1
|
Casanova M, Albert CM, Bautista F, Borinstein SC, Bradfield S, Bukowinski A, Campbell-Hewson Q, Hawkins DS, Kim A, Milano GM, Marshall LV, Pinto N, Pratilas CA, Rubio-San-Simón A, Windsor R, Majid O, Scott R, Jia Y, Paoletti C, Kontny U. Efficacy, safety, and pharmacokinetics of eribulin as monotherapy or in combination with irinotecan for patients with pediatric rhabdomyosarcoma, non-rhabdomyosarcoma soft tissue sarcoma, or Ewing sarcoma. ESMO Open 2025; 10:104129. [PMID: 39908698 PMCID: PMC11847257 DOI: 10.1016/j.esmoop.2024.104129] [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: 10/09/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND In this report, we present results from studies of eribulin as monotherapy (Study 223) and in combination with irinotecan (the phase II part of Study 213) for patients with relapsed/refractory pediatric rhabdomyosarcoma (RMS), non-rhabdomyosarcoma soft tissue sarcoma (NRSTS), or Ewing sarcoma (EWS). PATIENTS AND METHODS Studies 223 and 213 were phase II multicenter trials that enrolled pediatric patients with histologically confirmed disease. Treatment comprised 21-day cycles of eribulin mesylate 1.4 mg/m2 on days 1 and 8 (Study 223) or eribulin 1.4 mg/m2 on days 1 and 8 plus irinotecan 40 mg/m2 on days 1-5 (Study 213). For both studies, the primary endpoints were objective response rate (ORR) and duration of response (DOR); secondary endpoint included safety. RESULTS In Study 223, 21 patients (RMS, n = 8; NRSTS, n = 8; EWS, n = 5) were enrolled and treated. No responses were observed, resulting in early termination of enrollment. By the data cut-off date (22 February 2021), six patients (RMS, n = 3; NRSTS, n = 1; EWS, n = 2) had stable disease for ≥5 weeks. All patients had one or more treatment-emergent adverse event (TEAE), most commonly neutrophil count decreased (71.4%). In Study 213 (phase II part), 27 patients (RMS, n = 9; NRSTS, n = 9; EWS, n = 9) were enrolled/treated. By the data cut-off date (9 July 2021), three patients (one in each cohort) had had a response, resulting in an ORR of 11.1% and DORs of 2.9 (RMS), 1.4 (NRSTS), and 15.4 (EWS) months. All patients had one or more TEAE, most commonly diarrhea and neutrophil count decreased (51.9% each). CONCLUSIONS Eribulin, as monotherapy or combination therapy, exhibited a safety profile consistent with that observed previously in adult populations; however, efficacy in both studies was not considered adequate to advance investigation in these disease areas.
Collapse
Affiliation(s)
- M Casanova
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - C M Albert
- Division of Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy, Seattle Children's Hospital, Seattle; Department of Pediatrics, University of Washington School of Medicine, Seattle, USA
| | | | - S C Borinstein
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville
| | - S Bradfield
- Division of Pediatric Hematology/Oncology, Nemours Children's Health, Jacksonville
| | - A Bukowinski
- Department of Hematology/Oncology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Q Campbell-Hewson
- The Great North Children's Hospital, Royal Victoria Infirmary, Newcastle Upon Tyne, UK
| | - D S Hawkins
- Division of Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy, Seattle Children's Hospital, Seattle; Department of Pediatrics, University of Washington School of Medicine, Seattle, USA
| | - A Kim
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, USA
| | - G M Milano
- Division of Pediatric Hematology and Oncology, Gene and Cellular Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - L V Marshall
- Children and Young People's Unit, The Royal Marsden Hospital, and Divisions of Clinical Studies and Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - N Pinto
- Children's Hospital Colorado, Aurora
| | - C A Pratilas
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - A Rubio-San-Simón
- Department of Pediatric Oncology and Hematology, Hospital del Niño Jesús, Madrid, Spain
| | - R Windsor
- Children and Young People's Cancer Service, University College London Hospitals NHS Foundation Trust, London
| | | | | | | | | | - U Kontny
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Aachen, Aachen, Germany
| |
Collapse
|
2
|
