1
|
John TD, Maron G, Abraham A, Bertaina A, Bhoopalan SV, Bidgoli A, Bonfim C, Coleman Z, DeZern A, Li J, Louis C, Oved J, Pavel-Dinu M, Purtill D, Ruggeri A, Russell A, Wynn R, Boelens JJ, Prockop S, Sharma A. Strategic infection prevention after genetically modified hematopoietic stem cell therapies: recommendations from the International Society for Cell & Gene Therapy Stem Cell Engineering Committee. Cytotherapy 2024; 26:660-671. [PMID: 38483362 PMCID: PMC11213676 DOI: 10.1016/j.jcyt.2024.02.005] [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: 11/07/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/19/2024]
Abstract
There is lack of guidance for immune monitoring and infection prevention after administration of ex vivo genetically modified hematopoietic stem cell therapies (GMHSCT). We reviewed current infection prevention practices as reported by providers experienced with GMHSCTs across North America and Europe, and assessed potential immunologic compromise associated with the therapeutic process of GMHSCTs described to date. Based on these assessments, and with consensus from members of the International Society for Cell & Gene Therapy (ISCT) Stem Cell Engineering Committee, we propose risk-adapted recommendations for immune monitoring, infection surveillance and prophylaxis, and revaccination after receipt of GMHSCTs. Disease-specific and GMHSCT-specific considerations should guide decision making for each therapy.
Collapse
Affiliation(s)
- Tami D John
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Gabriela Maron
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Allistair Abraham
- Center for Cancer and Immunology Research, CETI, Children's National Hospital, Washington, District of Columbia, USA
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Senthil Velan Bhoopalan
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Alan Bidgoli
- Division of Blood and Marrow Transplantation, Children's Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, Georgia, USA
| | - Carmem Bonfim
- Pediatric Blood and Marrow Transplantation Division and Pelé Pequeno Príncipe Research Institute, Hospital Pequeno Príncipe, Curitiba, Brazil
| | - Zane Coleman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Amy DeZern
- Bone Marrow Failure and MDS Program, John Hopkins Medicine, Baltimore, Maryland, USA
| | - Jingjing Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Joseph Oved
- Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mara Pavel-Dinu
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Duncan Purtill
- Department of Haematology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | | | - Athena Russell
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert Wynn
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Susan Prockop
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
2
|
Mo CC, Hartley-Brown MA, Midha S, Richardson PG. Upfront or Deferred Autologous Stem Cell Transplantation for Newly Diagnosed Multiple Myeloma in the Era of Triplet and Quadruplet Induction and Minimal Residual Disease/Risk-Adapted Therapy. Cancers (Basel) 2023; 15:5709. [PMID: 38136255 PMCID: PMC10741557 DOI: 10.3390/cancers15245709] [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: 10/25/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
The standards of care for the initial treatment of patients with newly diagnosed multiple myeloma (NDMM) who are eligible for high-dose melphalan and autologous stem cell transplantation (HDM-ASCT) include highly active triplet and quadruplet regimens based on proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies. These regimens are resulting in improved outcomes and increasingly high rates of minimal residual disease (MRD)-negative responses without HDM-ASCT as part of the upfront therapy. Furthermore, recent randomized studies have shown that, while transplant-based approaches as a frontline therapy result in significantly longer progression-free survival compared to non-transplant approaches, this has not translated into an overall survival benefit. Given these developments, and in the context of the treatment burden of undergoing HDM-ASCT, in addition to the acute toxicities and long-term sequelae of HDM, which are associated with the genotoxicity of melphalan, there is an increasing rationale for considering deferring upfront HDM-ASCT in select transplant-eligible patients and saving it as a treatment option for later salvage therapy. Here, we review the latest clinical trial data on upfront or deferred HDM-ASCT and on the activity of quadruplet induction regimens, including rates of MRD-negative responses, and summarize emerging treatment approaches in the upfront setting such as the use of MRD-directed therapy and alternatives to HDM-ASCT.
