1
|
Xiao S, Yin H, Lv X, Wang Z, Jiang L, Xia Y, Liu Y. Inhibition of human UDP-glucuronosyltransferase (UGT) enzymes by darolutamide: Prediction of in vivo drug-drug interactions. Chem Biol Interact 2024; 403:111246. [PMID: 39278459 DOI: 10.1016/j.cbi.2024.111246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Darolutamide is a potent second-generation, selective nonsteroidal androgen receptor inhibitor (ARI), which has been approved by the US Food and Drug Administration (FDA) in treating castrate-resistant, non-metastatic prostate cancer (nmCRPC). Whether darolutamide affects the activity of UDP-glucuronosyltransferases (UGTs) is unknown. The purpose of the present study is to evaluate the inhibitory effect of darolutamide on recombinant human UGTs and pooled human liver microsomes (HLMs), and explore the potential for drug-drug interactions (DDIs) mediated by darolutamide through UGTs inhibition. The product formation rate of UGTs substrates with or without darolutamide was determined by HPLC or UPLC-MS/MS to estimate the inhibitory effect and inhibition modes of darolutamide on UGTs were evaluated by using the inhibition kinetics experiments. The results showed that 100 μM darolutamide exhibited inhibitory effects on most of the 12 UGTs tested. Inhibition kinetic studies of the enzyme revealed that darolutamide noncompetitively inhibited UGT1A1 and competitively inhibited UGT1A7 and 2B15, with the Ki of 14.75 ± 0.78 μM, 14.05 ± 0.42 μM, and 6.60 ± 0.08 μM, respectively. In particular, it also potently inhibited SN-38, the active metabolite of irinotecan, glucuronidation in HLMs with an IC50 value of 3.84 ± 0.46 μM. In addition, the in vitro-in vivo extrapolation (IVIVE) method was used to quantitatively predict the risk of darolutamide-mediated DDI via inhibiting UGTs. The prediction results showed that darolutamide may increase the risk of DDIs when administered in combination with substrates of UGT1A1, UGT1A7, or UGT2B15. Therefore, the combined administration of darolutamide and drugs metabolized by the above UGTs should be used with caution to avoid the occurrence of potential DDIs.
Collapse
Affiliation(s)
- Shichao Xiao
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Hang Yin
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Xin Lv
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhen Wang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Lili Jiang
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yangliu Xia
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China.
| | - Yong Liu
- School of Chemical Engineering, Ocean and Life Sciences, Dalian University of Technology, Panjin, 124221, China.
| |
Collapse
|
2
|
Wang C, Bai C, Zhang Z, Zhou H, Gao H, Wang S, Yuan Y. UGT1A7 altered HER2-positive breast cancer response to trastuzumab by affecting epithelial-to-mesenchymal transition: A potential biomarker to identify patients resistant to trastuzumab treatment. Cancer Gene Ther 2024; 31:1525-1535. [PMID: 39122832 DOI: 10.1038/s41417-024-00819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
HER2-positive (HER2+) breast cancer accounts for 20-30% of all breast cancers. Although trastuzumab has significantly improved the survival of patients with HER2+ breast cancer, more than 70% of patients develop drug resistance within one year of treatment. Differential-gene-expression analysis of trastuzumab-sensitive and resistant HER2+ breast cancer cell lines from GSE15043 was performed to identify the biomarkers associated with trastuzumab resistance. Differential biomarker expression was confirmed in FFPE tissues collected from clinical HER2+ breast cancer tumor samples that were sensitive or resistant to trastuzumab treatment. UGT1A7, a member of the uronic acid transferase family, was associated with trastuzumab resistance. UGT1A7 expression was downregulated in trastuzumab-resistant tumor tissues and in a cell line that developed trastuzumab resistance (BT474TR). Overexpressing UGT1A7 in BT474TR restored their sensitivity to trastuzumab treatment, whereas downregulating UGT1A7 expression in parental cells led to trastuzumab resistance. Importantly, UGT1A7 localized to the endoplasmic reticulum and altered stress responses. Furthermore, downregulating UGT1A7 expression promoted epithelial-to-mesenchymal transition (EMT) by affecting TWIST, SNAIL, and GRP78 expression and the AMP-activated protein kinase signaling pathway, thus contributing to trastuzumab resistance. This study demonstrated the important role and novel mechanisms of UGT1A7 in tumor responses to trastuzumab. Low UGT1A7 expression plays an important role in EMT and contributes to trastuzumab resistance. UGT1A7 has the potential to be developed as a biomarker for identifying patients who are resistant to trastuzumab treatment.
