1
|
Liu T, Yao Y, Liu B, Teng X, Zhang X, Dong M. Cost-effectiveness analysis of FOLFOXIRI plus bevacizumab versus mFOLFOX6 plus bevacizumab as first-line treatment of metastatic colorectal cancer. Expert Rev Pharmacoecon Outcomes Res 2025. [PMID: 40395124 DOI: 10.1080/14737167.2025.2509706] [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: 02/04/2025] [Revised: 04/28/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVES We first evaluated thecost-effectiveness of the FOLFOXIRI plus bevacizumab versus mFOLFOX6 plusbevacizumab as the first-line treatment of metastatic colorectal cancer fromhealthcare system perspective. METHODS A partitioned survivalmodel was developed to assess the costs and effects of FOLFOXIRI plusbevacizumab versus mFOLFOX6 plus bevacizumab. Health outcomes weremeasured in quality-adjusted life years (QALYs), and incrementalcost-effectiveness ratios (ICERs). The evaluation of the Chinese healthcarepayer perspective was performed across a lifetime horizon, encompassing directmedical expenses. RESULTS The estimated cost for FOLFOXIRI plus bevacizumab treatment was 9306.364USD, which was higher than 8218.436 USD estimated for mFOLFOX6 plusbevacizumab, leading to an ICER of 1961.857 USD per QALY. One-way sensitivityanalysis suggested the body surface area (BSA), the cost of irinotecan,and the cost of fluorouracilhad the largest impact on the ICER. The cost-effectiveness acceptability curvesshowed the probability of FOLFOXIRI plus bevacizumab being cost-effective was100% at a threshold of 12 300 USD per QALY. CONCLUSION FOLFOXIRI plus bevacizumab hadan economic advantage compared to mFOLFOX6 plus bevacizumab as the first-line treatment ofmetastatic CRC in China.
Collapse
Affiliation(s)
- Tong Liu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yao Yao
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bingjie Liu
- School of Health Management, Harbin Medical University, Harbin, China
| | - Xue Teng
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Zhang
- School of Health Management, Harbin Medical University, Harbin, China
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
2
|
Faisal MS, Hussain I, Ikram MA, Shah SB, Rehman A, Iqbal W. Irinotecan dosing and pharmacogenomics: a comprehensive exploration based on UGT1A1 variants and emerging insights. J Chemother 2025; 37:199-212. [PMID: 38706404 DOI: 10.1080/1120009x.2024.2349444] [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: 10/18/2023] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
Irinotecan is a critical anticancer drug used to treat metastatic colorectal cancer and advanced pancreatic ductal adenocarcinoma by obstructing topoisomerase 1; however, it can cause minor-to-severe and life-threatening adverse effects. UDP glucuronosyltransferase family 1 member A1 (UGT1A1) polymorphisms increase the risk of irinotecan-induced neutropenia and diarrhea. Hence, screening for UGT1A1 polymorphisms before irinotecan-based chemotherapy is recommended to minimize toxicity, whereas liposomes offer the potential to deliver irinotecan with fewer side effects in patients with pancreatic ductal adenocarcinoma. This review presents a comprehensive overview of the effects of genotype-guided dosing of irinotecan on UGT1A1*28 and UGT1A1*6 variants, incorporating pharmacogenomic research, optimal regimens for metastatic colorectal and pancreatic cancer treatment using irinotecan, guidelines for toxicity reduction, and an evaluation of the cost-effectiveness of UGT1A1 genotype testing.
Collapse
Affiliation(s)
- Muhammad Saleem Faisal
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Imran Hussain
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | | | - Syed Babar Shah
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Abdul Rehman
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Wajid Iqbal
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| |
Collapse
|
3
|
Yang X, Chen J, Wang Y, Wu Y, Zhang J. Managing Irinotecan-Induced Diarrhea: A Comprehensive Review of Therapeutic Interventions in Cancer Treatment. Pharmaceuticals (Basel) 2025; 18:359. [PMID: 40143136 PMCID: PMC11944746 DOI: 10.3390/ph18030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Irinotecan (CPT-11), an inhibitor of DNA topoisomerase I, stands as a pivotal therapeutic agent in oncology. However, its use is primarily constrained by side effects such as neutropenia and the onset of delayed diarrhea. Despite the effective management of neutropenia, CPT-11-induced diarrhea (CID) is often severe, leading to hospitalization, dosage adjustments, and in some cases, treatment discontinuation, which can significantly impact therapeutic outcomes. A multitude of pharmacological agents have been investigated in preclinical and clinical studies with the aim of reducing or preventing the onset of delayed diarrhea associated with CPT-11. This comprehensive review examines the underlying mechanisms of CPT-11-triggered delayed diarrhea and discusses the experimental medications and strategies that have been utilized to combat this adverse effect. This review encompasses an exploration of chemical formulations, the application of traditional Chinese medicine, and the advent of innovative drug delivery systems. It is anticipated that this article will serve as a valuable resource for both novice researchers in the realm of irinotecan chemotherapy and for those who are well-versed in the field, including experts and practicing clinicians.
Collapse
Affiliation(s)
- Xiaoqin Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.Y.); (J.C.)
| | - Jiamei Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.Y.); (J.C.)
| | - Yitao Wang
- State Key Laboratory of Quality Research in Traditional Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China;
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.Y.); (J.C.)
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.Y.); (J.C.)
| |
Collapse
|
4
|
Yuan M, Zheng Y, Wang F, Bai N, Zhang H, Bian Y, Liu H, He X. Discussion on the optimization of personalized medication using information systems based on pharmacogenomics: an example using colorectal cancer. Front Pharmacol 2025; 15:1516469. [PMID: 39877392 PMCID: PMC11772163 DOI: 10.3389/fphar.2024.1516469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025] Open
Abstract
Pharmacogenomics (PGx) is a powerful tool for clinical optimization of drug efficacy and safety. However, due to many factors affecting drugs in the real world, PGx still accounts for a small proportion of actual clinical application scenarios. Therefore, based on the information software, pharmacists use their professional advantages to integrate PGx into all aspects of pharmaceutical care, which is conducive to promoting the development of personalized medicine. In this paper, the establishment of an information software platform is summarized for the optimization of a personalized medication program based on PGx. Taking colorectal cancers (CRC) as an example, this paper also discusses the role of PGx in different working modes and participation in drug management of CRC patients by pharmacists with the help of information systems. Finally, we summarized the recommendations of different PGx guidelines to provide reference for the follow-up personalized pharmaceutical care.
Collapse
Affiliation(s)
- Mengying Yuan
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuankun Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fei Wang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Niuniu Bai
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Haoling Zhang
- Department of Pharmacy, Yuncheng Central Hospital, Yuncheng, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hao Liu
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xia He
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
5
|
Fourrier T, Truntzer C, Peroz M, Derangère V, Vincent J, Bengrine-Lefèvre L, Hennequin A, Palmier R, Orry D, Rabel T, Ghiringhelli F. Factors Influencing the Duration of Maintenance Therapy in Metastatic Colorectal Cancer. Cancers (Basel) 2024; 17:88. [PMID: 39796718 PMCID: PMC11720154 DOI: 10.3390/cancers17010088] [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: 10/17/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Metastatic colorectal cancer (mCRC) is mainly treated with 5-Fluoro-Uracil (5-FU), Oxaliplatin and Irinotecan chemotherapies and anti-Epidermal Growth Factor Receptor (EGFR) or anti-Vascular Endothelial Growth Factor (VEGF) targeted therapies. Due to chemotherapy-related toxicity, patients receive induction treatment to achieve tumour response followed by maintenance therapy with less cytotoxic molecules or a chemotherapy-free interval to reduce chemotherapy-related toxicity. In this study, the aim was to determine the patient, cancer and treatment factors that influence the duration of maintenance therapy (DMT). METHODS We collected retrospective data on a cohort of 133 patients treated at the Centre Georges François Leclerc (CGFL) cancer centre in Dijon between March 2014 and June 2022. Patients had unresectable or potentially resectable diseases. They received first-line induction treatment with chemotherapy and/or targeted therapy and maintenance treatment, defined as the interruption of at least one chemotherapy agent. RESULTS In the multivariate analysis, age (HR: 1.02, 95% CI 1.00-1.04, p = 0.031), N2 nodal status (HR: 1.78, 95% CI 1.09-2.89, p = 0.021) and the presence of peritoneal metastases (HR: 2.05, 95% CI 1.25-3.36, p = 0.004), as well as baseline carcino-embryonic antigen (CEA) level (HR: 1.10, 95% CI 1.00-1.20, p = 0.052), were significantly associated to poor DMT. Local treatment of liver metastases also significantly reduced the DMT (HR: 0.49, 95% CI 0.28-0.86, p = 0.013). In our cohort, induction triplet chemotherapy significantly increased the CEA delta (70% vs. 44%, p = 0.047) compared to doublet chemotherapy and led to a higher rate of liver surgery (40% vs. 21%, p = 0.014) and a trend for a higher rate of local treatment of metastases (62% vs. 45%, p = 0.059). CONCLUSIONS Duration of maintenance therapy is determined by the initial patient and colorectal cancer characteristics. However, it is significantly increased by local treatment of liver metastases. By reducing the tumour burden, a triplet induction chemotherapy regimen increases the rate of liver metastase resection.
Collapse
Affiliation(s)
- Théo Fourrier
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Caroline Truntzer
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| | - Morgane Peroz
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Valentin Derangère
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| | - Julie Vincent
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Leila Bengrine-Lefèvre
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Audrey Hennequin
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Rémi Palmier
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - David Orry
- Department of Surgical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Thomas Rabel
- Department of Surgical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - François Ghiringhelli
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| |
Collapse
|
6
|
Xu B, Wang S, Yan M, Sohn J, Li W, Tang J, Wang X, Wang Y, Im SA, Jiang D, Valdez T, Dasgupta A, Zhang Y, Yan Y, Komatsubara KM, Chung WP, Ma F, Dai MS. Sacituzumab govitecan in HR +HER2 - metastatic breast cancer: the randomized phase 3 EVER-132-002 trial. Nat Med 2024; 30:3709-3716. [PMID: 39354196 PMCID: PMC11645259 DOI: 10.1038/s41591-024-03269-z] [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: 04/05/2024] [Accepted: 08/23/2024] [Indexed: 10/03/2024]
Abstract
Sacituzumab govitecan (SG) significantly improved progression-free survival (PFS) and overall survival (OS) versus chemotherapy in hormone receptor-positive human epidermal growth factor receptor 2-negative (HR+HER2-) metastatic breast cancer (mBC) in the global TROPiCS-02 study. TROPiCS-02 enrolled few Asian patients. Here we report results of SG in Asian patients with HR+HER2- mBC from the EVER-132-002 study. Patients were randomized to SG (n = 166) or chemotherapy (n = 165). The primary endpoint was met: PFS was improved with SG versus chemotherapy (hazard ratio of 0.67, 95% confidence interval 0.52-0.87; P = 0.0028; median 4.3 versus 4.2 months). OS also improved with SG versus chemotherapy (hazard ratio of 0.64, 95% confidence interval 0.47-0.88; P = 0.0061; median 21.0 versus 15.3 months). The most common grade ≥3 treatment-emergent adverse events were neutropenia, leukopenia and anemia. SG demonstrated significant and clinically meaningful improvement in PFS and OS versus chemotherapy, with a manageable safety profile consistent with prior studies. SG represents a promising treatment option for Asian patients with HR+HER2- mBC (ClinicalTrials.gov identifier no. NCT04639986 ).
Collapse
Affiliation(s)
- Binghe Xu
- Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Shusen Wang
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Min Yan
- Henan Cancer Hospital/Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Wei Li
- The First Hospital of Jilin University, Changchun, China
| | - Jinhai Tang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Ying Wang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | - Yilin Yan
- Gilead Sciences Inc., Foster City, CA, USA
| | | | - Wei-Pang Chung
- National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Fei Ma
- Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | |
Collapse
|
7
|
Chen Y, Zhu D, Yu Y, Chang W, Ye L, Feng Q, Xu P, Chen M, Ji M, Wei Y, Liu T, Xu J. VIC Regimen (Vemurafenib/Irinotecan/Cetuximab) Versus Bevacizumab Plus Chemotherapy as First-Line Treatment for BRAF V600E-Mutated Unresectable or Metastatic Colorectal Cancer in Asian Patients: A Prospective Cohort Study. Clin Colorectal Cancer 2024; 23:354-363.e4. [PMID: 38845274 DOI: 10.1016/j.clcc.2024.05.006] [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: 04/12/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Colorectal cancers (CRC) with BRAF V600E mutation exhibit limited chemotherapy response and a poor prognosis. Safety and efficacy of the VIC (Vemurafenib/Irinotecan/Cetuximab) regimen in the first-line setting for patients with BRAF V600E-mutated CRC remain undetermined. METHODS In the prospective cohort study, the untreated, BRAF V600E-mutated, unresectable or metastatic CRC patients were enrolled. The VIC regimen and bevacizumab plus chemotherapy were compared in the first-line setting. The objective response rate (ORR), disease control rate (DCR), conversion resection rate, progression-free survival (PFS), and overall survival (OS) were evaluated. RESULTS In the intent-to-treat analysis, 38 patients received VIC regimen and 40 received bevacizumab plus chemotherapy. The ORR and DCR in the VIC group were significantly higher than in the bevacizumab-therapy group (ORR: 63.2% vs. 37.5%, P = .025; DCR: 94.7% vs. 75.0%, P = .019). The VIC regimen significantly outperformed bevacizumab plus chemotherapy in both PFS (11.9 vs. 7.7 months; hazard ratio [HR] = 0.51, 95% CI, 0.30-0.87; P = .010) and OS (25.3 vs. 14.6 months; HR = 0.43, 95% CI, 0.22-0.82; P = .011). In the VIC group, the conversion resection rate for liver metastases was 34.8% (8 of 23 patients), and for unresectable local CRC it was 54.5% (6 of 11 patients). The adverse events rates of Grade 3 to 4 were 34.2% and 32.5% for the VIC regimen and bevacizumab plus chemotherapy respectively. CONCLUSIONS Among Asian patients with BRAF V600E-mutated CRC, the VIC regimen showed favorable outcomes compared to bevacizumab plus chemotherapy in terms of tumor response and oncological survival, with a tolerable and manageable toxicity profile in the first-line setting.