Collapse
Affiliation(s)
| | | | | | - Paul G. Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, 450 Brookline Avenue, Dana 1B02, Boston, MA 02115, USA; (C.C.M.); (M.A.H.-B.); (S.M.)
| |
Collapse
|
3
|
Krishnatry AS, Hanze E, Bergsma T, Dhar A, Prohn M, Ferron‐Brady G. Exposure-response analysis of adverse events associated with molibresib and its active metabolites in patients with solid tumors. CPT Pharmacometrics Syst Pharmacol 2022; 11:556-568. [PMID: 34648693 PMCID: PMC9124358 DOI: 10.1002/psp4.12724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 11/15/2022] Open
Abstract
Molibresib (GSK525762) is an investigational orally bioavailable small-molecule bromodomain and extraterminal (BET) protein inhibitor for the treatment of advanced solid tumors. In the first-time-in-human BET115521 study of molibresib in patients with solid tumors, thrombocytopenia was the most frequent treatment-related adverse event (AE), QT prolongation was an AE of special interest based on preclinical signals, and gastrointestinal (GI) AEs (nausea, vomiting, diarrhea, and dysgeusia) were often observed. The aims of this analysis were the following: (i) develop a population pharmacokinetic (PK)/pharmacodynamic (PD) model capable of predicting platelet time courses in individual patients after administration of molibresib and identify covariates of clinical interest; (ii) evaluate the effects of molibresib (and/or its two active metabolites [GSK3529246]) exposure on cardiac repolarization by applying a systematic modeling approach using high-quality, intensive, PK time-matched 12-lead electrocardiogram measurements; (iii) evaluate the exposure-response (ER) relationship between molibresib and/or GSK3529246 exposures and the occurrence of Grade 2 or higher GI AEs. Overall, the PK/PD model (including a maximal drug effect model and molibresib concentration) adequately described platelet counts following molibresib treatment and was used to simulate the impact of molibresib dosing on thrombocytopenia at different doses and regimens. ER analyses showed no clinically meaningful QT interval prolongation with molibresib at up to 100 mg q.d., and no strong correlation between molibresib exposure and the occurrence of Grade 2 or higher GI AEs. The models described here can aid dosing/schedule and drug combination strategies and may support a thorough QT study waiver request for molibresib.
Collapse
Affiliation(s)
- Anu Shilpa Krishnatry
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKlineCollegevillePennsylvaniaUSA
| | - Eva Hanze
- qPharmetra LLCNijmegenthe Netherlands
| | | | - Arindam Dhar
- Epigenetics Research UnitGlaxoSmithKlineCollegevillePennsylvaniaUSA
| | | | - Geraldine Ferron‐Brady
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKlineCollegevillePennsylvaniaUSA
| |
Collapse
|
4
|
Optimization of G-CSF dosing schedule in patients treated with eribulin: a modeling approach. Cancer Chemother Pharmacol 2022; 89:197-208. [PMID: 34997290 DOI: 10.1007/s00280-021-04395-y] [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: 09/08/2021] [Accepted: 12/28/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND Granulocyte colony-stimulating factors (G-CSF) are commonly given to limit chemotherapy-induced neutropenia, but, in case of weekly chemotherapy such as eribulin, their administration schedules remain empirical. OBJECTIVES This pharmacokinetic/pharmacodynamic (PK/PD) study was conducted to establish the effect of different G-CSF regimens on neutropenia's incidence for patients treated by eribulin, to propose an optimal G-CSF dosing schedule. METHODS A population PK/PD model was developed to describe absolute neutrophil counts' (ANC) time course in 87 cancer patients receiving eribulin. The structural model considered ANC dynamics, neutropenic effect of eribulin and stimulating effect of G-CSF. Final model estimates were used to calculate neutropenia's incidence following different G-CSF dosing schedules for 1000 virtual subjects. RESULTS The final model successfully described most of the ANC time course for all patients. Simulations showed that a single G-CSF administration 48 h after each eribulin injection reduced the risk of severe neutropenia from 29.7 to 5.2%. Five days of G-CSF only after the second eribulin injection or no G-CSF administration induces similar incidence of neutropenia. CONCLUSION Simulations showed a single G-CSF administration 48 h after the end of each eribulin injection seems to be the optimal schedule to reduce eribulin-induced neutropenia. However, the new administration scheme should be tested in real life to evaluate its pertinence. TRIAL REGISTRATION Eudract 2015-001753-32, 2015/01/26.
