1
|
Molinelli AR, Cross SJ, Leggas M. Recent Advances in Therapeutic Drug Monitoring of Antineoplastic and Antimicrobial Agents in Children. Clin Lab Med 2025; 45:315-327. [PMID: 40348442 DOI: 10.1016/j.cll.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Therapeutic drug monitoring (TDM) is used to optimize drug therapy by ensuring efficacy or preventing toxicity. For a limited number of cytotoxic antineoplastic drugs, for aminoglycoside antibiotics, and for vancomycin the use of TDM is common practice. In this article, we summarize recent advances and indications for the TDM of antineoplastic agents in children, focusing on protein kinase inhibitors and the cytotoxic drug fludarabine. We also summarize recent recommendations for antimicrobial TDM of beta-lactam antibiotics and vancomycin.
Collapse
Affiliation(s)
- Alejandro R Molinelli
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop #150, Memphis, TN 38105, USA.
| | - Shane J Cross
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop #150, Memphis, TN 38105, USA. https://twitter.com/shane6cross
| | - Markos Leggas
- Center for Translational Pharmacology, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 313, I-5104, Memphis, TN 38105, USA
| |
Collapse
|
2
|
Götz KM, Turki AT, Och K, Selzer D, Brossette C, Graf N, Rauch J, Theobald S, Braun Y, Rohm K, Weiler G, Rüdesheim S, Schwab M, Eisenberg L, Pfeifer N, Kiefer S, Schwarz U, Riede C, Smola S, Beelen DW, Kaddu‐Mulindwa D, Rissland J, Bittenbring J, Lehr T. Model-Based Prediction of Clinically Relevant Thrombocytopenia after Allogeneic Hematopoietic Stem Cell Transplantation. Clin Pharmacol Ther 2025; 117:1413-1426. [PMID: 39912597 PMCID: PMC11993296 DOI: 10.1002/cpt.3580] [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: 06/06/2024] [Accepted: 01/21/2025] [Indexed: 02/07/2025]
Abstract
Platelet reconstitution after allogeneic hematopoietic cell transplantation (allo-HCT) is heterogeneous and influenced by various patient- and transplantation-related factors, associated with poor prognoses for poor graft function (PGF) and isolated thrombocytopenia. Tailored interventions could improve the outcome of patients with PGF and post-HCT thrombocytopenia. To provide individual predictions of 180-day platelet counts from early phase data, we developed a model of long-term platelet reconstitution after allo-HCT. A large cohort (n = 1949) of adult patients undergoing their first allo-HCT was included. Real-world data from 1,048 retrospective patients were used for non-linear mixed-effects model development. Bayesian forecasting was used to predict platelet-time profiles for 518 retrospective and 383 prospective patients during internal and external model validation, respectively. Thrombocytopenia was defined as mean platelet count < 75 × 109/L, derived from the last 12 platelet measurements within the first 180 days post-HCT. Thrombocytopenia affected 37% of all patients and was associated with significantly reduced overall survival (P-value < 0.0001). On days +7, +14, +21, and +28, the developed model achieved areas under the receiver-operating characteristic of ≥ 0.68, ≥ 0.75, ≥ 0.78, and 0.81 for the prediction of post-HCT thrombocytopenia, respectively, with anti-thymocyte globulin, donor relation, and total protein measurements representing prognostic markers for post-HCT platelet kinetics. A publicly accessible web-based demonstrator of the model was established (https://hsct.precisiondosing.de). In summary, the developed model predicts individual platelet counts from day +28 post-HCT adequately, utilizing internal and external datasets. The web-based demonstrator provides a basis to implement model-based predictions in clinical practice and to confirm these findings in future clinical studies.
Collapse
Affiliation(s)
- Katharina M. Götz
- Department of Clinical PharmacySaarland UniversitySaarbrückenGermany
| | - Amin T. Turki
- Department of Hematology and Stem Cell TransplantationUniversity Hospital EssenEssenGermany
- Department of Hematology and Oncology, Marienhospital University HospitalRuhr‐University BochumBochumGermany
| | - Katharina Och
- Department of Clinical PharmacySaarland UniversitySaarbrückenGermany
| | - Dominik Selzer
- Department of Clinical PharmacySaarland UniversitySaarbrückenGermany
| | - Christian Brossette
- Department of Pediatric Oncology and HematologySaarland UniversityHomburgGermany
| | - Norbert Graf
- Department of Pediatric Oncology and HematologySaarland UniversityHomburgGermany
| | - Jochen Rauch
- Department of Biomedical Data & BioethicsFraunhofer Institute for Biomedical Engineering (IBMT)SulzbachGermany
| | - Stefan Theobald
- Department of Pediatric Oncology and HematologySaarland UniversityHomburgGermany
| | - Yvonne Braun
- Department of Pediatric Oncology and HematologySaarland UniversityHomburgGermany
| | - Kerstin Rohm
- Department of Biomedical Data & BioethicsFraunhofer Institute for Biomedical Engineering (IBMT)SulzbachGermany
| | - Gabriele Weiler
- Department of Biomedical Data & BioethicsFraunhofer Institute for Biomedical Engineering (IBMT)SulzbachGermany
| | - Simeon Rüdesheim
- Department of Clinical PharmacySaarland UniversitySaarbrückenGermany
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgartGermany
| | - Matthias Schwab
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgartGermany
- Department of Clinical Pharmacology, Pharmacy and BiochemistryUniversity of TübingenTübingenGermany
- Cluster of Excellence iFIT (EXC2180) “Image‐guided and Functionally Instructed Tumor Therapies”University of TübingenTübingenGermany
| | - Lisa Eisenberg
- Department of Computer ScienceUniversity of TübingenTübingenGermany
| | - Nico Pfeifer
- Department of Computer ScienceUniversity of TübingenTübingenGermany
| | - Stephan Kiefer
- Department of Biomedical Data & BioethicsFraunhofer Institute for Biomedical Engineering (IBMT)SulzbachGermany
| | - Ulf Schwarz
- Institute for Formal Ontology and Medical Information Science (IFOMIS)Saarland UniversitySaarbrückenGermany
| | | | - Sigrun Smola
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | - Dietrich W. Beelen
- Department of Hematology and Stem Cell TransplantationUniversity Hospital EssenEssenGermany
| | | | - Jürgen Rissland
- Institute of VirologySaarland University Medical CenterHomburgGermany
| | - Jörg Bittenbring
- Department of Internal Medicine 1University Hospital SaarlandHomburgGermany
| | - Thorsten Lehr
- Department of Clinical PharmacySaarland UniversitySaarbrückenGermany
| |
Collapse
|
3
|
Boardman AP, Gutgarts V, Flynn J, Devlin SM, Goldman A, Tomas AA, Fein JA, Slingerland JB, Parascondola A, Lin RJ, Scordo M, Dahi PB, Giralt S, Palomba ML, Salles G, Nath K, Walji M, Corona M, Park JH, Shah GL, Perales MA, Jaffer-Sathick I, Shouval R. Predictors and implications of renal injury after CD19 chimeric antigen receptor T-cell therapy. Haematologica 2025; 110:651-664. [PMID: 39568416 PMCID: PMC11873692 DOI: 10.3324/haematol.2024.286021] [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: 06/06/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19 induce durable remissions in patients with relapsed or refractory non-Hodgkin lymphoma (NHL), but many patients experience treatment-related toxicity. Cytokine release syndrome and immune effector cell-associated neurologic syndrome are extensively characterized. However, limited data exist on the burden, predictors, and implications of acute kidney injury (AKI) after CAR T-cell therapy. On initial screening of the Food and Drug Administration adverse event reporting system, we identified a disproportionately high rate of renal adverse events among nearly 6,000 CAR T adverse event reports, suggesting it is clinically important in this patient population. In a subsequent single-center analysis of 399 NHL patients treated with CD19 CAR T cells, we found a substantial burden of AKI after CAR T infusion (10% and 5% of any grade and grade ≥2 AKI) with pre-renal causes being predominant (72%). Evolution to chronic kidney disease was rare, however, three patients required hemodialysis. Importantly, patients experiencing cytokine release syndrome and/or neurotoxicity as well as those with low serum albumin and high inflammatory cytokines, including IL-6 and TNF-α, were more likely to develop AKI. While pre-CAR T renal dysfunction was not associated with adverse outcomes, patients developing post-CAR T AKI had lower overall survival compared to their counterparts. Our findings indicate that renal dysfunction is a common toxicity of CAR T-cell therapy with meaningful prognostic impact. Notably, the link between systemic inflammation and renal dysfunction, suggests that readily available biomarkers may inform on renal injury risk after CAR T-cell therapy.
Collapse
Affiliation(s)
- Alexander P Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Victoria Gutgarts
- Department of Medicine, Weill Cornell Medical College, New York, NY; Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Adam Goldman
- Department of Medicine, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Aviv University, Israel
| | - Ana Alarcon Tomas
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Hematology and Hemotherapy Service, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Joshua A Fein
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - John B Slingerland
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Allison Parascondola
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Richard J Lin
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Michael Scordo
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Parastoo B Dahi
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Sergio Giralt
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - M Lia Palomba
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Gilles Salles
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Karthik Nath
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Moneeza Walji
- Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Magdalena Corona
- Hematology and Hemotherapy Service, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Jae H Park
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York
| | - Gunjan L Shah
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Miguel-Angel Perales
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Insara Jaffer-Sathick
- Department of Medicine, Weill Cornell Medical College, New York, NY; Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Roni Shouval
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medicine, Weill Cornell Medical College, New York, NY; Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Aviv University, Israel.
| |
Collapse
|
4
|
Toksvang LN, Brigitha LJ, van der Sluis IM, Brivio E, Raja R, Pontoppidan P, Buhl Rasmussen AS, Andres-Jensen L, Uhlving HH, Kielsen K, Als-Nielsen B, Elitzur S, Dalhoff K, Schmiegelow K, Rank CU. Therapeutic drug monitoring in acute lymphoblastic leukemia-a deep dive into pharmacokinetics, -dynamics, and -genetics of antileukemic drugs. Expert Rev Clin Pharmacol 2025; 18:131-149. [PMID: 39949259 DOI: 10.1080/17512433.2025.2465426] [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: 10/30/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Therapeutic drug monitoring (TDM) is important to optimize drug exposure and minimize toxicity for the individual patient. AREAS COVERED This narrative review covers the pharmacokinetics (PK), -dynamics (PD) and -genetics of classic chemotherapeutic drugs used in frontline therapy for acute lymphoblastic leukemia (ALL), including anthracyclines, asparaginase, busulfan, cyclophosphamide, cytarabine, glucocorticoids, methotrexate, nelarabine, thiopurines, tyrosine kinase inhibitors, and vincristine. Furthermore, novel immunotherapies including blinatumomab, inotuzumab ozogamicin, and chimeric antigen receptor T-cells that are rapidly moving into frontline therapy are addressed. This review focuses on TDM already used in clinical practice as well as the unused potential and feasibility of TDM. Finally, important factors affecting PK/PD such as obesity and transition to adolescence and young adulthood are discussed. EXPERT OPINION Investigation of TDM as standard of care for antileukemic agents is highly warranted to personalize curative yet toxic anticancer regimens within frontline ALL treatment. Some of the drugs have been used in ALL treatment regimens for decades, but a wide range of new compounds are being introduced, some like blinatumomab reaching standard-of-care designation. Not least, optimized drug efficacy and reduction of the risk of serious toxicities may render TDM implementation cost-effective.
Collapse
Affiliation(s)
- Linea N Toksvang
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Leiah J Brigitha
- Hemato-oncology Department, Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Inge M van der Sluis
- Hemato-oncology Department, Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Erica Brivio
- Hemato-oncology Department, Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Raheel Raja
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Peter Pontoppidan
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anna S Buhl Rasmussen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Liv Andres-Jensen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hilde Hylland Uhlving
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Katrine Kielsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Bodil Als-Nielsen
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Sarah Elitzur
- Pediatric Hematology-Oncology, Schneider Children's Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Kim Dalhoff
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg University Hospital, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Utke Rank
- Department of Hematology, University Hospital Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
5
|
Möhlmann J, van der Ploeg L, Langenhorst J, Bognàr T, van der Elst K, Bierings M, Huitema A, de Vries Schultink A, Lindemans C. Evaluation of standard fludarabine dosing and corresponding exposures in infants and young children undergoing hematopoietic cell transplantation. Bone Marrow Transplant 2025; 60:241-243. [PMID: 39537781 DOI: 10.1038/s41409-024-02467-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Julia Möhlmann
- Department of Clinical Pharmacy, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | - Lisanne van der Ploeg
- Department of Clinical Pharmacy, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | | | - Tim Bognàr
- Department of Clinical Pharmacy, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Kim van der Elst
- Department of Clinical Pharmacy, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Marc Bierings
- Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | - Alwin Huitema
- Department of Clinical Pharmacy, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands
- Department of Pharmacy and Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Aurelia de Vries Schultink
- Department of Clinical Pharmacy, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Caroline Lindemans
- Princess Máxima Centre for Paediatric Oncology, Utrecht, The Netherlands.