Hayato S, Hamuro L, Shimizu T, Yonemori K, Nishio S, Yunokawa M, Yoshida T, Nishio M, Matsumoto K, Takehara K, Hasegawa K, Kozuki T, Hirashima Y, Kato H, Miura T, Nomoto M, Zhao Y, Zhu L, Yasuda S. Pharmacokinetic and Exposure-Response Modeling Support Body Surface Area-Based Dosing of Farletuzumab Ecteribulin in Japanese Patients with Solid Tumors. J Clin Pharmacol 2025. [PMID: 39853764 DOI: 10.1002/jcph.6187] [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: 09/11/2024] [Accepted: 12/30/2024] [Indexed: 01/26/2025]
Abstract
The first-in-human, Phase 1 Study 101 showed antitumor activity and a tolerable safety profile of farletuzumab ecteribulin in Japanese patients with platinum-resistant ovarian and non-small cell lung cancer. A pharmacometric assessment evaluated farletuzumab ecteribulin pharmacokinetics and exposure-response (E-R) relationships for efficacy and safety to support dose optimization. Patients received 0.3-1.2 mg/kg of farletuzumab ecteribulin intravenously every 3 weeks. A pharmacokinetics (PK) model was developed and used for E-R analyses. Efficacy was assessed via tumor response and safety via known treatment-emergent adverse events (TEAEs) of farletuzumab ecteribulin, particularly pneumonitis/interstitial lung disease (ILD). Dosing scenarios were simulated to identify dosing that maximizes the probability of an objective response while minimizing the risk of ILD. The farletuzumab ecteribulin PK dataset included 1261 observations from 82 patients. The final model included an estimated population mean value for farletuzumab ecteribulin clearance of 0.0162 L/h. Body surface area (BSA) was a significant PK covariate and was included in the model. Body weight (BW) was associated with higher farletuzumab ecteribulin exposure. Using BW-based dosing, farletuzumab ecteribulin AUC (area under the serum concentration-time curve) was higher in patients with tumor response or stable disease versus patients with progressive disease and higher in patients with ILD and other TEAEs. Dosing simulations showed that BSA-based dosing (33 mg/m2) yielded similar tumor responses to BW-based dosing (0.9 mg/kg) and decreased ILD rates. This study showed that BW-based dosing resulted in higher risks of ILD events for patients with a high BW versus low BW, whereas BSA-based dosing is predicted to reduce this risk while maintaining clinical efficacy.
Collapse
Affiliation(s)
| | | | - Toshio Shimizu
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of New Experimental Therapeutics and International Cancer New Drug Development Center, Kansai Medical University Hospital, Osaka, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shin Nishio
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Fukuoka, Japan
| | - Mayu Yunokawa
- Department of Gynecologic Oncology, Cancer Institute Hospital, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Makoto Nishio
- Department of Thoracic Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koji Matsumoto
- Division of Medical Oncology, Hyogo Cancer Center, Hyogo, Japan
| | - Kazuhiro Takehara
- Department of Gynecologic Oncology, NHO Shikoku Cancer Center, Ehime, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | | | - Hidenori Kato
- Department of Gynecologic Oncology, Hokkaido Cancer Center, Sapporo, Japan
| | | | | | - Yue Zhao
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Li Zhu
- Bristol Myers Squibb, Princeton, NJ, USA
| | | |
Collapse
|
3
|
Briki M, Murisier A, Guidi M, Seydoux C, Buclin T, Marzolini C, Girardin FR, Thoma Y, Carrara S, Choong E, Decosterd LA. Liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) methods for the therapeutic drug monitoring of cytotoxic anticancer drugs: An update. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1236:124039. [PMID: 38490042 DOI: 10.1016/j.jchromb.2024.124039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 03/17/2024]
Abstract
In the era of precision medicine, there is increasing evidence that conventional cytotoxic agents may be suitable candidates for therapeutic drug monitoring (TDM)- guided drug dosage adjustments and patient's tailored personalization of non-selective chemotherapies. To that end, many liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) assays have been developed for the quantification of conventional cytotoxic anticancer chemotherapies, that have been comprehensively and critically reviewed. The use of stable isotopically labelled internal standards (IS) of cytotoxic drugs was strikingly uncommon, accounting for only 48 % of the methods found, although their use could possible to suitably circumvent patients' samples matrix effects variability. Furthermore, this approach would increase the reliability of cytotoxic drug quantification in highly multi-mediated cancer patients with complex fluctuating pathophysiological and clinical conditions. LC-MS/MS assays can accommodate multiplexed analyses of cytotoxic drugs with optimal selectivity and specificity as well as short analytical times and, when using stable-isotopically labelled IS for quantification, provide concentrations measurements with a high degree of certainty. However, there are still organisational, pharmacological, and medical constraints to tackle before TDM of cytotoxic drugs can be more largely adopted in the clinics for contributing to our ever-lasting quest to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- M Briki