Collapse
|
5
|
Abbiati RA, Pourdehnad M, Carrancio S, Pierce DW, Kasibhatla S, McConnell M, Trotter MWB, Loos R, Santini CC, Ratushny AV. Quantitative Systems Pharmacology Modeling of Avadomide-Induced Neutropenia Enables Virtual Clinical Dose and Schedule Finding Studies. AAPS J 2021; 23:103. [PMID: 34453265 PMCID: PMC8397660 DOI: 10.1208/s12248-021-00623-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023] Open
Abstract
Avadomide is a cereblon E3 ligase modulator and a potent antitumor and immunomodulatory agent. Avadomide trials are challenged by neutropenia as a major adverse event and a dose-limiting toxicity. Intermittent dosing schedules supported by preclinical data provide a strategy to reduce frequency and severity of neutropenia; however, the identification of optimal dosing schedules remains a clinical challenge. Quantitative systems pharmacology (QSP) modeling offers opportunities for virtual screening of efficacy and toxicity levels produced by alternative dose and schedule regimens, thereby supporting decision-making in translational drug development. We formulated a QSP model to capture the mechanism of avadomide-induced neutropenia, which involves cereblon-mediated degradation of transcription factor Ikaros, resulting in a maturation block of the neutrophil lineage. The neutropenia model was integrated with avadomide-specific pharmacokinetic and pharmacodynamic models to capture dose-dependent effects. Additionally, we generated a disease-specific virtual patient population to represent the variability in patient characteristics and response to treatment observed for a diffuse large B-cell lymphoma trial cohort. Model utility was demonstrated by simulating the avadomide effect in the virtual population for various dosing schedules and determining the incidence of high-grade neutropenia, its duration, and the probability of recovery to low-grade neutropenia.
Collapse
Affiliation(s)
- Roberto A Abbiati
- Bristol Myers Squibb, Center for Innovation and Translational Research Europe (CITRE), Seville, Spain.
| | | | | | | | | | | | - Matthew W B Trotter
- Bristol Myers Squibb, Center for Innovation and Translational Research Europe (CITRE), Seville, Spain
| | - Remco Loos
- Bristol Myers Squibb, Center for Innovation and Translational Research Europe (CITRE), Seville, Spain
| | - Cristina C Santini
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | |
Collapse
|
6
|
Choi YH, Zhang C, Liu Z, Tu MJ, Yu AX, Yu AM. A Novel Integrated Pharmacokinetic-Pharmacodynamic Model to Evaluate Combination Therapy and Determine In Vivo Synergism. J Pharmacol Exp Ther 2021; 377:305-315. [PMID: 33712506 PMCID: PMC8140393 DOI: 10.1124/jpet.121.000584] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/09/2021] [Indexed: 11/22/2022] Open
Abstract
Understanding pharmacokinetic (PK)-pharmacodynamic (PD) relationships is essential in translational research. Existing PK-PD models for combination therapy lack consideration of quantitative contributions from individual drugs, whereas interaction factor is always assigned arbitrarily to one drug and overstretched for the determination of in vivo pharmacologic synergism. Herein, we report a novel generic PK-PD model for combination therapy by considering apparent contributions from individual drugs coadministered. Doxorubicin (Dox) and sorafenib (Sor) were used as model drugs whose PK data were obtained in mice and fit to two-compartment model. Xenograft tumor growth was biphasic in mice, and PD responses were described by three-compartment transit models. This PK-PD model revealed that Sor (contribution factor = 1.62) had much greater influence on overall tumor-growth inhibition than coadministered Dox (contribution factor = 0.644), which explains the mysterious clinical findings on remarkable benefits for patients with cancer when adding Sor to Dox treatment, whereas there were none when adding Dox to Sor therapy. Furthermore, the combination index method was integrated into this predictive PK-PD model for critical determination of in vivo pharmacologic synergism that cannot be correctly defined by the interaction factor in conventional models. In addition, this new PK-PD model was able to identify optimal dosage combination (e.g., doubling experimental Sor dose and reducing Dox dose by 50%) toward much greater degree of tumor-growth inhibition (>90%), which was consistent with stronger synergy (combination index = 0.298). These findings demonstrated the utilities of this new PK-PD model and reiterated the use of valid method for the assessment of in vivo synergism. SIGNIFICANCE STATEMENT: A novel pharmacokinetic (PK)-pharmacodynamic (PD) model was developed for the assessment of combination treatment by considering contributions from individual drugs, and combination index method was incorporated to critically define in vivo synergism. A greater contribution from sorafenib to tumor-growth inhibition than that of coadministered doxorubicin was identified, offering explanation for previously inexplicable clinical observations. This PK-PD model and strategy shall have broad applications to translational research on identifying optimal dosage combinations with stronger synergy toward improved therapeutic outcomes.