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
6
|
Scordo M. The promises and potential pitfalls of pharmacokinetic model-based dosing in cellular therapy. Blood Adv 2024; 8:6015-6016. [PMID: 39368808 PMCID: PMC11635718 DOI: 10.1182/bloodadvances.2024012688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Affiliation(s)
- Michael Scordo
- Adult Bone Marrow Transplant Service, Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
7
|
Nath CE, Rosser SPA, Nath KK, Chung J, Larsen S, Gibson J, Gabriel M, Shaw PJ, Keogh SJ. The impact of age and renal function on the pharmacokinetics and protein binding characteristics of fludarabine in paediatric and adult patients undergoing allogeneic haematopoietic stem cell transplantation conditioning. Eur J Clin Pharmacol 2024; 80:1967-1987. [PMID: 39298000 PMCID: PMC11557628 DOI: 10.1007/s00228-024-03751-0] [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: 11/24/2023] [Accepted: 08/27/2024] [Indexed: 09/21/2024]
Abstract
AIM To evaluate the population pharmacokinetics of unbound F-Ara-A (the circulating metabolite of fludarabine) in 211 patients (age range, 0.1-63.4 years) undergoing allogeneic haematopoietic stem cell transplantation conditioning. METHODS Total (n = 2480) and unbound (n = 1403) F-Ara-A concentrations were measured in blood samples collected at timed intervals after fludarabine doses ranging from 10 to 50 mg/m2 and infused over 0.42-1.5 h. A three-compartment population pharmacokinetic model was developed based on unbound plasma concentrations and used to estimate F-Ara-A unbound pharmacokinetic parameters and fraction unbound (fu). A number of covariates, including glomerular filtration rate (GFR) and post-menstrual age (PMA), were evaluated for inclusion in the model. RESULTS The base population mean estimates ± relative standard error (%RSE) for unbound clearance from the central compartment (CLu) and inter-compartmental clearances (Q2u, Q3u) were 3.42 ± 3%, 6.54 ± 24% and 1.47 ± 16% L/h/70 kg, respectively. The population mean estimates (%RSE) for the unbound volume of distribution into the central (V1u) and peripheral compartments (V2u, V3u) were 9.65 ± 8%, 8.17 ± 9% and 16.4 ± 10% L/70 kg, respectively, and that for fu was 0.877 ± 1%. Covariate model development involved differentiating F-Ara-A CLu into non-renal (1.81 ± 9% L/h/70 kg) and renal components (1.02 ± 9%*GFR L/h/70 kg). A sigmoidal maturation factor was applied to renal CLu, with population mean estimates for the Hill exponent and PMA at 50% mature of 2.97 ± 4% and 69.1 ± 8% weeks, respectively. CONCLUSION Patient age and GFR are predictors of unbound F-Ara-A CLu. This has the potential to impact dose requirements. Dose individualisation by target concentration intervention will be facilitated by this model once it is externally validated.
Collapse
Affiliation(s)
- Christa E Nath
- Department of Biochemistry, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia.
- The Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia.
- Sydney Pharmacy School, University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Sebastian P A Rosser
- Department of Biochemistry, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
- The Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
- The Children's Hospital at Westmead Clinical School, University of Sydney, Westmead, NSW, 2145, Australia
| | - Kiran K Nath
- School of Psychology, Western Sydney University, Kingswood, NSW, 2747, Australia
| | - Jason Chung
- Department of Biochemistry, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Stephen Larsen
- Royal Prince Alfred Hospital, Camperdown, NSW, 2006, Australia
| | - John Gibson
- Royal Prince Alfred Hospital, Camperdown, NSW, 2006, Australia
| | - Melissa Gabriel
- The Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Peter J Shaw
- The Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
- The Children's Hospital at Westmead Clinical School, University of Sydney, Westmead, NSW, 2145, Australia
| | - Steven J Keogh
- The Cancer Centre for Children, The Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| |
Collapse
|
8
|
Ziętara KJ, Wróblewska K, Zajączkowska M, Taczała J, Lejman M. The Role of the JAK-STAT Pathway in Childhood B-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:6844. [PMID: 38999955 PMCID: PMC11241568 DOI: 10.3390/ijms25136844] [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: 05/14/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
B-cell lymphoblastic leukemia is a hematologic neoplasm that poses a serious health concern in childhood. Genetic aberrations, such as mutations in the genes IL-7, IL7R, JAK1, JAK2, TLSP, CRLF2, and KTM2A or gene fusions involving BCR::ABL1, ETV6::RUNX1, and PAX5::JAK2, often correlate with the onset of this disease. These aberrations can lead to malfunction of the JAK-STAT signaling pathway, which is implicated in various important biological processes, including those related to immunology. Understanding the mechanisms underlying the malfunction of the JAK-STAT pathway holds potential for research on drugs targeting its components. Available drugs that interfere with the JAK-STAT pathway include fludarabine, ruxolitinib, and fedratinib.
Collapse
Affiliation(s)
- Karolina Joanna Ziętara
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (K.J.Z.); (K.W.); (M.Z.)
| | - Kinga Wróblewska
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (K.J.Z.); (K.W.); (M.Z.)
| | - Monika Zajączkowska
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland; (K.J.Z.); (K.W.); (M.Z.)
| | - Joanna Taczała
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warszawa, Poland;
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
9
|
Frame DG, Geer M, Kasha S, Markstrom D, Scappaticci G, Feeney T, Hayduk A, Mansoor HM, Oberfeld A, D’Antonio H, Anand S, Choi SW, Maciejewski J, Pawarode A, Riwes MM, Tewari M, Magenau J, Ghosh M. Comparing 2-day vs 3-day flu-CY lymphodepleting regimens for CD19 CAR T-cell therapy in patients with non-hodgkin's lymphoma. Front Immunol 2024; 15:1403145. [PMID: 38947326 PMCID: PMC11211265 DOI: 10.3389/fimmu.2024.1403145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction Lymphodepleting chemotherapy (LDC) is critical to CAR T-cell expansion and efficacy. Despite this, there is not a consensus in the literature regarding the optimal LDC regimen, including dose and frequency. Methods We retrospectively reviewed consecutive patients at a single institution that received LDC prior to treatment with the CD19 directed CAR T-cell products axicabtagene ciloleucel and tisagenlecleucel. Patients treated at our center received fludarabine 30 mg/m2 and cyclophosphamide 500 mg/m2 for 3 consecutive days prior to May 2019. After this timepoint patients routinely received fludarabine 40 mg/m2 and cyclophosphamide 500 mg/m2 for 2 consecutive days. Clinical data from each cohort were obtained from the electronic medical record and compared for differences in CAR T-cell efficacy and toxicity. Results From June 2018 to August 2023, LDC was given to 92 patients prior to CD19 directed CAR T-cell therapy for relapsed non-Hodgkin's lymphoma. Twenty-eight patients received a 3-day regimen, and 64 patients received a 2-day regimen. In the total cohort, 75% of patients received axicabtagene ciloleucel and 25% received tisagenlecleucel. The overall response rates in both the 2-day regimen group and the 3-day regimen group were similar (69% vs 75%, p= 0.21) as were the complete response rates (50% vs 54%, p=0.82). There were no significant differences between the 2-day and 3-day regimens for grade 2-4 cytokine release syndrome (55% vs 50%, p=0.82), grade 2-4 immune effector cell associated-neurotoxicity syndrome (42% vs 29%, p=0.25), or time to resolution of neutropenia or thrombocytopenia. The rate of prolonged platelet recovery lasting greater than 60 days was higher with the 3-day regimen (9% vs 27%, p=0.026). Discussion As the number of patients eligible for CAR T-cell therapy continues to increase, optimizing each component of therapy is necessary. We show that a 2-day regimen of LDC with fludarabine and cyclophosphamide is feasible without significant impact on CAR T-cell efficacy or toxicity. Prospective studies are necessary to further determine the most effective LDC regimen.
Collapse
Affiliation(s)
- David G. Frame
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Marcus Geer
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Salena Kasha
- School of Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Denise Markstrom
- Department of Pharmacy Services and Clinical Pharmacy, Michigan Medicine, Ann Arbor, MI, United States
| | - Gianni Scappaticci
- Department of Pharmacy Services and Clinical Pharmacy, Michigan Medicine, Ann Arbor, MI, United States
| | - Tate Feeney
- Department of Pharmacy Services and Clinical Pharmacy, Michigan Medicine, Ann Arbor, MI, United States
| | - Andrew Hayduk
- School of Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Hilary M. Mansoor
- School of Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Avery Oberfeld
- School of Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Hannah D’Antonio
- School of Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Sarah Anand
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Sung Won Choi
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - John Maciejewski
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Attaphol Pawarode
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Mary Mansour Riwes
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Muneesh Tewari
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - John Magenau
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Monalisa Ghosh
- Department of Internal Medicine, College of Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Sánchez-Salinas MA, Miarons M, Troconiz IF, Navarro V, Varela J, Iacoboni G, Barba P. Is estimated exposure an accurate surrogate for measured fludarabine levels in patients with CAR T-cell therapy? Blood Adv 2024; 8:2130-2132. [PMID: 38231085 PMCID: PMC11059335 DOI: 10.1182/bloodadvances.2023011433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/18/2024] Open
Affiliation(s)
- Mario Andrés Sánchez-Salinas
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Marta Miarons
- Pharmacy Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Pharmacy Department, Consorci Hospitalari de Vic, Barcelona, Spain
| | - Iñaki F. Troconiz
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Víctor Navarro
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Javier Varela
- Pharmacy Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Gloria Iacoboni
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Pere Barba
- Department of Hematology, Vall d'Hebron University Hospital, Experimental Hematology, Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
11
|
Salvino MA, Mussetti A, Peña M, Paviglianiti A, Carreira AS, Rizky D, Sureda A. CAR T-cell therapy and the onco-nephrologist. FRONTIERS IN NEPHROLOGY 2024; 4:1378250. [PMID: 38706889 PMCID: PMC11066316 DOI: 10.3389/fneph.2024.1378250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
Cell therapy, specifically the revolutionary chimeric antigen receptor (CAR) T-cell therapy, has transformed the landscape of oncology, making substantial strides in practical treatment approaches. Today, established guidelines for diseases such as lymphomas, myelomas, and leukemias actively advocate the utilization of these once-unconventional therapies. The practical impact of these therapies is underscored by their unparalleled efficacy, reshaping the way we approach and implement treatments in the realm of oncology. However, CAR T-cell therapy, with its performance in anti-tumor aggression through cellular action and inflammatory response, also comes with various adverse events, one of which is kidney injury. Therefore, the management of these side effects is extremely important. The integration of knowledge between oncologists and specialized nephrologists has led to the emergence of a new sub-area of expertise for onco-nephrologists specializing in managing kidney complications from immune effector therapies.
Collapse
Affiliation(s)
- Marco Aurelio Salvino
- Programa Pos Graduacao Medicina Saude (PPGMS), Universidade Federal da Bahia, Salvador, Brazil
- L’Hospitalet, Institut Català de Oncologia, Barcelona, Spain
- Hematology Department, Instituto D´or de Pesquisa e Ensino-Bahia (IDOR Ba), Salvador, Brazil
| | | | - Marta Peña
- L’Hospitalet, Institut Català de Oncologia, Barcelona, Spain
| | | | | | - Daniel Rizky
- L’Hospitalet, Institut Català de Oncologia, Barcelona, Spain
- Hematology Medical Oncology, Dr. Kariadi General Hospital, Semarang, Indonesia
| | - Anna Sureda
- L’Hospitalet, Institut Català de Oncologia, Barcelona, Spain
- Institut d’Investigació Biomédica de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Knight E T, Oluwole O, Kitko C. The Implementation of Chimeric Antigen Receptor (CAR) T-cell Therapy in Pediatric Patients: Where Did We Come From, Where Are We Now, and Where are We Going? Clin Hematol Int 2024; 6:96-115. [PMID: 38817691 PMCID: PMC11108586 DOI: 10.46989/001c.94386] [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: 01/17/2024] [Accepted: 02/13/2024] [Indexed: 06/01/2024] Open
Abstract
CD19-directed Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized the treatment of patients with B-cell acute lymphoblastic leukemia (B-ALL). Somewhat uniquely among oncologic clinical trials, early clinical development occurred simultaneously in both children and adults. In subsequent years however, the larger number of adult patients with relapsed/refractory (r/r) malignancies has led to accelerated development of multiple CAR T-cell products that target a variety of malignancies, resulting in six currently FDA-approved for adult patients. By comparison, only a single CAR-T cell therapy is approved by the FDA for pediatric patients: tisagenlecleucel, which is approved for patients ≤ 25 years with refractory B-cell precursor ALL, or B-cell ALL in second or later relapse. Tisagenlecleucel is also under evaluation in pediatric patients with relapsed/refractory B-cell non-Hodgkin lymphoma, but is not yet been approved for this indication. All the other FDA-approved CD19-directed CAR-T cell therapies available for adult patients (axicabtagene ciloleucel, brexucabtagene autoleucel, and lisocabtagene maraleucel) are currently under investigations among children, with preliminary results available in some cases. As the volume and complexity of data continue to grow, so too does the necessity of rapid assimilation and implementation of those data. This is particularly true when considering "atypical" situations, e.g. those arising when patients do not precisely conform to the profile of those included in pivotal clinical trials, or when alternative treatment options (e.g. hematopoietic stem cell transplantation (HSCT) or bispecific T-cell engagers (BITEs)) are also available. We have therefore developed a relevant summary of the currently available literature pertaining to the use of CD19-directed CAR-T cell therapies in pediatric patients, and sought to provide guidance for clinicians seeking additional data about specific clinical situations.
