1
|
Lin K, Michaels E, Polley E, O'Donnell PH, Howard FM, Hahn O, Fleming GF, Nanda R, Chen N, Yang H. Retrospective evaluation of adjuvant capecitabine dosing patterns in triple negative breast cancer. J Oncol Pharm Pract 2024:10781552241289581. [PMID: 39397422 DOI: 10.1177/10781552241289581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
BACKGROUND The CREATE-X trial demonstrated that adjuvant capecitabine was effective in prolonging survival in high-risk triple-negative breast cancer (TNBC) patients. However, the recommended dose is generally not well tolerated by the US population. The goal of this study is to analyze dosing patterns in an ethnically diverse cohort to better characterize tolerability and inform future dosing guidelines. METHODS In our single-center retrospective study, we evaluated safety and tolerability in TNBC patients undergoing adjuvant capecitabine treatment. The primary endpoint, relative dose intensity (RDI) across eight cycles, was examined alongside subgroup analyses based on age, race, BMI, and initial dose. Secondary endpoints include capecitabine-related side effects and survival. RESULTS 67 patients who completed adjuvant capecitabine at University of Chicago Medicine (UCM) between January 2017 and November 2022 were eligible. The mean RDI across eight cycles of treatment was 60.2% (95% CI: 0.554-0.650). When compared to the CREATE-X trial, the RDI in our population was significantly lower (0.602 vs. 0.787, p < 0.001). There was no statistically significant difference in average RDI across eight cycles for patients stratified by age, BMI, race, or initial starting dose. The most frequently reported adverse events were hand-foot syndrome (73%), diarrhea (27%), and fatigue (22%), consistent with prior studies. CONCLUSIONS Our data demonstrates that a significant portion of patients have a lower tolerated dose of capecitabine in comparison to the recommended adjuvant dose. Acknowledging the limitations of our single-center analysis, RDI was not significantly affected by age, race, BMI, or initial starting dose.
Collapse
Affiliation(s)
- Kun Lin
- Department of Pharmacy, University of Chicago Medicine, Chicago, IL, USA
| | - Elena Michaels
- Department of Internal Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Eric Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | | | | | - Olwen Hahn
- University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Gini F Fleming
- University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Rita Nanda
- University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Nan Chen
- University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Heng Yang
- Department of Pharmacy, University of Chicago Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Lim JSJ, Ow SGW, Wong ALA, Lee MXW, Chan GHJ, Low JL, Sundar R, Choo JRE, Chong WQ, Ang YLE, Tai BC, Lee SC. Phase II study of trifluridine/tipiracil in metastatic breast cancers with or without prior exposure to fluoropyrimidines. Eur J Cancer 2023; 193:113311. [PMID: 37717281 DOI: 10.1016/j.ejca.2023.113311] [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: 04/21/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Fluoropyrimidines are commonly used in the treatment of metastatic breast cancer (MBC), and trifluridine/tipiracil (FTD/TPI) has shown activity in patients with colorectal and gastric cancers despite prior exposure to fluoropyrimidines. We investigate the role of FTD/TPI in patients with MBC with or without prior fluoropyridines in a single-arm phase II study. METHODS Patients with MBC were enroled first into a run-in dose confirmation phase, followed by two parallel cohorts including patients with (Cohort A) and without (Cohort B) prior exposure to fluoropyrimidines, where they were treated with FTD/TPI. Primary objectives for each cohort included determination of progression-free survival (PFS), and secondary objectives included determination of objective response rates (ORR), safety, and tolerability. RESULTS Seventy-four patients (42 Cohort A, 32 Cohort B) were enroled, all of whom were evaluable for toxicity and survival, with 72 evaluable for response. Median PFS was 5.7 months (95% confidence interval 3.8-8.3) and 9.4 months (95% CI 5.5-14.0) respectively in Cohorts A and B. Responses were observed regardless of prior exposure to fluoropyrimidines, with ORR of 19.5% (95% CI 8.8-34.9) and 16.1% (95% CI 5.5-33.7) in Cohorts A and B, and 6-month clinical benefit rates of 56.1% (95% CI 39.7-71.5) and 61.3% (95% CI 42.2-78.2) respectively. The safety profile was consistent with known toxicities of FTD/TPI, including neutropenia, fatigue, nausea, and anorexia, mitigated with dose modifications. CONCLUSION FTD/TPI showed promising antitumour activity with manageable toxicity and is a clinically valid option in patients with MBC.
Collapse
Affiliation(s)
- Joline S J Lim
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore; Experimental Therapeutics Programme, Cancer Science Institute, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Samuel G W Ow
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Andrea L A Wong
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore; Experimental Therapeutics Programme, Cancer Science Institute, Singapore, Singapore
| | - Matilda X W Lee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Gloria H J Chan
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Jia Li Low
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Raghav Sundar
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Joan R E Choo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Wan Qin Chong
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Yvonne L E Ang
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Bee Choo Tai
- Saw Swee Hock School of Public Health, National University Singapore, Singapore
| | - Soo Chin Lee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, Singapore; Experimental Therapeutics Programme, Cancer Science Institute, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore.