Collapse
Affiliation(s)
- Yijiao Chen
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dexiang Zhu
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiyi Yu
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Wenju Chang
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lechi Ye
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qingyang Feng
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pingping Xu
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive
| | - Miao Chen
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meiling Ji
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive
| | - Ye Wei
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianmin Xu
- Department of Colorectal Surgery, Colorectal Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China; Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Li X, Sabbatini D, Pegoraro E, Bello L, Clemens P, Guglieri M, van den Anker J, Damsker J, McCall J, Dang UJ, Hoffman EP, Jusko WJ. Assessing Pharmacogenomic loci Associated with the Pharmacokinetics of Vamorolone in Boys with Duchenne Muscular Dystrophy. J Clin Pharmacol 2024; 64:1130-1140. [PMID: 38682893 PMCID: PMC11357888 DOI: 10.1002/jcph.2446] [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: 11/28/2023] [Accepted: 03/27/2024] [Indexed: 05/01/2024]
Abstract
Human genetic variation (polymorphisms) in genes coding proteins involved in the absorption, distribution, metabolism, and elimination (ADME) of drugs can have a strong effect on drug exposure and downstream efficacy and safety outcomes. Vamorolone, a dissociative steroidal anti-inflammatory drug for treating Duchenne muscular dystrophy (DMD), primarily undergoes oxidation by CYP3A4 and CYP3A5 and glucuronidation by UDP-glucuronosyltransferases. This work assesses the pharmacokinetics (PKs) of vamorolone and sources of interindividual variability (IIV) in 81 steroid-naïve boys with DMD aged 4 to <7 years old considering the genetic polymorphisms of CYPS3A4 (CYP3A4*22, CYP3A4*1B), CYP3A5 (CYP3A5*3), and UGT1A1 (UGT1A1*60) utilizing population PK modeling. A one-compartment model with zero-order absorption (Tk0, duration of absorption), linear clearance (CL/F), and volume (V/F) describes the plasma PK data for boys with DMD receiving a wide range of vamorolone doses (0.25-6 mg/kg/day). The typical CL/F and V/F values of vamorolone were 35.8 L/h and 119 L, with modest IIV. The population Tk0 was 3.14 h yielding an average zero-order absorption rate (k0) of 1.16 mg/kg/h with similar absorption kinetics across subjects at the same vamorolone dose (i.e., no IIV on Tk0). The covariate analysis showed that none of the genetic covariates had any significant impact on the PKs of vamorolone in boys with DMD. Thus, the PKs of vamorolone is very consistent in these young boys with DMD.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | | | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Paula Clemens
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michela Guglieri
- John Walton Centre for Neuromuscular Disease, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | - John van den Anker
- Division of Clinical Pharmacology, Children’s National Hospital, Washington, DC, USA
- ReveraGen BioPharma, Rockville, MD, USA
| | | | | | - Utkarsh J. Dang
- Department of Health Sciences, Carleton University, Ottawa, Canada
| | - Eric P. Hoffman
- ReveraGen BioPharma, Rockville, MD, USA
- Department of Pharmaceutical Sciences, Binghamton University, State University of New York, Binghamton, NY, USA
| | - William J. Jusko
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
9
|
Nguyen CTT, Nguyen TMT, Phung TH. Meta-analysis revisiting the influence of UGT1A1*28 and UGT1A1*6 on irinotecan safety in colorectal cancer patients. Pharmacogenomics 2024; 25:469-477. [PMID: 39171626 PMCID: PMC11492647 DOI: 10.1080/14622416.2024.2385289] [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: 05/22/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Aim: To evaluate the association between irinotecan safety and the UGT1A1 gene polymorphism in colorectal cancer (CRC) patients.Materials & methods: The studies were systematically searched and identified from three databases (PubMed, Embase and The Cochrane Library) until 28 February 2023. The relationships were evaluated using pooled odds ratio (OR).Results: A total of 30 studies out of 600 were included, comprising 4471 patients. UGT1A1*28 was associated with a statistically significant increase in the OR for diarrhea (OR: 1.59, 95% CI = 1.24-2.06 in the additive model; OR = 3.24, 95% CI = 2.01-5.21 in the recessive model; and OR = 1.95, 95% CI = 1.42-2.69 in the dominant model) and neutropenia (OR = 1.70, 95% CI = 1.40-2.06 in the additive model; OR = 4.10, 95%CI = 2.69-6.23 in the recessive model; and OR = 1.93, 95% CI = 1.61-2.31 in the dominant model). Subgroup analysis indicated consistent associations in both Asian and non-Asian populations. UGT1A1*6 was associated with a statistically significant elevation in the OR for diarrhea (only in the recessive model, OR = 2.42; 95% CI = 1.14-5.11) and neutropenia (across all genetic models).Conclusion: The UGT1A1*28 and UGT1A1*6 alleles might be a crucial indicator for predicting irinotecan safety in CRC.
Collapse
Affiliation(s)
- Cuc Thi Thu Nguyen
- Faculty of Pharmaceutical Management & Economics, Hanoi University of Pharmacy, Hanoi, 10000, Vietnam
| | | | - Thanh Huong Phung
- Faculty of Biotechnology, Hanoi University of Pharmacy, Hanoi, 10000,Vietnam
| |
Collapse
|
10
|
Takakura T, Shimizu T, Yamamoto N. Antibody-drug conjugates in solid tumors; new strategy for cancer therapy. Jpn J Clin Oncol 2024; 54:837-846. [PMID: 38704241 PMCID: PMC11322887 DOI: 10.1093/jjco/hyae054] [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: 12/30/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have emerged as a novel class of anticancer treatment. ADCs are composed of three parts: a monoclonal antibody, a linker and a payload. A monoclonal antibody binds to the specific antigen present at the cancer cells, allowing selective delivery of the cytotoxic agents to the tumor site. Several ADCs are approved by the US Food and Drug Administration for the treatment of hematologic cancers and solid tumors with clinically meaningful survival benefit. However, the development of ADCs faces a lot of challenges and there is a need to get better understanding of ADCs in order to improve patient outcomes. Here, we briefly discuss the structure and mechanism of ADCs, as well as the clinical data of current approved ADCs in solid tumors.
Collapse
Affiliation(s)
- Toshiaki Takakura
- Department of Pulmonary Medicine and Medical Oncology, Wakayama Medical University Faculty of Medicine, 811-1 Kimiidera, Wakayama, Wakayama 641-8510, Japan
| | - Toshio Shimizu
- Department of Pulmonary Medicine and Medical Oncology, Wakayama Medical University Faculty of Medicine, 811-1 Kimiidera, Wakayama, Wakayama 641-8510, Japan
| | - Nobuyuki Yamamoto
- Department of Pulmonary Medicine and Medical Oncology, Wakayama Medical University Faculty of Medicine, 811-1 Kimiidera, Wakayama, Wakayama 641-8510, Japan
| |
Collapse
|
11
|
Haberkorn BCM, Hoogendijk L, Loosveld OS, Thijs AMJ, Verstijnen J. Real Life Data and Outcome of FOLFIRINOX Use in Metastatic Pancreatic Cancer Patients in General Hospitals in the Netherlands. J Gastrointest Cancer 2024; 55:838-844. [PMID: 38319559 DOI: 10.1007/s12029-023-01006-2] [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] [Accepted: 12/21/2023] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Pancreatic cancer is one of the five most common causes of cancer mortality in developed countries. In patients with metastatic disease, the most frequent treatment used is FOLFIRINOX, which is associated with moderate toxicity which could influence quality of life. The efficacy of FOLFIRINOX in a general population in the Netherlands has not been subject of research before, and therefore, this research has been set up in order to investigate what the real-life benefits of FOLFIRINOX are in a population with metastatic pancreatic cancer (mPC) treated in three general hospitals in the Netherlands. METHODS The data used in this study was collected by patient records leading to a noninterventional retrospective cohort study. Eighty-six patients, over 18 years of age, diagnosed with mPC between the years 2015 and 2022 and treated with FOLFIRINOX at Maasstad Hospital in Rotterdam, Amphia Hospital in Breda, or Catharina Hospital in Eindhoven, were included in the study. Kaplan-Meier models were used in order to represent survival outcomes. RESULTS The results showed a median overall survival of 228 days (IQR 118-355). Only 14.0% (n = 12) completed the first-line treatment, and 51.2% (n = 44) of patients stopped treatment before or during cycle 6. Toxicity is highest, grade 3, after the first cycle but remains high for grade 1 and 2 during all treatment cycles. CONCLUSION Survival rates for patient with metastatic pancreatic cancer treated with FOLFIRINOX were worse in our study population than in comparative studies.
Collapse
Affiliation(s)
- B C M Haberkorn
- Department of Medical Oncology, Maasstad Ziekenhuis Rotterdam, Maasstadweg 21, 3079 DZ, Rotterdam, the Netherlands.
| | | | - O S Loosveld
- Department of Medical Oncology, Amphia Ziekenhuis Breda, Breda, Netherlands
| | - A M J Thijs
- Department of Medical Oncology, Catharina Ziekenhuis Eindhoven, Eindhoven, Netherlands
| | - J Verstijnen
- Department of Medical Oncology, Maasstad Ziekenhuis Rotterdam, Maasstadweg 21, 3079 DZ, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Ascione L, Guidi L, Prakash A, Trapani D, LoRusso P, Lou E, Curigliano G. Unlocking the Potential: Biomarkers of Response to Antibody-Drug Conjugates. Am Soc Clin Oncol Educ Book 2024; 44:e431766. [PMID: 38828973 DOI: 10.1200/edbk_431766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Antibody-drug conjugates (ADCs) have reshaped the cancer treatment landscape across a variety of different tumor types. ADCs' peculiar pharmacologic design combines the cytotoxic properties of chemotherapeutic agents with the selectivity of targeted therapies. At present, the approval of many ADCs used in clinical practice has not always been biomarker-driven. Indeed, predicting ADCs' activity and toxicity through the demonstration of specific biomarkers is still a great unmet need, and the identification of patients who can derive significant benefit from treatment with ADCs may often be uncertain. With the lack of robust predictive biomarkers to anticipate primary, intrinsic resistance to ADCs and no consolidated biomarkers to aid in the early identification of treatment resistance (ie, acquired resistance), the determination of precise biologic mechanisms of ADC activity and safety becomes priority in the quest for better patient-centric outcomes. Of great relevance, whether the target antigen expression is a determinant of ADCs' primary activity is still to be clarified, and available data remain quite controversial. Antigen expression assessment is typically performed on tissue biopsy, hence only providing information on a specific tumor site, therefore unable to capture heterogeneous patterns of tumor antigen expression. Quantifying the expression of the target antigen across all tumor sites would help better understand tumor heterogeneity, whereas molecularly characterizing tumor-intrinsic features over time might provide information on resistance mechanisms. In addition, toxicity can represent a critical concern, since most ADCs have a safety profile that resembles that of chemotherapies, with often unique adverse events requiring special management, possibly because of the differential in pharmacokinetics between the small-molecule agent versus payload of a similar class (eg, deruxtecan conjugate-related interstitial lung disease). As such, the identification of robust predictive biomarkers of safety and activity of ADCs has the potential to improve patient selection and enrich the population of patients most likely to derive a substantial clinical benefit, especially in those disease settings where different ADCs happen to be approved in competing clinical indications, with undefined biomarkers to make precise decision making and unclear data on how to sequence ADCs. At this point, the identification of clinically actionable biomarkers in the space of ADCs remains a top research priority.
Collapse
Affiliation(s)
- Liliana Ascione
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| | - Lorenzo Guidi
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| | - Ajay Prakash
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Dario Trapani
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| | - Patricia LoRusso
- Yale University School of Medicine, Yale Cancer Center, New Haven, CT
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Giuseppe Curigliano
- Division of Early Drug Development, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hematology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
13
|
Ginzac A, Thivat E, Petorin C, Richard D, Herviou P, Molnar I, Devaud H, Creveaux I, Ferrer F, Authier N, Jary M, Pezet D, Durando X. A phase-II study based on dose adjustment according to UGT1A1 polymorphism: is irinotecan underdosed in first-line FOLFIRI regimen for mCRC? Cancer Chemother Pharmacol 2024; 93:225-236. [PMID: 37932443 PMCID: PMC10901933 DOI: 10.1007/s00280-023-04603-x] [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: 07/06/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Irinotecan has considerable importance in the treatment of metastatic colorectal cancer (mCRC). UDP-glucoronyltransferase (UGT) 1A1 is responsible for the inactivation of SN-38, a metabolite of irinotecan. Depending on UGT1A1 polymorphism, the activity of the UGT enzyme can be reduced leading to more frequent occurrence of adverse events related to irinotecan. The present study aimed to assess the safety and efficacy of different doses of irinotecan adjusted according to UGT1A1 polymorphism. METHODS Thirty-four patients treated with FOLFIRI as first-line treatment for mCRC were included in this study. The irinotecan dosage was adapted on the basis of UGT1A1 polymorphisms: *1/*1 (370 mg/m2); *1/*28 (310 mg/m2), and *28/*28 (180 mg/m2). The incidence of grades 3 and 4 toxicities (neutropenia, febrile neutropenia, and diarrhoea) was recorded. Response was assessed according to the RECIST 1.1 criteria. RESULTS On the basis of UGT1A1 genotyping, 20 patients were *1/*1 (58.8%), 12 were *1/*28 (35.3%) and 2 were *28/*28 (5.9%). Seven patients experienced at least one severe toxicity, i.e., 21% of the population, amounting to eleven adverse events. Concerning the response rate, 15 patients (44%) had partial or complete response. CONCLUSION This study demonstrates that mCRC patients treated with FOLFIRI can tolerate a higher dose of irinotecan than the standard dose, i.e., > 180 mg/m2, on the basis of their UGT1A1 genotype, without increased toxicities. TRIAL REGISTRATION NCT01963182 (registered on 16/10/2013, Clermont-Ferrand, France).
