For the remaining drugs, including commonly used agents such as cisplatin, cytarabine, ifosfamide, and methotrexate, pharmacological evidence for dosing in infants was limited or non-existent: grades C and D were scored for 10 and 2 drugs, respectively. The review provides clinically relevant evidence-based dosing guidance for cytotoxic drugs in neonates and infants.
Collapse
Affiliation(s)
- A Laura Nijstad
- Department of Clinical Pharmacy, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands.
| | - Shelby Barnett
- Newcastle University Centre for Cancer, Newcastle University, NE2 4HH Newcastle Upon Tyne, UK
| | - Arief Lalmohamed
- Department of Clinical Pharmacy, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands; Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands
| | - Inez M Bérénos
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands
| | - Elizabeth Parke
- Newcastle University Centre for Cancer, Newcastle University, NE2 4HH Newcastle Upon Tyne, UK
| | - Vickyanne Carruthers
- Newcastle University Centre for Cancer, Newcastle University, NE2 4HH Newcastle Upon Tyne, UK
| | - Deborah A Tweddle
- Newcastle University Centre for Cancer, Newcastle University, NE2 4HH Newcastle Upon Tyne, UK; Great North Children's Hospital, NE1 4LP Newcastle Upon Tyne, UK
| | - Jordon Kong
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Alwin D R Huitema
- Department of Clinical Pharmacy, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands; Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Gareth J Veal
- Newcastle University Centre for Cancer, Newcastle University, NE2 4HH Newcastle Upon Tyne, UK.
| |
Collapse
|
20
|
Effect of GSTA1 Variants on Busulfan-Based Conditioning Regimen Prior to Allogenic Hematopoietic Stem-Cell Transplantation in Pediatric Asians. Pharmaceutics 2022; 14:pharmaceutics14020401. [PMID: 35214132 PMCID: PMC8880478 DOI: 10.3390/pharmaceutics14020401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Busulfan is widely used as a chemotherapy treatment before hematopoietic stem-cell transplantation (HSCT). However, the response of busulfan is highly variable and unpredictable, whereby the pharmacogenetic interference of glutathione S-transferase (GST) has strong evidence in Caucasians and some adult Asians but not in pediatric Asian patients. This study was aimed at investigating the associations of GST genetic polymorphisms with variations in the pharmacokinetic (PK) properties of busulfan in pediatric Asian patients. This retrospective cohort study recruited 92 pediatric patients. The polymorphism of GSTA1 was genotyped by Sanger sequencing, and GSTM1 and GSTP1 were genotyped by real-time PCR. Drug concentration and PK estimation were identified using an LC-MS/MS method and a noncompartmental model. Statistical analysis was performed by R software. Out of 92 patients, 48 (53%) were males, the mean age was 8.4 ± 5.12 years old, and the average weight was 26.52 ± 14.75 kg. The allele frequencies of GSTA1*B and of GSTM1 and GSTP1* deletions were 16.9%, 68.5%, and 21.2%, respectively. Patients with GSTA1*B had a statistically significant impact on the PK of busulfan, whereas those with GSTM1 and GSTP1 did not (p > 0.05). The carriers of GSTA1*B showed a significant difference compared to noncarriers in terms of t1/2 (for first dose: 161.9 vs. 134.3 min, p = 0.0016; for second dose: 156.1 vs. 129.8, p = 0.012), CL (88.74 vs. 124.23 mL/min, p = 0.0089), Cmax (4232.6 vs. 3675.5 ng/mL, p = 0.0021), and AUC (5310.6 vs. 4177.1 µM/min, p = 0.00033). The augmentation of AUC was around 27.1% in patients carrying the GSTA1*B variant. The GSTA1 polymorphism was significantly associated with variations of the pharmacokinetic properties of busulfan treatment in pediatric Asian patients.
Collapse
|
21
|
Waespe N, Strebel S, Nava T, Uppugunduri CRS, Marino D, Mattiello V, Otth M, Gumy-Pause F, Von Bueren AO, Baleydier F, Mader L, Spoerri A, Kuehni CE, Ansari M. Cohort-based association study of germline genetic variants with acute and chronic health complications of childhood cancer and its treatment: Genetic Risks for Childhood Cancer Complications Switzerland (GECCOS) study protocol. BMJ Open 2022; 12:e052131. [PMID: 35074812 PMCID: PMC8788194 DOI: 10.1136/bmjopen-2021-052131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Childhood cancer and its treatment may lead to various health complications. Related impairment in quality of life, excess in deaths and accumulated healthcare costs are relevant. Genetic variations are suggested to contribute to the wide inter-individual variability of complications but have been used only rarely to risk-stratify treatment and follow-up care. This study aims to identify germline genetic variants associated with acute and late complications of childhood cancer. METHODS AND ANALYSIS The Genetic Risks for Childhood Cancer Complications Switzerland (GECCOS) study is a nationwide cohort study. Eligible are patients and survivors who were diagnosed with childhood cancers or Langerhans cell histiocytosis before age 21 years, were registered in the Swiss Childhood Cancer Registry (SCCR) since 1976 and have consented to the Paediatric Biobank for Research in Haematology and Oncology, Geneva, host of the national Germline DNA Biobank Switzerland for Childhood Cancer and Blood Disorders (BISKIDS).GECCOS uses demographic and clinical data from the SCCR and the associated Swiss Childhood Cancer Survivor Study. Clinical outcome data consists of organ function testing, health conditions diagnosed by physicians, second primary neoplasms and self-reported information from participants. Germline genetic samples and sequencing data are collected in BISKIDS. We will perform association analyses using primarily whole-exome or whole-genome sequencing to identify genetic variants associated with specified health conditions. We will use clustering and machine-learning techniques and assess multiple health conditions in different models. DISCUSSION GECCOS will improve knowledge of germline genetic variants associated with childhood cancer-associated health conditions and help to further individualise cancer treatment and follow-up care, potentially resulting in improved efficacy and reduced side effects. ETHICS AND DISSEMINATION The Geneva Cantonal Commission for Research Ethics has approved the GECCOS study.Research findings will be disseminated through national and international conferences, publications in peer-reviewed journals and in lay language online. TRIAL REGISTRATION NUMBER NCT04702321.
Collapse
Affiliation(s)
- Nicolas Waespe
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Paediatrics, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Sven Strebel
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences (GHS), University of Bern, Bern, Switzerland
| | - Tiago Nava
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Women, Children, and Adolescents, University Hospitals of Geneva, Geneve, Switzerland
| | - Chakradhara Rao S Uppugunduri
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Denis Marino
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Veneranda Mattiello
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Women, Children, and Adolescents, University Hospitals of Geneva, Geneve, Switzerland
| | - Maria Otth
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Division of Oncology-Hematology, Department of Pediatrics, Kantonsspital Aarau AG, Aarau, Switzerland
| | - Fabienne Gumy-Pause
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Women, Children, and Adolescents, University Hospitals of Geneva, Geneve, Switzerland
| | - André O Von Bueren
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Women, Children, and Adolescents, University Hospitals of Geneva, Geneve, Switzerland
| | - Frederic Baleydier
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Women, Children, and Adolescents, University Hospitals of Geneva, Geneve, Switzerland
| | - Luzius Mader
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Adrian Spoerri
- SwissRDL - Medical Registries and Data Linkage, Institute of Social and Preventive Medicine, Universitat Bern, Bern, Switzerland
| | - Claudia E Kuehni
- Childhood Cancer Research Group, Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Paediatrics, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Marc Ansari
- CANSEARCH Research Platform for Paediatric Oncology and Haematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Paediatric Oncology and Haematology, Department of Women, Children, and Adolescents, University Hospitals of Geneva, Geneve, Switzerland
| |
Collapse
|
22
|
Robin S, Hassine KB, Muthukumaran J, Jurkovic Mlakar S, Krajinovic M, Nava T, Uppugunduri CRS, Ansari M. A potential implication of UDP-glucuronosyltransferase 2B10 in the detoxification of drugs used in pediatric hematopoietic stem cell transplantation setting: an in silico investigation. BMC Mol Cell Biol 2022; 23:5. [PMID: 35062878 PMCID: PMC8781437 DOI: 10.1186/s12860-021-00402-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022] Open
Abstract
Background Sinusoidal occlusion syndrome (SOS) is a potentially severe complication following hematopoietic stem cell transplantation (HSCT) in pediatric patients. Treatment related risk factors such as intensity of conditioning, hepatotoxic co-medication and patient related factors such as genetic variants predispose individuals to develop SOS. The variant allele for SNP rs17146905 in UDP-glucuronosyl transferase 2B10 (UGT2B10) gene was correlated with the occurrence of SOS in an exome-wide association study. UGT2B10 is a phase II drug metabolizing enzyme involved in the N-glucuronidation of tertiary amine containing drugs. Methods To shed light on the functionality of UGT2B10 enzyme in the metabolism of drugs used in pediatric HSCT setting, we performed in silico screening against custom based library of putative ligands. First, a list of potential substrates for in silico analysis was prepared using a systematic consensus-based strategy. The list comprised of drugs and their metabolites used in pediatric HSCT setting. The three-dimensional structure of UGT2B10 was not available from the Research Collaboratory Structural Bioinformatics - Protein Data Bank (RCSB - PDB) repository and thus we predicted the first human UGT2B10 3D model by using multiple template homology modeling with MODELLER Version 9.2 and molecular docking calculations with AutoDock Vina Version 1.2 were implemented to quantify the estimated binding affinity between selected putative substrates or ligands and UGT2B10. Finally, we performed molecular dynamics simulations using GROMACS Version 5.1.4 to confirm the potential UGT2B10 ligands prioritized after molecular docking (exhibiting negative free binding energy). Results Four potential ligands for UGT2B10 namely acetaminophen, lorazepam, mycophenolic acid and voriconazole n-oxide intermediate were identified. Other metabolites of voriconazole satisfied the criteria of being possible ligands of UGT2B10. Except for bilirubin and 4-Hydroxy Voriconazole, all the ligands (particularly voriconazole and hydroxy voriconazole) are oriented in substrate binding site close to the co-factor UDP (mean ± SD; 0.72 ± 0.33 nm). Further in vitro screening of the putative ligands prioritized by in silico pipeline is warranted to understand the nature of the ligands either as inhibitors or substrates of UGT2B10. Conclusions These results may indicate the clinical and pharmacological relevance UGT2B10 in pediatric HSCT setting. With this systematic computational methodology, we provide a rational-, time-, and cost-effective way to identify and prioritize the interesting putative substrates or inhibitors of UGT2B10 for further testing in in vitro experiments. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00402-5.