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Bio/CMOS Interfaces Laboratory, École Polytechnique Fédérale de Lausanne-EPFL, 2002 Neuchâtel, Switzerland
| | - A Murisier
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - M Guidi
- Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, 1206 Geneva, Switzerland; Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - C Seydoux
- Internal Medicine Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - T Buclin
- Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - C Marzolini
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - F R Girardin
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Y Thoma
- School of Engineering and Management Vaud, HES-SO University of Applied Sciences and Arts Western Switzerland, 1401 Yverdon-les-Bains, Switzerland
| | - S Carrara
- Bio/CMOS Interfaces Laboratory, École Polytechnique Fédérale de Lausanne-EPFL, 2002 Neuchâtel, Switzerland
| | - E Choong
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - L A Decosterd
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland.
| |
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
|
Macpherson IR, He Y, Palmieri C. Eribulin, Child-Pugh score, and liver-function tests: lessons from pivotal breast cancer studies 301 and 305. Breast Cancer Res 2021; 23:33. [PMID: 33736675 PMCID: PMC7977154 DOI: 10.1186/s13058-021-01407-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/17/2021] [Indexed: 11/12/2022] Open
Abstract
Background The recommended starting dose of eribulin in patients with hepatic impairment is based on the Child-Pugh score, largely informed by a pharmacokinetic study of 18 patients. In the pivotal studies of eribulin in metastatic breast cancer (Study 301 and Study 305 [EMBRACE]), entry criteria and dose modifications were based on liver-function test (LFT) results rather than Child-Pugh score. In populations such as patients with metastatic breast cancer, in which metastatic infiltration is the predominant cause of hepatic impairment, using Child-Pugh score may be problematic; in clinical practice, it has been more common for oncologists to make dosing decisions based on LFTs. To address this, the effects of abnormal baseline LFT results on eribulin efficacy and safety were investigated. Methods In this pooled post hoc analysis, 1062 patients who were randomized to receive eribulin in Studies 301 and 305 were divided into 4 groups: (A) no elevated LFT results (no liver impairment); (B) increased levels of aspartate aminotransferase and/or alanine aminotransferase; (C) decreased albumin and/or increased levels of aspartate aminotransferase and/or alanine aminotransferase but not increased bilirubin; and (D) increased bilirubin. Patients were subcategorized by presence of liver metastasis. Drug exposure, dose intensity, and treatment-emergent adverse events (TEAEs) were analyzed. Results Eribulin mesylate mean dosage was 0.82 (group A)–0.65 mg/m2/week (group D). Group D had shorter treatment, more dose reductions/delays, more TEAEs leading to dose modifications, and numerically lower objective response rates and clinical benefit rates versus groups A–C. TEAE rates leading to dose modification were similar between group D (45.5%) and groups A–C (range, 43.5–54.9%) in the absence of liver metastases, but higher in group D (91.3%) compared with groups A–C (range, 41.7–54.3%) if liver metastases were present. Conclusions Mild elevations in bilirubin levels were associated with increased toxicity and a greater requirement for dose modifications. Based both on these study data and existing recommendations, we propose a novel scheme to guide initial dose selection in patients with metastatic breast cancer and hepatic impairment that is based on LFTs rather than Child-Pugh score. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01407-w.