Collapse
Affiliation(s)
- Angeline Ginzac
- INSERM U1240 Imagerie Moléculaire et Stratégies Théranostiques (IMoST), Université Clermont Auvergne, 63000, Clermont-Ferrand, France
- Centre d'Investigation Clinique, UMR501, 63000, Clermont-Ferrand, France
- Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean PERRIN, 63000, Clermont-Ferrand, France
| | - Emilie Thivat
- INSERM U1240 Imagerie Moléculaire et Stratégies Théranostiques (IMoST), Université Clermont Auvergne, 63000, Clermont-Ferrand, France.
- Centre d'Investigation Clinique, UMR501, 63000, Clermont-Ferrand, France.
- Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean PERRIN, 63000, Clermont-Ferrand, France.
| | - Caroline Petorin
- Département de Chirurgie Digestive et Hépatobiliaire, Hôpital Estaing, 63000, Clermont-Ferrand, France
| | - Damien Richard
- Service de Pharmacologie Médicale, Unité de Pharmacologie et de Toxicologie Biologique, CHU Gabriel MONTPIED, 63000, Clermont-Ferrand, France
| | - Pauline Herviou
- Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean PERRIN, 63000, Clermont-Ferrand, France
| | - Ioana Molnar
- INSERM U1240 Imagerie Moléculaire et Stratégies Théranostiques (IMoST), Université Clermont Auvergne, 63000, Clermont-Ferrand, France
- Centre d'Investigation Clinique, UMR501, 63000, Clermont-Ferrand, France
- Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean PERRIN, 63000, Clermont-Ferrand, France
| | - Hervé Devaud
- Service d'oncologie Médicale, Centre Jean PERRIN, 63000, Clermont-Ferrand, France
| | - Isabelle Creveaux
- Département de Biochimie et Génétique Moléculaire, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Florent Ferrer
- Service de Pharmacologie Médicale, Unité de Pharmacologie et de Toxicologie Biologique, CHU Gabriel MONTPIED, 63000, Clermont-Ferrand, France
| | - Nicolas Authier
- Université Clermont Auvergne, CHU de Clermont-Ferrand, Inserm, Pharmacologie Médicale/Centre Evaluation et Traitement de La Douleur, Observatoire Français des Médicaments Antalgiques, Institut Analgesia, 63001, Clermont-Ferrand, France
| | - Marine Jary
- Service de Chirurgie Digestive, U1071, M2iSH, USC-INRA 2018, CHU Clermont-Ferrand, Université Clermont Auvergne, INSERM, INRA, F-63000, Clermont-Ferrand, France
| | - Denis Pezet
- Service de Chirurgie Digestive, U1071, M2iSH, USC-INRA 2018, CHU Clermont-Ferrand, Université Clermont Auvergne, INSERM, INRA, F-63000, Clermont-Ferrand, France
| | - Xavier Durando
- INSERM U1240 Imagerie Moléculaire et Stratégies Théranostiques (IMoST), Université Clermont Auvergne, 63000, Clermont-Ferrand, France
- Centre d'Investigation Clinique, UMR501, 63000, Clermont-Ferrand, France
- Département de Recherche Clinique, Délégation Recherche Clinique et Innovation, Centre Jean PERRIN, 63000, Clermont-Ferrand, France
- Service d'oncologie Médicale, Centre Jean PERRIN, 63000, Clermont-Ferrand, France
| |
Collapse
|
14
|
Van Quang H, Vuong NB, Trang BNL, Toan NL, Van Tong H. Association of UGT1A1 gene variants, expression levels, and enzyme concentrations with 2,3,7,8-TCDD exposure in individuals exposed to Agent Orange/Dioxin. Sci Rep 2024; 14:3315. [PMID: 38332122 PMCID: PMC10853243 DOI: 10.1038/s41598-024-54004-0] [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: 04/06/2023] [Accepted: 02/07/2024] [Indexed: 02/10/2024] Open
Abstract
Among the congener of dioxin, 2,3,7,8-TCDD is the most toxic, having a serious long-term impact on the environment and human health. UDP-glucuronosyltransferase 1A1 (UGT1A1) plays a crucial role in the detoxification and excretion of endogenous and exogenous lipophilic compounds, primarily in the liver and gastrointestinal tract. This study aimed to investigate the association of UGT1A1 gene polymorphisms, expression levels, and enzyme concentration with Agent Orange/Dioxin exposure. The study included 100 individuals exposed to Agent Orange/Dioxin nearby Da Nang and Bien Hoa airports in Vietnam and 100 healthy controls. UGT1A1 SNP rs10929303, rs1042640 and rs8330 were determined by Sanger sequencing, mRNA expression was quantified by RT-qPCR and plasma UGT1A1 concentrations were measured by ELISA. The results showed that UGT1A1 polymorphisms at SNPs rs10929303, rs1042640 and rs8330 were associated with Agent Orange/Dioxin exposure (OR = 0.55, P = 0.018; OR = 0.55, P = 0.018 and OR = 0.57, P = 0.026, respectively). UGT1A1 mRNA expression levels and enzyme concentration were significantly elevated in individuals exposed to Agent Orange/Dioxin compared to controls (P < 0.0001). Benchmark dose (BMD) analyses showed that chronic exposure to 2,3,7,8-TCDD contamination affects the UGT1A1 mRNA and protein levels. Furthermore, UGT1A1 polymorphisms affected gene expression and enzyme concentrations in individuals exposed to Agent Orange/Dioxin. In conclusion, UGT1A1 gene polymorphisms, UGT1A gene expression levels and UGT1A1 enzyme concentrations were associated with Agent Orange/Dioxin exposure. The metabolism of 2,3,7,8-TCDD may influence UGT1A gene expression and enzyme concentrations.
Collapse
Affiliation(s)
- Ha Van Quang
- Department of Haematology, Toxicology, Radiation, and Occupation, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
- The Center of Toxicological and Radiological Training and Research, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nguyen Ba Vuong
- Department of Haematology, Toxicology, Radiation, and Occupation, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| | - Bui Ngoc Linh Trang
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, 222 Phung Hung, Ha Dong, Hanoi, Vietnam
| | - Nguyen Linh Toan
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Hoang Van Tong
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, 222 Phung Hung, Ha Dong, Hanoi, Vietnam.
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam.
| |
Collapse
|
15
|
Glewis S, Lingaratnam S, Krishnasamy M, H Martin J, Tie J, Alexander M, Michael M. Pharmacogenetics testing (DPYD and UGT1A1) for fluoropyrimidine and irinotecan in routine clinical care: Perspectives of medical oncologists and oncology pharmacists. J Oncol Pharm Pract 2024; 30:30-37. [PMID: 37021580 DOI: 10.1177/10781552231167554] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
BACKGROUND Despite robust evidence and international guidelines, to support routine pharmacogenetic (PGx) testing, integration in practice has been limited. This study explored clinicians' views and experiences of pre-treatment DPYD and UGT1A1 gene testing and barriers to and enablers of routine clinical implementation. METHODS A study-specific 17-question survey was emailed (01 February-12 April 2022) to clinicians from the Medical Oncology Group of Australia (MOGA), the Clinical Oncology Society of Australia (COSA) and International Society of Oncology Pharmacy Practitioners (ISOPP). Data were analysed and reported using descriptive statistics. RESULTS Responses were collected from 156 clinicians (78% medical oncologists, 22% pharmacists). Median response rate of 8% (ranged from 6% to 24%) across all organisations. Only 21% routinely test for DPYD and 1% for UGT1A1. For patients undergoing curative/palliative intent treatments, clinicians reported intent to implement genotype-guided dosing by reducing FP dose for DPYD intermediate metabolisers (79%/94%), avoiding FP for DPYD poor metabolisers (68%/90%), and reducing irinotecan dose for UGT1A1 poor metabolisers (84%, palliative setting only). Barriers to implementation included: lack of financial reimbursements (82%) and perceived lengthy test turnaround time (76%). Most Clinicians identified a dedicated program coordinator, i.e., PGx pharmacist (74%) and availability of resources for education/training (74%) as enablers to implementation. CONCLUSION PGx testing is not routinely practised despite robust evidence for its impact on clinical decision making in curative and palliative settings. Research data, education and implementation studies may overcome clinicians' hesitancy to follow guidelines, especially for curative intent treatments, and may overcome other identified barriers to routine clinical implementation.
Collapse
Affiliation(s)
- Sarah Glewis
- Department of Pharmacy, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | | | - Mei Krishnasamy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Academic Nursing Unit, Peter MacCallum Cancer Centre, Melbourne, Australia
- VCCC Alliance, Parkville, Australia
| | - Jennifer H Martin
- School of Medicine and Public Health, University of Newcastle, New South Wales, Australia
| | - Jeanne Tie
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Marliese Alexander
- Department of Pharmacy, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Michael Michael
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
16
|
Hulshof EC, Deenen MJ, Nijenhuis M, Soree B, de Boer-Veger NJ, Buunk AM, Houwink EJF, Risselada A, Rongen GAPJM, van Schaik RHN, Touw DJ, van der Weide J, van Westrhenen R, Deneer VHM, Guchelaar HJ, Swen JJ. Dutch pharmacogenetics working group (DPWG) guideline for the gene-drug interaction between UGT1A1 and irinotecan. Eur J Hum Genet 2023; 31:982-987. [PMID: 36443464 PMCID: PMC10474017 DOI: 10.1038/s41431-022-01243-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/05/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Abstract
The Dutch Pharmacogenetics Working Group (DPWG) aims to facilitate PGx implementation by developing evidence-based pharmacogenetics guidelines to optimize pharmacotherapy. This guideline describes the starting dose optimization of the anti-cancer drug irinotecan to decrease the risk of severe toxicity, such as (febrile) neutropenia or diarrhoea. Uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1 encoded by the UGT1A1 gene) enzyme deficiency increases risk of irinotecan-induced toxicity. Gene variants leading to UGT1A1 enzyme deficiency (e.g. UGT1A1*6, *28 and *37) can be used to optimize an individual's starting dose thereby preventing carriers from toxicity. Homozygous or compound heterozygous carriers of these allele variants are defined as UGT1A1 poor metabolisers (PM). DPWG recommends a 70% starting dose in PM patients and no dose reduction in IM patients who start treatment with irinotecan. Based on the DPWG clinical implication score, UGT1A1 genotyping is considered "essential", indicating that UGT1A1 testing must be performed prior to initiating irinotecan treatment.
Collapse
Affiliation(s)
- Emma C Hulshof
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marga Nijenhuis
- Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands.
| | - Bianca Soree
- Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands
| | | | | | - Elisa J F Houwink
- Department of Public Health and Primary Care (PHEG), Leiden University Medical Centre, Leiden, The Netherlands
- National eHealth Living Lab (NELL), Leiden, The Netherlands
| | - Arne Risselada
- Department of Clinical Pharmacy, Wilhelmina Hospital, Assen, The Netherlands
| | - Gerard A P J M Rongen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy & Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of PharmacoTherapy, -Epidemiology & -Economics, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Jan van der Weide
- Department of Clinical Chemistry, St. Jansdal Hospital, Harderwijk, The Netherlands
| | - Roos van Westrhenen
- Department of Psychiatry, Parnassia Group, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, London, UK
| | - Vera H M Deneer
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
17
|
Tarantino P, Ricciuti B, Pradhan SM, Tolaney SM. Optimizing the safety of antibody-drug conjugates for patients with solid tumours. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00783-w. [PMID: 37296177 DOI: 10.1038/s41571-023-00783-w] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
Over the past 5 years, improvements in the design of antibody-drug conjugates (ADCs) have enabled major advances that have reshaped the treatment of several advanced-stage solid tumours. Considering the intended rationale behind the design of ADCs, which is to achieve targeted delivery of cytotoxic molecules by linking them to antibodies targeting tumour-specific antigens, ADCs would be expected to be less toxic than conventional chemotherapy. However, most ADCs are still burdened by off-target toxicities that resemble those of the cytotoxic payload as well as on-target toxicities and other poorly understood and potentially life-threatening adverse effects. Given the rapid expansion in the clinical indications of ADCs, including use in curative settings and various combinations, extensive efforts are ongoing to improve their safety. Approaches currently being pursued include clinical trials optimizing the dose and treatment schedule, modifications of each ADC component, identification of predictive biomarkers for toxicities, and the development of innovative diagnostic tools. In this Review, we describe the determinants of the toxicities of ADCs in patients with solid tumours, highlighting key strategies that are expected to improve tolerability and enable improvements in the treatment outcomes of patients with advanced-stage and those with early stage cancers in the years to come.
Collapse
Affiliation(s)
- Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Biagio Ricciuti
- Harvard Medical School, Boston, MA, USA
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shan M Pradhan
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Biswas M, Jinda P, Sukasem C. Pharmacogenomics in Asians: Differences and similarities with other human populations. Expert Opin Drug Metab Toxicol 2023; 19:27-41. [PMID: 36755439 DOI: 10.1080/17425255.2023.2178895] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/07/2023] [Indexed: 02/10/2023]
Abstract
INTRODUCTION Various pharmacogenomic (PGx) variants differ widely in different ethnicities. and clinical outcomes associated with these variants may also be substantially varied. Literature was searched in different databases, i.e. PubMed, ScienceDirect, Web of Science, and PharmGKB, from inception to 30 June 2022 for this review. AREAS COVERED Certain PGx variants were distinctly varied in Asian populations compared to the other human populations, e.g. CYP2C19*2,*3,*17; CYP2C9*2,*3; CYP2D6*4,*5,*10,*41; UGT1A1*6,*28; HLA-B*15:02, HLA-B*15:21, HLA-B*58:01, and HLA-A*31:01. However, certain other variants do not vary greatly between Asian and other ethnicities, e.g. CYP3A5*3; ABCB1, and SLCO1B1*5. As evident in this review, the risk of major adverse cardiovascular events (MACE) was much stronger in Asian patients taking clopidogrel and who inherited the CYP2C19 loss-of-function alleles, e.g. CYP2C19*2 and*3, when compared to the western/Caucasian patients. Additionally, the risk of carbamazepine-induced severe cutaneous adverse drug reactions (SCARs) for the patients inheriting HLA-B*15:02 and HLA-B*15:21 alleles varied significantly between Asian and other ethnicities. In contrast, both Caucasian and Asian patients inheriting the SLCO1B1*5 variant possessed a similar magnitude of muscle toxicity, i.e. myopathy. EXPERT OPINION Asian countries should take measures toward expanding PGx research, as well as initiatives for the purposes of obtaining clinical benefits from this newly evolving and economically viable treatment model.