Collapse
|
23
|
Association study of candidate DNA-repair gene variants and acute graft versus host disease in pediatric patients receiving allogeneic hematopoietic stem-cell transplantation. THE PHARMACOGENOMICS JOURNAL 2022; 22:9-18. [PMID: 34711928 PMCID: PMC8794787 DOI: 10.1038/s41397-021-00251-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/26/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023]
Abstract
Acute Graft versus Host Disease (aGvHD) grades 2-4 occurs in 15-60% of pediatric patients undergoing allogeneic haematopoietic stem-cell transplantation (allo-HSCT). The collateral damage to normal tissue by conditioning regimens administered prior to allo-HSCT serve as an initial trigger for aGvHD. DNA-repair mechanisms may play an important role in mitigating this initial damage, and so the variants in corresponding DNA-repair protein-coding genes via affecting their quantity and/or function. We explored 51 variants within 17 DNA-repair genes for their association with aGvHD grades 2-4 in 60 pediatric patients. The cumulative incidence of aGvHD 2-4 was 12% (n = 7) in the exploratory cohort. MGMT rs10764881 (G>A) and EXO rs9350 (c.2270C>T) variants were associated with aGvHD 2-4 [Odds ratios = 14.8 (0 events out of 40 in rs10764881 GG group) and 11.5 (95% CI: 2.3-191.8), respectively, multiple testing corrected p ≤ 0.001]. Upon evaluation in an extended cohort (n = 182) with an incidence of aGvHD 2-4 of 22% (n = 40), only MGMT rs10764881 (G>A) remained significant (adjusted HR = 2.05 [95% CI: 1.06-3.94]; p = 0.03) in the presence of other clinical risk factors. Higher MGMT expression was seen in GG carriers for rs10764881 and was associated with higher IC50 of Busulfan in lymphoblastoid cells. MGMT rs10764881 carrier status could predict aGvHD occurrence in pediatric patients undergoing allo-HSCT.
Collapse
|
24
|
Teslenko I, Watson CJW, Xia Z, Chen G, Lazarus P. Characterization of Cytosolic Glutathione S-Transferases Involved in the Metabolism of the Aromatase Inhibitor, Exemestane. Drug Metab Dispos 2021; 49:1047-1055. [PMID: 34593616 PMCID: PMC11025106 DOI: 10.1124/dmd.121.000635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Exemestane (EXE) is a hormonal therapy used to treat estrogen receptor-positive breast cancer by inhibiting the final step of estrogen biosynthesis catalyzed by the enzyme aromatase. Cysteine conjugates of EXE and its active metabolite 17β-dihydro-EXE (DHE) are the major metabolites found in both the urine and plasma of patients taking EXE. The initial step in cysteine conjugate formation is glutathione conjugation catalyzed by the glutathione S-transferase (GST) family of enzymes. The goal of the present study was to identify cytosolic hepatic GSTs active in the GST-mediated metabolism of EXE and 17β-DHE. Twelve recombinant cytosolic hepatic GSTs were screened for their activity against EXE and 17β-DHE, and glutathionylated EXE and 17β-DHE conjugates were detected by ultra-performance liquid chromatography tandem mass spectrometry. GST α (GSTA) isoform 1, GST μ (GSTM) isoform 3 and isoform 1 were active against EXE, whereas only GSTA1 exhibited activity against 17β-DHE. GSTM1 exhibited the highest affinity against EXE with a Michaelis-Menten constant (KM) value that was 3.8- and 7.1-fold lower than that observed for GSTA1 and GSTM3, respectively. Of the three GSTs, GSTM3 exhibited the highest intrinsic clearance against EXE (intrinsic clearance = 0.14 nl·min-1·mg-1). The KM values observed for human liver cytosol against EXE (46 μM) and 17β-DHE (77 μM) were similar to those observed for recombinant GSTA1 (53 and 30 μM, respectively). Western blot analysis revealed that GSTA1 and GSTM1 composed 4.3% and 0.57%, respectively, of total protein in human liver cytosol; GSTM3 was not detected. These data suggest that GSTA1 is the major hepatic cytosolic enzyme involved in the clearance of EXE and its major active metabolite, 17β-DHE. SIGNIFICANCE STATEMENT: Most previous studies related to the metabolism of the aromatase inhibitor exemestane (EXE) have focused mainly on phase I metabolic pathways and the glucuronidation phase II metabolic pathway. However, recent studies have indicated that glutathionylation is the major metabolic pathway for EXE. The present study is the first to characterize hepatic glutathione S-transferase (GST) activity against EXE and 17β-dihydro-EXE and to identify GST α 1 and GST μ 1 as the major cytosolic GSTs involved in the hepatic metabolism of EXE.
Collapse
Affiliation(s)
- Irina Teslenko
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Christy J W Watson
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Zuping Xia
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Gang Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|
25
|
GSTM1 and GSTT1 double null genotypes determining cell fate and proliferation as potential risk factors of relapse in children with hematological malignancies after hematopoietic stem cell transplantation. J Cancer Res Clin Oncol 2021; 148:71-86. [PMID: 34499222 PMCID: PMC8752561 DOI: 10.1007/s00432-021-03769-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/14/2021] [Indexed: 10/29/2022]
Abstract
PURPOSE This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models. METHODS GSTM1- and GSTT1-null alleles were genotyped using germline DNA from whole blood prior to a conditioning BU-based regimen. Association of GSTM1- and GSTT1-null variants with relapse incidence was analyzed using multivariable competing risk analysis. BU-induced cell death studies were conducted in GSTs- null and non-null LCLs and CRISPR-Cas9 gene-edited THP1 leukemia cell lines. RESULTS Carrying GSTM1/GSTT1 double null genotype was found to be an independent risk factor for post-HSCT relapse in 86 children (adjusted HR: 6.52 [95% Cl, 2.76-15.42; p = 1.9 × 10-5]). BU-induced cell death preferentially in THP1GSTM1(non-null) and LCLsGSTM1(non-null) as shown by decreased viability, increased necrosis and levels of the oxidized form of glutathione compared to null cells, while GSTT1 non-null cells showed increased baseline proliferation. CONCLUSION The clinical association suggests that GSTM1/GSTT1 double null genotype could serve as genetic stratification biomarker for the high risk of post-HSCT relapse. Functional studies have indicated that GSTM1 status modulates BU-induced cell death. On the other hand, GSTT1 is proposed to be involved in baseline cell proliferation.
Collapse
|
26
|
Ben Hassine K, Nava T, Théoret Y, Nath CE, Daali Y, Kassir N, Lewis V, Bredius RGM, Shaw PJ, Bittencourt H, Krajinovic M, Uppugunduri CRS, Ansari M. Precision dosing of intravenous busulfan in pediatric hematopoietic stem cell transplantation: Results from a multicenter population pharmacokinetic study. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1043-1056. [PMID: 34453497 PMCID: PMC8452291 DOI: 10.1002/psp4.12683] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/14/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022]
Abstract
Busulfan (Bu) is a common component of conditioning regimens before hematopoietic stem cell transplantation (HSCT) and is known for high interpatient pharmacokinetic (PK) variability. This study aimed to develop and externally validate a multicentric, population PK (PopPK) model for intravenous Bu in pediatric patients before HSCT to first study the influence of glutathione‐s‐transferase A1 (GSTA1) polymorphisms on Bu's PK in a large multicentric pediatric population while accounting for fludarabine (Flu) coadministration and, second, to establish an individualized, model‐based, first‐dose recommendation for intravenous Bu that can be widely used in pediatric patients. The model was built using data from 302 patients from five transplantation centers who received a Bu‐based conditioning regimen. External model validation used data from 100 patients. The relationship between body weight and Bu clearance (CL) was best described by an age‐dependent allometric scaling of a body weight model. A stepwise covariate analysis identified Day 1 of Bu conditioning, GSTA1 metabolic groups based on GSTA1 polymorphisms, and Flu coadministration as significant covariates influencing Bu CL. The final model adequately predicted Bu first‐dose CL in the external cohort, with 81% of predicted area under the curves within the therapeutic window. The final model showed minimal bias (mean prediction error, −0.5%; 95% confidence interval [CI], −3.1% to 2.0%) and acceptable precision (mean absolute prediction error percentage, 18.7%; 95% CI, 17.0%–20.5%) in Bu CL prediction for dosing. This multicentric PopPK study confirmed the influence of GSTA1 polymorphisms and Flu coadministration on Bu CL. The developed model accurately predicted Bu CL and first doses in an external cohort of pediatric patients.