Collapse
Affiliation(s)
- Iain R Macpherson
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Yaohua He
- Formerly of Eisai Inc., Woodcliff Lake, NJ, USA
| | - Carlo Palmieri
- Department of Molecular and Clinical Cancer Medicine, The Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK. .,Academic Department of Medical Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
6
|
Tarcsa E, Guffroy MR, Falahatpisheh H, Phipps C, Kalvass JC. Antibody-drug conjugates as targeted therapies: Are we there yet? A critical review of the current clinical landscape. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 37:13-22. [PMID: 34895651 DOI: 10.1016/j.ddtec.2020.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/15/2020] [Indexed: 06/14/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted therapies with the expectation of broadened therapeutic window due to tumor-specific drug delivery. Recent approvals, including ADCs with a novel payload class, topoisomerase-1 inhibitors, generated renewed excitement in the field. We provide a critical review of approved and late-stage molecules, discuss strategies in solid tumors and ADCs outside oncology. Our pharmacokinetics-based assessment of targeting suggests that ADCs, especially in solid tumors, rely on additional mechanisms for efficacy including slow-release of the payload to the circulation at potentially efficacious levels. Further adjustments in the technology are needed to fulfill the promise of true targeted drug delivery.
Collapse
Affiliation(s)
- Edit Tarcsa
- AbbVie Bioresearch Center, 100 Research Dr., Worcester, MA, 01605, USA.
| | | | | | - Colin Phipps
- AbbVie Inc., 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| | - John C Kalvass
- AbbVie Inc., 1 North Waukegan Rd, North Chicago, IL, 60064, USA
| |
Collapse
|
7
|
Yin A, Moes DJAR, van Hasselt JGC, Swen JJ, Guchelaar HJ. A Review of Mathematical Models for Tumor Dynamics and Treatment Resistance Evolution of Solid Tumors. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:720-737. [PMID: 31250989 PMCID: PMC6813171 DOI: 10.1002/psp4.12450] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/17/2019] [Indexed: 12/19/2022]
Abstract
Increasing knowledge of intertumor heterogeneity, intratumor heterogeneity, and cancer evolution has improved the understanding of anticancer treatment resistance. A better characterization of cancer evolution and subsequent use of this knowledge for personalized treatment would increase the chance to overcome cancer treatment resistance. Model‐based approaches may help achieve this goal. In this review, we comprehensively summarized mathematical models of tumor dynamics for solid tumors and of drug resistance evolution. Models displayed by ordinary differential equations, algebraic equations, and partial differential equations for characterizing tumor burden dynamics are introduced and discussed. As for tumor resistance evolution, stochastic and deterministic models are introduced and discussed. The results may facilitate a novel model‐based analysis on anticancer treatment response and the occurrence of resistance, which incorporates both tumor dynamics and resistance evolution. The opportunities of a model‐based approach as discussed in this review can be of great benefit for future optimizing and personalizing anticancer treatment.