Collapse
Affiliation(s)
- Mohitosh Biswas
- Department of Pharmacy, University of Rajshahi, 6205, Rajshahi, Bangladesh
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 10400, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Ramathibodi Hospital, Somdech Phra Debaratana Medical Center SDMC, 10400, Bangkok, Thailand
| | - Pimonpan Jinda
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 10400, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Ramathibodi Hospital, Somdech Phra Debaratana Medical Center SDMC, 10400, Bangkok, Thailand
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 10400, Bangkok, Thailand
- Laboratory for Pharmacogenomics, Ramathibodi Hospital, Somdech Phra Debaratana Medical Center SDMC, 10400, Bangkok, Thailand
- Pharmacogenomics and Precision Medicine Clinic, Bumrungrad Genomic Medicine Institute (BGMI), Bumrungrad International Hospital, 10110, Bangkok, Thailand
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L69 3GL, Liverpool, UK
| |
Collapse
|
19
|
The Somatic Mutation Landscape of UDP-Glycosyltransferase ( UGT) Genes in Human Cancers. Cancers (Basel) 2022; 14:cancers14225708. [PMID: 36428799 PMCID: PMC9688768 DOI: 10.3390/cancers14225708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The human UDP-glycosyltransferase (UGTs) superfamily has a critical role in the metabolism of anticancer drugs and numerous pro/anti-cancer molecules (e.g., steroids, lipids, fatty acids, bile acids and carcinogens). Recent studies have shown wide and abundant expression of UGT genes in human cancers. However, the extent to which UGT genes acquire somatic mutations within tumors remains to be systematically investigated. In the present study, our comprehensive analysis of the somatic mutation profiles of 10,069 tumors from 33 different TCGA cancer types identified 3427 somatic mutations in UGT genes. Overall, nearly 18% (1802/10,069) of the assessed tumors had mutations in UGT genes with huge variations in mutation frequency across different cancer types, ranging from over 25% in five cancers (COAD, LUAD, LUSC, SKCM and UCSC) to less than 5% in eight cancers (LAML, MESO, PCPG, PAAD, PRAD, TGCT, THYM and UVM). All 22 UGT genes showed somatic mutations in tumors, with UGT2B4, UGT3A1 and UGT3A2 showing the largest number of mutations (289, 307 and 255 mutations, respectively). Nearly 65% (2260/3427) of the mutations were missense, frame-shift and nonsense mutations that have been predicted to code for variant UGT proteins. Furthermore, about 10% (362/3427) of the mutations occurred in non-coding regions (5' UTR, 3' UTR and splice sites) that may be able to alter the efficiency of translation initiation, miRNA regulation or the splicing of UGT transcripts. In conclusion, our data show widespread somatic mutations of UGT genes in human cancers that may affect the capacity of cancer cells to metabolize anticancer drugs and endobiotics that control pro/anti-cancer signaling pathways. This highlights their potential utility as biomarkers for predicting therapeutic efficacy and clinical outcomes.
Collapse
|
20
|
Irisawa A, Takeno M, Watanabe K, Takahashi H, Mitsunaga S, Ikeda M. Incidence of and risk factors for severe neutropenia during treatment with the modified FOLFIRINOX therapy in patients with advanced pancreatic cancer. Sci Rep 2022; 12:15574. [PMID: 36114233 PMCID: PMC9481868 DOI: 10.1038/s41598-022-18669-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/17/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractAlthough FOLFIRINOX (l-Leucovorin/5-FU/Irinotecan/Oxaliplatin) is established as one of the standard therapies for patients with metastatic pancreatic cancer, the modified FOLFIRINOX (mFOLFIRINOX) is often used in clinical practice to reduce the incidence of toxicities. Febrile neutropenia (FN) and severe neutropenia during FOLFIRINOX are especially frequently observed in Japanese patients. In this study, we evaluated the incidence of FN and severe neutropenia, and explored the risk factors for severe neutropenia in patients receiving treatment with mFOLFIRINOX. The data of patients who had received mFOLFIRINOX between December 2013 and December 2014 at the National Cancer Center Hospital East were reviewed retrospectively. We graded the neutropenia severity and defined ≥ Grade 3 neutropenia as severe neutropenia. Univariate and multivariate analysis were undertaken to evaluate the associations with risk of development of severe neutropenia. A total of 122 patients were enrolled in this study. Sixty two patients (51%) and 10 patients (8%) developed severe neutropenia and FN, respectively. Multivariate analysis identified a low baseline white blood cell count (odds ratio [OR], 14.50; 95% confidence interval (CI), 3.27–111.14; p = 0.002) and presence of heterozygosity for UGT1A1*28 or UGT1A1*6 polymorphism (OR, 2.84; 95% CI, 1.18–7.17; p = 0.023) as independent risk factors for severe neutropenia. The incidences of severe neutropenia and FN in patients receiving mFOLFIRINOX in our clinical practice were comparable to previous reports. The risk factors for severe neutropenia in patients receiving mFOLFIRINOX were a low baseline white blood cell count and presence of heterozygosity for UGT1A1*28 or UGT1A1*6 polymorphism.
Collapse
|
21
|
Hulshof EC, de With M, Creemers GJ, Guchelaar HJ, Mathijssen RHJ, Gelderblom H, Deenen MJ. Response to letter entitled re: UGT1A1 genotype-guided dosing of irinotecan: A prospective safety and cost analysis in poor metaboliser patients. Eur J Cancer 2022; 172:231-233. [DOI: 10.1016/j.ejca.2022.05.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 11/26/2022]
|
22
|
Rugo HS, Tolaney SM, Loirat D, Punie K, Bardia A, Hurvitz SA, O'Shaughnessy J, Cortés J, Diéras V, Carey LA, Gianni L, Piccart MJ, Loibl S, Goldenberg DM, Hong Q, Olivo M, Itri LM, Kalinsky K. Safety analyses from the phase 3 ASCENT trial of sacituzumab govitecan in metastatic triple-negative breast cancer. NPJ Breast Cancer 2022; 8:98. [PMID: 36038616 PMCID: PMC9424318 DOI: 10.1038/s41523-022-00467-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/28/2022] [Indexed: 11/08/2022] Open
Abstract
Sacituzumab govitecan (SG) is an anti-Trop-2 antibody-drug conjugate with an SN-38 payload. In the ASCENT study, patients with metastatic triple-negative breast cancer (mTNBC) relapsed/refractory to ≥2 prior chemotherapy regimens (≥1 in the metastatic setting), received SG or single-agent treatment of physician's choice (eribulin, vinorelbine, capecitabine, or gemcitabine). This ASCENT safety analysis includes the impact of age and UGT1A1 polymorphisms, which hinder SN-38 detoxification. SG demonstrated a manageable safety profile in patients with mTNBC, including those ≥65 years; neutropenia/diarrhea are key adverse events (AE). Patients with UGT1A1 *28/*28 genotype versus those with 1/*28 and *1/*1 genotypes had higher rates of grade ≥3 SG-related neutropenia (59% vs 47% and 53%), febrile neutropenia (18% vs 5% and 3%), anemia (15% vs 6% and 4%), and diarrhea (15% vs 9% and 10%), respectively. Individuals with UGT1A1 *28/*28 genotype should be monitored closely; active monitoring and routine AE management allow optimal therapeutic exposure of SG.
Collapse
Affiliation(s)
- Hope S Rugo
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA.
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Delphine Loirat
- Department of Medical Oncology and D3i, Institut Curie, Paris, France
| | - Kevin Punie
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Aditya Bardia
- Department of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Sara A Hurvitz
- Medical Oncology, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, US Oncology, Dallas, TX, USA
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Quiron Group, Madrid & Barcelona, Barcelona, Spain
- Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - Véronique Diéras
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Lisa A Carey
- Department of Hematology and Oncology, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | | | - Sibylle Loibl
- Department of Medicine and Research, Hämatologisch-Onkologische Gemeinschaftspraxis am Bethanien-Krankenhaus, Frankfurt, Germany
| | - David M Goldenberg
- Department of Clinical Development, Immunomedics, Inc., Morris Plains, NJ, USA
| | - Quan Hong
- Department of Clinical Development, Immunomedics, Inc., Morris Plains, NJ, USA
| | - Martin Olivo
- Department of Clinical Development, Immunomedics, Inc., Morris Plains, NJ, USA
| | - Loretta M Itri
- Department of Clinical Development, Immunomedics, Inc., Morris Plains, NJ, USA
| | - Kevin Kalinsky
- Columbia University Irving Medical Center, New York, NY, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
23
|
Atasilp C, Biswas M, Jinda P, Nuntharadthanaphong N, Rachanakul J, Hongkaew Y, Vanwong N, Saokaew S, Sukasem C. Association of UGT1A1*6, UGT1A1*28, or ABCC2 c.3972C>T genetic polymorphisms with irinotecan-induced toxicity in Asian cancer patients: Meta-analysis. Clin Transl Sci 2022; 15:1613-1633. [PMID: 35506159 PMCID: PMC9283744 DOI: 10.1111/cts.13277] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 11/30/2022] Open
Abstract
Effects of UGT1A1*6 and UGT1A1*28 genetic polymorphisms on irinotecan-induced severe toxicities in Asian cancer patients are inconclusive. Also, ABCC2 c.3972C>T may affect toxicity of irinotecan. The aim was to assess the aggregated risk of neutropenia or diarrhea in Asian cancer patients taking irinotecan and inherited UGT1A1*6, UGT1A1*28, or ABCC2 c.3972C>T genetic variants. A PubMed literature search for eligible studies was conducted. Odds ratios (ORs) were measured using RevMan software where p values <0.05 were statistically significant. Patients that inherited both UGT1A1*6 and UGT1A1*28 genetic variants (heterozygous: UGT1A1*1/*6 + *1/*28 and homozygous: UGT1A1*6/*6 + *28/*28) were significantly associated with increased risk of neutropenia and diarrhea compared to patients with UGT1A1*1/*1 (neutropenia: OR 2.89; 95% CI 1.97-4.23; p < 0.00001; diarrhea: OR 2.26; 95% CI 1.71-2.99; p < 0.00001). Patients carrying homozygous variants had much stronger effects in developing toxicities (neutropenia: OR 6.23; 95% CI 3.11-12.47; p < 0.00001; diarrhea: OR 3.21; 95% CI 2.13-4.85; p < 0.00001) than those with heterozygous variants. However, patients carrying the ABCC2 c.3972C>T genetic variant were not significantly associated with neutropenia (OR 1.67; 95% CI 0.98-2.84; p = 0.06) and were significantly associated with a reduction in irinotecan-induced diarrhea (OR 0.31; 95% CI 0.11-0.81; p = 0.02). Asian cancer patients should undergo screening for both UGT1A1*6 and UGT1A1*28 genetic variants to reduce substantially irinotecan-induced severe toxicities.
Collapse
Affiliation(s)
- Chalirmporn Atasilp
- Chulabhorn International College of MedicineThammasat UniversityPathum ThaniThailand
| | - Mohitosh Biswas
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
- Department of PharmacyUniversity of RajshahiRajshahiBangladesh
| | - Pimonpan Jinda
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
| | - Nutthan Nuntharadthanaphong
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
| | - Jiratha Rachanakul
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
| | - Yaowaluck Hongkaew
- Advance Research and Development LaboratoryBumrungrad International HospitalBangkokThailand
| | - Natchaya Vanwong
- Department of Clinical Chemistry, Faculty of Allied Health SciencesChulalongkorn UniversityBangkokThailand
| | - Surasak Saokaew
- Division of Pharmacy Practice, Department of Pharmaceutical CareSchool of Pharmaceutical Sciences, University of PhayaoPhayaoThailand
- Center of Health Outcomes Research and Therapeutic Safety (COHORTS)School of Pharmaceutical Sciences, University of PhayaoPhayaoThailand
- Unit of Excellence on Clinical Outcomes Research and Integration (UNICORN)School of Pharmaceutical Sciences, University of PhayaoPhayaoThailand
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of PathologyFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Laboratory for Pharmacogenomics, Somdech Phra Debaratana Medical Center (SDMC), Ramathibodi HospitalBangkokThailand
- Pharmacogenomics and Precision Medicine, The Preventive Genomics & Family Check‐up Services CenterBumrungrad International HospitalBangkokThailand
| |
Collapse
|
24
|
Emery LP, Brooks GA. Revisiting UGT1A1 Pharmacogenetic Testing Before Irinotecan-Why Not? JCO Oncol Pract 2022; 18:281-282. [PMID: 35108028 PMCID: PMC9014453 DOI: 10.1200/op.21.00840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/12/2021] [Indexed: 01/07/2023] Open
Affiliation(s)
- Lukas P. Emery
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Gabriel A. Brooks
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH
- Geisel School of Medicine, Lebanon, NH
| |
Collapse
|
25
|
LI Q, SUN T, ZHANG H, LIU W, XIAO Y, SUN H, YIN W, YAO Y, GU Y, LIU Y, YI F, WANG Q, YU J, CAO B, LIANG L. Characteristics and Clinical Implication of UGT1A1 Heterozygous Mutation in Tumor. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:137-146. [PMID: 35340156 PMCID: PMC8976199 DOI: 10.3779/j.issn.1009-3419.2022.101.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND The literature recommends that reduced dosage of CPT-11 should be applied in patients with UGT1A1 homozygous mutations, but the impact of UGT1A1 heterozygous mutations on the adverse reactions of CPT-11 is still not fully clear. METHODS A total of 107 patients with UGT1A1 heterozygous mutation or wild-type, who were treated with CPT-11 from January 2018 to September 2021 in Peking University Third Hospital, were retrospectively enrolled. The adverse reaction spectra of patients with UGT1A1*6 and UGT1A1*28 mutations were analyzed. Adverse reactions were evaluated according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) 5.0. The efficacy was evaluated according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. The genotypes of UGT1A1*6 and UGT1A1*28 were detected by digital fluorescence molecular hybridization. RESULTS There were 43 patients with UGT1A1*6 heterozygous mutation, 26 patients with UGT1A1*28 heterozygous mutation, 8 patients with UGT1A1*6 and UGT1A1*28 double heterozygous mutations, 61 patients with heterozygous mutation at any gene locus of UGT1A1*6 and UGT1A1*28. Logistic regression analysis showed that the presence or absence of vomiting (P=0.013) and mucositis (P=0.005) was significantly correlated with heterozygous mutation of UGT1A1*28, and the severity of vomiting (P<0.001) and neutropenia (P=0.021) were significantly correlated with heterozygous mutation of UGT1A1*6. In colorectal cancer, UGT1A1*6 was significantly correlated to diarrhea (P=0.005), and the other adverse reactions spectrum was similar to that of the whole patient cohort, and efficacy and prognosis were similar between patients with different genotypes and patients treated with reduced CPT-11 dosage or not. CONCLUSIONS In clinical use, heterozygous mutations of UGT1A1*6 and UGT1A1*28 are related to the risk and severity of vomiting, diarrhea, neutropenia and mucositis in patients with Pan-tumor and colorectal cancer post CPT-11 therpy. In colorectal cancer, UGT1A1*6 is significantly related to diarrhea post CPT-11 use, efficacy and prognosis is not affected by various genotypes or CPT-11 dosage reduction.