Collapse
Affiliation(s)
- Khalil Ben Hassine
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.,Division of Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Tiago Nava
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.,Division of Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Yves Théoret
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.,Clinical Pharmacology Unit, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Christa E Nath
- Department of Biochemistry, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,The Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Youssef Daali
- Clinical Pharmacology and Toxicology Division, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty of Medicine & Sciences, University of Geneva, Geneva, Switzerland
| | - Nastya Kassir
- Genentech/Roche, Clinical Pharmacology, South San Francisco, California, USA
| | - Victor Lewis
- Department of Pediatrics, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Robbert G M Bredius
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter J Shaw
- The Cancer Centre for Children, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Henrique Bittencourt
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.,Clinical Pharmacology Unit, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Maja Krajinovic
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.,Clinical Pharmacology Unit, Sainte-Justine University Health Center (SJUHC), Montreal, Quebec, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Chakradhara Rao Satyanarayana Uppugunduri
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.,Division of Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Marc Ansari
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland.,Division of Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
27
|
Remy A, Théorêt Y, Ansari M, Bittencourt H, Ducruet T, Nava T, Pastore Y, Rezgui MA, Krajinovic M, Kleiber N. Is Busulfan Clearance Different in Patients With Sickle Cell Disease? Let's Clear Up That Case With Some Controls. J Pediatr Hematol Oncol 2021; 43:e867-e872. [PMID: 33661168 DOI: 10.1097/mph.0000000000002106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 12/28/2020] [Indexed: 11/26/2022]
Abstract
In busulfan-based conditioning regimen for hematopoietic stem cell transplantation in children, accurate a priori determination of the first dose is important because of its narrow therapeutic window. Sickle cell disease (SCD) influences pharmacokinetics of the commonly used drugs by affecting organs responsible for drug metabolism and elimination. This pharmacokinetics study assesses the influence of SCD on the metabolic pathway of busulfan that is mainly metabolized in the liver. In this retrospective cross-sectional case-control study, 16 patients with SCD were matched to 50 patients without SCD on known busulfan clearance's covariates (glutathione-S-transferase alpha1 polymorphisms, age, weight). Clearance of the first dose of busulfan was not significantly different independently of genetic or anthropometric factors in patients with or without SCD.
Collapse
Affiliation(s)
- Amandine Remy
- Department of Pediatric Hematology, Univ. Lille, CHU Lille, Lille, France
- Unité de pharmacologie clinique
| | - Yves Théorêt
- Unité de pharmacologie clinique
- Departments of Pharmacology and Physiology
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Université de Montréal
| | - Marc Ansari
- CANSEARCH Research Laboratory, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva
- Department of Women-Children-Adolescents, Division of General Pediatrics, Pediatric Onco-Hematology Unit, Geneva University Hospital, Geneva, Switzerland
| | - Henrique Bittencourt
- Pediatrics
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Université de Montréal
| | - Thierry Ducruet
- Applied Clinical Research Unit, CHU Sainte-Justine, Montreal, QC, Canada
| | - Tiago Nava
- CANSEARCH Research Laboratory, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva
- Department of Women-Children-Adolescents, Division of General Pediatrics, Pediatric Onco-Hematology Unit, Geneva University Hospital, Geneva, Switzerland
| | - Yves Pastore
- Pediatrics
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Université de Montréal
| | - Mohamed A Rezgui
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Université de Montréal
| | - Maja Krajinovic
- Departments of Pharmacology and Physiology
- Pediatrics
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Université de Montréal
| | - Niina Kleiber
- Departments of Pharmacology and Physiology
- Pediatrics
- Research Center, CHU Sainte-Justine
| |
Collapse
|
28
|
Ansari M, Petrykey K, Rezgui MA, Del Vecchio V, Cortyl J, Ameur M, Nava T, Beaulieu P, St-Onge P, Mlakar SJ, Uppugunduri CRS, Théoret Y, Bartelink IH, Boelens JJ, Bredius RGM, Dalle JH, Lewis V, Kangarloo BS, Corbacioglu S, Sinnett D, Bittencourt H, Krajinovic M. Genetic susceptibility to acute graft versus host disease in pediatric patients undergoing HSCT. Bone Marrow Transplant 2021; 56:2697-2704. [PMID: 34215854 DOI: 10.1038/s41409-021-01386-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 11/09/2022]
Abstract
The most frequent complication of allogeneic hematopoietic stem cell transplantation is acute Graft versus Host Disease (aGVHD). Proliferation and differentiation of donor T cells initiate inflammatory response affecting the skin, liver, and gastrointestinal tract. Besides recipient-donor HLA disparities, disease type, and the conditioning regimen, variability in the non-HLA genotype have an impact on aGVHD onset, and genetic variability of key cytokines and chemokines was associated with increased risk of aGVHD. To get further insight into the recipient genetic component of aGVHD grades 2-4 in pediatric patients, we performed an exome-wide association study in a discovery cohort (n = 87). Nine loci sustained correction for multiple testing and were analyzed in a validation group (n = 168). Significant associations were replicated for ERC1 rs1046473, PLEK rs3816281, NOP9 rs2332320 and SPRED1 rs11634702 variants through the interaction with non-genetic factors. The ERC1 variant was significant among patients that received the transplant from HLA-matched related individuals (p = 0.03), bone marrow stem cells recipients (p = 0.007), and serotherapy-negative patients (p = 0.004). NOP9, PLEK, and SPRED1 effects were modulated by stem cell source, and serotherapy (p < 0.05). Furthermore, ERC1 and PLEK SNPs correlated with aGVHD 3-4 independently of non-genetic covariates (p = 0.02 and p = 0.003). This study provides additional insight into the genetic component of moderate to severe aGVHD.
Collapse
Affiliation(s)
- Marc Ansari
- Cansearch research platform for paediatric oncology and haematology, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Women, Child and Adolescent, Onco-Hematology Unit, Geneva University Hospital, Geneva, Switzerland
| | - Kateryna Petrykey
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Mohamed Aziz Rezgui
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada
| | - Veronica Del Vecchio
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Jacques Cortyl
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada
| | - Milad Ameur
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Tiago Nava
- Cansearch research platform for paediatric oncology and haematology, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Women, Child and Adolescent, Onco-Hematology Unit, Geneva University Hospital, Geneva, Switzerland
| | - Patrick Beaulieu
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada
| | - Pascal St-Onge
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada
| | - Simona Jurkovic Mlakar
- Cansearch research platform for paediatric oncology and haematology, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Women, Child and Adolescent, Onco-Hematology Unit, Geneva University Hospital, Geneva, Switzerland
| | - Chakradhara Rao S Uppugunduri
- Cansearch research platform for paediatric oncology and haematology, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Women, Child and Adolescent, Onco-Hematology Unit, Geneva University Hospital, Geneva, Switzerland
| | - Yves Théoret
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada.,Clinical Pharmacology Unit, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada
| | - Imke H Bartelink
- Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Pharmacology and Pharmacy, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jaap-Jan Boelens
- Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, The Netherlands.,Stem cell transplantation and cellular therapy program, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robbert G M Bredius
- Department of Pediatrics, Division of Immunology, Infectious Diseases and SCT, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean-Hugues Dalle
- Pediatric Hematology Department, Robert Debré Hospital, Assistance Publique, Hôpitaux de Paris and Paris-Diderot University, Paris, France
| | - Victor Lewis
- Department of Oncology, Alberta Children's Hospital, Calgary, AB, Canada
| | - Bill S Kangarloo
- Department of Oncology, Alberta Children's Hospital, Calgary, AB, Canada
| | - Selim Corbacioglu
- Department of Hematology, Oncology, and Stem Cell Transplantation University Children's Hospital Regensburg, Regensburg, Germany
| | - Daniel Sinnett
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Henrique Bittencourt
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Maja Krajinovic
- Charles-Bruneau Cancer Center, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada. .,Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC, Canada. .,Clinical Pharmacology Unit, Sainte-Justine University Health Center (SJUHC), Montreal, QC, Canada. .,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
29
|
Dilo A, Daali Y, Desmeules J, Chalandon Y, Uppugunduri CRS, Ansari M. Comparing Dried Blood Spots and Plasma Concentrations for Busulfan Therapeutic Drug Monitoring in Children. Ther Drug Monit 2021; 42:111-117. [PMID: 31348116 DOI: 10.1097/ftd.0000000000000673] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Busulfan (Bu) is one of the conditioning regimen components for pediatric hematopoietic stem cell transplantation. Bu therapeutic drug monitoring (TDM) is essential for a successful treatment outcome and toxicity evasion. Dried blood spot (DBS) sampling is a rapid and simple method for Bu TDM, compared with conventional plasma sampling. This study evaluated the feasibility of using the DBS method for Bu TDM. The hematocrit (Hct) and conditioning day were also examined for their impact on the DBS method's performance. METHODS Venous blood collected from 6 healthy volunteers was diluted, using their plasma into 4 samples of varying Hct values. Each sample was spiked with Bu calibrators (300, 600, and 1400 ng/mL), prepared using DBS and dried plasma spot (DPS) sampling and analyzed using a validated liquid-chromatography tandem-mass spectrometry method. Clinical blood samples (n = 153) from pediatric patients (n = 15) treated with Bu (mainly from doses 1, 2, 5, and 9) were used to prepare paired volumetric DBS and DPS samples. A Bland-Altman plot and Deming regression were used to define the agreement between the paired DBS and DPS measurements. Passing-Bablok regression analyses investigated the effects of Hct and conditioning day on the linearity between both methods. RESULTS In vitro analyses showed good agreement between DBS and DPS measurements, with a mean difference of -5.4% and a 95% confidence interval on the limits of agreement of -15.3% to 4.6%. Clinical samples showed good correlation (Pearson correlation coefficient = 0.96; slope = 1.00) between the DBS and DPS methods. The DBS method met the clinical acceptance limits for clinical samples, with a bias <±20%. Bland-Altman plots showed good agreement, with only 5.8% of paired measurements exceeding the limits of agreement (±1.96 SD), although within its 95% confidence interval. Hct observations ranged from 21.7% to 34.7% and did not affect Bu concentrations measured from DBS in either the in vitro or in vivo studies. CONCLUSIONS These results show that DBS is a useful method for Bu TDM, provided samples are analyzed on the collection day. DBS sampling offers advantages over traditional plasma sampling in infants and younger children because only small volumes of blood are required.