Collapse
Affiliation(s)
- Anyue Yin
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan G C van Hasselt
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
8
|
Nair S, LLerena A. New perspectives in personalised medicine for ethnicity in cancer: population pharmacogenomics and pharmacometrics. Drug Metab Pers Ther 2018; 33:61-64. [PMID: 29688886 DOI: 10.1515/dmpt-2018-0008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sujit Nair
- Amrita Cancer Discovery Biology Laboratory, Amrita Vishwa Vidyapeetham University, Amritapuri, Clappana P.O., Kollam - 690525, Kerala, India
| | - Adrián LLerena
- CICAB Clinical Research Centre at Extremadura University Hospital and Medical School, Universidad de Extremadura, Badajoz, Spain
| |
Collapse
|
9
|
Kawamura T, Kasai H, Fermanelli V, Takahashi T, Sakata Y, Matsuoka T, Ishii M, Tanigawara Y. Pharmacodynamic analysis of eribulin safety in breast cancer patients using real-world postmarketing surveillance data. Cancer Sci 2018; 109:2822-2829. [PMID: 29933506 PMCID: PMC6125471 DOI: 10.1111/cas.13708] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 01/26/2023] Open
Abstract
Postmarketing surveillance is useful to collect safety data in real-world clinical settings. In this study, we applied postmarketing real-world data on a mechanistic model analysis for neutropenic profiles of eribulin in patients with recurrent or metastatic breast cancer. Demographic and safety data were collected using an active surveillance method from eribulin-treated recurrent or metastatic breast cancer patients. Changes in neutrophil counts over time were analyzed using a mechanistic pharmacodynamic model. Pathophysiological factors that might affect the severity of neutropenia were investigated, and neutropenic patterns were simulated for different treatment schedules. Clinical and laboratory data were collected from 401 patients (5199 neutrophil count measurements) who had not received granulocyte colony-stimulating factor and were eligible for pharmacodynamic analysis. The estimated mean parameters were as follows: mean transit time = 104.5 h, neutrophil proliferation rate constant = 0.0377 h-1 , neutrophil elimination rate constant = 0.0295 h-1 , and linear coefficient of drug effect = 0.0413 mL/ng. Low serum albumin levels and low baseline neutrophil counts were associated with severe neutropenia. The probability of grade ≥3 neutropenia was predicted to be 69%, 27%, and 27% for patients on standard, biweekly, and triweekly treatment scenarios, respectively, based on virtual simulations using the developed pharmacodynamic model. In conclusion, this is the first application of postmarketing surveillance data to a model-based safety analysis. This analysis of safety data reflecting authentic clinical settings will provide useful information on the safe use and potential risk factors of eribulin.
Collapse
Affiliation(s)
- Takahisa Kawamura
- Department of Clinical Pharmacokinetics and PharmacodynamicsKeio University School of MedicineTokyoJapan
- Division of Thoracic OncologyShizuoka Cancer CenterShizuokaJapan
| | | | - Valentina Fermanelli
- Department of Clinical Pharmacokinetics and PharmacodynamicsKeio University School of MedicineTokyoJapan
- Mathematical SciencesChalmers University of TechnologyGöteborgSweden
- University of GothenburgGöteborgSweden
| | | | | | | | | | - Yusuke Tanigawara
- Department of Clinical Pharmacokinetics and PharmacodynamicsKeio University School of MedicineTokyoJapan
| |
Collapse
|
10
|
Nair S, Kong ANT. Emerging roles for clinical pharmacometrics in cancer precision medicine. ACTA ACUST UNITED AC 2018; 4:276-283. [PMID: 30345221 DOI: 10.1007/s40495-018-0139-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Purpose of review Although significant progress has been made in cancer research, there exist unmet needs in patient care as reflected by the 'Cancer Moonshot' goals. This review appreciates the potential utility of quantitative pharmacology in cancer precision medicine. Recent findings Precision oncology has received federal funding largely due to 'The Precision Medicine Initiative'. Precision medicine takes into account the inter-individual variability, and allows for tailoring the right medication or the right dose of drug to the best subpopulation of patients who will likely respond to the intervention, thus enhancing therapeutic success and reducing "financial toxicity" to patients, families and caregivers. The National Cancer Institute (NCI) committed US$ 70 million from its fiscal year 2016 budget to advance precision oncology research. Through the 'Critical Path Initiative', pharmacometrics has gained an important role in drug development; however, it is yet to find widespread clinical applicability. Summary Stakeholders including clinicians and pharmacometricians need to work in concert to ensure that benefits of model-based approaches are harnessed to personalize cancer care to the individual needs of the patient via better dosing strategies, companion diagnostics, and predictive biomarkers. In medical oncology, where immediate patient care is the clinician's primary concern, pharmacometric approaches can be tailored to build models that rely on patient data already digitally available in the Electronic Health Record (EHR) to facilitate quick collaboration and avoid additional funding needs. Taken together, we offer a roadmap for the future of precision oncology which is fraught with both challenges and opportunities for pharmacometricians and clinicians alike.