Collapse
Affiliation(s)
| | - Olalekan Oluwole
- Medicine Hematology and Oncology, Vanderbilt University Medical Center
| | | |
Collapse
|
13
|
Wicha SG, Wansing EMA, Dadkhah A, Ayuk FA, Kröger NM, Langebrake C. Chimeric antigen receptor T-cell therapy and fludarabine: precision dosing imperatives. Blood Adv 2024; 8:797-798. [PMID: 38191740 PMCID: PMC10847728 DOI: 10.1182/bloodadvances.2023012068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/15/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024] Open
Affiliation(s)
- Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Eva M. A. Wansing
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, Hospital Pharmacy, Hamburg, Germany
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Adrin Dadkhah
- University Medical Center Hamburg-Eppendorf, Hospital Pharmacy, Hamburg, Germany
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis A. Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus M. Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Langebrake
- University Medical Center Hamburg-Eppendorf, Hospital Pharmacy, Hamburg, Germany
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Ligon JA, Ramakrishna S, Ceppi F, Calkoen FGJ, Diorio C, Davis KL, Jacoby E, Gottschalk S, Schultz LM, Capitini CM. INSPIRED Symposium Part 4B: Chimeric Antigen Receptor T Cell Correlative Studies-Established Findings and Future Priorities. Transplant Cell Ther 2024; 30:155-170. [PMID: 37863355 PMCID: PMC12047531 DOI: 10.1016/j.jtct.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of B cell malignancies, with multiple CAR T cell products approved for numerous indications by regulatory agencies worldwide. However, significant work remains to be done to enhance these treatments. In March 2023, a group of experts in CAR T cell therapy assembled at the National Institutes of Health in Bethesda, Maryland at the Insights in Pediatric CAR T Cell Immunotherapy: Recent Advances and Future Directions (INSPIRED) Symposium to identify key areas for research for the coming years. In session 4B, correlative studies to be incorporated into future clinical trials and real-world settings were discussed. Active areas of research identified included (1) optimizing CAR T cell product manufacturing; (2) ensuring adequate lymphodepletion prior to CAR T cell administration; (3) overcoming immunoregulatory cells and tumor stroma present in the tumor microenvironment, particularly in solid tumors; (4) understanding tumor intrinsic properties that lead to CAR T cell immunotherapy resistance; and (5) uncovering biomarkers predictive of treatment resistance, treatment durability, or immune-related adverse events. Here we review the results of previously published clinical trials and real-world studies to summarize what is currently known about each of these topics. We then outline priorities for future research that we believe will be important for improving our understanding of CAR T cell therapy and ultimately leading to better outcomes for patients.
Collapse
Affiliation(s)
- John A Ligon
- Department of Pediatrics, Division of Hematology/Oncology, University of Florida, Gainesville, Florida; University of Florida Health Cancer Center, Gainesville, Florida.
| | - Sneha Ramakrishna
- Stanford Center for Cancer Cell Therapy, Stanford University School of Medicine, Stanford, California; Department of Pediatrics, Stanford University, Stanford, California
| | - Francesco Ceppi
- Division of Pediatrics, Department of Woman-Mother-Child, Pediatric Hematology-Oncology Unit, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Friso G J Calkoen
- Division of Pediatric Oncology, Princess Maxima Center, Utrecht, The Netherlands
| | - Caroline Diorio
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kara L Davis
- Stanford Center for Cancer Cell Therapy, Stanford University School of Medicine, Stanford, California; Department of Pediatrics, Stanford University, Stanford, California
| | - Elad Jacoby
- Pediatric Hemato-Oncology, Sheba Medical Center and Tel Aviv University, Tel Aviv, Israel
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Liora M Schultz
- Stanford Center for Cancer Cell Therapy, Stanford University School of Medicine, Stanford, California; Department of Pediatrics, Stanford University, Stanford, California
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| |
Collapse
|
15
|
Hutcherson SM, Schofield RC, Carlow DC. Determination of Treosulfan and Fludarabine in Plasma by Turbulent Flow Liquid Chromatography-Tandem Mass Spectrometry (TFLC-MS/MS). Methods Mol Biol 2024; 2737:453-463. [PMID: 38036846 DOI: 10.1007/978-1-0716-3541-4_42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Treosulfan is a structural analog of the alkylating agent busulfan which has been shown in clinical trials to exhibit comparable myeloablative activity while causing fewer serious side effects. Treosulfan is currently being considered for FDA approval in combination with fludarabine, one of the most commonly used myeloablative agents, as a conditioning regimen prior to hematopoietic stem cell transplantation (HSCT). Because plasma concentrations of both treosulfan and fludarabine exhibit significant interindividual variability, therapeutic drug monitoring (TDM) is indicated to ensure dosages are administered that maximize efficacy while minimizing toxicity. In this chapter, we describe a rapid, accurate assay to detect treosulfan and fludarabine simultaneously in human plasma using turbulent flow liquid chromatography coupled to electrospray ionization tandem mass spectrometry (TFLC-ESI-MS/MS). Treosulfan and fludarabine are extracted from only 100 μL of acidified plasma via protein precipitation with methanol containing isotope-labeled internal standards. The extract is injected into the TFLC-ESI-MS/MS system, and the analytes are quantified using multiple reaction monitoring and a six-point calibration curve.
Collapse
Affiliation(s)
- Shelby M Hutcherson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan C Schofield
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dean C Carlow
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
16
|
Schofield RC, Scordo M, Shah G, Carlow DC. Quantification of Clofarabine and Fludarabine in Plasma by High-Performance Liquid Chromatography-Tandem Mass Spectrometry. Methods Mol Biol 2024; 2737:175-184. [PMID: 38036821 DOI: 10.1007/978-1-0716-3541-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative therapeutic treatment for many patients with high-risk hematologic malignancies and bone marrow failure syndromes. While allo-HCT can be highly effective, it is met with significant bone marrow conditioning regimen-related toxicities and complications such as infections related to poor immune reconstitution. This chapter describes the measurement of clofarabine and fludarabine concentrations to support clinical trials whose goal is to determine the optimal therapeutic ranges in order to maximize effectiveness while minimizing variability and regimen-related adverse events and toxicities. Moreover, the same holds true for patients receiving fludarabine as part of their lymphodepleting chemotherapy for chimeric antigen receptor T-cells (CAR T-cells). It is believed that one of the causes of variable outcomes after CAR T-cell therapy is lymphodepletion due to the variable fludarabine concentrations.This chapter describes a HPLC-MS/MS method to measure both compounds simultaneously. Clofarabine and fludarabine are extracted with solvent from plasma by the addition of deuterated internal standards prepared in methanol. Chromatographic separation is attained using a reversed-phase column followed by mass spectrometry which is performed in the positive ion mode. Herein, the described method to quantify both compounds in plasma is fast, accurate, and sensitive and allows for rapid drug concentration monitoring and timely dose adjustments.
Collapse
Affiliation(s)
- Ryan C Schofield
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Gunjan Shah
- Adult Bone Marrow Transplant Service, Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Dean C Carlow
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
17
|
Lickefett B, Chu L, Ortiz-Maldonado V, Warmuth L, Barba P, Doglio M, Henderson D, Hudecek M, Kremer A, Markman J, Nauerth M, Negre H, Sanges C, Staber PB, Tanzi R, Delgado J, Busch DH, Kuball J, Luu M, Jäger U. Lymphodepletion - an essential but undervalued part of the chimeric antigen receptor T-cell therapy cycle. Front Immunol 2023; 14:1303935. [PMID: 38187393 PMCID: PMC10770848 DOI: 10.3389/fimmu.2023.1303935] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Lymphodepletion (LD) or conditioning is an essential step in the application of currently used autologous and allogeneic chimeric antigen receptor T-cell (CAR-T) therapies as it maximizes engraftment, efficacy and long-term survival of CAR-T. Its main modes of action are the depletion and modulation of endogenous lymphocytes, conditioning of the microenvironment for improved CAR-T expansion and persistence, and reduction of tumor load. However, most LD regimens provide a broad and fairly unspecific suppression of T-cells as well as other hematopoietic cells, which can also lead to severe side effects, particularly infections. We reviewed 1271 published studies (2011-2023) with regard to current LD strategies for approved anti-CD19 CAR-T products for large B cell lymphoma (LBCL). Fludarabine (Flu) and cyclophosphamide (Cy) (alone or in combination) were the most commonly used agents. A large number of different schemes and combinations have been reported. In the respective schemes, doses of Flu and Cy (range 75-120mg/m2 and 750-1.500mg/m2) and wash out times (range 2-5 days) differed substantially. Furthermore, combinations with other agents such as bendamustine (benda), busulfan or alemtuzumab (for allogeneic CAR-T) were described. This diversity creates a challenge but also an opportunity to investigate the impact of LD on cellular kinetics and clinical outcomes of CAR-T. Only 21 studies explicitly investigated in more detail the influence of LD on safety and efficacy. As Flu and Cy can potentially impact both the in vivo activity and toxicity of CAR-T, a more detailed analysis of LD outcomes will be needed before we are able to fully assess its impact on different T-cell subsets within the CAR-T product. The T2EVOLVE consortium propagates a strategic investigation of LD protocols for the development of optimized conditioning regimens.
Collapse
Affiliation(s)
- Benno Lickefett
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Lulu Chu
- Cell Therapy Clinical Pharmacology and Modeling, Takeda, Boston, MA, United States
| | | | - Linda Warmuth
- Institut für Med. Mikrobiologie, Immunologie und Hygiene, Technische Universität Munich, Munich, Germany
| | - Pere Barba
- Hematology Department, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Matteo Doglio
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - David Henderson
- Bayer Aktiengesellschaft (AG), Business Development & Licensing & Open Innovation (OI), Pharmaceuticals, Berlin, Germany
| | - Michael Hudecek
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Andreas Kremer
- ITTM S.A. (Information Technology for Translational Medicine), Esch-sur-Alzette, Luxembourg
| | - Janet Markman
- Cell Therapy Clinical Pharmacology and Modeling, Takeda, Boston, MA, United States
| | - Magdalena Nauerth
- Institut für Med. Mikrobiologie, Immunologie und Hygiene, Technische Universität Munich, Munich, Germany
| | - Helene Negre
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Carmen Sanges
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Philipp B. Staber
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Rebecca Tanzi
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Julio Delgado
- Department of Hematology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Dirk H. Busch
- Institut für Med. Mikrobiologie, Immunologie und Hygiene, Technische Universität Munich, Munich, Germany
| | - Jürgen Kuball
- Legal and Regulatory Affairs Committee of the European Society for Blood and Marrow Transplantation, Leiden, Netherlands
| | - Maik Luu
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Ulrich Jäger
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Filioglou D, Husnain M, Khurana S, Simpson RJ, Katsanis E. Has the shortage of fludarabine altered the current paradigm of lymphodepletion in favor of bendamustine? Front Immunol 2023; 14:1329850. [PMID: 38077398 PMCID: PMC10702755 DOI: 10.3389/fimmu.2023.1329850] [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: 10/30/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
The most common lymphodepletion regimen used prior to infusion of chimeric antigen receptor-T cells (CAR-T) is cyclophosphamide (CY) in combination with fludarabine (Flu) (CY-FLU). While cyclophosphamide (CY) possesses lymphotoxic effects, it concurrently preserves regulatory T cell activity, potentially affecting the efficacy of CAR-T cells. Moreover, the use of fludarabine (FLU) has been linked to neurotoxicity, which could complicate the early detection of immune effector cell-associated neurotoxicity syndrome (ICANS) observed in CAR-T cell therapy. Given the ongoing shortage of FLU, alternative lymphodepleting agents have become necessary. To date, only a limited number of studies have directly compared different lymphodepleting regimens, and most of these comparisons have been retrospective in nature. Herein, we review the current literature on lymphodepletion preceding CAR-T cell therapies for lymphoid hematologic malignancies, with a specific focus on the use of bendamustine (BEN). Recent evidence suggests that administering BEN before CAR-T cell infusion yields comparable efficacy, possibly with a more favorable toxicity profile when compared to CY-FLU. This warrants further investigation through randomized prospective studies.