Collapse
Affiliation(s)
- Ana Dilo
- Clinical Pharmacology and Toxicology Unit, Geneva University Hospitals, University of Geneva
| | - Youssef Daali
- Clinical Pharmacology and Toxicology Unit, Geneva University Hospitals, University of Geneva
| | - Jules Desmeules
- Clinical Pharmacology and Toxicology Unit, Geneva University Hospitals, University of Geneva
| | - Yves Chalandon
- Division of Hematology, Department of Oncology, Geneva University Hospitals
| | - Chakradhara R S Uppugunduri
- Onco-Hematology Unit, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospitals; and.,CANSEARCH Research Laboratory, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marc Ansari
- Onco-Hematology Unit, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospitals; and.,CANSEARCH Research Laboratory, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
30
|
Waespe N, Strebel S, Jurkovic Mlakar S, Krajinovic M, Kuehni CE, Nava T, Ansari M. Genetic Predictors for Sinusoidal Obstruction Syndrome-A Systematic Review. J Pers Med 2021; 11:jpm11050347. [PMID: 33925809 PMCID: PMC8145271 DOI: 10.3390/jpm11050347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/18/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Sinusoidal obstruction syndrome (SOS) is a potentially life-threatening complication after hematopoietic stem cell transplantation (HSCT) or antineoplastic treatment without HSCT. Genetic variants were investigated for their association with SOS, but the evidence is inconclusive. We performed a systematic literature review to identify genes, gene variants, and methods of association analyses of genetic markers with SOS. We identified 23 studies after HSCT and 4 studies after antineoplastic treatment without HSCT. One study (4%) performed whole-exome sequencing (WES) and replicated the analysis in an independent cohort, 26 used a candidate-gene approach. Three studies included >200 participants (11%), and six were of high quality (22%). Variants in 34 genes were tested in candidate gene studies after HSCT. Variants in GSTA1 were associated with SOS in three studies, MTHFR in two, and CPS1, CTH, CYP2B6, GSTM1, GSTP1, HFE, and HPSE in one study each. UGT2B10 and LNPK variants were identified in a WES analysis. After exposure to antineoplastic agents without HSCT, variants in six genes were tested and only GSTM1 was associated with SOS. There was a substantial heterogeneity of populations within and between studies. Future research should be based on sufficiently large homogenous samples, adjust for covariates, and replicate findings in independent cohorts.
Collapse
Affiliation(s)
- Nicolas Waespe
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, University of Geneva, 1205 Geneva, Switzerland; (N.W.); (S.S.); (S.J.M.); (T.N.)
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland;
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Sven Strebel
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, University of Geneva, 1205 Geneva, Switzerland; (N.W.); (S.S.); (S.J.M.); (T.N.)
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland;
- Graduate School for Health Sciences (GHS), University of Bern, 3012 Bern, Switzerland
| | - Simona Jurkovic Mlakar
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, University of Geneva, 1205 Geneva, Switzerland; (N.W.); (S.S.); (S.J.M.); (T.N.)
| | - Maja Krajinovic
- Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Department of Pediatrics, Montreal, QC H3T 1C5, Canada;
- Clinical Pharmacology Unit, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Department of Pharmacology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Claudia Elisabeth Kuehni
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland;
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland
| | - Tiago Nava
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, University of Geneva, 1205 Geneva, Switzerland; (N.W.); (S.S.); (S.J.M.); (T.N.)
- Department of Women, Children and Adolescents, Division of Pediatric Oncology and Hematology, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Marc Ansari
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, University of Geneva, 1205 Geneva, Switzerland; (N.W.); (S.S.); (S.J.M.); (T.N.)
- Department of Women, Children and Adolescents, Division of Pediatric Oncology and Hematology, Geneva University Hospital, 1205 Geneva, Switzerland
- Correspondence: ; Tel.: +41-79-553-6100
| |
Collapse
|
31
|
Mlakar V, Curtis PHD, Armengol M, Ythier V, Dupanloup I, Hassine KB, Lesne L, Murr R, Mlakar SJ, Nava T, Ansari M. The analysis of GSTA1 promoter genetic and functional diversity of human populations. Sci Rep 2021; 11:5038. [PMID: 33658540 PMCID: PMC7930039 DOI: 10.1038/s41598-021-83996-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/10/2021] [Indexed: 12/30/2022] Open
Abstract
GSTA1 encodes a member of a family of enzymes that function to add glutathione to target electrophilic compounds, including carcinogens, therapeutic drugs, environmental toxins, and products of oxidative stress. GSTA1 has several functional SNPs within its promoter region that are responsible for a change in its expression by altering promoter function. This study aims to investigate distributions of GSTA1 promoter haplotypes across different human populations and to assess their impact on the expression of GSTA1. PHASE 2.1.1 was used to infer haplotypes and diplotypes of six GSTA1 promoter SNPs on 2501 individuals from 26 populations classified by the 1000 Genomes Project into five super-populations that included Africa (N = 660), America (N = 347), East Asia (N = 504), Europe (N = 502), and South Asia (N = 488). We used pairwise FST analysis to compare sub-populations and luciferase reporter assay (LRA) to evaluate the impact of each SNP on activation of transcription and interaction with other SNPs. The distributions of GSTA1 promoter haplotypes and diplotypes were significantly different among the different human populations. Three new promoter haplotypes were found in the African super-population. LRA demonstrated that SNPs at -52 and -69 has the most impact on GSTA1 expression, however other SNPs have a significant impact on transcriptional activity. Based on LRA, a new model of cis-elements interaction is presented. Due to the significant differences in GSTA1 diplotype population frequencies, future pharmacogenomics or disease-related studies would benefit from the inclusion of the complete GSTA1 promoter haplotype based on the newly proposed metabolic grouping derived from the LRA results.
Collapse
Affiliation(s)
- Vid Mlakar
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Patricia Huezo-Diaz Curtis
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Marc Armengol
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Victor Ythier
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1211, Geneva, Switzerland
| | | | - Khalil Ben Hassine
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Laurence Lesne
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Rabih Murr
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1211, Geneva, Switzerland
| | - Simona Jurkovic Mlakar
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Tiago Nava
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Marc Ansari
- Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, University of Geneva, Avenue de la Roseraie 64, 1205, Geneva, Switzerland. .,Pediatric Oncology and Hematology Unit, Department of Women, Children and Adolescents, Geneva University Hospital, Rue Willy-Donzé 6, 1205, Geneva, Switzerland.
| |
Collapse
|
32
|
Yuan J, Sun N, Feng X, He H, Mei D, Zhu G, Zhao L. Optimization of Busulfan Dosing Regimen in Pediatric Patients Using a Population Pharmacokinetic Model Incorporating GST Mutations. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:253-268. [PMID: 33623415 PMCID: PMC7894888 DOI: 10.2147/pgpm.s289834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/11/2021] [Indexed: 12/28/2022]
Abstract
Purpose The aim of this study was to develop a novel busulfan dosing regimen, based on a population pharmacokinetic (PPK) model in Chinese children, and to achieve better area under the concentration-time curve (AUC) targeting. Patients and Methods We collected busulfan concentration-time samples from 69 children who received intravenous busulfan prior to allogeneic hematopoietic stem cell transplantation (allo-HSCT). A population pharmacokinetic model for busulfan was developed by nonlinear mixed effect modelling and was validated by an external dataset (n=14). A novel busulfan dosing regimen was developed through simulated patients, and has been verified on real patients. Limited sampling strategy (LSS) was established by Bayesian forecasting. Mean absolute prediction error (MAPE) and relative root mean Squared error (rRMSE) were calculated to evaluate predictive accuracy. Results A one-compartment model with first-order elimination best described the data. GSTA1 genotypes, body surface area (BSA) and aspartate aminotransferase (AST) were found to be significant covariates of Bu clearance, and BSA had significant impact of the volume of distribution. Moreover, two equations were obtained for recommended dose regimens: dose (mg)=34.14×BSA (m2)+3.75 (for GSTA1 *A/*A), Dose (mg)=30.99×BSA (m2)+3.21 (for GSTA1 *A/*B). We also presented a piecewise dosage based on BSA categories for each GSTA1 mutation. A two-point LSS, two hours and four hours after dosing, behaved well with acceptable prediction precision (rRMSE=1.026%, MAPE=6.55%). Conclusion We recommend a GSTA1-BSA and BSA-based dosing (Q6 h) based on a PPK model for personalizing busulfan therapy in pediatric population. Additionally, an optimal LSS (C2h and C4h) provides convenience for therapeutic drug monitoring (TDM) in the future.