Collapse
Affiliation(s)
- Qian LI
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Tao SUN
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Hua ZHANG
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
| | - Wei LIU
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Yu XIAO
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Hongqi SUN
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Wencheng YIN
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Yanhong YAO
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Yangchun GU
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Yan'e LIU
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Fumei YI
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Qiqi WANG
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Jinyu YU
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Baoshan CAO
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Li LIANG
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China,Li LIANG, E-mail:
| |
Collapse
|
26
|
The Value of Pharmacogenetics to Reduce Drug-Related Toxicity in Cancer Patients. Mol Diagn Ther 2022; 26:137-151. [PMID: 35113367 PMCID: PMC8975257 DOI: 10.1007/s40291-021-00575-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
Many anticancer drugs cause adverse drug reactions (ADRs) that negatively impact safety and reduce quality of life. The typical narrow therapeutic range and exposure-response relationships described for anticancer drugs make precision dosing critical to ensure safe and effective drug exposure. Germline mutations in pharmacogenes contribute to inter-patient variability in pharmacokinetics and pharmacodynamics of anticancer drugs. Patients carrying reduced-activity or loss-of-function alleles are at increased risk for ADRs. Pretreatment genotyping offers a proactive approach to identify these high-risk patients, administer an individualized dose, and minimize the risk of ADRs. In the field of oncology, the most well-studied gene-drug pairs for which pharmacogenetic dosing recommendations have been published to improve safety are DPYD-fluoropyrimidines, TPMT/NUDT15-thiopurines, and UGT1A1-irinotecan. Despite the presence of these guidelines, the scientific evidence showing the benefits of pharmacogenetic testing (e.g., improved safety and cost-effectiveness) and the development of efficient multi-gene genotyping panels, routine pretreatment testing for these gene-drug pairs has not been implemented widely in the clinic. Important considerations required for widespread clinical implementation include pharmacogenetic education of physicians, availability or allocation of institutional resources to build an efficient clinical infrastructure, international standardization of guidelines, uniform adoption of guidelines by regulatory agencies leading to genotyping requirements in drug labels, and development of cohesive reimbursement policies for pretreatment genotyping. Without clinical implementation, the potential of pharmacogenetics to improve patient safety remains unfulfilled.
Collapse
|
27
|
Yap TA, Hamilton E, Bauer T, Dumbrava EE, Jeselsohn R, Enke A, Hurley S, Lin KK, Habeck J, Giordano H, Shapiro GI. Phase Ib SEASTAR Study: Combining Rucaparib and Sacituzumab Govitecan in Patients With Cancer With or Without Mutations in Homologous Recombination Repair Genes. JCO Precis Oncol 2022; 6:e2100456. [PMID: 35138920 PMCID: PMC8865521 DOI: 10.1200/po.21.00456] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Timothy A. Yap
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Erika Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | - Todd Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hulshof EC, de With M, de Man FM, Creemers GJ, Deiman BALM, Swen JJ, Houterman S, Koolen SLW, Bins S, Thijs AMJ, Laven MMJ, Hövels AM, Luelmo SAC, Houtsma D, Shulman K, McLeod HL, van Schaik RHN, Guchelaar HJ, Mathijssen RHJ, Gelderblom H, Deenen MJ. UGT1A1 genotype-guided dosing of irinotecan: A prospective safety and cost analysis in poor metaboliser patients. Eur J Cancer 2022; 162:148-157. [PMID: 34998046 DOI: 10.1016/j.ejca.2021.12.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/25/2021] [Accepted: 12/05/2021] [Indexed: 12/23/2022]
Abstract
AIM To determine the safety, feasibility, pharmacokinetics, and cost of UGT1A1 genotype-guided dosing of irinotecan. PATIENTS AND METHODS In this prospective, multicentre, non-randomised study, patients intended for treatment with irinotecan were pre-therapeutically genotyped for UGT1A1∗28 and UGT1A1∗93. Homozygous variant carriers (UGT1A1 poor metabolisers; PMs) received an initial 30% dose reduction. The primary endpoint was incidence of febrile neutropenia in the first two cycles of treatment. Toxicity in UGT1A1 PMs was compared to a historical cohort of UGT1A1 PMs treated with full dose therapy, and to UGT1A1 non-PMs treated with full dose therapy in the current study. Secondary endpoints were pharmacokinetics, feasibility, and costs. RESULTS Of the 350 evaluable patients, 31 (8.9%) patients were UGT1A1 PM and received a median 30% dose reduction. The incidence of febrile neutropenia in this group was 6.5% compared to 24% in historical UGT1A1 PMs (P = 0.04) and was comparable to the incidence in UGT1A1 non-PMs treated with full dose therapy. Systemic exposure of SN-38 of reduced dosing in UGT1A1 PMs was still slightly higher compared to a standard-dosed irinotecan patient cohort (difference: +32%). Cost analysis showed that genotype-guided dosing was cost-saving with a cost reduction of €183 per patient. CONCLUSION UGT1A1 genotype-guided dosing significantly reduces the incidence of febrile neutropenia in UGT1A1 PM patients treated with irinotecan, results in a therapeutically effective systemic drug exposure, and is cost-saving. Therefore, UGT1A1 genotype-guided dosing of irinotecan should be considered standard of care in order to improve individual patient safety.
Collapse
Affiliation(s)
- Emma C Hulshof
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, the Netherlands; Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mirjam de With
- Department of Medical Oncology, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Clinical Chemistry, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Femke M de Man
- Department of Medical Oncology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Geert-Jan Creemers
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | - Birgit A L M Deiman
- Department of Molecular Biology, Catharina Hospital, Eindhoven, the Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Saskia Houterman
- Department of Education and Research, Catharina Hospital, Eindhoven, the Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus University Medical Centre, Rotterdam, the Netherlands; Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Anna M J Thijs
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | - Marjan M J Laven
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | - Anke M Hövels
- Hovels Consultancy HTA and Health Economics, Bilthoven, the Netherlands
| | - Saskia A C Luelmo
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Danny Houtsma
- Department of Medical Oncology, Haga Hospital, The Hague, the Netherlands
| | - Katerina Shulman
- Department of Medical Oncology, Carmel Medical Centre and Clalit Haifa District Regional Oncology Clinics, Haifa, Israel
| | - Howard L McLeod
- University of South Florida Taneja College of Pharmacy, Tampa, FL, USA
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, the Netherlands; Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
29
|
Emelyanova M, Pokataev I, Shashkov I, Kopantseva E, Lyadov V, Heydarov R, Mikhailovich V. TYMS 3'-UTR Polymorphism: A Novel Association with FOLFIRINOX-Induced Neurotoxicity in Pancreatic Cancer Patients. Pharmaceutics 2021; 14:pharmaceutics14010077. [PMID: 35056973 PMCID: PMC8779442 DOI: 10.3390/pharmaceutics14010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/17/2021] [Accepted: 12/25/2021] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal malignancy that has the worst 5-year survival rate of all of the common malignant tumors. Surgery, chemotherapy, and/or chemoradiation remain the main tactics for PDAC treatment. The efficacy of chemotherapy is often compromised because of the substantial risk of severe toxicities. In our study, we focused on identification of polymorphisms in the genes involved in drug metabolism, DNA repair and replication that are associated with inter-individual differences in drug-induced toxicities. Using the microarray, we genotyped 12 polymorphisms in the DPYD, XPC, GSTP1, MTHFR, ERCC1, UGT1A1, and TYMS genes in 78 PDAC patients treated with FOLFIRINOX. It was found that the TYMS rs11280056 polymorphism (6 bp-deletion in TYMS 3'-UTR) predicted grade 1-2 neurotoxicity (p = 0.0072 and p = 0.0019, according to co-dominant (CDM) and recessive model (RM), respectively). It is the first report on the association between TYMS rs11280056 and peripheral neuropathy. We also found that PDAC patients carrying the GSTP1 rs1695 GG genotype had a decreased risk for grade 3-4 hematological toxicity as compared to those with the AA or AG genotypes (p = 0.032 and p = 0.014, CDM and RM, respectively). Due to relatively high p-values, we consider that the impact of GSTP1 rs1695 requires further investigation in a larger sample size.
Collapse
Affiliation(s)
- Marina Emelyanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.E.); (I.S.); (E.K.); (R.H.)
| | - Ilya Pokataev
- Department of Oncology, Moscow Clinical Oncology Hospital No.1, Moscow City Health Department, 105005 Moscow, Russia; (I.P.); (V.L.)
| | - Igor Shashkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.E.); (I.S.); (E.K.); (R.H.)
- Federal Research Centre ‘Fundamentals of Biotechnology’, Russian Academy of Sciences, 119071 Moscow, Russia
| | - Elena Kopantseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.E.); (I.S.); (E.K.); (R.H.)
| | - Vladimir Lyadov
- Department of Oncology, Moscow Clinical Oncology Hospital No.1, Moscow City Health Department, 105005 Moscow, Russia; (I.P.); (V.L.)
- Department of Oncology and Palliative Medicine, Russian Medical Academy of Continuous Professional Education, 123242 Moscow, Russia
- Department of Oncology, Novokuznetsk State Institute for Continuous Medical Education, 654005 Novokuznetsk, Russia
| | - Rustam Heydarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.E.); (I.S.); (E.K.); (R.H.)
| | - Vladimir Mikhailovich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.E.); (I.S.); (E.K.); (R.H.)
- Correspondence: or ; Tel./Fax: +7-499-1351177
| |
Collapse
|
30
|
Karas S, Innocenti F. All You Need to Know About UGT1A1 Genetic Testing for Patients Treated With Irinotecan: A Practitioner-Friendly Guide. JCO Oncol Pract 2021; 18:270-277. [PMID: 34860573 DOI: 10.1200/op.21.00624] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Irinotecan is an anticancer agent widely used for the treatment of solid tumors, including colorectal and pancreatic cancers. Severe neutropenia and diarrhea are common dose-limiting toxicities of irinotecan-based therapy, and UGT1A1 polymorphisms are one of the major risk factors of these toxicities. In 2005, the US Food and Drug Administration revised the drug label to indicate that patients with UGT1A1*28 homozygous genotype should receive a decreased dose of irinotecan. However, UGT1A1*28 testing is not routinely used in the clinic, and specific reasons include lack of access to concise information on this wide issue as well as mixed recommendations by regulatory and professional entities. To assist oncologists in assessing whether and when to use UGT1A1 genetic testing in patients receiving irinotecan-based therapies, this article provided (1) essential knowledge of UGT1A1 polymorphisms; (2) an update on the impact of UGT1A1 polymorphisms on efficacy and toxicity of contemporary irinotecan-based regimens; (3) dosing adjustments based upon the UGT1A1 genotypes, and (4) recommendations from currently available guidelines from the US and international scientific consortia and major oncology societies.
Collapse
Affiliation(s)
- Spinel Karas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
31
|
Hamdeh S, Micic D, Hanauer S. Review article: drug-induced small bowel injury. Aliment Pharmacol Ther 2021; 54:1370-1388. [PMID: 34668591 DOI: 10.1111/apt.16642] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/05/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Drug-induced gastrointestinal injury has been increasingly reported, but its exact incidence is not known. The small and large intestines represent the most affected sites of injury, accounting for 20%-40% of all gastrointestinal side effects. AIM To provide an updated literature review detailing medications linked to the development of small bowel injury. METHODS We conducted a literature search on PubMed from its inception to May 1, 2021. We included English-language original studies, meta-analyses, systematic reviews, review articles and case reports. RESULTS Drug-induced enteropathy can range from asymptomatic histological changes resulting in a subtle, self-limited disease to a chronic inflammatory condition mimicking inflammatory bowel disease, or bowel perforation. Endoscopy can demonstrate erythema, mucosal friability, oedema, erosions, ulcers or strictures in severe cases. Histology may include mucosal erosions and ulcerations, focal active enteritis, villous atrophy, epithelial apoptosis or necrotising enteritis. A well-established association has been found with the use of nonsteroidal anti-inflammatory drugs, immunosuppressants, chemotherapeutic agents, antibiotics, immunotherapies, etanercept and olmesartan. Possible associations have been reported with other biologic agents, medications used for glycemic control, antihypertensives, cholinesterase inhibitors, potassium and iron supplements, with conflicting data regarding contraceptives/hormonal therapy and isotretinoin. CONCLUSION Physicians should be aware of the manifestations of drug-induced enteropathy as early recognition can lead to prompt discontinuation of the offending therapy and, therefore, a reduced risk of future complications.