Collapse
Affiliation(s)
| | - Muhammad Husnain
- Department of Medicine, University of Arizona, Tucson, AZ, United States
- The University of Arizona Cancer Center, Tucson, AZ, United States
| | - Sharad Khurana
- Department of Medicine, University of Arizona, Tucson, AZ, United States
- The University of Arizona Cancer Center, Tucson, AZ, United States
| | - Richard J. Simpson
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States
| | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
- Department of Medicine, University of Arizona, Tucson, AZ, United States
- The University of Arizona Cancer Center, Tucson, AZ, United States
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States
- Department of Pathology, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
19
|
Li X, Kalwak K, Beier R, Kehne J, Möller AK, Baumgart J, Beelen DW, Hilger RA, Vora A, Sykora KW. Population pharmacokinetic modeling of treosulfan and rationale for dose recommendation in children treated for conditioning prior to allogeneic hematopoietic stem cell transplantation. Drug Metab Pharmacokinet 2023; 52:100515. [PMID: 37481830 DOI: 10.1016/j.dmpk.2023.100515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/10/2023] [Accepted: 05/21/2023] [Indexed: 07/25/2023]
Abstract
Intravenously infused treosulfan was evaluated in adult and pediatric patients for conditioning regimen prior to allogeneic hematopoietic stem cell transplantation. A population pharmacokinetic (PK) model was initially developed on 116 adult and pediatric PK profiles from historical trials, to support treosulfan dose recommendations for children in 2 prospective trials. The aim was to assess and update the initial population PK model by inclusion of additional 83 pediatric PK profiles from these 2 trials. The final population PK model was 2-compartmental with dosing in the central compartment, linear elimination, and inter-compartmental clearance. Inter-individual variability was included on clearance (CL), central volume (V1), peripheral volume (V2), and inter-compartmental clearance (Q). The final model described an effect of the body surface area (BSA) on CL, V1, V2, and Q. The final model resulted in a modified dose recommendation for children and advises treosulfan doses of 10 g/m2, 12 g/m2, and 14 g/m2 for BSAs of <0.4 m2, ≥0.4 to <0.9 m2, and ≥0.9 m2, respectively. This simplified BSA-dependent dose recommendation was developed for children, ensuring a well comparable treosulfan exposure as a dose of 14 g/m2 in adults - irrespective of their age and without applying individual therapeutic drug monitoring.
Collapse
Affiliation(s)
- Xieran Li
- medac GmbH, Theaterstraße 6, 22880, Wedel, Germany.
| | - Krzysztof Kalwak
- Wroclaw Medical University, Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Wybrzeze Ludwika Pasteura 1, 50-367, Wroclaw, Poland
| | - Rita Beier
- Hannover Medical School, Department of Paediatric Haematology and Oncology, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Jochen Kehne
- medac GmbH, Theaterstraße 6, 22880, Wedel, Germany
| | | | | | - Dietrich W Beelen
- University Hospital Essen, Department of Haematology and Stem Cell Transplantation, West-German Cancer Centre, Hufelandstraße 55, 45147, Essen, Germany
| | - Ralf A Hilger
- University Hospital Essen, West-German Cancer Center, Department of Medical Oncology, Hufelandstraße 55, 45147, Essen, Germany
| | - Ajay Vora
- Great Ormond Street Hospital for Children NHS Foundation, Great Ormond Street, WC1N 3JH, London, United Kingdom
| | - Karl-Walter Sykora
- Hannover Medical School, Department of Paediatric Haematology and Oncology, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| |
Collapse
|
20
|
Scordo M, Flynn JR, Gonen M, Devlin SM, Parascondola A, Tomas AA, Shouval R, Brower J, Porter DL, Schuster SJ, Bachanova V, Maakaron J, Maziarz RT, Chen AI, Nastoupil LJ, McGuirk JP, Oluwole OO, Ip A, Leslie LA, Bishop MR, Riedell PA, Perales MA. Identifying an optimal fludarabine exposure for improved outcomes after axi-cel therapy for aggressive B-cell non-Hodgkin lymphoma. Blood Adv 2023; 7:5579-5585. [PMID: 37522731 PMCID: PMC10514205 DOI: 10.1182/bloodadvances.2023010302] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/01/2023] Open
Abstract
Fludarabine is one of the most common agents given for lymphodepletion before CD19 chimeric antigen receptor T cells, but its optimal therapeutic intensity is unknown. Using data from a multicenter consortium, we estimated fludarabine exposure (area under the curve [AUC]) using a population pharmacokinetic (PK) model in 199 adult patients with aggressive B-cell non-Hodgkin lymphomas who received commercial axicabtagene ciloleucel (Axi-cel). We evaluated the association of estimated fludarabine AUC with key outcomes, aiming to find an AUC that optimized efficacy and tolerability. We identified low (<18 mg × hour/L [mgh/L]), optimal (18-20 mgh/L), and high (>20 mgh/L) AUC groups for analyses; the 6-month cumulative incidences of relapse/progression of disease (relapse/POD) by AUC groups were 54% (45%-62%), 28% (15%-44%), and 30% (14%-47%), respectively; and the 1-year progression-free survival (PFS) rates were 39% (31%-48%), 66% (52%-84%), and 46% (30%-70%) and the overall survival (OS) rates were 58% (50%-67%), 77% (64%-92%), and 66% (50%-87%), respectively. In multivariable analyses compared with low AUC, an optimal AUC was associated with the highest PFS (hazard ratio [HR], 0.52; 0.3-0.91; P = .02) and lowest risk of relapse/POD (HR, 0.46; 0.25-0.84; P = .01) without an increased risk of any-grade cytokine release syndrome (HR, 1.1; 0.7-1.6; P = .8) or and immune effector cell-associated neurotoxicity syndrome (ICANS) (HR, 1.36; 0.83-2.3; P = .2). A high AUC was associated with the greatest risk of any-grade ICANS (HR, 1.9; 1.1-3.2; P = .02). Although the main cause of death in all groups was relapse/POD, nonrelapse-related deaths, including 3 deaths from ICANS, were more frequent in the high AUC group. These findings suggest that PK-directed fludarabine dosing to achieve an optimal AUC may result in improved outcomes for patients receiving axi-cel.
Collapse
Affiliation(s)
- Michael Scordo
- Adult Bone Marrow Transplant Service, Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jessica R. Flynn
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mithat Gonen
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sean M. Devlin
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - Roni Shouval
- Adult Bone Marrow Transplant Service, Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jamie Brower
- Cell Therapy and Transplant and Lymphoma Programs, Division of Hematology-Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - David L. Porter
- Cell Therapy and Transplant and Lymphoma Programs, Division of Hematology-Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Stephen J. Schuster
- Cell Therapy and Transplant and Lymphoma Programs, Division of Hematology-Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Joseph Maakaron
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Richard T. Maziarz
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Andy I. Chen
- Center for Hematologic Malignancies, Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Loretta J. Nastoupil
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Joseph P. McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Olalekan O. Oluwole
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
- Department of Oncology, Hackensack Meridian School of Medicine, Nutley, NJ
| | - Lori A. Leslie
- Division of Lymphoma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
- Department of Oncology, Hackensack Meridian School of Medicine, Nutley, NJ
| | - Michael R. Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Peter A. Riedell
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
21
|
Rejeski K, Jain MD, Smith EL. Mechanisms of Resistance and Treatment of Relapse after CAR T-cell Therapy for Large B-cell Lymphoma and Multiple Myeloma. Transplant Cell Ther 2023; 29:418-428. [PMID: 37076102 PMCID: PMC10330792 DOI: 10.1016/j.jtct.2023.04.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Although chimeric antigen receptor (CAR) T cell therapy (CAR-T) has altered the treatment landscape for relapsed/refractory B cell malignancies and multiple myeloma, only a minority of patients attain long-term disease remission. The underlying reasons for CAR-T resistance are multifaceted and can be broadly divided into host-related, tumor-intrinsic, microenvironmental and macroenvironmental, and CAR-T-related factors. Emerging host-related determinants of response to CAR-T relate to gut microbiome composition, intact hematopoietic function, body composition, and physical reserve. Emerging tumor-intrinsic resistance mechanisms include complex genomic alterations and mutations to immunomodulatory genes. Furthermore, the extent of systemic inflammation prior to CAR-T is a potent biomarker of response and reflects a proinflammatory tumor micromilieu characterized by infiltration of myeloid-derived suppressor cells and regulatory T cell populations. The tumor and its surrounding micromilieu also can shape the response of the host to CAR-T infusion and the subsequent expansion and persistence of CAR T cells, a prerequisite for efficient eradication of tumor cells. Here, focusing on both large B cell lymphoma and multiple myeloma, we review resistance mechanisms, explore therapeutic avenues to overcome resistance to CAR-T, and discuss the management of patients who relapse after CAR-T.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III – Hematology/Oncology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, USA
| | | |
Collapse
|
22
|
Slatter M, Lum SH. Personalized hematopoietic stem cell transplantation for inborn errors of immunity. Front Immunol 2023; 14:1162605. [PMID: 37090739 PMCID: PMC10113466 DOI: 10.3389/fimmu.2023.1162605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Patients with inborn errors of immunity (IEI) have been transplanted for more than 50 years. Many long-term survivors have ongoing medical issues showing the need for further improvements in how hematopoietic stem cell transplantation (HSCT) is performed if patients in the future are to have a normal quality of life. Precise genetic diagnosis enables early treatment before recurrent infection, autoimmunity and organ impairment occur. Newborn screening for severe combined immunodeficiency (SCID) is established in many countries. For newly described disorders the decision to transplant is not straight-forward. Specific biologic therapies are effective for some diseases and can be used as a bridge to HSCT to improve outcome. Developments in reduced toxicity conditioning and methods of T-cell depletion for mismatched donors have made transplant an option for all eligible patients. Further refinements in conditioning plus precise graft composition and additional cellular therapy are emerging as techniques to personalize the approach to HSCT for each patient.
Collapse
Affiliation(s)
- Mary Slatter
- Paediatric Immunology and HSCT, Newcastle University, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Great North Children’s Hospital, Newcastle upon Tyne, United Kingdom
| | - Su Han Lum
- Paediatric Immunology and HSCT, Newcastle University, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Great North Children’s Hospital, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
23
|
Owen K, Ghaly R, Shohdy KS, Thistlethwaite F. Lymphodepleting chemotherapy practices and effect on safety and efficacy outcomes in patients with solid tumours undergoing T cell receptor-engineered T cell (TCR-T) Therapy: a systematic review and meta-analysis. Cancer Immunol Immunother 2023; 72:805-814. [PMID: 36315268 PMCID: PMC9628360 DOI: 10.1007/s00262-022-03287-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND T cell receptor-engineered T cell (TCR-T) therapy has shown promising efficacy in advanced solid tumours. Lymphodepleting (LD) chemotherapy improves TCR-T cell therapy efficacy but is associated with significant toxicities. Evidence is sparse regarding the optimum LD regimen for TCR-T cell therapy in solid tumours. METHODS A systematic review was conducted of interventional, prospective clinical trials describing LD practices prior to TCR-T cell therapy in patients with advanced solid tumours. The objective was to define LD regimens administered prior to TCR-T cell therapy and their effects on specific safety and efficacy outcomes in this patient population. RESULTS Searches returned 484 studies, 19 (231 patients) met the eligibility criteria. Cyclophosphamide (cyclo) 60 mg/kg daily (2 days), plus fludarabine (fludara) 25 mg/m2 daily (5 days) was the most common LD regimen (38% of studies). Higher dose LD regimens were associated with increased pooled incidence rates of febrile neutropaenia compared to low dose (0.64, [95% Confidence interval (CI): 0.50-0.78], vs. 0.39 [95% CI: 0.25-0.53], respectively) but were not significantly associated with higher objective responses (odds ratio: 1.05, 95%CI: 0.60-1.82, p = 0.86). A major shortfall in safety data reporting was identified; determination of LD regimen effects on many safety outcomes was not possible. CONCLUSION Standard consensus guidelines for the design and reporting of adoptive cell therapy (ACT) studies would facilitate accurate risk-benefit analysis for optimising LD regimens in patients with advanced solid tumours.
Collapse
Affiliation(s)
- Kathryn Owen
- ATMP Master Programme, The University of Manchester, Manchester, UK
| | - Ramy Ghaly
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kyrillus S Shohdy
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Fiona Thistlethwaite
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.
- Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|
24
|
Chen RL, Ip PP, Shaw JJ, Wang YH, Fan LH, Shen YL, Joseph NA, Chen TE, Chen LY. Anti-Thymocyte Globulin (ATG)-Free Nonmyeloablative Haploidentical PBSCT Plus Post-Transplantation Cyclophosphamide Is a Safe and Efficient Treatment Approach for Pediatric Acquired Aplastic Anemia. Int J Mol Sci 2022; 23:ijms232315192. [PMID: 36499545 PMCID: PMC9739033 DOI: 10.3390/ijms232315192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/11/2022] Open
Abstract
Most cases of acquired aplastic anemia (AA) arise from autoimmune destruction of hematopoietic stem and progenitor cells. Human leukocyte antigen (HLA)-haploidentical nonmyeloablative hematopoietic stem cell transplantation (HSCT) plus post-transplantation cyclophosphamide (PTCy) is increasingly applied to salvage AA using bone marrow as graft and anti-thymocyte globulin (ATG) in conditioning. Herein, we characterize a cohort of twelve AA patients clinically and molecularly, six who possessed other immunological disorders (including two also carrying germline SAMD9L mutations). Each patient with SAMD9L mutation also carried an AA-related rare BCORL1 variant or CTLA4 p.T17A GG genotype, respectively, and both presented short telomere lengths. Six of the ten patients analyzed harbored AA-risky HLA polymorphisms. All patients recovered upon non-HSCT (n = 4) or HSCT (n = 8) treatments. Six of the eight HSCT-treated patients were subjected to a modified PTCy-based regimen involving freshly prepared peripheral blood stem cells (PBSC) as graft and exclusion of ATG. All patients were engrafted between post-transplantation days +13 and +18 and quickly reverted to normal life, displaying a sustained complete hematologic response and an absence of graft-versus-host disease. These outcomes indicate most AA cases, including of the SAMD9L-inherited subtype, are immune-mediated and the modified PTCy-based regimen we present is efficient and safe for salvage.
Collapse
Affiliation(s)
- Rong-Long Chen
- Department of Pediatric Hematology and Oncology, Koo Foundation Sun Yat-sen Cancer Center, Taipei 11259, Taiwan
- Correspondence:
| | - Peng Peng Ip
- Institute of Molecular Biology, Academia Sinica, Taipei 115024, Taiwan
| | - Jy-juinn Shaw
- School of Law, National Yang Ming Chiao Tung University, Hsinchu City 30093, Taiwan
| | - Yun-Hsin Wang
- Department of Chemistry, Tamkang University, Tamsui, New Taipei City 251301, Taiwan
| | - Li-Hua Fan
- Department of Pharmacy, Koo Foundation Sun Yat-sen Cancer Center, Taipei 11259, Taiwan
| | - Yi-Ling Shen
- Institute of Molecular Biology, Academia Sinica, Taipei 115024, Taiwan
| | - Nithila A. Joseph
- Institute of Molecular Biology, Academia Sinica, Taipei 115024, Taiwan
| | - Tsen-Erh Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 115024, Taiwan
| | - Liuh-Yow Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 115024, Taiwan
| |
Collapse
|
25
|
Brooks JT, Solans BP, Lu Y, Kharbanda S, Dvorak CC, Lalefar N, Long S, Gupta AO, Horn B, Lamba JK, Huang L, Apsel-Winger B, Keizer RJ, Savic R, Long-Boyle J. Prospective Validation and Refinement of a Population Pharmacokinetic Model of Fludarabine in Children and Young Adults Undergoing Hematopoietic Cell Transplantation. Pharmaceutics 2022; 14:pharmaceutics14112462. [PMID: 36432661 PMCID: PMC9694406 DOI: 10.3390/pharmaceutics14112462] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
Fludarabine is a nucleoside analog with antileukemic and immunosuppressive activity commonly used in allogeneic hematopoietic cell transplantation (HCT). Several fludarabine population pharmacokinetic (popPK) and pharmacodynamic models have been published enabling the movement towards precision dosing of fludarabine in pediatric HCT; however, developed models have not been validated in a prospective cohort of patients. In this multicenter pharmacokinetic study, fludarabine plasma concentrations were collected via a sparse-sampling strategy. A fludarabine popPK model was evaluated and refined using standard nonlinear mixed effects modelling techniques. The previously described fludarabine popPK model well-predicted the prospective fludarabine plasma concentrations. Individuals who received model-based dosing (MBD) of fludarabine achieved significantly more precise overall exposure of fludarabine. The fludarabine popPK model was further improved by both the inclusion of fat-free mass instead of total body weight and a maturation function on fludarabine clearance. The refined popPK model is expected to improve dosing recommendations for children younger than 2 years and patients with higher body mass index. Given the consistency of fludarabine clearance and exposure across its multiple days of administration, therapeutic drug monitoring is not likely to improve targeted exposure attainment.
Collapse
Affiliation(s)
- Jordan T. Brooks
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Bioengineering and Therapeutics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Belen P. Solans
- Department of Bioengineering and Therapeutics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ying Lu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sandhya Kharbanda
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Christopher C. Dvorak
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nahal Lalefar
- Benioff Children’s Hospital of Oakland, University of California San Francisco, Oakland, CA 94609, USA
| | - Susie Long
- Department of Pediatrics, University of Minnesota Masonic Children’s Hospital, Minneapolis, MN 55454, USA
| | - Ashish O. Gupta
- Department of Pediatrics, University of Minnesota Masonic Children’s Hospital, Minneapolis, MN 55454, USA
| | - Biljana Horn
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Department of Pediatrics, University of Florida, Gainesville, FL 32603, USA
| | - Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Department of Pediatrics, University of Florida, Gainesville, FL 32603, USA
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Beth Apsel-Winger
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
| | | | - Rada Savic
- Department of Bioengineering and Therapeutics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Janel Long-Boyle
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
- Correspondence:
| |
Collapse
|
26
|
Xiao X, Wang Y, Zou Z, Yang Y, Wang X, Xin X, Tu S, Li Y. Combination strategies to optimize the efficacy of chimeric antigen receptor T cell therapy in haematological malignancies. Front Immunol 2022; 13:954235. [PMID: 36091028 PMCID: PMC9460961 DOI: 10.3389/fimmu.2022.954235] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/01/2022] [Indexed: 02/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the therapeutic landscape of haematological malignancies. However, resistance and relapse remain prominent limitations, and they are related to the limited persistence and efficacy of CAR T cells, downregulation or loss of tumour antigens, intrinsic resistance of tumours to death signalling, and immune suppressive microenvironment. Rational combined modality treatments are regarded as a promising strategy to further unlock the antitumor potential of CAR T cell therapy, which can be applied before CAR T cell infusion as a conditioning regimen or in ex vivo culture settings as well as concomitant with or after CAR T cell infusion. In this review, we summarize the combinatorial strategies, including chemotherapy, radiotherapy, haematopoietic stem cell transplantation, targeted therapies and other immunotherapies, in an effort to further enhance the effectiveness of this impressive therapy and benefit more patients.
Collapse
Affiliation(s)
- Xinyi Xiao
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yazhuo Wang
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Zhengbang Zou
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yufei Yang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyu Wang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Xin
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sanfang Tu
- Department of Haematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China,*Correspondence: Sanfang Tu, ; Yuhua Li,
| | - Yuhua Li
- Department of Haematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China,*Correspondence: Sanfang Tu, ; Yuhua Li,
| |
Collapse
|
27
|
Relationship of iothalamate clearance and NRM in patients receiving fludarabine and melphalan reduced-intensity conditioning. Blood Adv 2022; 6:3844-3849. [PMID: 35522968 PMCID: PMC9278281 DOI: 10.1182/bloodadvances.2021006395] [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: 10/18/2021] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
The reduced-intensity conditioning regimen, fludarabine and melphalan, is frequently used in allogeneic hematopoietic stem cell transplantation (HSCT). Melphalan and the active metabolite of fludarabine, F-ara-A, are excreted via the kidneys. Existing methods to assess clearance in this setting are based on serum creatinine, which has known limitations for glomerular filtration rate (GFR) estimation in patients with malignancy. Measured GFR (mGFR) may better predict drug dosing to mitigate toxicity and increase the chances of successful engraftment. The primary objective of this study was to assess the association between mGFR and risk for non-relapse mortality (NRM) in allogeneic HSCT patients receiving conditioning with fludarabine and melphalan. In the 109 included patients, mGFR < 65 ml/min/1.73m2 predicted a significantly higher rate of overall NRM (HR 2.13, 95% CI, 1.03-4.35, P = 0.04) and 1-year incidence of infection (HR 2.63, 95% CI, 1.54-4.55, P < 0.001) in addition to a significantly lower 2-year survival (P = 0.019). Kidney function estimated via eGFR and eCrCl did not correlate with post-transplant outcomes. These results suggest that mGFR is a promising approach for assessing clearance in allogeneic HSCT patients and may be preferred to standard creatinine-based eGFR strategies.
Collapse
|
28
|
Fabrizio VA, Boelens JJ, Mauguen A, Baggott C, Prabhu S, Egeler E, Mavroukakis S, Pacenta H, Phillips CL, Rossoff J, Stefanski HE, Talano JA, Moskop A, Margossian SP, Verneris MR, Myers GD, Karras NA, Brown PA, Qayed M, Hermiston M, Satwani P, Krupski C, Keating AK, Wilcox R, Rabik CA, Chinnabhandar V, Kunicki M, Goksenin AY, Mackall CL, Laetsch TW, Schultz LM, Curran KJ. Optimal fludarabine lymphodepletion is associated with improved outcomes after CAR T-cell therapy. Blood Adv 2022; 6:1961-1968. [PMID: 34788386 PMCID: PMC9006295 DOI: 10.1182/bloodadvances.2021006418] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 11/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells provide a therapeutic option in hematologic malignancies. However, treatment failure after initial response approaches 50%. In allogeneic hematopoietic cell transplantation, optimal fludarabine exposure improves immune reconstitution, resulting in lower nonrelapse mortality and increased survival. We hypothesized that optimal fludarabine exposure in lymphodepleting chemotherapy before CAR T-cell therapy would improve outcomes. In a retrospective analysis of patients with relapsed/refractory B-cell acute lymphoblastic leukemia undergoing CAR T-cell (tisagenlecleucel) infusion after cyclophosphamide/fludarabine lymphodepleting chemotherapy, we estimated fludarabine exposure as area under the curve (AUC; mg × h/L) using a validated population pharmacokinetic (PK) model. Fludarabine exposure was related to overall survival (OS), cumulative incidence of relapse (CIR), and a composite end point (loss of B-cell aplasia [BCA] or relapse). Eligible patients (n = 152) had a median age of 12.5 years (range, <1 to 26), response rate of 86% (n = 131 of 152), 12-month OS of 75.1% (95% confidence interval [CI], 67.6% to 82.6%), and 12-month CIR of 36.4% (95% CI, 27.5% to 45.2%). Optimal fludarabine exposure was determined as AUC ≥13.8 mg × h/L. In multivariable analyses, patients with AUC <13.8 mg × h/L had a 2.5-fold higher CIR (hazard ratio [HR], 2.45; 95% CI, 1.34-4.48; P = .005) and twofold higher risk of relapse or loss of BCA (HR, 1.96; 95% CI, 1.19-3.23; P = .01) compared with those with optimal fludarabine exposure. High preinfusion disease burden was also associated with increased risk of relapse (HR, 2.66; 95% CI, 1.45-4.87; P = .001) and death (HR, 4.77; 95% CI, 2.10-10.9; P < .001). Personalized PK-directed dosing to achieve optimal fludarabine exposure should be tested in prospective trials and, based on this analysis, may reduce disease relapse after CAR T-cell therapy.