Collapse
Affiliation(s)
- Sujit Nair
- Amrita Cancer Discovery Biology Laboratory, Amrita Vishwa Vidyapeetham University, Amritapuri, Clappana P.O., Kollam - 690525, Kerala, India
| | - Ah-Ng Tony Kong
- Center for Cancer Chemoprevention Research and Department of Pharmaceutics, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ-08854, USA
| |
Collapse
|
11
|
Koliou P, Karavasilis V, Theochari M, Pollack SM, Jones RL, Thway K. Advances in the treatment of soft tissue sarcoma: focus on eribulin. Cancer Manag Res 2018; 10:207-216. [PMID: 29440930 PMCID: PMC5798537 DOI: 10.2147/cmar.s143019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Eribulin mesylate is a synthetic derivative of halichondrin B isolated from a marine sponge. Its mechanism of action is through microtubule inhibition, which is different from that of taxanes. Eribulin has been approved for the treatment of metastatic breast cancer and more recently for non-operable or metastatic liposarcoma in patients who have received prior anthracycline chemotherapy. The major side effects of eribulin are bone marrow suppression including neutropenia, leukopenia, anemia, and fatigue/weakness, which can be well managed. In this article, we reviewed evidence from the latest published data on eribulin and its use in the treatment of soft tissue sarcomas. We explored the drug’s mechanism of action, pharmacodynamics, pharmacokinetics, and metabolism. Lastly, we reviewed all preclinical studies as well as clinical trials that investigated eribulin.
Collapse
Affiliation(s)
- Panagiotis Koliou
- The London Sarcoma Service, University College London Hospital, London, UK
| | | | | | - Seth M Pollack
- Division of Oncology, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
12
|
Maeda S, Saimura M, Minami S, Kurashita K, Nishimura R, Kai Y, Yano H, Mashino K, Mitsuyama S, Shimokawa M, Tamura K. Efficacy and safety of eribulin as first- to third-line treatment in patients with advanced or metastatic breast cancer previously treated with anthracyclines and taxanes. Breast 2017; 32:66-72. [DOI: 10.1016/j.breast.2016.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 10/20/2022] Open
|
13
|
Tan AR, Sarantopoulos J, Lee L, Reyderman L, He Y, Olivo M, Goel S. Pharmacokinetics of eribulin mesylate in cancer patients with normal and impaired renal function. Cancer Chemother Pharmacol 2015; 76:1051-61. [PMID: 26433580 PMCID: PMC4612322 DOI: 10.1007/s00280-015-2878-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 09/16/2015] [Indexed: 02/01/2023]
Abstract
Purpose
To evaluate the effect of renal impairment on eribulin mesylate pharmacokinetics following a single dose in adults with advanced solid tumors. Methods Patients were grouped by renal function: moderate impairment (creatinine clearance [CrCl] 30–50 mL/min), severe impairment (CrCl 15–29 mL/min), or normal (CrCl ≥80 mL/min). During each 21-day cycle, eribulin mesylate doses (days 1 and 8) were administered intravenously: moderate, 1.1 mg/m2 (except cycle 1 day 1, 1.4 mg/m2); severe, 0.7 mg/m2; normal, 1.4 mg/m2. Results Nineteen patients were enrolled (normal, n = 6; moderate, n = 7; severe, n = 6). Renal impairment was associated with an increased mean dose-normalized area under the concentration–time curve (ratios for moderate/normal and severe/normal: 1.49; 90 % confidence interval [CI] 0.9, 2.45). CrCl and renal function correlated positively, with a numerically small slope (0.0184; 90 % CI −0.00254, 0.0394). A simulated dose reduction to eribulin 1.1 mg/m2 in patients with moderate or severe renal impairment achieved the same exposure as 1.4 mg/m2 in those with normal renal function. All groups had similar toxicity profiles, with no unexpected adverse events. Conclusions Renal impairment decreased eribulin clearance and increased exposure. Pharmacokinetic evaluation supports an eribulin dose reduction to 1.1 mg/m2 in patients with moderate or severe renal impairment. ClinicalTrials.gov Identifier
NCT01418677.