Collapse
Affiliation(s)
- Shadi Hamdeh
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Motility, University of Kansas, Lawrence, KS, USA
| | - Dejan Micic
- Department of Internal Medicine, Section of Gastroenterology, Hepatology and Nutrition, University of Chicago, Chicago, IL, USA
| | - Stephen Hanauer
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
32
|
Czauderna C, Luley K, von Bubnoff N, Marquardt JU. Tailored Systemic Therapy for Colorectal Cancer Liver Metastases. Int J Mol Sci 2021; 22:11780. [PMID: 34769209 PMCID: PMC8584068 DOI: 10.3390/ijms222111780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 11/29/2022] Open
Abstract
Liver metastases are the most common site of metastatic spread in colorectal cancer. Current treatment approaches involve effective systemic therapies in combination with surgical and/or interventional strategies. Multimodal strategies greatly improved clinical outcomes of patients with metastatic colorectal cancer over the last decades. Identification of predictive and prognostic biomarkers helped to comprehensively refine individual targeted treatment approaches and resulted in median overall survival rates of 30 months or longer. Current guidelines, thus, recommend treatment selection according to patients' performance status, tumor localization and stage as well as the tumor's molecular and genetic status. Here, we outline the latest developments in molecular decision-making for patients with upfront resectable, potentially or initially unresectable and non/never-resectable colorectal cancer liver metastases.
Collapse
Affiliation(s)
- Carolin Czauderna
- Department of Medicine I, University Medical Center Schleswig-Holstein—Campus Lübeck, 23558 Lübeck, Germany;
| | - Kim Luley
- Department of Hemato-Oncology, University Medical Center Schleswig-Holstein—Campus Lübeck, 23558 Lübeck, Germany; (K.L.); (N.v.B.)
| | - Nikolas von Bubnoff
- Department of Hemato-Oncology, University Medical Center Schleswig-Holstein—Campus Lübeck, 23558 Lübeck, Germany; (K.L.); (N.v.B.)
| | - Jens U. Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein—Campus Lübeck, 23558 Lübeck, Germany;
| |
Collapse
|
33
|
Mathew JM, Mpangase PT, Sengupta D, Kwenda S, Mavri-Damelin D, Ramsay M. UGT1A1 regulatory variant with potential effect on efficacy of HIV and cancer drugs commonly prescribed in South Africa. Pharmacogenomics 2021; 22:963-972. [PMID: 34528449 DOI: 10.2217/pgs-2021-0062] [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: 11/21/2022] Open
Abstract
Aim: Despite the high disease burden of human immunodeficiency virus (HIV) infection and colorectal cancer (CRC) in South Africa (SA), treatment-relevant pharmacogenetic variants are understudied. Materials & methods: Using publicly available genotype and gene expression data, a bioinformatic pipeline was developed to identify liver expression quantitative trait loci (eQTLs). Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. Conclusion: As increased UGT1A1 expression could affect the clinical outcome of dolutegravir and irinotecan treatment by increasing drug clearance, patients with the rs28967009A variant may require increased drug doses to reach therapeutic levels or should be prescribed alternative drugs.
Collapse
Affiliation(s)
- Jenny Mary Mathew
- Division of Human Genetics, National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2000, South Africa.,Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa
| | - Phelelani Thokozani Mpangase
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa
| | - Dhriti Sengupta
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa
| | - Stanford Kwenda
- Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, 2192, South Africa
| | - Demetra Mavri-Damelin
- School of Molecular & Cell Biology, Faculty of Science, University of the Witwatersrand, Johannesburg, 2050, South Africa
| | - Michèle Ramsay
- Division of Human Genetics, National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2000, South Africa.,Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2050, South Africa
| |
Collapse
|
34
|
Riera P, Páez D. Elucidating the role of pharmacogenetics in irinotecan efficacy and adverse events in metastatic colorectal cancer patients. Expert Opin Drug Metab Toxicol 2021; 17:1157-1163. [PMID: 34486919 DOI: 10.1080/17425255.2021.1974397] [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] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Irinotecan is a cytotoxic agent that is widely used in the treatment of several types of solid tumors. However, although it is generally well tolerated, approximately 20% to 35% of patients develop severe toxicity, particularly delayed-type diarrhea and neutropenia. As the incidence of such toxicities is often associated with the UGT1A1 *28/*28, *6/*28 and *6/*6 genotypes, individualized dosing could reduce these adverse events. Furthermore, prospective trials have shown that patients harboring the UGT1A1 *1/*1 and *1/*28 genotypes can tolerate higher doses of irinotecan, which may in turn impact on a better outcome. Upfront UGT1A1 genotyping could therefore be a usefulness strategy in order to individualize irinotecan dosing, but consensus on the recommended dose based on the UGT1A1 genotype is still lacking. AREAS COVERED This review summarizes the results of the main pharmacogenetic studies focused on irinotecan. We provide an overview of current evidence and recommendations for individualized dosing of irinotecan in metastatic colorectal cancer patients. EXPERT OPINION Implementation of UGT1A1*28 and UGT1A1*6 genotyping in clinical practice is a first step toward personalizing irinotecan therapy. This approach is likely to improve patient care and reduce healthcare costs. Future large and prospective studies will help to clarify the clinical value of other genetic markers in irinotecan treatment personalization.
Collapse
Affiliation(s)
- Pau Riera
- Pharmacy Department, Hospital De La Santa Creu I Sant Pau, Barcelona, Spain.,U705, Isciii Center for Biomedical Research on Rare Diseases (Ciberer), Barcelona, Spain
| | - David Páez
- U705, Isciii Center for Biomedical Research on Rare Diseases (Ciberer), Barcelona, Spain.,Medical Oncology Department, Hospital De La Santa Creu I Sant Pau, Barcelona, Spain
| |
Collapse
|
35
|
Jia Z, Zhu Z, Wang Y, Ding J, Lin Z, Zhang Y, Li Z. The prognostic value of serum bilirubin in colorectal cancer patients with surgical resection. Int J Biol Markers 2021; 36:17246008211036128. [PMID: 34374580 DOI: 10.1177/17246008211036128] [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] [Indexed: 11/17/2022]
Abstract
PURPOSE Serum bilirubin plays an important role in antioxidant and anticancer processes. The inverse association between serum bilirubin and cancer risk have been widely reported in multiple cancers. The aim of this retrospective study was to investigate the prognostic impact of serum bilirubin in colorectal cancer patients undergoing surgical resection. METHODS The value of serum bilirubin including total bilirubin, direct bilirubin, and indirect bilirubin were tested at pre-operatively in 330 colorectal cancer patients. The optimal cut-off values for these three biomarkers were determined by X-tile program. The relationship between serum bilirubin and outcomes were examined using Kaplan-Meier curves log-rank test, univariate and multivariate cox regression. Moreover, a number of risk factors were used to form a nomogram for evaluating risk of survival. RESULTS The optimal cut-off points of serum total bilirubin, direct bilirubin, and indirect bilirubin were 19.5 μmol/L, 5.0 μmol/L and 8.1 μmol/L, respectively. Elevated total bilirubin and direct bilirubin were significantly associated with overall survival in surgical colorectal cancer patients. Additionally, predictive nomogram including total bilirubin and direct bilirubin for overall survival was established for predicting overall survival in surgical colorectal cancer patients. CONCLUSIONS These findings indicated that preoperative elevated total bilirubin and direct bilirubin could be considered as independent prognostic biomarkers for poor overall survival of colorectal cancer patients.
Collapse
Affiliation(s)
- Zhangjun Jia
- Jiangsu Cancer Hospital & 26481Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210009, PR China
| | - Zeyu Zhu
- Huaian Hospital, Huaian, Jiangsu Province, 223200, PR China
| | - Ying Wang
- Jiangsu Cancer Hospital & 26481Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210009, PR China
| | - Jing Ding
- Jiangsu Cancer Hospital & 26481Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210009, PR China
| | - Zhenzhong Lin
- Jiangsu Cancer Hospital & 26481Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210009, PR China
| | - Yanyan Zhang
- Jiangsu Cancer Hospital & 26481Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210009, PR China
| | - Zhipeng Li
- Jiangsu Cancer Hospital & 26481Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210009, PR China
| |
Collapse
|
36
|
Pattanaik S, Jain A, Ahluwalia J. Evolving Role of Pharmacogenetic Biomarkers to Predict Drug-Induced Hematological Disorders. Ther Drug Monit 2021; 43:201-220. [PMID: 33235023 DOI: 10.1097/ftd.0000000000000842] [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: 06/26/2020] [Accepted: 09/21/2020] [Indexed: 11/26/2022]
Abstract
ABSTRACT Drug-induced hematological disorders constitute up to 30% of all blood dyscrasias seen in the clinic. Hematologic toxicity from drugs may range from life-threatening marrow aplasia, agranulocytosis, hemolysis, thrombosis to mild leukopenia, and thrombocytopenia. Pathophysiologic mechanisms underlying these disorders vary from an extension of the pharmacological effect of the drug to idiosyncratic and immune-mediated reactions. Predicting these reactions is often difficult, and this makes clinical decision-making challenging. Evidence supporting the role of pharmacogenomics in the management of these disorders in clinical practice is rapidly evolving. Despite the Clinical Pharmacology Implementation Consortium and Pharmacogenomics Knowledge Base recommendations, few tests have been incorporated into routine practice. This review aims to provide a comprehensive summary of the various drugs which are implicated for the hematological adverse events, their underlying mechanisms, and the current evidence and practical recommendations to incorporate pharmacogenomic testing in clinical care for predicting these disorders.
Collapse
Affiliation(s)
| | - Arihant Jain
- Internal Medicine, Hematology and Bone Marrow Transplantation, and
| | - Jasmina Ahluwalia
- Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
37
|
Nelson RS, Seligson ND, Bottiglieri S, Carballido E, Cueto AD, Imanirad I, Levine R, Parker AS, Swain SM, Tillman EM, Hicks JK. UGT1A1 Guided Cancer Therapy: Review of the Evidence and Considerations for Clinical Implementation. Cancers (Basel) 2021; 13:1566. [PMID: 33805415 PMCID: PMC8036652 DOI: 10.3390/cancers13071566] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
Multi-gene assays often include UGT1A1 and, in certain instances, may report associated toxicity risks for irinotecan, belinostat, pazopanib, and nilotinib. However, guidance for incorporating UGT1A1 results into therapeutic decision-making is mostly lacking for these anticancer drugs. We summarized meta-analyses, genome-wide association studies, clinical trials, drug labels, and guidelines relating to the impact of UGT1A1 polymorphisms on irinotecan, belinostat, pazopanib, or nilotinib toxicities. For irinotecan, UGT1A1*28 was significantly associated with neutropenia and diarrhea, particularly with doses ≥ 180 mg/m2, supporting the use of UGT1A1 to guide irinotecan prescribing. The drug label for belinostat recommends a reduced starting dose of 750 mg/m2 for UGT1A1*28 homozygotes, though published studies supporting this recommendation are sparse. There was a correlation between UGT1A1 polymorphisms and pazopanib-induced hepatotoxicity, though further studies are needed to elucidate the role of UGT1A1-guided pazopanib dose adjustments. Limited studies have investigated the association between UGT1A1 polymorphisms and nilotinib-induced hepatotoxicity, with data currently insufficient for UGT1A1-guided nilotinib dose adjustments.
Collapse
Affiliation(s)
- Ryan S. Nelson
- Department of Consultative Services, ARUP Laboratories, Salt Lake City, UT 84108, USA;
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Nathan D. Seligson
- Department of Pharmacotherapy and Translational Research, The University of Florida, Jacksonville, FL 32610, USA;
- Department of Hematology and Oncology, Nemours Children’s Specialty Care, Jacksonville, FL 32207, USA
| | - Sal Bottiglieri
- Department of Pharmacy, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Estrella Carballido
- Department of Oncological Sciences, University of South Florida, Tampa, FL 33612, USA; (E.C.); (I.I.); (R.L.)
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Alex Del Cueto
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Iman Imanirad
- Department of Oncological Sciences, University of South Florida, Tampa, FL 33612, USA; (E.C.); (I.I.); (R.L.)
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Richard Levine
- Department of Oncological Sciences, University of South Florida, Tampa, FL 33612, USA; (E.C.); (I.I.); (R.L.)
- Department of Satellite and Community Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Sandra M. Swain
- Georgetown University Medical Center, MedStar Health, Washington, DC 20007, USA;
| | - Emma M. Tillman
- Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - J. Kevin Hicks
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL 33612, USA;
- Department of Oncological Sciences, University of South Florida, Tampa, FL 33612, USA; (E.C.); (I.I.); (R.L.)
| |
Collapse
|
38
|
Ruiz-Bañobre J, Goel A. Genomic and epigenomic biomarkers in colorectal cancer: From diagnosis to therapy. Adv Cancer Res 2021; 151:231-304. [PMID: 34148615 PMCID: PMC10338180 DOI: 10.1016/bs.acr.2021.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths in the United States. Despite ongoing efforts aimed at increasing screening for CRC and early detection, and development of more effective therapeutic regimens, the overall morbidity and mortality from this malignancy remains a clinical challenge. Therefore, identifying and developing genomic and epigenomic biomarkers that can improve CRC diagnosis and help predict response to current therapies are of paramount importance for improving survival outcomes in CRC patients, sparing patients from toxicity associated with current regimens, and reducing the economic burden associated with these treatments. Although efforts to develop biomarkers over the past decades have achieved some success, the recent availability of high-throughput analytical tools, together with the use of machine learning algorithms, will likely hasten the development of more robust diagnostic biomarkers and improved guidance for clinical decision-making in the coming years. In this chapter, we provide a systematic and comprehensive overview on the current status of genomic and epigenomic biomarkers in CRC, and comment on their potential clinical significance in the management of patients with this fatal malignancy, including in the context of precision medicine.
Collapse
Affiliation(s)
- Juan Ruiz-Bañobre
- Medical Oncology Department, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), CIBERONC, Santiago de Compostela, Spain; Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), CIBERONC, Santiago de Compostela, Spain
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
39
|
Benson AB, Venook AP, Al-Hawary MM, Arain MA, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Farkas L, Garrido-Laguna I, Grem JL, Gunn A, Hecht JR, Hoffe S, Hubbard J, Hunt S, Johung KL, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Miller ED, Mulcahy MF, Nurkin S, Overman MJ, Parikh A, Patel H, Pedersen K, Saltz L, Schneider C, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Gregory KM, Gurski LA. Colon Cancer, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19:329-359. [PMID: 33724754 DOI: 10.6004/jnccn.2021.0012] [Citation(s) in RCA: 914] [Impact Index Per Article: 228.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This selection from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Colon Cancer focuses on systemic therapy options for the treatment of metastatic colorectal cancer (mCRC), because important updates have recently been made to this section. These updates include recommendations for first-line use of checkpoint inhibitors for mCRC, that is deficient mismatch repair/microsatellite instability-high, recommendations related to the use of biosimilars, and expanded recommendations for biomarker testing. The systemic therapy recommendations now include targeted therapy options for patients with mCRC that is HER2-amplified, or BRAF V600E mutation-positive. Treatment and management of nonmetastatic or resectable/ablatable metastatic disease are discussed in the complete version of the NCCN Guidelines for Colon Cancer available at NCCN.org. Additional topics covered in the complete version include risk assessment, staging, pathology, posttreatment surveillance, and survivorship.