Collapse
Affiliation(s)
- Jinjie Yuan
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, People's Republic of China.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ning Sun
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xinying Feng
- Phase I Clinical Trials Centre, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, People's Republic of China
| | - Huan He
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Dong Mei
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Guanghua Zhu
- Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Libo Zhao
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
33
|
Ben Hassine K, Powys M, Svec P, Pozdechova M, Versluys B, Ansari M, Shaw PJ. Total Body Irradiation Forever? Optimising Chemotherapeutic Options for Irradiation-Free Conditioning for Paediatric Acute Lymphoblastic Leukaemia. Front Pediatr 2021; 9:775485. [PMID: 34956984 PMCID: PMC8705537 DOI: 10.3389/fped.2021.775485] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
Total-body irradiation (TBI) based conditioning prior to allogeneic hematopoietic stem cell transplantation (HSCT) is generally regarded as the gold-standard for children >4 years of age with acute lymphoblastic leukaemia (ALL). Retrospective studies in the 1990's suggested better survival with irradiation, confirmed in a small randomised, prospective study in the early 2000's. Most recently, this was reconfirmed by the early results of the large, randomised, international, phase III FORUM study published in 2020. But we know survivors will suffer a multitude of long-term sequelae after TBI, including second malignancies, neurocognitive, endocrine and cardiometabolic effects. The drive to avoid TBI directs us to continue optimising irradiation-free, myeloablative conditioning. In chemotherapy-based conditioning, the dominant myeloablative effect is provided by the alkylating agents, most commonly busulfan or treosulfan. Busulfan with cyclophosphamide is a long-established alternative to TBI-based conditioning in ALL patients. Substituting fludarabine for cyclophosphamide reduces toxicity, but may not be as effective, prompting the addition of a third agent, such as thiotepa, melphalan, and now clofarabine. For busulfan, it's wide pharmacokinetic (PK) variability and narrow therapeutic window is well-known, with widespread use of therapeutic drug monitoring (TDM) to individualise dosing and control the cumulative busulfan exposure. The development of first-dose selection algorithms has helped achieve early, accurate busulfan levels within the targeted therapeutic window. In the future, predictive genetic variants, associated with differing busulfan exposures and toxicities, could be employed to further tailor individualised busulfan-based conditioning for ALL patients. Treosulfan-based conditioning leads to comparable outcomes to busulfan-based conditioning in paediatric ALL, without the need for TDM to date. Future PK evaluation and modelling may optimise therapy and improve outcome. More recently, the addition of clofarabine to busulfan/fludarabine has shown encouraging results when compared to TBI-based regimens. The combination shows activity in ALL as well as AML and deserves further evaluation. Like busulfan, optimization of chemotherapy conditioning may be enhanced by understanding not just the PK of clofarabine, fludarabine, treosulfan and other agents, but also the pharmacodynamics and pharmacogenetics, ideally in the context of a single disease such as ALL.
Collapse
Affiliation(s)
- Khalil Ben Hassine
- Cansearch Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Madeleine Powys
- Blood Transplant and Cell Therapies, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Peter Svec
- Department of Pediatric Hematology and Oncology, Comenius University, Bratislava, Slovakia.,Bone Marrow Transplantation Unit, National Institute of Children's Diseases, Bratislava, Slovakia
| | - Miroslava Pozdechova
- Department of Pediatric Hematology and Oncology, Comenius University, Bratislava, Slovakia.,Bone Marrow Transplantation Unit, National Institute of Children's Diseases, Bratislava, Slovakia
| | | | - Marc Ansari
- Cansearch Research Platform for Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Pediatric Oncology and Hematology, Department of Women, Child and Adolescent, University Geneva Hospitals, Geneva, Switzerland
| | - Peter J Shaw
- Blood Transplant and Cell Therapies, Children's Hospital at Westmead, Sydney, NSW, Australia.,Speciality of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
34
|
Seydoux C, Medinger M, Gerull S, Halter J, Heim D, Chalandon Y, Levrat SM, Schanz U, Nair G, Ansari M, Simon P, Passweg JR, Cantoni N. Busulfan-cyclophosphamide versus cyclophosphamide-busulfan as conditioning regimen before allogeneic hematopoietic cell transplantation: a prospective randomized trial. Ann Hematol 2021; 100:209-216. [PMID: 33098041 PMCID: PMC7782401 DOI: 10.1007/s00277-020-04312-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/15/2020] [Indexed: 01/20/2023]
Abstract
Busulfan and cyclophosphamide (BuCy) is a frequently used myeloablative conditioning regimen for allogeneic hematopoietic cell transplantation (allo-HCT). Theoretical considerations and pharmacological data indicate that application of busulfan prior to subsequent cyclophosphamide (BuCy) may trigger liver toxicity. Reversing the order of application to cyclophosphamide-busulfan (CyBu) might be preferable, a hypothesis supported by animal data and retrospective studies. We performed a prospective randomized trial to determine impact of order of application of Bu and Cy before allo-HCT in 70 patients with hematological malignancy, 33 patients received BuCy and 37 CyBu for conditioning. In the short term, there were minimal differences in liver toxicity favoring CyBu over BuCy, significant only for alanine amino transferase at day 30 (p = 0.03). With longer follow-up at 4 years, non-relapse mortality (6% versus 27%, p = 0.05) was lower and survival (63% versus 43%, p = 0.06) was higher with CyBu compared to BuCy. Other outcomes, such as engraftment (p = 0.21), acute and chronic graft-versus-host disease (p = 0.40; 0.36), and relapse (p = 0.79), were similar in both groups. We prospectively show evidence that the order of application of Cy and Bu in myeloablative conditioning in allo-HCT patients has impact on outcome.
Collapse
Affiliation(s)
- Claire Seydoux
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital of Basel, Petersgraben 4, CH-4031, Basel, Switzerland
| | - Michael Medinger
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital of Basel, Petersgraben 4, CH-4031, Basel, Switzerland.
| | - Sabine Gerull
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital of Basel, Petersgraben 4, CH-4031, Basel, Switzerland
| | - Joerg Halter
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital of Basel, Petersgraben 4, CH-4031, Basel, Switzerland
| | - Dominik Heim
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital of Basel, Petersgraben 4, CH-4031, Basel, Switzerland
| | - Yves Chalandon
- Division of Hematology, Bone Marrow Transplant Unit, University Hospital of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stavroula Masouridi Levrat
- Division of Hematology, Bone Marrow Transplant Unit, University Hospital of Geneva and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Urs Schanz
- Department of Medical Oncology and Hematology, Stem-/Immune- cell-transplant Unit, University Hospital of Zurich, Zurich, Switzerland
| | - Gayathri Nair
- Department of Medical Oncology and Hematology, Stem-/Immune- cell-transplant Unit, University Hospital of Zurich, Zurich, Switzerland
| | - Marc Ansari
- Department Women, Children and Adolescent, Unit of Oncology and Hematology Pediatric, University Hospital of Geneva, Geneva, Switzerland
- Cansearch Research Laboratory, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Simon
- Clinical Trials Unit, Department of Clinical Research, Basel University, Basel, Switzerland
| | - Jakob R Passweg
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital of Basel, Petersgraben 4, CH-4031, Basel, Switzerland.
| | - Nathan Cantoni
- Division of Oncology, Hematology and Transfusion Medicine, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
35
|
Review of the Pharmacokinetics and Pharmacodynamics of Intravenous Busulfan in Paediatric Patients. Clin Pharmacokinet 2020; 60:17-51. [PMID: 33128207 DOI: 10.1007/s40262-020-00947-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
We aimed to review the pharmacokinetics (PK) of intravenous busulfan in paediatric patients, identify covariate factors influencing exposure, investigate evidence of changes in PK behaviour over time, and correlate exposure with efficacy and toxicity outcomes. A literature review was undertaken of original research published between 2007 and 2019, investigating the PK and pharmacodynamics (PD) of intravenous busulfan in patients ≤ 18 years of age. The review identified 41 publications characterising the PK, and 45 publications describing the PD, of busulfan. Median typical clearance (CL) was 0.22 L/h/kg and median typical volume of distribution was 0.69 L/kg. Patient weight, age, glutathione-S-transferase A1 (GSTA1) genotype and busulfan dosing day/time were the most commonly identified factors affecting CL. Of nine studies investigating changes in CL, seven reported reduced CL over the 4-day course of treatment. Exposure monitoring methods and therapeutic targets were heterogeneous across studies. Relationships between busulfan exposure and patient outcomes were observed in five studies. One study observed a cumulative area under the concentration-time curve over all days of treatment of between 78 and 101 mg/L·h, and two studies observed an average concentration at first dose of < 600 ng/mL improved overall survival, transplant-related mortality, or relapse. One study observed increased sinusoidal obstructive syndrome with maximum busulfan concentration > 1.88 ng/mL. Patient weight, age and GSTA1 genotype are important covariates to consider when individualising busulfan therapy. Reduced busulfan CL over time may need to be accounted for, particularly in patients not receiving phenytoin co-therapy. Standardised monitoring of busulfan exposure over the entire course of treatment and further investigation of the role of busulfan metabolites and pharmacogenomics is warranted.