Collapse
Affiliation(s)
- Young Hee Choi
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California (Y.H.C., C.Z., Z.L., M.-J.T., A.-M.Y.); College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.); and Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (A.-X.Y.)
| | - Chao Zhang
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California (Y.H.C., C.Z., Z.L., M.-J.T., A.-M.Y.); College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.); and Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (A.-X.Y.)
| | - Zhenzhen Liu
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California (Y.H.C., C.Z., Z.L., M.-J.T., A.-M.Y.); College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.); and Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (A.-X.Y.)
| | - Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California (Y.H.C., C.Z., Z.L., M.-J.T., A.-M.Y.); College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.); and Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (A.-X.Y.)
| | - Ai-Xi Yu
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California (Y.H.C., C.Z., Z.L., M.-J.T., A.-M.Y.); College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.); and Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (A.-X.Y.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, University of California (UC) Davis School of Medicine, Sacramento, California (Y.H.C., C.Z., Z.L., M.-J.T., A.-M.Y.); College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Ilsandong-gu, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.); and Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China (A.-X.Y.)
| |
Collapse
|
7
|
Macaire P, Paris J, Vincent J, Ghiringhelli F, Bengrine-Lefevre L, Schmitt A. Impact of granulocyte colony-stimulating factor on FOLFIRINOX-induced neutropenia prevention: A population pharmacokinetic/pharmacodynamic approach. Br J Clin Pharmacol 2020; 86:2473-2485. [PMID: 32386071 DOI: 10.1111/bcp.14356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/21/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS Granulocyte colony-stimulating factor (G-CSF) is frequently prescribed to prevent chemotherapy-induced neutropenia, but the administration schedule remains empirical in case of bimonthly chemotherapy such as FOLFIRINOX regimen. This pharmacokinetic/pharmacodynamic (PK/PD) study was performed to determine the effect of different G-CSF regimens on the incidence and duration of neutropenia following FOLFIRINOX administration in order to propose an optimal G-CSF dosing schedule. METHODS A population PK/PD model was developed to describe individual neutrophil time course from absolute neutrophil counts (ANC) obtained in 40 advanced cancer patients receiving FOLFIRINOX regimen. The structural model considered ANC dynamics, neutropenic effect of cytotoxics and the stimulating effect of G-CSF on neutrophils. Final model estimates were used to simulate different G-CSF dosing schedules for 1000 virtual subjects. The incidence and duration of neutropenia were then calculated for different G-CSF dosing schedules. RESULTS The final model successfully described the myelosuppressive effect induced by the 3 cytotoxics for all patients. Simulations showed that pegfilgrastim administration reduced the risk of severe neutropenia by 22.9% for subjects with low ANC at the start of chemotherapy. Median duration in this group was also shortened by 3.1 days when compared to absence of G-CSF. Delayed G-CSF administration was responsible for higher incidence and longer duration of neutropenia compared to absence of administration. CONCLUSION The PK/PD model well described our population's ANC data. Simulations showed that pegylated-G-CSF administration 24 hours after the end of chemotherapy seems to be the optimal schedule to reduce FOLFIRINOX-induced neutropenia. We also underline the potential negative effect of G-CSF maladministration.