Collapse
Affiliation(s)
- Al B Benson
- 1Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Alan P Venook
- 2UCSF Helen Diller Family Comprehensive Cancer Center
| | | | | | | | | | - Stacey Cohen
- 6Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | - Linda Farkas
- 9UT Southwestern Simmons Comprehensive Cancer Center
| | | | | | | | | | | | | | - Steven Hunt
- 16Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | - Smitha Krishnamurthi
- 19Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | - Eric D Miller
- 22The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Mary F Mulcahy
- 1Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | - Katrina Pedersen
- 16Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Vivaldi C, Crucitta S, Catanese S, Cucchiara F, Arrigoni E, Pecora I, Rofi E, Fornaro L, Salani F, Massa V, Vasile E, Morganti R, Danesi R, Del Re M. Comprehensive pharmacogenetic analysis of DPYD, UGT, CDA, and ABCB1 polymorphisms in pancreatic cancer patients receiving mFOLFIRINOX or gemcitabine plus nab-paclitaxel. THE PHARMACOGENOMICS JOURNAL 2021; 21:233-242. [PMID: 33462346 DOI: 10.1038/s41397-020-00203-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 11/09/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022]
Abstract
Modified FOLFIRINOX (mFOLFIRINOX) and gemcitabine + nab-paclitaxel (GemNab) regimens represent a standard treatment in advanced pancreatic cancer (aPC). DPYD and UGT1A1 variants are relevant predictors of fluoropyrimidine and irinotecan-associated adverse events (AEs). Furthermore, data about the associations between polymorphisms in ABCB and CDA genes and GemNab-related toxicities are still controversial. The present study analyzes the association between DPYD, UGT, ABCB1, CDA variants, and AEs in aPC patients (pts) treated with mFOLFIRINOX or GemNab. Blood samples collected from 104 aPC pts treated with mFOLFIRINOX and 63 with GemNab were tested for DPYD c.1679T>G, IVS14+1G>A, c.2194G>A, c.2846A>T, UGT1A1*28, CDA c.79A>C, and ABCB1 c.1236C>T, c.2677G>T/A, c.3435C>T by real-time PCR and automatic sequencing. In mFOLFIRINOX cohort, DPYD IVS14+1GA genotype was associated with G4 hematological AEs, while the UGT1A1*28 significantly correlated with the risk of thrombocytopenia (p = 0.006). In the GemNab cohort, a significant association between CDA c.79CC and high-grade nausea was observed (p = 0.002). Moreover, the presence of at least a mutant allele in ABCB1 increased the risk of overall hematological AEs (p = 0.01), both further strengthened by the presence of CDA c.79CC (p = 0.0002). DPYD IVS14+1A allele is confirmed to be associated with fluoropyrimidine life-threatening toxicities, and UGT1A1*28 is related with a higher risk of hematologic AEs following irinotecan treatment. CDA c.79C and ABCB1 c.1236T, c.2677T/A, and c.3435T mutant alleles are predictive biomarkers of GemNab-related AEs. All these variants should be considered in aPC pts candidate to mFOLFIRINOX or GemNab treatments.
Collapse
Affiliation(s)
- Caterina Vivaldi
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Silvia Catanese
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Arrigoni
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Irene Pecora
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Eleonora Rofi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lorenzo Fornaro
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Francesca Salani
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Valentina Massa
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Enrico Vasile
- Medical Oncology Unit 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Riccardo Morganti
- Departmental Section of Statistical Support for Clinical Trials, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
41
|
Roberto M, Rossi A, Panebianco M, Pomes LM, Arrivi G, Ierinò D, Simmaco M, Marchetti P, Mazzuca F. Drug-Drug Interactions and Pharmacogenomic Evaluation in Colorectal Cancer Patients: The New Drug-PIN ® System Comprehensive Approach. Pharmaceuticals (Basel) 2021; 14:ph14010067. [PMID: 33467633 PMCID: PMC7830292 DOI: 10.3390/ph14010067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/17/2022] Open
Abstract
Drug–drug interactions (DDIs) can affect both treatment efficacy and toxicity. We used Drug-PIN® (Personalized Interactions Network) software in colorectal cancer (CRC) patients to evaluate drug–drug–gene interactions (DDGIs), defined as the combination of DDIs and individual genetic polymorphisms. Inclusion criteria were: (i) stage II-IV CRC; (ii) ECOG PS (Performance status sec. Eastern coperative oncology group) ≤2; (iii) ≥5 concomitant drugs; and (iv) adequate renal, hepatic, and bone marrow function. The Drug-PIN® system analyzes interactions between active and/or pro-drug forms by integrating biochemical, demographic, and genomic data from 110 SNPs. We selected DDI, DrugPin1, and DrugPin2 scores, resulting from concomitant medication interactions, concomitant medications, and SNP profiles, and DrugPin1 added to chemotherapy drugs, respectively. Thirty-four patients, taking a median of seven concomitant medications, were included. The median DrugPin1 and DrugPin2 scores were 42.6 and 77.7, respectively. In 13 patients, the DrugPin2 score was two-fold higher than the DrugPin1 score, with 7 (54%) of these patients experiencing severe toxicity that required hospitalization. On chi-squared testing for any toxicity, a doubled DrugPin2 score (p = 0.001) was significantly related to G3–G4 toxicity. Drug-PIN® software may prevent severe adverse events, decrease hospitalizations, and improve survival in cancer patients.
Collapse
Affiliation(s)
- Michela Roberto
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy; (M.R.); (A.R.); (G.A.); (D.I.); (P.M.); (F.M.)
- Department of Medical-Surgical Sciences and Translation Medicine, Sapienza University, Sant’Andrea University Hospital, 00187 Rome, Italy
| | - Alessandro Rossi
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy; (M.R.); (A.R.); (G.A.); (D.I.); (P.M.); (F.M.)
| | - Martina Panebianco
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy; (M.R.); (A.R.); (G.A.); (D.I.); (P.M.); (F.M.)
- Correspondence:
| | - Leda Marina Pomes
- Department of Neuroscience, Mental Health, 00187 Rome, Italy; (L.M.P.); (M.S.)
- and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, Sant’Andrea University Hospital, 00187 Rome, Italy
| | - Giulia Arrivi
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy; (M.R.); (A.R.); (G.A.); (D.I.); (P.M.); (F.M.)
| | - Debora Ierinò
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy; (M.R.); (A.R.); (G.A.); (D.I.); (P.M.); (F.M.)
- Department of Medical-Surgical Sciences and Translation Medicine, Sapienza University, Sant’Andrea University Hospital, 00187 Rome, Italy
| | - Maurizio Simmaco
- Department of Neuroscience, Mental Health, 00187 Rome, Italy; (L.M.P.); (M.S.)
- and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, Sant’Andrea University Hospital, 00187 Rome, Italy
- Department of Advanced Molecular Diagnostics, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy
| | - Paolo Marchetti
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy; (M.R.); (A.R.); (G.A.); (D.I.); (P.M.); (F.M.)
| | - Federica Mazzuca
- Oncology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, University “La Sapienza”, 00187 Rome, Italy; (M.R.); (A.R.); (G.A.); (D.I.); (P.M.); (F.M.)
| |
Collapse
|
42
|
Simões AR, Fernández-Rozadilla C, Maroñas O, Carracedo Á. The Road so Far in Colorectal Cancer Pharmacogenomics: Are We Closer to Individualised Treatment? J Pers Med 2020; 10:E237. [PMID: 33228198 PMCID: PMC7711884 DOI: 10.3390/jpm10040237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
In recent decades, survival rates in colorectal cancer have improved greatly due to pharmacological treatment. However, many patients end up developing adverse drug reactions that can be severe or even life threatening, and that affect their quality of life. These remain a limitation, as they may force dose reduction or treatment discontinuation, diminishing treatment efficacy. From candidate gene approaches to genome-wide analysis, pharmacogenomic knowledge has advanced greatly, yet there is still huge and unexploited potential in the use of novel technologies such as next-generation sequencing strategies. This review summarises the road of colorectal cancer pharmacogenomics so far, presents considerations and directions to be taken for further works and discusses the path towards implementation into clinical practice.
Collapse
Affiliation(s)
- Ana Rita Simões
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Ceres Fernández-Rozadilla
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
| | - Olalla Maroñas
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain; (A.R.S.); (O.M.); (Á.C.)
- Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica; SERGAS, 15706 Santiago de Compostela, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Raras—CIBERER, 28029 Madrid, Spain
| |
Collapse
|
43
|
Bruera G, Ricevuto E. Pharmacogenomic Assessment of Patients with Colorectal Cancer and Potential Treatments. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:601-617. [PMID: 33235483 PMCID: PMC7678498 DOI: 10.2147/pgpm.s253586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022]
Abstract
Evolving intensiveness of colorectal cancer (CRC) treatment, including chemotherapeutics and targeted agents associations, in adjuvant and metastatic CRC (MCRC) settings, increased overall survival (OS) with individual variability of toxicity. Pharmacogenomic guidelines recommended pre-treatment identification of at-risk patients suggesting dose adjustment of fluoropyrimidines based on dihydropyrimidine dehydrogenase (DPYD), and irinotecan on UDP glucuronosyl-transferase 1 family polypeptide A1 (UGT1A1) genetic variants, but they are poorly applied in clinical practice. This review highlighted clinically validated pharmacogenetic markers, to underline the need of their implementation in the multidisciplinary molecular board for individual CRC patients in clinical practice. Five clinically relevant DPYD variants with different prevalence impair enzymatic effectiveness and significantly increase toxicity: c.1236 G>A (c.1129–5923 C>G, HapB3), 4.1–4.8%; c.1679 T>G (DPYD*13), c.1905+1G>A (DPYD*2A), c.2846 A>T, c.2194 A>T (DPYD*6) 1% each. c.1679T>G and c.1905+1G>A are most deleterious on DPD effectiveness, moderately reduced in c.1236/HapB3 and c.2846A>T. Cumulatively, these variants explain approximately half of the estimated 10–15% fluoropyrimidine-related gastrointestinal and hematological toxicities due to DPD. Prevalent UGT1A1 gene [TA]7TAA promoter allelic variant UGT1A1*28, characterized by an extra TA repeat, is associated with low transcriptional and reduced enzymatic effectiveness, decreased SN38 active irinotecan metabolite glucuronidation, vs wild-type UGT1A1*1 [A(TA)6TAA]. Homozygote UGT1A1*28 alleles patients are exposed to higher hematological and gastrointestinal toxicities, even more than heterozygote, at >150 mg/m2 dose. Dose reduction is recommended for homozygote variant. Wild-type UGT1A1*28 alleles patients could tolerate increased doses, potentially affecting favorable outcomes. Implementation of up-front evaluation of the five validated DPYD variants and UGT1A1*28 in the multidisciplinary molecular tumor board, also including CRC genetic characterization, addresses potential treatments with fluoropyrimidines and irinotecan associations at proper doses and schedules, particularly for early CRC, MCRC patients fit for intensive regimens or unfit for conventional regimens, requiring treatment modulations, and also for patients who experience severe, unexpected toxicities. Integration of individual evaluation of toxicity syndromes (TS), specifically limiting TS (LTS), an innovative indicator of toxicity burden in individual patients, may be useful to better evaluate relationships between pharmacogenomic analyses with safety profiles and clinical outcomes.
Collapse
Affiliation(s)
- Gemma Bruera
- Oncology Territorial Care, S. Salvatore Hospital, Oncology Network ASL1 Abruzzo, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Enrico Ricevuto
- Oncology Territorial Care, S. Salvatore Hospital, Oncology Network ASL1 Abruzzo, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | |
Collapse
|
44
|
Hulshof EC, Deenen MJ, Guchelaar HJ, Gelderblom H. Pre-therapeutic UGT1A1 genotyping to reduce the risk of irinotecan-induced severe toxicity: Ready for prime time. Eur J Cancer 2020; 141:9-20. [PMID: 33125947 DOI: 10.1016/j.ejca.2020.09.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pre-therapeutic UGT1A1 genotyping is not yet routinely performed in most hospitals in patients starting irinotecan chemotherapy. The aim of this position paper was to evaluate the available evidence and to assess the potential value of genotyping of UGT1A1∗28 and UGT1A1*6 in patients before starting treatment with irinotecan to reduce the risk of severe toxicity. METHODS The literature was selected and assessed based on five pre-specified criteria: 1) the level of evidence for associations between UGT1A1 polymorphisms and irinotecan-induced severe toxicity, 2) clinical validity and utility of pre-therapeutic genotyping of UGT1A1, 3) safety and tolerability of irinotecan in carriers of UGT1A1 polymorphisms, 4) availability of specific dose recommendations for irinotecan in carriers of UGT1A1 polymorphisms, 5) evidence of cost benefits of pre-therapeutic genotyping of UGT1A1. RESULTS On all five criteria, study results were favourable for pre-therapeutic genotyping of UGT1A1. A high level of evidence (level I) was found for a higher incidence of irinotecan-induced severe toxicity in homozygous carriers of UGT1A1*28 or UGT1A1*6. The clinical validity and utility of this genetic test proved to be acceptable. Dose-finding studies showed a lower maximum tolerated dose in homozygous variant allele carriers, and most of the drug labels and guidelines recommend a dose reduction of 25-30% in these patients. In addition, pre-therapeutic genotyping of UGT1A1 is likely to save costs. CONCLUSION Pre-therapeutic genotyping of UGT1A1 in patients initiating treatment with irinotecan improves patient safety, is likely to be cost-saving, and should, therefore, become standard of care.