Collapse
Affiliation(s)
- Vanessa A. Fabrizio
- Colorado Children’s Hospital, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christina Baggott
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Snehit Prabhu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Emily Egeler
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Sharon Mavroukakis
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Holly Pacenta
- Department of Pediatrics, University of Texas Southwestern Medical Center/Children’s Health, Dallas, TX
- Division of Hematology and Oncology, Cook Children’s Medical Center, Fort Worth, TX
| | - Christine L. Phillips
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
- Cancer and Blood Disease Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jenna Rossoff
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL
| | - Heather E. Stefanski
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Julie-An Talano
- Department of Pediatric Hematology Oncology, Medical College of Wisconsin, Wauwatosa, WI
| | - Amy Moskop
- Department of Pediatric Hematology Oncology, Medical College of Wisconsin, Wauwatosa, WI
| | - Steven P. Margossian
- Pediatric Hematology-Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Michael R. Verneris
- Colorado Children’s Hospital, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | | | - Nicole A. Karras
- Department of Pediatrics, City of Hope National Medical Center, Duarte, CA
| | - Patrick A. Brown
- Department of Oncology, Sidney Kimmel Cancer Center, John Hopkins School of Medicine, Baltimore, MD
| | - Muna Qayed
- Children’s Healthcare of Atlanta, Emory University, Atlanta, GA
| | - Michelle Hermiston
- Benioff Children’s Hospital, University of California San Francisco, San Francisco, CA
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Christa Krupski
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| | - Amy K. Keating
- Colorado Children’s Hospital, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | | | - Cara A. Rabik
- Department of Oncology, Sidney Kimmel Cancer Center, John Hopkins School of Medicine, Baltimore, MD
| | - Vasant Chinnabhandar
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Michael Kunicki
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - A. Yasemin Goksenin
- Benioff Children’s Hospital, University of California San Francisco, San Francisco, CA
| | - Crystal L. Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Division of Stem Cell Transplantation and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Theodore W. Laetsch
- Department of Pediatrics, University of Texas Southwestern Medical Center/Children’s Health, Dallas, TX
- Department of Pediatrics and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
- Division of Oncology, Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Liora M. Schultz
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Kevin J. Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Weill Cornell Medical College, New York, NY
| |
Collapse
|
29
|
van der Stoep MYEC, Oostenbrink LVE, Bredius RGM, Moes DJAR, Guchelaar HJ, Zwaveling J, Lankester AC. Therapeutic Drug Monitoring of Conditioning Agents in Pediatric Allogeneic Stem Cell Transplantation; Where do We Stand? Front Pharmacol 2022; 13:826004. [PMID: 35330826 PMCID: PMC8940165 DOI: 10.3389/fphar.2022.826004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is an established curative treatment that has significantly improved clinical outcome of pediatric patients with malignant and non-malignant disorders. This is partly because of the use of safer and more effective combinations of chemo- and serotherapy prior to HSCT. Still, complications due to the toxicity of these conditioning regimens remains a major cause of transplant-related mortality (TRM). One of the most difficult challenges to further improve HSCT outcome is reducing toxicity while maintaining efficacy. The use of personalized dosing of the various components of the conditioning regimen by means of therapeutic drug monitoring (TDM) has been the topic of interest in the last decade. TDM could play an important role, especially in children who tend to show greater pharmacokinetic variability. However, TDM should only be performed when it has clear added value to improve clinical outcome or reduce toxicity. In this review, we provide an overview of the available evidence for the relationship between pharmacokinetic parameters and clinical outcome or toxicities of the most commonly used conditioning agents in pediatric HSCT.
Collapse
Affiliation(s)
- M. Y. Eileen C. van der Stoep
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: M. Y. Eileen C. van der Stoep,
| | - Lisa V. E. Oostenbrink
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Robbert G. M. Bredius
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Dirk Jan A. R. Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Juliette Zwaveling
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Arjan C. Lankester
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
30
|
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
|
31
|
Fludarabine Exposure Predicts Outcome after CD19 CAR T-Cell Therapy in Children and Young Adults with Acute Leukemia. Blood Adv 2022; 6:1969-1976. [PMID: 35134115 PMCID: PMC9006280 DOI: 10.1182/bloodadvances.2021006700] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/03/2022] [Indexed: 12/04/2022] Open
Abstract
A cumulative fludarabine AUCT0−∞ ≥14 mg*h/L correlates with improved LFS after CD19 CAR T-cell infusion. Clinical outcome in patients receiving CD19 CAR T cells might be improved by optimizing fludarabine exposure in the lymphodepleting regimen.
The addition of fludarabine to cyclophosphamide as a lymphodepleting regimen prior to CD19 chimeric antigen receptor (CAR) T-cell therapy significantly improved outcomes in patients with relapsed/refractory (r/r) B-cell acute lymphoblastic leukemia (B-ALL). Fludarabine exposure, previously shown to be highly variable when dosing is based on body surface area (BSA), is a predictor for survival in allogeneic hematopoietic cell transplantation (allo-HCT). Hence, we hypothesized that an optimal exposure of fludarabine might be of clinical importance in CD19 CAR T-cell treatment. We examined the effect of cumulative fludarabine exposure during lymphodepletion, defined as concentration-time curve (AUC), on clinical outcome and lymphocyte kinetics. A retrospective analysis was conducted with data from 26 patients receiving tisagenlecleucel for r/r B-ALL. Exposure of fludarabine was shown to be a predictor for leukemia-free survival (LFS), B-cell aplasia, and CD19-positive relapse following CAR T-cell infusion. Minimal event probability was observed at a cumulative fludarabine AUCT0−∞ ≥14 mg*h/L, and underexposure was defined as an AUCT0−∞ <14 mg*h/L. In the underexposed group, the median LFS was 1.8 months, and the occurrence of CD19-positive relapse within 1 year was 100%, which was higher compared with the group with an AUCT0−∞ ≥14 mg*h/L (12.9 months; P < .001; and 27.4%; P = .0001, respectively). Furthermore, the duration of B-cell aplasia within 6 months was shorter in the underexposed group (77.3% vs 37.3%; P = .009). These results suggest that optimizing fludarabine exposure may have a relevant impact on LFS following CAR T-cell therapy, which needs to be validated in a prospective clinical trial.
Collapse
|
32
|
Individualized Dosage Optimization for Myeloablative Conditioning before Unrelated Cord Blood Transplantation in a Diamond–Blackfan Anemia Patient with Germline RPL11 Mutation: A Case Study. Processes (Basel) 2022. [DOI: 10.3390/pr10020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022] Open
Abstract
Unrelated cord blood transplantation (CBT) for Diamond–Blackfan anemia (DBA), a systemic ribosomopathy affecting the disposition of conditioning agents, has resulted in outcomes inferior to those by transplantations from matched donors. We report the experience of the pharmacokinetics-guided myeloablative unrelated CBT in a DBA patient with a germline RPL11 mutation. The conditioning consisted of individualized dosing of fludarabine (based on weight and renal function with a target area under the curve (AUC) of 17.5 mg·h/L) and busulfan (based on therapeutic drug monitoring with a target AUC of 90 mg·h/L), as well as dosing and timing of thymoglobulin (based on body weight and pre-dose lymphocyte count to target pre-CBT AUC of 30.7 AU·day/mL and post-CBT AUC of 4.3 AU·day/mL, respectively). The pharmacokinetic measures resulted in a 27.5% reduction in busulfan and a 35% increase in fludarabine, as well as an over three-fold increase in thymoglobulin dosage with the start time changed to day-9 instead of day-2 compared to regular regimens. The transplantation resulted in rapid, complete, and sustained hematopoietic engraftment. The patient is now healthy over 3 years after CBT. A pharmacokinetics-guided individualized dosing strategy for conditioning might be a feasible option to improve the outcomes of DBA patients receiving unrelated myeloablative CBT.
Collapse
|
33
|
de Koning C, Tao W, Lacna A, van Veghel K, Horwitz ME, Sanz G, Jagasia MH, Wagner JE, Stiff PJ, Hanna R, Cilloni D, Valcárcel D, Peled T, Galamidi Cohen E, Goshen U, Pandit A, Lindemans CA, Jan Boelens J, Nierkens S. Lymphoid and myeloid immune cell reconstitution after nicotinamide-expanded cord blood transplantation. Bone Marrow Transplant 2021; 56:2826-2833. [PMID: 34312498 PMCID: PMC8563413 DOI: 10.1038/s41409-021-01417-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Omidubicel (nicotinamide-expanded cord blood) is a potential alternative source for allogeneic hematopoietic cell transplantation (HCT) when an HLA-identical donor is lacking. A phase I/II trial with standalone omidubicel HCT showed rapid and robust neutrophil and platelet engraftment. In this study, we evaluated the immune reconstitution (IR) of patients receiving omidubicel grafts during the first 6 months post-transplant, as IR is critical for favorable outcomes of the procedure. Data was collected from the omidubicel phase I-II international, multicenter trial. The primary endpoint was the probability of achieving adequate CD4+ T-cell IR (CD4IR: > 50 × 106/L within 100 days). Secondary endpoints were the recovery of T-cells, natural killer (NK)-cells, B-cells, dendritic cells (DC), and monocytes as determined with multicolor flow cytometry. LOESS-regression curves and cumulative incidence plots were used for data description. Thirty-six omidubicel recipients (median 44; 13-63 years) were included, and IR data was available from 28 recipients. Of these patients, 90% achieved adequate CD4IR. Overall, IR was complete and consisted of T-cell, monocyte, DC, and notably fast NK- and B-cell reconstitution, compared to conventional grafts. Our data show that transplantation of adolescent and adult patients with omidubicel results in full and broad IR, which is comparable with IR after HCT with conventional graft sources.
Collapse
Affiliation(s)
- Coco de Koning
- University Medical Center Utrecht, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Weiyang Tao
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Amelia Lacna
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Guillermo Sanz
- Hospital Universitario y Politécnico la Fe, València, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | - Caroline A Lindemans
- University Medical Center Utrecht, Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jaap Jan Boelens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stefan Nierkens
- University Medical Center Utrecht, Utrecht, The Netherlands.
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Nijstad AL, Nierkens S, Lindemans CA, Boelens JJ, Bierings M, Versluys AB, van der Elst KC, Huitema AD. Population pharmacokinetics of clofarabine for allogeneic hematopoietic cell transplantation in paediatric patients. Br J Clin Pharmacol 2021; 87:3218-3226. [PMID: 33444472 PMCID: PMC8359279 DOI: 10.1111/bcp.14738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 12/17/2022] Open
Abstract
AIMS Clofarabine has recently been evaluated as part of the conditioning regimen for allogeneic hematopoietic stem cell transplantation (HCT) in children. Pharmacokinetic (PK) exposure of different agents commonly used in conditioning regimens is strongly related to HCT outcome. Consequently, the PK of clofarabine may be important for outcome. This report describes the population PK of clofarabine in paediatric patients and one adult. METHODS From 80 paediatric (0.5-18 years) and 1 adult patient (37 years), 805 plasma concentrations were included in pharmacokinetic analyses using nonlinear mixed effects modelling. RESULTS A two-compartment model adequately described the PK of clofarabine. Body weight and estimated glomerular filtration rate (eGFR) were included as covariates. Clearance was differentiated into nonrenal and renal clearance (approximately 55% of total clearance), resulting in population estimates of 24.0 L/h (95% confidence interval [CI] 13.7-34.4) and 29.8 L/h (95% CI 23.9-36.1) for a patient of 70 kg with normal renal function, respectively. Unexplained interindividual variability in clearance was 17.8% (95% CI 14.6-22.4). A high variability in exposure was observed (range area under the curveT0-inf 1.8-6.0 mg/L*h) after body surface area (BSA) based dosing. Interestingly, children with low body weight had a lower exposure than children with a higher body weight, which indicates that the currently practised BSA-based dosing is not adequate for clofarabine. CONCLUSION A clofarabine dosing algorithm based on this PK model, using body weight and eGFR, results in a more predictable exposure than BSA-based dosing. However, the exact target exposure needs to be further investigated.
Collapse
Affiliation(s)
- A. Laura Nijstad
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
- Pediatric Blood and Bone Marrow TransplantationPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
| | - Caroline A. Lindemans
- Pediatric Blood and Bone Marrow TransplantationPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
- Department of Pediatrics, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - Jaap Jan Boelens
- Pediatric Blood and Bone Marrow TransplantationPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
- Stem Cell Transplantation and Cellular Therapies, MSK KidsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Marc Bierings
- Pediatric Blood and Bone Marrow TransplantationPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
- Department of Pediatrics, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - A. Birgitta Versluys
- Pediatric Blood and Bone Marrow TransplantationPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
- Department of Pediatrics, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - Kim C.M. van der Elst
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
| | - Alwin D.R. Huitema
- Department of Clinical Pharmacy, Division of Laboratory Medicine and Pharmacy, University Medical Center UtrechtUtrecht UniversityUtrechtthe Netherlands
- Department of Pharmacy & PharmacologyNetherlands Cancer InstituteAmsterdamthe Netherlands
- Department of PharmacologyPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
| |
Collapse
|
35
|
CD4+ T-cell reconstitution predicts survival outcomes after acute graft-versus-host-disease: a dual-center validation. Blood 2021; 137:848-855. [PMID: 33150379 DOI: 10.1182/blood.2020007905] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/04/2020] [Indexed: 11/20/2022] Open
Abstract
Acute graft-versus-host-Disease (aGVHD) is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). We previously showed that early CD4+ T-cell immune reconstitution (IR; CD4+ IR) predicts survival after HCT. Here, we studied the relation between CD4+ IR and survival in patients developing aGVHD. Pediatric patients undergoing first allogeneic HCT at University Medical Center Utrecht (UMC)/Princess Máxima Center (PMC) or Memorial Sloan Kettering Cancer Center (MSK) were included. Primary outcomes were nonrelapse mortality (NRM) and overall survival (OS), stratified for aGVHD and CD4+ IR, defined as ≥50 CD4+ T cells per μL within 100 days after HCT or before aGVHD onset. Multivariate and time-to-event Cox proportional hazards models were applied, and 591 patients (UMC/PMC, n = 276; MSK, n = 315) were included. NRM in patients with grade 3 to 4 aGVHD with or without CD4+ IR within 100 days after HCT was 30% vs 80% (P = .02) at UMC/PMC and 5% vs 67% (P = .02) at MSK. This was associated with lower OS without CD4+ IR (UMC/PMC, 61% vs 20%; P = .04; MSK, 75% vs 33%; P = .12). Inadequate CD4+ IR before aGVHD onset was associated with significantly higher NRM (74% vs 12%; P < .001) and inferior OS (24% vs 78%; P < .001). In this retrospective analysis, we demonstrate that early CD4+ IR, a simple and robust marker predictive of outcomes after HCT, is associated with survival after moderate to severe aGVHD. This association must be confirmed prospectively but suggests strategies to improve T-cell recovery after HCT may influence survival in patients developing aGVHD.