Collapse
Affiliation(s)
- Antoinette R Tan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.,Department of Medical Oncology, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, 28204, USA
| | - John Sarantopoulos
- Institute for Drug Development, Cancer Therapy and Research Center, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA
| | - Lucy Lee
- Department of Clinical Pharmacology and Translational Medicine, Eisai Inc., Woodcliff Lake, NJ, 07677, USA
| | - Larisa Reyderman
- Department of Clinical Pharmacology and Translational Medicine, Oncology, Eisai Inc., Woodcliff Lake, NJ, 07677, USA
| | - Yi He
- Department of Biostatistics, Oncology, Eisai Inc., Woodcliff Lake, NJ, 07677, USA
| | - Martin Olivo
- Department of Oncology Clinical Development, Eisai Inc., Woodcliff Lake, NJ, 07677, USA
| | - Sanjay Goel
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Road, Bronx, NY, 10461, USA.
| |
Collapse
|
14
|
van Hasselt JGC, Gupta A, Hussein Z, Beijnen JH, Schellens JHM, Huitema ADR. Disease Progression/Clinical Outcome Model for Castration-Resistant Prostate Cancer in Patients Treated With Eribulin. CPT Pharmacometrics Syst Pharmacol 2015; 4:386-95. [PMID: 26312162 PMCID: PMC4544052 DOI: 10.1002/psp4.49] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 04/22/2015] [Indexed: 12/16/2022] Open
Abstract
Frameworks that associate cancer dynamic disease progression models with parametric survival models for clinical outcome have recently been proposed to support decision making in early clinical development. Here we developed such a disease progression clinical outcome model for castration-resistant prostate cancer (CRPC) using historical phase II data of the anticancer agent eribulin. Disease progression was captured using the dynamics of prostate-specific antigen (PSA). For clinical outcome, overall survival (OS) was used. The model for PSA dynamics comprised parameters for baseline PSA (23.2 ng/ml, relative standard error (RSE) 16.5%), growth rate (0.00879 day(-1), RSE 12.6%), drug effect (0.241 µg·h·l(-1) day(-1), RSE 32.6%), and resistance development (0.0113 day(-1), RSE 44.3%). OS was modeled according to a Weibull distribution. Predictors for survival included model-predicted PSA time to nadir (TTN), PSA growth rate, Eastern Cooperative Oncology Group (ECOG) score, and baseline PSA. The developed framework can be considered to support informative design and analysis of drugs developed for CRPC.