Collapse
Affiliation(s)
- Emma C Hulshof
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, the Netherlands; Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, the Netherlands; Department of Clinical Pharmacy and Toxicology, 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 of Personalized Therapeutics, the Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
45
|
Kee PS, Chin PKL, Kennedy MA, Maggo SDS. Pharmacogenetics of Statin-Induced Myotoxicity. Front Genet 2020; 11:575678. [PMID: 33193687 PMCID: PMC7596698 DOI: 10.3389/fgene.2020.575678] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
Statins, a class of lipid-lowering medications, have been a keystone treatment in cardiovascular health. However, adverse effects associated with statin use impact patient adherence, leading to statin discontinuation. Statin-induced myotoxicity (SIM) is one of the most common adverse effects, prevalent across all ages, genders, and ethnicities. Although certain demographic cohorts carry a higher risk, the impaired quality of life attributed to SIM is significant. The pathogenesis of SIM remains to be fully elucidated, but it is clear that SIM is multifactorial. These factors include drug-drug interactions, renal or liver dysfunction, and genetics. Genetic-inferred risk for SIM was first reported by a landmark genome-wide association study, which reported a higher risk of SIM with a polymorphism in the SLCO1B1 gene. Since then, research associating genetic factors with SIM has expanded widely and has become one of the foci in the field of pharmacogenomics. This review provides an update on the genetic risk factors associated with SIM.
Collapse
Affiliation(s)
- Ping Siu Kee
- Gene Structure and Function Laboratory, Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | | | - Martin A. Kennedy
- Gene Structure and Function Laboratory, Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Simran D. S. Maggo
- Gene Structure and Function Laboratory, Carney Centre for Pharmacogenomics, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
46
|
Badée J, Qiu N, Collier AC, Takahashi RH, Forrest WF, Parrott N, Schmidt S, Fowler S. Characterization of the Ontogeny of Hepatic UDP-Glucuronosyltransferase Enzymes Based on Glucuronidation Activity Measured in Human Liver Microsomes. J Clin Pharmacol 2020; 59 Suppl 1:S42-S55. [PMID: 31502688 DOI: 10.1002/jcph.1493] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
An understanding of the postnatal development of hepatic UDP-glucuronosyltransferase (UGT) enzymes is required for accurate prediction of the age-dependent changes in pharmacokinetics of many drugs used in children. However, the maturation rate of hepatic UGT isoforms remains a major knowledge gap. This study aimed to establish the age-associated changes in glucuronidation activity of 10 major hepatic UGT isoforms in humans, namely, UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2B4, UGT2B7, UGT2B10, UGT2B15, and UGT2B17. Human liver microsomes from pediatric and adult donors were incubated under optimized incubation conditions to assess the activity rates of hepatic UGT isoforms using a panel of 19 in vitro UGT probe substrates and clinically used drugs. Statistically strong correlations of glucuronidation activities allowed the ontogeny of UGT1A1, UGT1A4, UGT2B7, UGT2B10, and UGT2B15 to be established using multiple selective UGT substrates and matched human liver microsome samples. The postnatal development of hepatic UGTs is isoform-dependent using either individual or cross-correlated selective isoform substrates. Maximal adult activity was reached at different times ranging from within a month (UGT1A1, UGT2B4, UGT2B7, UGT2B10, and UGT2B15), during infancy (UGT1A3, UGT1A4, and UGT1A9), to adolescence (UGT1A6 and UGT2B17). This study provides an extensive characterization of the postnatal ontogeny profiles of hepatic UGT enzymes that are instrumental for predicting drug disposition via in vitro-in vivo extrapolation algorithms and verifying pharmacokinetic predictions against in vivo observations via pediatric physiologically based pharmacokinetic modeling in pediatric patients.
Collapse
Affiliation(s)
- Justine Badée
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida, USA
| | - Nahong Qiu
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | - Abby C Collier
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ryan H Takahashi
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - William F Forrest
- Department of Bioinformatics and Computational Biology, Genentech, Inc., South San Francisco, California, USA
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida, USA
| | - Stephen Fowler
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
47
|
Federico SM, Pappo AS, Sahr N, Sykes A, Campagne O, Stewart CF, Clay MR, Bahrami A, McCarville MB, Kaste SC, Santana VM, Helmig S, Gartrell J, Shelat A, Brennan RC, Hawkins D, Godwin K, Bishop MW, Furman WL, Stewart E. A phase I trial of talazoparib and irinotecan with and without temozolomide in children and young adults with recurrent or refractory solid malignancies. Eur J Cancer 2020; 137:204-213. [PMID: 32795876 DOI: 10.1016/j.ejca.2020.06.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Talazoparib combined with irinotecan and temozolomide demonstrated efficacy in a murine Ewing sarcoma model. Based on these data, we conducted a phase I trial of talazoparib and irinotecan with/without temozolomide in paediatric patients with recurrent/refractory solid malignancies. PATIENTS AND METHODS Cohorts of 3-6 patients with recurrent/refractory solid malignancies received escalating doses of oral talazoparib and intravenous irinotecan (arm A) and oral talazoparib, oral temozolomide and intravenous irinotecan (arm B) in a 3 + 3 design. Talazoparib was administered on days 1-6, and intravenous irinotecan and oral temozolomide were administered on days 2-6, of a 21-day course. Serum for talazoparib and irinotecan pharmacokinetics was obtained during course 1. UGT1A1 polymorphism and Schlafen family member 11 (SLFN11) immunohistochemical staining were performed. RESULTS Forty-one patients (20 males; median age, 14.6 years; 24 with recurrent disease) were evaluable for dose escalation. Twenty-nine and 12 patients were treated on arm A and arm B, respectively, for a total of 208 courses. The most common diagnosis was Ewing sarcoma (53%). The most common ≥grade III haematologic toxicities in arms A and B included neutropenia (78% and 31%, respectively) and thrombocytopenia (42% and 31%, respectively). In arms A and B, febrile neutropenia (24% and 14%, respectively) and diarrhoea (21% and 7%, respectively) were the most common ≥grade III non-hematologic toxicities. Six patients (Ewing sarcoma [5 patients] and synovial sarcoma [1 patient]) had a response (1 with a complete response, 5 with a partial response). The objective response rates were 10.3% (arm A) and 25% (arm B). Pharmacokinetic testing demonstrated no evidence of drug-drug interaction between talazoparib and irinotecan. UGT1A1 was not related to response. SLFN11 positivity was associated with best response to therapy. CONCLUSIONS The combination of talazoparib and irinotecan with/without temozolomide is feasible and active in Ewing sarcoma, and further investigation is warranted.
Collapse
Affiliation(s)
- Sara M Federico
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - Alberto S Pappo
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Natasha Sahr
- Departments of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - April Sykes
- Departments of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Olivia Campagne
- Departments of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Clinton F Stewart
- Departments of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael R Clay
- Departments of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Armita Bahrami
- Departments of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mary B McCarville
- Departments of Radiological Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sue C Kaste
- Departments of Radiological Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Victor M Santana
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Sara Helmig
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Jessica Gartrell
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anang Shelat
- Departments of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Rachel C Brennan
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Dana Hawkins
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kimberly Godwin
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael W Bishop
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Wayne L Furman
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Elizabeth Stewart
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Departments of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; The Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
48
|
Kalthoff S, Paulusch S, Rupp A, Holdenrieder S, Hartmann G, Strassburg CP. The coffee ingredients caffeic acid and caffeic acid phenylethyl ester protect against irinotecan-induced leukopenia and oxidative stress response. Br J Pharmacol 2020; 177:4193-4208. [PMID: 32548889 PMCID: PMC7443465 DOI: 10.1111/bph.15162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 05/26/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022] Open
Abstract
Background and Purpose Irinotecan, used in colorectal cancer therapy, is metabolized by glucuronidation involving different UDP‐glucuronosyltransferase (UGT)1A isoforms leading to facilitated elimination from the body. Individuals homozygous for the genetic variants UGT1A1*28 (Gilbert syndrome) and UGT1A7*3 are more susceptible to irinotecan side effects, severe diarrhoea and leukopenia. The aim of this study was to investigate the protective effects and active constituents of coffee during irinotecan therapy using humanized transgenic (htg)UGT1A‐WT and htgUGT1A‐SNP (carry UGT1A1*28 and UGT1A7*3 polymorphisms) mice. Experimental Approach HtgUGT1A mice were pretreated with coffee or caffeic acid (CA) + caffeic acid phenylethyl ester (CAPE) and injected with irinotecan. The effects of coffee and CA + CAPE were investigated using reporter gene assays, immunoblot, TaqMan‐PCR, siRNA analyses and blood counts. Key Results Only the combination of the two coffee ingredients, CA and CAPE, mediates protective effects of coffee in a model of irinotecan toxicity by activation of UGT1A genes. Coffee and CA + CAPE significantly increased UGT1A expression and activity along with SN‐38 glucuronide excretion in irinotecan‐injected htgUGT1A mice, resulting in significant improvement of leukopenia, intestinal oxidative stress and inflammation. Conclusion and Implications In this study, we identify the compounds responsible for mediating the previously reported coffee‐induced activation of UGT1A gene expression. CA and CAPE represent key factors for the protective properties of coffee which are capable of reducing irinotecan toxicity, exerting antioxidant and protective effects. Provided that CA + CAPE do not affect irinotecan efficacy, they might represent a novel strategy for the treatment of irinotecan toxicity.
Collapse
Affiliation(s)
- Sandra Kalthoff
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Stefan Paulusch
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Alexander Rupp
- Institute of Laboratory Medicine, German Heart Center of the Technical University Munich, Munich, Germany
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Center of the Technical University Munich, Munich, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
49
|
Carr DF, Turner RM, Pirmohamed M. Pharmacogenomics of anticancer drugs: Personalising the choice and dose to manage drug response. Br J Clin Pharmacol 2020; 87:237-255. [PMID: 32501544 DOI: 10.1111/bcp.14407] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/11/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
The field of pharmacogenomics has made great strides in oncology over the last 20 years and indeed a significant number of pre-emptive genetic tests are now routinely undertaken prior to anticancer drug administration. Many of these gene-drug interactions are the fruits of candidate gene and genome-wide association studies, which have largely focused on common genetic variants (allele frequency>1%). Examples where there is clinical utility include genotyping or phenotyping for G6PD to prevent rasburicase-induced RBC haemolysis, and TPMT to prevent thiopurine-induced bone marrow suppression. Other associations such as CYP2D6 status in determining the efficacy of tamoxifen are more controversial because of contradictory evidence from different sources, which has led to variability in the implementation of testing. As genomic technology becomes ever cheaper and more accessible, we must look to the additional data our genome can provide to explain interindividual variability in anticancer drug response. Clearly genes do not act on their own and it is therefore important to investigate genetic factors in conjunction with clinical factors, interacting concomitant drug therapies and other factors such as the microbiome, which can all affect drug disposition. Taking account of all of these factors, in conjunction with the somatic genome, is more likely to provide better predictive accuracy in determining anticancer drug response, both efficacy and safety. This review summarises the existing knowledge related to the pharmacogenomics of anticancer drugs and discusses areas of opportunity for further advances in personalisation of therapy in order to improve both drug safety and efficacy.
Collapse
Affiliation(s)
- Daniel F Carr
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Richard M Turner
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Munir Pirmohamed
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| |
Collapse
|
50
|
Antelo G, Hierro C, Fernández JP, Baena E, Bugés C, Layos L, Manzano JL, Caro M, Mesia R. Rectal neuroendocrine carcinoma: case report of a rare entity and perspective review of promising agents. Drugs Context 2020; 9:dic-2020-2-4. [PMID: 32477420 PMCID: PMC7233296 DOI: 10.7573/dic.2020-2-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/07/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) comprise a heterogeneous group of tumours, which can be classified into neuroendocrine tumours (NETs), neuroendocrine carcinomas (NECs) and mixed neuroendocrine non-neuroendocrine neoplasms (MiNENs). To date, there is no consensus regarding the optimal therapy, which usually depends on the primary location and classification, according to morphological features of differentiation and proliferation rates. Nevertheless, multidisciplinary strategies combining medical treatments and locoregional strategies have yielded better efficacy results. Here, we report the case of a patient diagnosed with a nonfunctional rectal NECs with metastatic widespread to pelvic lymph nodes and bilateral lung metastases. The patient received three cycles of platinum-etoposide, concomitantly with palliative radiotherapy. Although CT scan after three cycles showed a significant partial response, there was an early fatal progression only 3 months after having stopped systemic therapy. As formerly described in the literature, this case highlights the aggressive behaviour of NECs, rare tumours that often present in advanced stages at diagnosis. Lately, new insights into the molecular biology of NECs have unveiled the possibility of using novel drugs, such as targeted agents or immunotherapy, in molecularly selected subgroups of patients. In this review, we discuss the current management of this rare entity and provide an overview of the most relevant molecular findings, whilst illustrating the potential value that prescreening panels can offer, searching for actionable targets (MSI/dMMR, PD-L1, BRAFv600E) to guide therapy with promising agents that could fill a void in this disease.
Collapse
Affiliation(s)
- Gabriela Antelo
- Radiation Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - Cinta Hierro
- Medical Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Badalona-Applied Research Group in Oncology (B-ARGO)-Germans Trias i Pujol Institute (IGTP); Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - Juan Pablo Fernández
- Pathology Department, Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - Eduardo Baena
- Radiation Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - Cristina Bugés
- Medical Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Badalona-Applied Research Group in Oncology (B-ARGO)-Germans Trias i Pujol Institute (IGTP); Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - Laura Layos
- Medical Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Badalona-Applied Research Group in Oncology (B-ARGO)-Germans Trias i Pujol Institute (IGTP); Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - José Luis Manzano
- Medical Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Badalona-Applied Research Group in Oncology (B-ARGO)-Germans Trias i Pujol Institute (IGTP); Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - Mónica Caro
- Radiation Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| | - Ricard Mesia
- Medical Oncology Department, Catalan Institute of Oncology (ICO)-Badalona; Badalona-Applied Research Group in Oncology (B-ARGO)-Germans Trias i Pujol Institute (IGTP); Germans Trias i Pujol University Hospital (HUGTiP), Badalona, Barcelona, Spain
| |
Collapse
|