Collapse
|
36
|
Chen RL, Fang LH, Yang XY, El Amrani M, Uijtendaal EV, Chen YF, Ku WC. Therapeutic Drug Monitoring of Busulfan in Patients Undergoing Hematopoietic Cell Transplantation: A Pilot Single-Center Study in Taiwan. Pharmaceuticals (Basel) 2021; 14:613. [PMID: 34206798 PMCID: PMC8308703 DOI: 10.3390/ph14070613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/24/2021] [Indexed: 01/02/2023] Open
Abstract
(1) Background: Busulfan has been used as a conditioning regimen in allogeneic hematopoietic cell stem transplantation (HSCT). Owing to a large inter-individual variation in pharmacokinetics, therapeutic drug monitoring (TDM)-guided busulfan dosing is necessary to reduce graft failure and relapse rate. As there exists no TDM of busulfan administration for HCT in Taiwan, we conducted a pilot study to assess the TDM-dosing of busulfan in the Taiwanese population; (2) Methods: Seven patients with HCT from The Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan, received conditioning regimens consisting of intravenous busulfan and other chemotherapies. After the initial busulfan dose, blood samples were collected for busulfan TDM at 5 min, 1 h, 2 h, and 3 h. Busulfan was extracted and detected by performing stable-isotope dilution LC-MS/MS. Plasma busulfan concentration was quantified and used for dose adjustment. Potential adverse effects of busulfan, such as mucositis and hepatic veno-occlusive disease (VOD), were also evaluated; (3) Results: The LC-MS/MS method was validated with an analyte recovery of 88-99%, within-run and between-run precision of <15%, and linearity ranging from 10 to 10,000 ng/mL. Using TDM-guided busulfan dosing, dose adjustment was necessary and performed in six out of seven patients (86%) with successful engraftments in all patients (100%). Mild mucositis was observed, and VOD was diagnosed in only one patient; (4) Conclusions: This single-center study in Taiwan demonstrated the importance of busulfan TDM in increasing the success rate of HCT transplantation. It is also necessary to further investigate the optimal busulfan target value in the Taiwanese population in the future.
Collapse
Affiliation(s)
- Rong-Long Chen
- Department of Pediatric Hematology and Oncology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei City 112019, Taiwan;
| | - Li-Hua Fang
- Department of Pharmacy, Koo Foundation Sun Yat-Sen Cancer Center, Taipei City 112019, Taiwan; (L.-H.F.); (X.-Y.Y.)
| | - Xin-Yi Yang
- Department of Pharmacy, Koo Foundation Sun Yat-Sen Cancer Center, Taipei City 112019, Taiwan; (L.-H.F.); (X.-Y.Y.)
| | - Mohsin El Amrani
- Department of Clinical Pharmacy, Division Laboratory, Medicine and Pharmacy, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (M.E.A.); (E.V.U.)
| | - Esther Veronique Uijtendaal
- Department of Clinical Pharmacy, Division Laboratory, Medicine and Pharmacy, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (M.E.A.); (E.V.U.)
| | - Yen-Fu Chen
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| |
Collapse
|
37
|
de Witte M, Daenen LGM, van der Wagen L, van Rhenen A, Raymakers R, Westinga K, Kuball J. Allogeneic Stem Cell Transplantation Platforms With Ex Vivo and In Vivo Immune Manipulations: Count and Adjust. Hemasphere 2021; 5:e580. [PMID: 34095763 PMCID: PMC8171366 DOI: 10.1097/hs9.0000000000000580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/14/2021] [Indexed: 01/16/2023] Open
Abstract
Various allogeneic (allo) stem cell transplantation platforms have been developed over the last 2 decades. In this review we focus on the impact of in vivo and ex vivo graft manipulation on immune reconstitution and clinical outcome. Strategies include anti-thymocyte globulin- and post-transplantation cyclophosphamide-based regimens, as well as graft engineering, such as CD34 selection and CD19/αβT cell depletion. Differences in duration of immune suppression, reconstituting immune repertoires, and associated graft-versus-leukemia effects and toxicities mediated through viral reactivations are highlighted. In addition, we discuss the impact of different reconstituting repertoires on donor lymphocyte infusions and post allo pharmacological interventions to enhance tumor control. We advocate for precisely counting all graft ingredients and therapeutic drug monitoring during conditioning in the peripheral blood, and for adjusting dosing accordingly on an individual basis. In addition, we propose novel trial designs to better assess the impact of variations in transplantation platforms in order to better learn from our diversity of "counts" and potential "adjustments." This will, in the future, allow daily clinical practice, strategic choices, and future trial designs to be based on data guided decisions, rather than relying on dogma and habits.
Collapse
Affiliation(s)
- Moniek de Witte
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Laura G. M. Daenen
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Lotte van der Wagen
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Anna van Rhenen
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Reiner Raymakers
- Department of Hematology, University Medical Center Utrecht, The Netherlands
| | - Kasper Westinga
- Cell Therapy Facility, University Medical Center Utrecht, The Netherlands
| | - Jürgen Kuball
- Department of Hematology, University Medical Center Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
38
|
Takahashi T, Scheibner A, Cao Q, Pearson R, Sanghavi K, Weisdorf DJ, Brunstein CG, Rogosheske J, Bachanova V, Warlick ED, Wiseman A, Jacobson PA. Higher Fludarabine and Cyclophosphamide Exposures Lead to Worse Outcomes in Reduced-Intensity Conditioning Hematopoietic Cell Transplantation for Adult Hematologic Malignancy. Transplant Cell Ther 2021; 27:773.e1-773.e8. [PMID: 34044184 DOI: 10.1016/j.jtct.2021.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/28/2021] [Accepted: 05/13/2021] [Indexed: 11/15/2022]
Abstract
Reduced-intensity conditioning regimens using fludarabine (Flu) and cyclophosphamide (Cy) have been widely used in hematopoietic cell transplantation (HCT) recipients. The optimal exposure of these agents remains to be determined. We aimed to delineate the exposure-outcome associations of Flu and Cy separately and then both combined on HCT outcomes. This is a single-center, observational, pharmacokinetic (PK)-pharmacodynamic (PD) study of Flu and Cy in HCT recipients age ≥18 years who received Cy (50 mg/kg in a single dose), Flu (150 to 200 mg/m2 given as 5 daily doses), and total body irradiation (TBI; 200 cGy). We measured trough concentrations of 9-β-D-arabinosyl-2-fluoradenine (F-ara-A), an active metabolite of Flu, on days -5 and -4 (F-ara-ADay-5 and F-ara-ADay-4, respectively), and measured phosphoramide mustard (PM), the final active metabolite of Cy, and estimated the area under the curve (AUC). The 89 enrolled patients had a nonrelapse mortality (NRM) of 9% (95% confidence interval [CI], 3% to 15%) at day +100 and 15% (95% CI, 7% to 22%) at day +180, and an overall survival (OS) of 73% (95% CI, 63% to 81%) at day +180. In multivariate analysis, higher PM area under the curve (AUC) for 0 to 8 hours (PM AUC0-8 hr) was an independent predictor of worse NRM (P < .01 at both day +100 and day +180) and worse day +180 OS (P < .01), but no associations were identified for F-ara-A trough levels. We observed lower day +100 NRM in those with both high F-ara-ADay-4 trough levels (≥40 ng/mL; >25th percentile) and low PM AUC0-8 hr (<34,235 hr ng/mL; <75th percentile), compared with high exposures to both agents (hazard ratio, 0.06; 95% CI, 0.01 to 0.48). No patients with low F-ara-ADay-4 (<40 ng/mL; <25th percentile) had NRM by day +100, regardless of PM AUC. The interpatient PK variability was large in F-ara-ADay-4 trough and PM AUC0-8 hr (29-fold and 5.0-fold, respectively). Flu exposure alone was not strongly associated with NRM or OS in this reduced Flu dose regimen; however, high exposure to both Flu and Cy was associated with a >16-fold higher NRM. These results warrant further investigation to optimize reduced-intensity regimens based on better PK-PD understanding and possible adaptation to predictable factors influencing drug clearance.
Collapse
Affiliation(s)
- Takuto Takahashi
- Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, University of Minnesota, Minneapolis, Minnesota; Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Aileen Scheibner
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Qing Cao
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Rachael Pearson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Kinjal Sanghavi
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Daniel J Weisdorf
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Claudio G Brunstein
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - John Rogosheske
- Department of Pharmacy, M Health Fairview, Minneapolis, Minnesota
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Erica D Warlick
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Anthony Wiseman
- University of Minnesota Medical School, Minneapolis, Minnesota
| | - Pamala A Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
39
|
αβ T-cell graft depletion for allogeneic HSCT in adults with hematological malignancies. Blood Adv 2021; 5:240-249. [PMID: 33570642 DOI: 10.1182/bloodadvances.2020002444] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
We conducted a multicenter prospective single-arm phase 1/2 study that assesses the outcome of αβ T-cell depleted allogeneic hematopoietic stem cell transplantation (allo-HSCT) of peripheral blood derived stem cells from matched related, or unrelated donors (10/10 and 9/10) in adults, with the incidence of acute graft-versus-host disease (aGVHD) as the primary end point at day 100. Thirty-five adults (median age, 59; range, 19-69 years) were enrolled. Conditioning consisted of antithymocyte globulin, busulfan, and fludarabine, followed by 28 days of mycophenolic acid after allo-HSCT. The minimal follow-up time was 24 months. The median number of infused CD34+ cells and αβ T cells were 6.1 × 106 and 16.3 × 103 cells per kg, respectively. The cumulative incidence (CI) of aGVHD grades 2-4 and 3-4 at day 100 was 26% and 14%. One secondary graft failure was observed. A prophylactic donor lymphocyte infusion (DLI) (1 × 105 CD3+ T cells per kg) was administered to 54% of the subjects, resulting in a CI of aGVHD grades 2-4 and 3-4 to 37% and 17% at 2 years. Immune monitoring revealed an early reconstitution of natural killer (NK) and γδ T cells. Cytomegalovirus reactivation associated with expansion of memory-like NK cells. The CI of relapse was 29%, and the nonrelapse mortality 32% at 2 years. The 2-year CI of chronic GVHD (cGVHD) was 23%, of which 17% was moderate. We conclude that only 26% of patients developed aGVHD 2-4 after αβ T-cell-depleted allo-HSCT within 100 days and was associated with a low incidence of cGVHD after 2 years. This trial was registered at www.trialregister.nl as #NL4767.
Collapse
|
40
|
Potential Risk Factors Associated With Graft Failure of Haploidentical Hematopoietic Stem Cell Transplantation in Children With Sickle Cell Disease. J Pediatr Hematol Oncol 2021; 43:e583-e586. [PMID: 32604335 DOI: 10.1097/mph.0000000000001873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/28/2020] [Indexed: 11/25/2022]
Abstract
Nonmyeloablative (NMA) haploidentical hematopoietic stem cell transplantation for sickle cell disease has significantly increased donor availability for transplant and is increasingly used as curative treatment. The authors describe 3 pediatric patients who rejected grafts after an NMA regimen, previously reported to result in good engraftment rates in the mainly adult population. In this manuscript, potential factors contributing to rejection are described and discussed. The authors emphasize the need to further optimize the NMA regimens in pediatric patients and perform haploidentical transplants for sickle cell disease on clinical trials.
Collapse
|
41
|
Al-Khouja A, Park K, Anderson DJ, Young C, Wang J, Huang SM, Khurana M, Burckart GJ. Dosing Recommendations for Pediatric Patients With Renal Impairment. J Clin Pharmacol 2020; 60:1551-1560. [PMID: 32542790 PMCID: PMC8670561 DOI: 10.1002/jcph.1676] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/22/2020] [Indexed: 01/10/2023]
Abstract
A treatment gap exists for pediatric patients with renal impairment. Alterations in renal clearance and metabolism of drugs render standard dosage regimens inappropriate and may lead to drug toxicity, but these studies are not routinely conducted during drug development. The objective of this study was to examine the clinical evidence behind current renal impairment dosage recommendations for pediatric patients in a standard pediatric dosing handbook. The sources of recommendations and comparisons included the pediatric dosing handbook (Lexicomp), the U.S. Food and Drug Administration-approved manufacturer's labels, and published studies in the literature. One hundred twenty-six drugs in Lexicomp had pediatric renal dosing recommendations. Only 14% (18 of 126) of Lexicomp pediatric renal dosing recommendations referenced a pediatric clinical study, and 15% of manufacturer's labels (19 of 126) described specific dosing regimens for renally impaired pediatric patients. Forty-two products had published information on pediatric renal dosing, but 19 (45%) were case studies. When pediatric clinical studies were not referenced in Lexicomp, the renal dosing recommendations followed the adult and pediatric dosing recommendations on the manufacturer's label. Clinical evidence in pediatric patients does not exist for most renal dosing recommendations in a widely used pediatric dosing handbook, and the adult renal dosing recommendations from the manufacturer's label are currently the primary source of pediatric renal dosing information.