Collapse
Affiliation(s)
- JGC van Hasselt
- Department of Clinical Pharmacology, Netherlands Cancer InstituteAmsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer InstituteAmsterdam, The Netherlands
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden UniversityLeiden, The Netherlands
| | | | | | - JH Beijnen
- Department of Clinical Pharmacology, Netherlands Cancer InstituteAmsterdam, The Netherlands
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Clinical Pharmacology and Pharmacoepidemiology, Utrecht UniversityUtrecht, The Netherlands
| | - JHM Schellens
- Department of Pharmacy & Pharmacology, Netherlands Cancer InstituteAmsterdam, The Netherlands
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Clinical Pharmacology and Pharmacoepidemiology, Utrecht UniversityUtrecht, The Netherlands
| | - ADR Huitema
- Department of Clinical Pharmacology, Netherlands Cancer InstituteAmsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer InstituteAmsterdam, The Netherlands
| |
Collapse
|
15
|
van Hasselt JGC, Gupta A, Hussein Z, Beijnen JH, Schellens JHM, Huitema ADR. Integrated Simulation Framework for Toxicity, Dose Intensity, Disease Progression, and Cost Effectiveness for Castration-Resistant Prostate Cancer Treatment With Eribulin. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:374-85. [PMID: 26312161 PMCID: PMC4544051 DOI: 10.1002/psp4.48] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 04/22/2015] [Indexed: 01/08/2023]
Abstract
Quantitative model-based analyses are helpful to support decision-making in drug development. In oncology, disease progression/clinical outcome (DPCO) models have been used for early predictions of clinical outcome, but most of such approaches did not include adverse events or dose intensity. In addition, cost-effectiveness evaluations of investigational compounds are becoming increasingly important. Here, we developed an integrated model-based framework including relevant treatment effects for patients with castration-resistant prostate cancer treated with the anticancer agent eribulin. The framework included (i) a DPCO model relating prostate-specific antigen (PSA) dynamics to survival; (ii) models for adverse events including dose-limiting neutropenia and other graded toxicities; (iii) a model for Eastern Cooperative Oncology Group (ECOG) performance score; (iv) a model for dropout; (v) the consideration of cost effectiveness. The model allowed simulation of realistic treatment courses. Subsequently, simulations evaluating alternative treatment protocols or patient characteristics were performed in order to derive inferences on expected efficacy and cost effectiveness.
Collapse
Affiliation(s)
- J G C van Hasselt
- Department of Clinical Pharmacology, Netherlands Cancer Institute Amsterdam, The Netherlands ; Department of Pharmacy & Pharmacology, Netherlands Cancer Institute Amsterdam, The Netherlands ; Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University Leiden, The Netherlands
| | | | | | - J H Beijnen
- Department of Clinical Pharmacology, Netherlands Cancer Institute Amsterdam, The Netherlands ; Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht University Utrecht, The Netherlands
| | - J H M Schellens
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute Amsterdam, The Netherlands ; Division of Pharmacoepidemiology & Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht University Utrecht, The Netherlands
| | - A D R Huitema
- Department of Clinical Pharmacology, Netherlands Cancer Institute Amsterdam, The Netherlands ; Department of Pharmacy & Pharmacology, Netherlands Cancer Institute Amsterdam, The Netherlands
| |
Collapse
|
16
|
Joy A, Ghosh M, Fernandes R, Clemons M. Systemic treatment approaches in her2-negative advanced breast cancer-guidance on the guidelines. Curr Oncol 2015; 22:S29-42. [PMID: 25848337 PMCID: PMC4381789 DOI: 10.3747/co.22.2360] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite advancements in the treatment of early-stage breast cancer, many patients still develop disease recurrence; others present with de novo metastatic disease. For most patients with advanced breast cancer, the primary treatment intent is noncurative-that is, palliative-in nature. The goals of treatment should therefore focus on maximizing symptom control and extending survival. Treatments should be evaluated on an individualized basis in terms of evidence, but also with full respect for the wishes of the patient in terms of acceptable toxicity. Given the availability of extensive reviews on the roles of endocrine therapy and her2 (human epidermal growth factor receptor 2)-targeted therapies for advanced disease, we focus here mainly on treatment guidelines for the non-endocrine management of her2-negative advanced breast cancer in a Canadian health care context.
Collapse
Affiliation(s)
- A.A. Joy
- Department of Oncology, Division of Medical Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB
| | - M. Ghosh
- Department of Oncology, Division of Medical Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB
| | - R. Fernandes
- Division of Medical Oncology, University of Ottawa and The Ottawa Hospital Research Institute, The Ottawa Hospital Cancer Centre, Ottawa, ON
| | - M.J. Clemons
- Division of Medical Oncology, University of Ottawa and The Ottawa Hospital Research Institute, The Ottawa Hospital Cancer Centre, Ottawa, ON
| |
Collapse
|