Collapse
Affiliation(s)
- Pauline Macaire
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| | - Justine Paris
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| | - Julie Vincent
- Oncology Department, Centre Georges-François Leclerc, Dijon, France
| | - François Ghiringhelli
- INSERM U1231, University of Burgundy Franche-Comté, Dijon, France.,Oncology Department, Centre Georges-François Leclerc, Dijon, France
| | | | - Antonin Schmitt
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| |
Collapse
|
8
|
Pai AA, Devasia AJ, Panetta JC, Mani S, Stallon Illangeswaran RS, Mohanan E, Balakrishnan B, Lakshmi KM, Kulkarni U, Aboobacker FN, Korula A, Abraham A, Srivastava A, Mathews V, George B, Balasubramanian P. Pharmacokinetics and Efficacy of Generic Melphalan Is Comparable to Innovator Formulation in Patients With Multiple Myeloma Undergoing Autologous Stem Cell Transplantation. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 20:130-135.e1. [PMID: 31791694 DOI: 10.1016/j.clml.2019.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/15/2019] [Accepted: 08/22/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND High-dose melphalan (MEL) is the standard conditioning regimen used for autologous stem cell transplantation (ASCT) in patients with multiple myeloma (MM). Generic MEL is routinely used in various transplant centers across the world including ours due to its reduced cost and ease of availability. We compared the pharmacokinetics (PK) and the clinical efficacy of generic MEL with that of the innovator formulation in MM patients undergoing ASCT. PATIENTS AND METHODS Sixty-three patients diagnosed with MM receiving high-dose MEL were included in this study. MEL levels in plasma were measured using a liquid chromatography tandem mass spectrometry (HPLC/MS-MS) protocol and non-linear mixed effects modeling was used to evaluate the PK of the data. RESULTS The interindividual variability (IIV) in MEL area under the concentration versus time curve (AUC) and clearance (CL) were 4.39, 5.88-fold for generic, and 4.34, 6.85-fold for the innovator formulation, respectively. The median MEL AUC and CL were comparable between the 2 formulations. The population PK analysis showed age and creatinine CL as the only significant covariates explaining IIV in MEL AUC/CL. Analysis of MEL PK parameters with clinical outcome showed no significant differences in terms of onset and severity of mucositis, day to neutrophil and platelet engraftment, as well as response status on day 100 post ASCT between patients receiving generic or innovator formulations of MEL. In addition, neither MEL AUC nor CL was found to be associated with day +100 response. CONCLUSION Our study suggests that the PK and efficacy of the generic MEL is comparable to the innovator formulation.
Collapse
Affiliation(s)
- Aswin Anand Pai
- Department of Hematology, Christian Medical College, Vellore, India
| | - Anup J Devasia
- Department of Hematology, Christian Medical College, Vellore, India
| | - John Carl Panetta
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN
| | - Sathya Mani
- Department of Hematology, Christian Medical College, Vellore, India
| | | | | | | | | | - Uday Kulkarni
- Department of Hematology, Christian Medical College, Vellore, India
| | | | - Anu Korula
- Department of Hematology, Christian Medical College, Vellore, India
| | - Aby Abraham
- Department of Hematology, Christian Medical College, Vellore, India
| | - Alok Srivastava
- Department of Hematology, Christian Medical College, Vellore, India
| | - Vikram Mathews
- Department of Hematology, Christian Medical College, Vellore, India
| | - Biju George
- Department of Hematology, Christian Medical College, Vellore, India
| | | |
Collapse
|
9
|
Persaud AK, Li J, Johnson JA, Seligson N, Sborov DW, Duah E, Cho YK, Wang D, Phelps MA, Hofmeister CC, Poi MJ. XRCC1‐mediated DNA repair is associated with progression‐free survival of multiple myeloma patients after autologous stem cell transplant. Mol Carcinog 2019; 58:2327-2339. [DOI: 10.1002/mc.23121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Avinash K. Persaud
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of PharmacyThe Ohio State University Columbus Ohio
| | - Junan Li
- Division of Pharmacy Practice and Science, College of PharmacyThe Ohio State University Columbus Ohio
- Comprehensive Cancer CenterThe Ohio State University Columbus Ohio
| | - Jasmine A. Johnson
- Division of Pharmacy Practice and Science, College of PharmacyThe Ohio State University Columbus Ohio
| | - Nathan Seligson
- Department of PharmacyThe Ohio State University Wexner Medical Center Columbus Ohio
| | - Douglas W. Sborov
- Division of Hematology and Hematologic MalignanciesUniversity of Utah—Huntsman Cancer Institute Salt Lake City Utah
| | - Ernest Duah
- Division of Pharmacy Practice and Science, College of PharmacyThe Ohio State University Columbus Ohio
| | - Yu Kyoung Cho
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of PharmacyThe Ohio State University Columbus Ohio
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research, College of PharmacyUniversity of Florida Gainesville Florida
| | - Mitch A. Phelps
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of PharmacyThe Ohio State University Columbus Ohio
- Comprehensive Cancer CenterThe Ohio State University Columbus Ohio
| | - Craig C. Hofmeister
- Department of Hematology and OncologyWinship Cancer Institute of Emory University Atlanta Georgia
| | - Ming J. Poi
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of PharmacyThe Ohio State University Columbus Ohio
- Division of Pharmacy Practice and Science, College of PharmacyThe Ohio State University Columbus Ohio
- Department of PharmacyThe Ohio State University Wexner Medical Center Columbus Ohio
| |
Collapse
|