Collapse
Affiliation(s)
- Amer Al-Khouja
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kyunghun Park
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Daijha J.C. Anderson
- Eshelman School of Pharmacy,University of North Carolina,Chapel Hill, North Carolina, USA
| | - Caitlyn Young
- University of Southern California, Los Angeles, California, USA
| | - Jian Wang
- Office of Drug Evaluation IV, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Shiew Mei Huang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mona Khurana
- Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gilbert J. Burckart
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
42
|
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
|
43
|
Fludarabine exposure in the conditioning prior to allogeneic hematopoietic cell transplantation predicts outcomes. Blood Adv 2020; 3:2179-2187. [PMID: 31324638 DOI: 10.1182/bloodadvances.2018029421] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/30/2019] [Indexed: 12/11/2022] Open
Abstract
Fludarabine is the most frequently used agent in conditioning regimens for allogeneic hematopoietic cell transplantation (HCT). Body surface area-based dosing leads to highly variable fludarabine exposure. We studied the relation between fludarabine exposure and clinical outcomes. A retrospective, pharmacokinetic-pharmacodynamic analysis was conducted with data from patients undergoing HCT with fludarabine (160 mg/m2) as part of a myeloablative conditioning (busulfan targeted to an area under the plasma-concentration-time curve [AUC] of 90 mg*h/L) and rabbit antithymocyte globulin (6-10 mg/kg; from day -9/-12) between 2010 and 2016. Fludarabine exposure as AUC was calculated for each patient using a previously published population pharmacokinetic model and related to 2-year event-free survival (EFS) by means of (parametric) time-to-event models. Relapse, nonrelapse mortality (NRM), and graft failure were considered events. One hundred ninety-two patients were included (68 benign and 124 malignant disorders). The optimal fludarabine exposure was determined as an AUC of 20 mg*h/L. In the overexposed group, EFS was lower (hazard ratio [HR], 2.0; 95% confidence interval [CI], 1.1-3.5; P = .02), due to higher NRM (HR, 3.4; 95% CI, 1.6-6.9; P <001) associated with impaired immune reconstitution (HR, 0.43; 95% CI, 0.26-0.70; P <001). The risks of NRM and graft failure were increased in the underexposed group (HR, 3.3; 95% CI, 1.2-9.4; P = .02; HR, 4.8; 95% CI, 1.2-19; P = .02, respectively). No relationship with relapse was found. Fludarabine exposure is a strong predictor of survival after HCT, stressing the importance of optimum fludarabine dosing. Individualized dosing, based on weight and "renal function" or "therapeutic drug monitoring," to achieve optimal fludarabine exposure might improve survival.
Collapse
|
44
|
van Roessel I, Prockop S, Klein E, Boulad F, Scaradavou A, Spitzer B, Kung A, Curran K, O'Reilly RJ, Kernan NA, Cancio M, Boelens JJ. Early CD4+ T cell reconstitution as predictor of outcomes after allogeneic hematopoietic cell transplantation. Cytotherapy 2020; 22:503-510. [PMID: 32622752 DOI: 10.1016/j.jcyt.2020.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/05/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND An association between early CD4+ T cell immune reconstitution (CD4+ IR) and survival after T-replete allogeneic hematopoietic cell transplantation (HCT) has been previously reported. Here we report validation of this relationship in a separate cohort that included recipients of ex vivo T-cell-depleted (TCD) HCT. We studied the relationship between CD4+ IR and clinical outcomes. METHODS A retrospective analysis of children/young adults receiving their first allogeneic HCT for any indication between January 2008 and December 2017 was performed. We related early CD4+ IR (defined as achieving >50 CD4+ T cells/µL on two consecutive measures within 100 days of HCT) to overall survival (OS), relapse, non-relapse mortality (NRM), event-free survival (EFS) and acute graft-versus-host disease (aGVHD). Fine and Gray competing risk models and Cox proportional hazard models were used. RESULTS In this analysis, 315 patients with a median age of 10.4 years (interquartile range 5.0-16.5 years) were included. The cumulative incidence of CD4+ IR at 100 days was 66.7% in the entire cohort, 54.7% in TCD (N = 208, hazard ratio [HR] 0.47, P < 0.001), 90.0% in uCB (N = 40) and 89.6% in T-replete (N = 47) HCT recipients. In multi-variate analyses, not achieving early CD4+ IR was a predictor of inferior OS (HR 2.35, 95% confidence interval [CI] 1.46-3.79, P < 0.001) and EFS (HR 1.80, 95% CI 1.20-2.69, P = 0.004) and increased NRM (HR 6.58, 95% CI 2.82-15.38, P < 0.001). No impact of CD4+ IR on relapse or aGVHD was found. Within the TCD group, similar associations were observed. CONCLUSION In this HCT cohort, including recipients of TCD HCT, we confirmed that early CD4+ IR was an excellent predictor of outcomes. Finding strategies to predict or improve CD4+ IR may influence outcomes.
Collapse
Affiliation(s)
- Ichelle van Roessel
- Department of Pediatrics, UMC Utrecht and Princess Maxima Centrum for Pediatric Oncology, Utrecht, the Netherlands
| | - Susan Prockop
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA.
| | - Elizabeth Klein
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Farid Boulad
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andromachi Scaradavou
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Barbara Spitzer
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andrew Kung
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kevin Curran
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Richard J O'Reilly
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nancy A Kernan
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria Cancio
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
45
|
Langenhorst JB, Dorlo TPC, van Kesteren C, van Maarseveen EM, Nierkens S, de Witte MA, Boelens JJ, Huitema ADR. Clinical Trial Simulation To Optimize Trial Design for Fludarabine Dosing Strategies in Allogeneic Hematopoietic Cell Transplantation. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:272-281. [PMID: 31957334 PMCID: PMC7239337 DOI: 10.1002/psp4.12486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Optimal fludarabine exposure has been associated with improved treatment outcome in allogeneic hematopoietic cell transplantation, suggesting potential benefit of individualized dosing. A randomized controlled trial (RCT) comparing alternative fludarabine dosing strategies to current practice may be warranted, but should be sufficiently powered for a relevant end point, while still feasible to enroll. To find the optimal design, we simulated RCTs comparing current practice (160 mg/m2 ) to either covariate-based or therapeutic drug monitoring (TDM)-guided dosing with potential outcomes being nonrelapse mortality (NRM), graft failure, or relapse, and ultimately overall survival (covering all three aforementioned outcomes). The inclusion in each treatment arm (n) required to achieve 80% power was calculated for all combinations of end points and dosing comparisons. The trial requiring the lowest n for sufficient power compared TDM-guided dosing to current practice with NRM as primary outcome (n = 70, NRM decreasing from 21% to 5.7%). We conclude that a superiority trial is feasible.
Collapse
Affiliation(s)
- Jurgen B Langenhorst
- Pediatric Blood and Marrow Transplant Program, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Centre Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands.,Model-informed drug development consultant, Pharmetheus AB, Uppsala, Sweden
| | - Thomas P C Dorlo
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Charlotte van Kesteren
- Pediatric Blood and Marrow Transplant Program, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Erik M van Maarseveen
- Department of Clinical Pharmacy, University Medical Centre Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | - Stefan Nierkens
- Pediatric Blood and Marrow Transplant Program, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Centre Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | - Moniek A de Witte
- Department of Hematology, University Medical Centre Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| | - Jaap Jan Boelens
- Pediatric Blood and Marrow Transplant Program, Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Stem Cell Transplant and Cellular Therapies, Pediatrics, Memorial Sloan Kettering Cancer Centre, New York, New York, USA
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Centre Utrecht (UMCU), Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
46
|
Boelens JJ, Hosszu KK, Nierkens S. Immune Monitoring After Allogeneic Hematopoietic Cell Transplantation: Toward Practical Guidelines and Standardization. Front Pediatr 2020; 8:454. [PMID: 32974239 PMCID: PMC7472532 DOI: 10.3389/fped.2020.00454] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic cell transplantation (HCT) is often a last resort, but potentially curative treatment option for children suffering from hematological malignancies and a variety of non-malignant disorders, such as bone marrow failure, inborn metabolic disease or immune deficiencies. Although efficacy and safety of the HCT procedure has increased significantly over the last decades, the majority of the patients still suffer from severe acute toxicity, viral reactivation, acute or chronic graft-versus-host disease (GvHD) and/or, in case of malignant disease, relapses. Factors influencing HCT outcomes are numerous and versatile. For example, there is variation in the selected graft sources, type of infused cell subsets, cell doses, and the protocols used for conditioning, as well as immune suppression and treatment of adverse events. Moreover, recent pharmacokinetic studies show that medications used in the conditioning regimen (e.g., busulphan, fludarabine, anti-thymocyte globulin) should be dosed patient-specific to achieve optimal exposure in every individual patient. Due to this multitude of variables and site-specific policies/preferences, harmonization between HCT centers is still difficult to achieve. Literature shows that adequate immune recovery post-HCT limits both relapse and non-relapse mortality (death due to viral reactivations and GvHD). Monitoring immune parameters post-HCT may facilitate a timely prediction of outcome. The use of standardized assays to measure immune parameters would facilitate a fast comparison between different strategies tested in different centers or between different clinical trials. We here discuss immune cell markers that may contribute to clinical decision making and may be worth to standardize in multicenter collaborations for future trials.
Collapse
Affiliation(s)
- Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kinga K Hosszu
- Stem Cell Transplantation and Cellular Therapies, MSK Kids, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology and UMC Utrecht, Utrecht, Netherlands
| |
Collapse
|
47
|
Pai SY. Treatment of primary immunodeficiency with allogeneic transplant and gene therapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:457-465. [PMID: 31808905 PMCID: PMC6913427 DOI: 10.1182/hematology.2019000052] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The treatment of primary immunodeficiency disorders with allogeneic hematopoietic cell transplantation (HCT) has a history dating back to 1968 with the first successful transplant for a patient with severe combined immunodeficiency (SCID). The omission of conditioning for patients with SCID owing to their inability to reject allogeneic grafts and the increasing use of reduced intensity conditioning regimens often result in a state of mixed or split donor-recipient chimerism. The use of gene therapy (GT) via retroviral or lentiviral transduction of autologous CD34+ hematopoietic stem and progenitor cells is expected to correct only a portion of the hematopoietic stem cell compartment. The consequences of partial correction after either form of cellular therapy differ according to how the genetic deficiency affects immune cell development and function. Moreover, the conditioning regimen or lack thereof impacts the cell lineages at risk of partial correction. Advances in our understanding of immune reconstitution after HCT and GT for SCID, Wiskott-Aldrich syndrome, and chronic granulomatous disease are discussed.
Collapse
Affiliation(s)
- Sung-Yun Pai
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA; and
- Harvard Medical School, Boston, MA
| |
Collapse
|
48
|
Frequency of lethal central nervous system neurotoxicity in patients undergoing allogeneic stem cell transplantation: a retrospective registry analysis. Bone Marrow Transplant 2019; 55:1642-1646. [PMID: 31695171 DOI: 10.1038/s41409-019-0738-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 11/09/2022]
|
49
|
Consensus recommendations for the role and competencies of the EBMT clinical pharmacist and clinical pharmacologist involved in hematopoietic stem cell transplantation. Bone Marrow Transplant 2019; 55:62-69. [PMID: 31101890 DOI: 10.1038/s41409-019-0538-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/24/2019] [Accepted: 04/27/2019] [Indexed: 11/09/2022]
|
50
|
Admiraal R, Boelens JJ. Pharmacotherapy in Pediatric Hematopoietic Cell Transplantation. Handb Exp Pharmacol 2019; 261:471-489. [PMID: 31375921 DOI: 10.1007/164_2019_247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hematopoietic cell transplantation (HCT) is a curative treatment option for both malignant and nonmalignant diseases. Success of the procedure mainly depends on disease control and treatment-related complications. Pharmacotherapy plays a major role in HCT and significantly impacts the outcomes. Main drug use within HCT includes conditioning, GvHD prophylaxis, and prevention/treatment of infections.Increasing evidence suggests individualized dosing in (pediatric) HCT may improve outcome. Dose individualization may result in a better predictable drug treatment in terms of safety and efficacy, including timely immune reconstitution after HCT and optimal tumor or disease control, which may result in improved survival chances.
Collapse
Affiliation(s)
- R Admiraal
- Blood and Marrow Transplantation Program, Prinses Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J J Boelens
- Stem Cell Transplantation and Cellular Therapies Pediatrics, New York, NY, USA. .,Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|