| |
Collapse
|
3
|
Al-Mahayri ZN, Patrinos GP, Ali BR. Toxicity and Pharmacogenomic Biomarkers in Breast Cancer Chemotherapy. Front Pharmacol 2020; 11:445. [PMID: 32351390 PMCID: PMC7174767 DOI: 10.3389/fphar.2020.00445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/20/2020] [Indexed: 02/05/2023] Open
Abstract
Breast cancer (BC) is one of the most prevalent types of cancer worldwide with high morbidity and mortality rates. Treatment modalities include systemic therapy, in which chemotherapy is a major component in many cases. Several chemotherapeutic agents are used in combinations or as single agents with many adverse events occurring in variable frequencies. These events can be a significant barrier in completing the treatment regimens. Germline genomic variants are thought of as potential determinants in chemotherapy response and the development of side effects. Some pharmacogenomic studies were designed to explore germline variants that can be used as biomarkers for predicting developing toxicity or adverse events during chemotherapy in BC. In this review, we reassess and summarize the major findings of pharmacogenomic studies of chemotherapy toxicity during BC management. In addition, deficiencies hampering utilizing these findings and the potential targets of future research are emphasized. Main insufficiencies in toxicity pharmacogenomics studies originate from study design, sample limitations, heterogeneity of selected genes, variants, and toxicity definitions. With the advent of high throughput genotyping techniques, researchers are expected to explore the identified as well as the potential genetic biomarkers of toxicity and efficacy to improve BC management. However, to achieve this, the limitations of previous work should be evaluated and avoided to reach more conclusive and translatable evidence for personalizing BC chemotherapy.
Collapse
Affiliation(s)
- Zeina N Al-Mahayri
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - George P Patrinos
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece.,Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
4
|
Bolaji OO, Adehin A, Adeagbo BA. Pharmacogenomics in the Nigerian population: the past, the present and the future. Pharmacogenomics 2019; 20:915-926. [DOI: 10.2217/pgs-2019-0046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Nigerian population exhibits huge ethnic and genetic diversity, typical of African populations, which can be harnessed for improved drug-response and disease management. Existing data on genes relevant to drug response, so far generated for the population, indeed confirm the prevalence of some clinically significant pharmacogenes. These reports detail prevailing genetic alleles and metabolic phenotypes of vital drug metabolizing monooxygenases, transferases and drug transporters. While the utilization of existing pharmacogenomic data for healthcare delivery remains unpopular, several past and on-going studies suggest that a future shift toward genotype-stratified dosing of drugs and disease management in the population is imminent. This review discusses the present state of pharmacogenomics in Nigeria and the potential benefits of sustained research in this field for the population.
Collapse
Affiliation(s)
- Oluseye O Bolaji
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Ayorinde Adehin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
- Institute of Biomedical & Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, PR China
| | - Babatunde A Adeagbo
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
| |
Collapse
|
5
|
Heong V, Syn NL, Lee XW, Sapari NS, Koh XQ, Adam Isa ZF, Sy Lim J, Lim D, Pang B, Thian YL, Ng LK, Wong AL, Soo RA, Yong WP, Chee CE, Lee SC, Goh BC, Soong R, Tan DSP. Value of a molecular screening program to support clinical trial enrollment in Asian cancer patients: The Integrated Molecular Analysis of Cancer (IMAC) Study. Int J Cancer 2017; 142:1890-1900. [PMID: 28994108 DOI: 10.1002/ijc.31091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/15/2017] [Accepted: 09/27/2017] [Indexed: 02/01/2023]
Abstract
The value of precision oncology initiatives in Asian contexts remains unresolved. Here, we review the institutional implementation of prospective molecular screening to facilitate accrual of patients into biomarker-driven clinical trials, and to explore the mutational landscape of advanced tumors occurring in a prospective cohort of Asian patients (n = 396) with diverse cancer types. Next-generation sequencing (NGS) and routine clinicopathological assays, such as immunohistochemistry, copy number analysis and in situ hybridization tests, were performed on tumor samples. Actionable biomarker results were used to identify eligibility for early-phase, biomarker-driven clinical trials. Overall, NGS was successful in 365 of 396 patients (92%), achieving a mean depth of 1,943× and coverage uniformity of 96%. The median turnaround time from sample receipt to return of genomic results was 26.0 days (IQR, 19.0-39.0 days). Reportable mutations were found in 300 of 365 patients (82%). Ninety-one percent of patients at study enrollment indicated consent to receive incidental findings and willingness to undergo genetic counseling if required. The most commonly mutated oncogenes included KRAS (19%), PIK3CA (16%), EGFR (5%), BRAF (3%) and KIT (3%); while the most frequently mutated tumor suppressor genes included TP53 (40%), SMARCB1 (12%), APC (8%), PTEN (6%) and SMAD4 (5%). Among 23 patients enrolled in genotype-matched trials, median progression-free survival was 2.9 months (IQR, 1.5-4.0 months). Nine of 20 evaluable patients (45%; 95% CI, 23.1-68.5%) derived clinical benefit, including 3 partial responses and 6 with stable disease lasting ≥ 8 weeks.
Collapse
Affiliation(s)
- Valerie Heong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Nicholas L Syn
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xiao Wen Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Nur Sabrina Sapari
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xue Qing Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Zul Fazreen Adam Isa
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Joey Sy Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Diana Lim
- Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Department of Pathology, National University Hospital, National University Health System, Singapore
| | - Brendan Pang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Department of Pathology, National University Hospital, National University Health System, Singapore
| | - Yee Liang Thian
- Department of Diagnostic Imaging, National University Hospital, National University Health System, Singapore
| | - Lai Kuan Ng
- Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Andrea L Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ross Andrew Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Cheng Ean Chee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Soo-Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Boon-Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - David S P Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, Singapore
| |
Collapse
|