Collapse
|
36
|
Bradford KL, Liu S, Krajinovic M, Ansari M, Garabedian E, Tse J, Wang X, Shaw KL, Gaspar HB, Candotti F, Kohn DB. Busulfan Pharmacokinetics in Adenosine Deaminase-Deficient Severe Combined Immunodeficiency Gene Therapy. Biol Blood Marrow Transplant 2020; 26:1819-1827. [PMID: 32653625 PMCID: PMC7529956 DOI: 10.1016/j.bbmt.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/28/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
The pharmacokinetics of low-dose busulfan (BU) were investigated as a nonmyeloablative conditioning regimen for autologous gene therapy (GT) in pediatric subjects with adenosine deaminase-deficient severe combined immunodeficiency disease (ADA SCID). In 3 successive clinical trials, which included either γ-retroviral (γ-RV) or lentiviral (LV) vectors, subjects were conditioned with BU using different dosing nomograms. The first cohort received BU doses based on body surface area (BSA), the second cohort received doses based on actual body weight (ABW), and in the third cohort, therapeutic drug monitoring (TDM) was used to target a specific area under the concentration-time curve (AUC). Neither BSA-based nor ABW-based dosing achieved a consistent cumulative BU AUC; in contrast, TDM-based dosing led to more consistent AUC. BU clearance increased as subject age increased from birth to 18 months. However, weight and age alone were insufficient to accurately predict the dose that would consistently achieve a target AUC. Furthermore, various clinical, laboratory, and genetic factors (eg, genotypes for glutathione-S-transferase isozymes known to participate in BU metabolism) were analyzed, but no single finding predicted subjects with rapid versus slow clearance. Analysis of BU AUC and the postengraftment vector copy number (VCN) in granulocytes, a surrogate marker of the level of engrafted gene-modified hematopoietic stem and progenitor cells (HSPCs), demonstrated gene marking at levels sufficient for therapeutic benefit in the subjects who had achieved the target BU AUC. Although many factors determine the ultimate engraftment following GT, this work demonstrates that the BU AUC correlated with the eventual level of engrafted gene-modified HSPCs within a vector group (γ-RV versus LV), with significantly higher levels of granulocyte VCN in the recipients of LV-modified grafts compared to recipients of γ-RV-transduced grafts. Taken together, these findings provide insight into low-dose BU pharmacokinetics in the unique setting of autologous GT for ADA SCID, and these dosing principles may be applied to future GT trials using low-dose BU to open the bone marrow niche.
Collapse
Affiliation(s)
- Kathryn L Bradford
- Department of Pediatric Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Siyu Liu
- Department of Population Sciences, City of Hope/Beckman Research Institute, Duarte, California; Department of Hematology and Hematopoietic Cell Transplantation, City of Hope/Beckman Research Institute, Duarte, California
| | - Maja Krajinovic
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada; Department of Pharmacology and Physiology, University of Montreal, Montreal, Quebec, Canada
| | - Marc Ansari
- Hematology-Oncology Unit, Department of Pediatrics, Geneva University Hospital & CANSEARCH Research Laboratory, University of Geneva, Geneva, Switzerland
| | - Elizabeth Garabedian
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - John Tse
- Department of Pharmaceutical Services, Ronald Reagan Medical Center, UCLA, Los Angeles, California
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, UCLA Health, Los Angeles, California
| | - Kit L Shaw
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California
| | - H Bobby Gaspar
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Orchard Therapeutics, London, United Kingdom
| | - Fabio Candotti
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland; Division of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | - Donald B Kohn
- Department of Pediatric Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California; The Broad Stem Cell Research Center, University of California, Los Angeles, California.
| |
Collapse
|
37
|
Kishimoto K, Hasegawa D, Irie K, Okada A, Nakamura S, Tamura A, Yamamoto N, Kozaki A, Saito A, Ishida T, Fukushima S, Kosaka Y. Pharmacokinetic analysis for model-supported therapeutic drug monitoring of busulfan in Japanese pediatric hematopoietic stem cell transplantation recipients. Pediatr Transplant 2020; 24:e13696. [PMID: 32196880 DOI: 10.1111/petr.13696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/09/2020] [Accepted: 02/21/2020] [Indexed: 12/22/2022]
Abstract
This prospective observational study analyzed the pharmacokinetics of busulfan in Japanese children and evaluated the predicting accuracy of previous pediatric PPK models of busulfan. This study enrolled five patients (aged 2-12 years, BW 14-48 kg) receiving a busulfan-based conditioning regimen for hematopoietic stem cell transplantation at our hospital between January 2017 and December 2018. All patients received a 2-hour intravenous busulfan infusion four times daily for a total of 16 doses. After the infusions, 51 plasma samples were collected with the plasma busulfan concentration measured by liquid chromatography-tandem mass spectrometry. PPK model fitting was analyzed using the (%MPE) and the (%MAPE). Limited sampling strategies for estimating busulfan AUC were also evaluated. High interpatient variability was observed in the PK parameters. The most suitable PPK model that reflected our data was McCune's two-compartment model (%MPE -8.7, %MAPE 19.3). A combination sampling method using the busulfan concentration at 2 and 6 hours after the start of the first busulfan dose was found to be able to estimate AUC4 day . These results provide useful information on busulfan therapeutic drug monitoring in the Japanese pediatric population.
Collapse
Affiliation(s)
- Kenji Kishimoto
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Daiichiro Hasegawa
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Kei Irie
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kobe Gakuin University, Kobe, Japan.,Department of Pharmacy, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Kobe, Japan
| | - Akira Okada
- Department of Regulatory Science, Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Sayaka Nakamura
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Akihiro Tamura
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Nobuyuki Yamamoto
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Aiko Kozaki
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Atsuro Saito
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Toshiaki Ishida
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| | - Shoji Fukushima
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Science, Kobe Gakuin University, Kobe, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology & Oncology, Children's Cancer Center, Kobe Children's Hospital, Kobe, Japan
| |
Collapse
|
38
|
Marsit H, Philippe M, Neely M, Rushing T, Bertrand Y, Ducher M, Leclerc V, Guitton J, Bleyzac N, Goutelle S. Intra-individual Pharmacokinetic Variability of Intravenous Busulfan in Hematopoietic Stem Cell-Transplanted Children. Clin Pharmacokinet 2020; 59:1049-1061. [PMID: 32157629 DOI: 10.1007/s40262-020-00877-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Busulfan therapeutic drug monitoring (TDM) is necessary to better achieve the target exposure in children before hematopoietic stem cell transplantation (HSCT). However, TDM-based dosing may be challenging if intra-individual pharmacokinetic variability (also denoted inter-occasion variability [IOV]) occurs during therapy. OBJECTIVES The objectives of this study were to describe and quantify busulfan IOV in children, and to investigate its potential determinants. METHODS We performed a new analysis of published data from children who received intravenous busulfan over 4 days before HSCT. We calculated individual pharmacokinetic parameters on each day of therapy using a published population pharmacokinetic model of busulfan and analyzed their changes. Population estimation of IOV was also performed with non-linear mixed effects (NLME) modeling. Potential predictors of significant decrease in busulfan clearance (CL) were assessed by using machine learning approaches. RESULTS IOV could be assessed in 136 children. Between day (D) 1 and D2, most patients (80%) experienced a decrease in busulfan CL, with a median change of - 7.9%. However, both large decreases (minimum, - 48.5%) and increases in CL (maximum, + 44%) were observed. Over D1-D3 of therapy, mean CL significantly decreased (- 15%), with a decrease of ≥ 20% in 22% of patients. Some patients also showed unstable CL from day to day. NLME modeling of IOV provided a coefficient of variation of 10.6% and 13.1% for volume of distribution (Vd) and CL, respectively. Some determinants of significant decreases in busulfan CL were identified, but predictive performance of the models was limited. CONCLUSIONS Significant busulfan intra-individual variability may occur in children who receive a HSCT and is hardly predictable. The main risk is busulfan overexposure. Performing TDM repeatedly over therapy appears to be the best way to accurately estimate busulfan exposure and perform precision dosing.
Collapse
Affiliation(s)
- Hanen Marsit
- Univ Lyon, Université Lyon 1, UMR CNRS 5558, Laboratoire de Biométrie et Biologie Evolutive, Villeurbanne, France.,Université de Monastir, Faculté de Pharmacie, Monastir, Tunisia
| | | | - Michael Neely
- Laboratory of Applied Pharmacokinetics and Bioinformatics, Division of Pediatric Infectious Diseases, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, USA
| | - Teresa Rushing
- Pharmacy Department, Children's Hospital Los Angeles and the University of Southern California, Los Angeles, CA, USA
| | - Yves Bertrand
- Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France
| | - Michel Ducher
- Service de Pharmacie, Hospices Civils de Lyon, Groupement Hospitalier Nord, Hôpital Pierre Garraud, 136 rue du Commandant Charcot, 69005, Lyon, France.,Univ Lyon, Université Lyon 1, EMR 3738 PK/PD Modeling in Oncology and Hematology, Lyon, France
| | - Vincent Leclerc
- Service de Pharmacie, Hospices Civils de Lyon, Groupement Hospitalier Nord, Hôpital Pierre Garraud, 136 rue du Commandant Charcot, 69005, Lyon, France.,Univ Lyon, Université Lyon 1, EMR 3738 PK/PD Modeling in Oncology and Hematology, Lyon, France
| | - Jérôme Guitton
- Univ Lyon, Université Lyon 1, EMR 3738 PK/PD Modeling in Oncology and Hematology, Lyon, France.,Laboratoire de Pharmacologie et Toxicologie, Hospices Civils de Lyon, Groupement Hospitalier Sud, Lyon, France
| | - Nathalie Bleyzac
- Service de Pharmacie, Hospices Civils de Lyon, Groupement Hospitalier Nord, Hôpital Pierre Garraud, 136 rue du Commandant Charcot, 69005, Lyon, France.,Univ Lyon, Université Lyon 1, EMR 3738 PK/PD Modeling in Oncology and Hematology, Lyon, France
| | - Sylvain Goutelle
- Univ Lyon, Université Lyon 1, UMR CNRS 5558, Laboratoire de Biométrie et Biologie Evolutive, Villeurbanne, France. .,Service de Pharmacie, Hospices Civils de Lyon, Groupement Hospitalier Nord, Hôpital Pierre Garraud, 136 rue du Commandant Charcot, 69005, Lyon, France. .,Univ Lyon, Université Lyon 1, ISPB, Faculté de Pharmacie de Lyon, Lyon, France.
| |
Collapse
|
39
|
Sun Y, Huang J, Hao C, Li Z, Liang W, Zhang W, Chen B, Yang W, Hu J. Population pharmacokinetic analysis of intravenous busulfan: GSTA1 genotype is not a predictive factor of initial dose in Chinese adult patients undergoing hematopoietic stem cell transplantation. Cancer Chemother Pharmacol 2019; 85:293-308. [PMID: 31834435 DOI: 10.1007/s00280-019-04001-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE This study aimed to develop a population pharmacokinetic (PPK) model to investigate the impact of GSTA1, GSTP1, and GSTM1 genotypes on busulfan pharmacokinetic (PK) variability in Chinese adult patients. METHODS Forty-three and 19 adult patients who underwent hematopoietic stem cell transplantation (HSCT) were enrolled for modeling group and validation group, respectively. All patients received twice-daily intravenous busulfan as part of conditioning regimen before HSCT. The PPK model was developed by nonlinear mixed-effect modeling. Covariates investigated were age, sex, actual body weight, body surface area, diagnoses, hepatic function markers, GST genotypes and conditioning regimen. RESULTS A total of 488 busulfan concentrations from 43 patients were obtained for the PPK model. The PK of intravenous busulfan was described by one-compartment model with first-order elimination with estimated clearance (CL) of 14.2 L/h and volume of distribution of 64.1 L. Inclusion of GSTA1 genotype as a covariate accounted for 1.1% of the inter-individual variability of busulfan CL (from 17.8% in the basic model to 16.7% in the final model). The accuracy and applicability of the final model were externally validated in the independent group. The difference of busulfan PK between Chinese patients and Caucasian patients existed because of the rarity of haplotype *B in Chinese population. CONCLUSIONS Although the GSTA1 genotype-based PPK model of intravenous busulfan was successfully developed and externally validated, the GSTA1 genotype was not considered to be clinically relevant to busulfan CL. We did not suggest the guidance of GSTA1 genotype on initial busulfan dose in Chinese adult patients.
Collapse
Affiliation(s)
- Yidan Sun
- Department of Bone Marrow Transplantation, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 20025, China
| | - Jingjing Huang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chenxia Hao
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ziwei Li
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wu Liang
- NeoTrident Co. Ltd., Beijing, China
| | - Weixia Zhang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bing Chen
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wanhua Yang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiong Hu
- Department of Bone Marrow Transplantation, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, 20025, China.
| |
Collapse
|
40
|
Genetic Susceptibility to Hepatic Sinusoidal Obstruction Syndrome in Pediatric Patients Undergoing Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 26:920-927. [PMID: 31790828 DOI: 10.1016/j.bbmt.2019.11.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/29/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
Sinusoidal obstruction syndrome (SOS) is a well-recognized and potentially life-threatening complication of hematopoietic stem cell transplantation (HSCT). SOS arises from endothelial cell damage and hepatocellular injury mostly due to the transplantation conditioning regimens but also to other patient, disease, and treatment-related factors. Understanding risk factors associated with the development of SOS is critical for early initiation of treatment or prophylaxis. The knowledge about genetic contribution is limited; few studies investigated so far selected a set of genes. To get more comprehensive insight in the genetic component, we performed an exome-wide association study using genetic variants derived from whole-exome sequencing. The analyses were performed in a discovery cohort composed of 87 pediatric patients undergoing HSCT following a busulfan-containing conditioning regimen. Eight lead single-nucleotide polymorphisms (SNPs) were identified after correction for multiple testing and subsequently analyzed in a validation cohort (n = 182). Three SNPs were successfully replicated, including rs17146905 (P = .001), rs16931326 (P = .04), and rs2289971 (P = .03), located respectively in the UGT2B10, BHLHE22, and KIAA1715 genes. UGT2B10 and KIAA1715 were retained in a multivariable model while controlling for nongenetic covariates and previously identified risk variants in the GSTA1 promoter. The modulation of associations by conditioning regimens was noted; KIAA1715 was dependent on the intensity of the conditioning regimen, whereas the effect of UGT2B10 was equally applicable to all of them. Combined effect of associated loci was also observed (P = .00006) with a genotype-related SOS risk of 9.8. To our knowledge, this is the first study addressing the genetic component of SOS at an exome-wide level and identifying novel genetic variations conferring a higher risk of SOS, which might be useful for personalized prevention and treatment strategies.
Collapse
|
41
|
Afsar NA, Bruckmueller H, Werk AN, Nisar MK, Ahmad HR, Cascorbi I. Implications of genetic variation of common Drug Metabolizing Enzymes and ABC Transporters among the Pakistani Population. Sci Rep 2019; 9:7323. [PMID: 31086207 PMCID: PMC6514210 DOI: 10.1038/s41598-019-43736-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/10/2019] [Indexed: 01/09/2023] Open
Abstract
Genetic polymorphism of drug metabolizing enzymes and transporters may influence drug response. The frequency varies substantially between ethnicities thus having implications on appropriate selection and dosage of various drugs in different populations. The distribution of genetic polymorphisms in healthy Pakistanis has so far not been described. In this study, 155 healthy adults (98 females) were included from all districts of Karachi. DNA was extracted from saliva and genotyped for relevant SNVs in CYP1A1, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and CYP3A5 as well as ALDH3A1, GSTA1, ABCB1 and ABCC2. About 64% of the participants were born to parents who were unrelated to each other. There was generally a higher prevalence (p < 0.05) of variant alleles of CYP450 1A2, 2B6, 2C19, 3A5, ALDH3A1, GSTM1 as well as ABCB1 and ABCC2 in this study cohort than in other ethnicities reported in the HapMap database. In contrast, the prevalence of variant alleles was lower in GSTA1. Therefore, in the Pakistani population sample from Karachi a significantly different prevalence of variant drug metabolizing enzymes and ABC transporters was observed as compared to other ethnicities, which could have putative clinical consequences on drug efficacy and safety.
Collapse
Affiliation(s)
- Nasir Ali Afsar
- Jinnah Medical and Dental College, Sohail University, 22-23 Shaheed-e-Millat Road, Karachi, 75400, Pakistan.
| | - Henrike Bruckmueller
- Institute of Experimental and Clinical Pharmacology, Christian Albrechts University Kiel, Hospitalstr. 4, Kiel, 24105, Germany
| | - Anneke Nina Werk
- Institute of Experimental and Clinical Pharmacology, Christian Albrechts University Kiel, Hospitalstr. 4, Kiel, 24105, Germany.,Department of Internal Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Muhammad Kashif Nisar
- Jinnah Medical and Dental College, Sohail University, 22-23 Shaheed-e-Millat Road, Karachi, 75400, Pakistan.,Liaquat National Hospital & Medical College, Karachi, Pakistan
| | - H R Ahmad
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan.,Sindh Institute of Urology and Transplantation, Karachi, Pakistan
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, Christian Albrechts University Kiel, Hospitalstr. 4, Kiel, 24105, Germany
| |
Collapse
|
42
|
Kim MG, Kwak A, Choi B, Ji E, Oh JM, Kim K. Effect of glutathione S-transferase genetic polymorphisms on busulfan pharmacokinetics and veno-occlusive disease in hematopoietic stem cell transplantation: A meta-analysis. Basic Clin Pharmacol Toxicol 2018; 124:691-703. [PMID: 30511436 DOI: 10.1111/bcpt.13185] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/28/2018] [Indexed: 12/25/2022]
Abstract
This meta-analysis was conducted to derive an integrated conclusion about the influence of glutathione S-transferase (GST) genetic polymorphisms on busulfan pharmacokinetic (PK) parameters and veno-occlusive disease (VOD). Studies which analysed the effect of GST genetic polymorphisms on area under the curve (AUC), clearance (CL) or VOD were searched for and selected. A pooled analysis was conducted using Comprehensive Meta-Analysis programme. Nineteen studies were included in this meta-analysis. GSTA1*B and GSTM1 null genotypes significantly decreased CLIV of busulfan (standardized difference in means (SDM) = -1.103; P = 0.019 and SDM = -0.418; P = 0.002, respectively). GSTA1*B significantly increased AUCIV of busulfan (SDM = 0.832; P = 0.046), whereas GSTM1 did not (SDM = 0.155; P = 0.478). The PK parameters of oral busulfan did not differ according to GST genotype. GSTA1, GSTM1 and GSTP1 were not significantly associated with VOD occurrence. GSTA1 and GSTM1 genotypes affected CLIV of busulfan, but only GSTA1 affected AUCIV . There was no significant difference in the PK parameters of oral busulfan (CLPO and AUCPO ) and VOD when only GST genotypes were considered.
Collapse
Affiliation(s)
- Myeong Gyu Kim
- Graduate School of Clinical Pharmacy, CHA University, Pocheon, Korea
| | - Arim Kwak
- College of Pharmacy, Korea University, Sejong, Korea
| | - Boyoon Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Eunhee Ji
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Incheon, Korea
| | - Jung Mi Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Kyungim Kim
- College of Pharmacy, Korea University, Sejong, Korea
| |
Collapse
|
43
|
Nava T, Kassir N, Rezgui MA, Uppugunduri CRS, Huezo-Diaz Curtis P, Duval M, Théoret Y, Daudt LE, Litalien C, Ansari M, Krajinovic M, Bittencourt H. Incorporation of GSTA1 genetic variations into a population pharmacokinetic model for IV busulfan in paediatric hematopoietic stem cell transplantation. Br J Clin Pharmacol 2018; 84:1494-1504. [PMID: 29469189 DOI: 10.1111/bcp.13566] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 01/12/2018] [Accepted: 02/09/2018] [Indexed: 11/26/2022] Open
Abstract
AIMS The aim of this study is to develop a population pharmacokinetic (PopPK) model for intravenous busulfan in children that incorporates variants of GSTA1, gene coding for the main enzyme in busulfan metabolism. METHODS Busulfan concentration-time data was collected from 112 children and adolescents (median 5.4 years old, range: 0.1-20) who received intravenous busulfan during the conditioning regimen prior to stem cell transplantation. Weight, sex, baseline disease (malignant vs. non-malignant), age, conditioning regimen and GSTA1 diplotypes were evaluated as covariates of pharmacokinetic parameters by using nonlinear mixed effects analysis. The ability to achieve the target AUC24h (3600-6000 μM min-1 ) was assessed by estimating the first dose based on the present PopPK model and by comparing the results with other available models in children. RESULTS A one-compartment model with first-order elimination best described the data. Allometric scaling of weight and a factor of busulfan metabolism maturation were included in the base model. GSTA1 diplotypes were found to be a significant covariate of busulfan clearance, which was 7% faster in rapid metabolizers and 12% slower in poor metabolizers, in comparison with normal ones. Busulfan doses calculated using the parameters of the proposed PopPK model were estimated to achieve the target AUC in 85.2% of the cases (95% CI 78.7-91.7%). CONCLUSION This is the first PopPK for busulfan that successfully incorporated GSTA1 genotype in a paediatric population. Its use may contribute to better prediction of busulfan exposure in children and adolescents since the first dose, by tailoring the dose according to the individual metabolic capacity.
Collapse
Affiliation(s)
- Tiago Nava
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,CANSEARCH Research Laboratory, Department of Pediatrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pediatrics, Onco-Hematology Unit, University Hospital of Geneva, Geneva, Switzerland.,Post Graduate Program in Child and Adolescent Health, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nastya Kassir
- Certara Strategic Consulting, Montreal, Quebec, Canada
| | - Mohamed Aziz Rezgui
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Chakradhara Rao Satyanarayana Uppugunduri
- CANSEARCH Research Laboratory, Department of Pediatrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pediatrics, Onco-Hematology Unit, University Hospital of Geneva, Geneva, Switzerland
| | - Patricia Huezo-Diaz Curtis
- CANSEARCH Research Laboratory, Department of Pediatrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pediatrics, Onco-Hematology Unit, University Hospital of Geneva, Geneva, Switzerland
| | - Michel Duval
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Yves Théoret
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,Clinical Pharmacology Unit, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec, Canada.,Department of Pharmacology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Liane E Daudt
- Post Graduate Program in Child and Adolescent Health, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Catherine Litalien
- Clinical Pharmacology Unit, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Marc Ansari
- CANSEARCH Research Laboratory, Department of Pediatrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Pediatrics, Onco-Hematology Unit, University Hospital of Geneva, Geneva, Switzerland
| | - Maja Krajinovic
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,Clinical Pharmacology Unit, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec, Canada.,Department of Pharmacology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Henrique Bittencourt
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
44
|
Nava T, Rezgui MA, Uppugunduri CRS, Curtis PHD, Théoret Y, Duval M, Daudt LE, Ansari M, Krajinovic M, Bittencourt H. GSTA1 Genetic Variants and Conditioning Regimen: Missing Key Factors in Dosing Guidelines of Busulfan in Pediatric Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2017; 23:1918-1924. [PMID: 28807770 DOI: 10.1016/j.bbmt.2017.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/23/2017] [Indexed: 12/14/2022]
Abstract
Busulfan (Bu) is a key component of conditioning regimens used before hematopoietic stem cell transplantation (SCT) in children. Different predictive methods have been used to calculate the first dose of Bu. To evaluate the necessity of further improvements, we retrospectively analyzed the currently available weight- and age-based guidelines to calculate the first doses in 101 children who underwent allogenic SCT in CHU Sainte-Justine, Montreal, after an intravenous Bu-containing conditioning regimen according to genetic and clinical factors. The measured areas under the curve (AUCs) were within target (900 to 1500 µM/min) in 38.7% of patients after the administration of the first dose calculated based on age and weight, as locally recommended. GSTA1 diplotypes linked to poor Bu metabolism (G3) and fludarabine-containing regimens were the only factors associated with AUC within target (OR, 4.7 [95% CI, 1.1 to 19.8, P = .04]; and OR, 9.9 [95% CI, 1.6 to 61.7, P = .01], respectively). From the 11 methods selected for dose calculation, the percentage of AUCs within the target varied between 16% and 74%. In some models G3 was associated with AUCs within the therapeutic and the toxic range, whereas rapid metabolizers (G1) were correlated with subtherapeutic AUCs when different methods were used. These associations were confirmed by clearance-prediction analysis, in which GSTA1 diplotypes consistently influenced the prediction errors of the methods. These findings suggest that these factors should be considered in Bu dose prediction in addition to the anthropometric data from patients. Furthermore, our data indicated that GSTA1 diplotypes was a factor that should be included in future population pharmacokinetic models, including similar conditioning regiments, to improve the prediction of Bu exposure after its initial dose.
Collapse
Affiliation(s)
- Tiago Nava
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada; Department of Pediatrics, Faculty of Medicine, CANSEARCH Research Laboratory, Geneva, Switzerland; Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospital, Geneva University, Geneva, Switzerland; Post-Graduate Program in Child and Adolescent Health, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| | - Mohamed A Rezgui
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Chakradhara R S Uppugunduri
- Department of Pediatrics, Faculty of Medicine, CANSEARCH Research Laboratory, Geneva, Switzerland; Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospital, Geneva University, Geneva, Switzerland
| | - Patricia Huezo-Diaz Curtis
- Department of Pediatrics, Faculty of Medicine, CANSEARCH Research Laboratory, Geneva, Switzerland; Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospital, Geneva University, Geneva, Switzerland
| | - Yves Théoret
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada; Department of Pharmacology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada; Clinical Pharmacology Unit, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Michel Duval
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada; Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Liane E Daudt
- Post-Graduate Program in Child and Adolescent Health, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Marc Ansari
- Department of Pediatrics, Faculty of Medicine, CANSEARCH Research Laboratory, Geneva, Switzerland; Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospital, Geneva University, Geneva, Switzerland
| | - Maja Krajinovic
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada; Department of Pharmacology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada; Clinical Pharmacology Unit, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Henrique Bittencourt
- Department of Pediatrics, Charles-Bruneau Cancer Center, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada; Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|