Collapse
Affiliation(s)
- Cong Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Chenguang Bai
- Department of Radiology, Jiangsu Cancer Hospital &Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhe Zhang
- Department of Pathology, Jiangsu Cancer Hospital &Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Zhou
- Department of Chemotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Siwei Wang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital &Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan Yuan
- Department of Chemotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Zhang Q, Chen Y, Li J, Xia H, Tong Y, Liu Y. Recent Advances in Hepatic Metabolic Regulation by the Nuclear Factor Rev-erbɑ. Curr Drug Metab 2024; 25:2-12. [PMID: 38409696 DOI: 10.2174/0113892002290055240212074758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/28/2024]
Abstract
Rev-erbɑ (NR1D1) is a nuclear receptor superfamily member that plays a vital role in mammalian molecular clocks and metabolism. Rev-erbɑ can regulate the metabolism of drugs and the body's glucose metabolism, lipid metabolism, and adipogenesis. It is even one of the important regulatory factors regulating the occurrence of metabolic diseases (e.g., diabetes, fatty liver). Metabolic enzymes mediate most drug metabolic reactions in the body. Rev-erbɑ has been recognized to regulate drug metabolic enzymes (such as Cyp2b10 and Ugt1a9). Therefore, this paper mainly reviewed that Rev-erbɑ regulates I and II metabolic enzymes in the liver to affect drug pharmacokinetics. The expression of these drug metabolic enzymes (up-regulated or down-regulated) is related to drug exposure and effects/ toxicity. In addition, our discussion extends to Rev-erbɑ regulating some transporters (such as P-gp, Mrp2, and Bcrp), as they also play an essential role in drug metabolism. Finally, we briefly describe the role and mechanism of nuclear receptor Rev-erbɑ in lipid and glucose homeostasis, obesity, and metabolic disorders syndrome. In conclusion, this paper aims to understand better the role and mechanism of Rev-erbɑ in regulating drug metabolism, lipid, glucose homeostasis, obesity, and metabolic disorders syndrome, which explores how to target Rev-erbɑ to guide the design and development of new drugs and provide scientific reference for the molecular mechanism of new drug development, rational drug use, and drug interaction.
Collapse
Affiliation(s)
- Qi Zhang
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yutong Chen
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Jingqi Li
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Haishan Xia
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yongbin Tong
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yuyu Liu
- College of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
4
|
Babayeva M, Loewy ZG. Cannabis Pharmacogenomics: A Path to Personalized Medicine. Curr Issues Mol Biol 2023; 45:3479-3514. [PMID: 37185752 PMCID: PMC10137111 DOI: 10.3390/cimb45040228] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Cannabis and related compounds have created significant research interest as a promising therapy in many disorders. However, the individual therapeutic effects of cannabinoids and the incidence of side effects are still difficult to determine. Pharmacogenomics may provide the answers to many questions and concerns regarding the cannabis/cannabinoid treatment and help us to understand the variability in individual responses and associated risks. Pharmacogenomics research has made meaningful progress in identifying genetic variations that play a critical role in interpatient variability in response to cannabis. This review classifies the current knowledge of pharmacogenomics associated with medical marijuana and related compounds and can assist in improving the outcomes of cannabinoid therapy and to minimize the adverse effects of cannabis use. Specific examples of pharmacogenomics informing pharmacotherapy as a path to personalized medicine are discussed.
Collapse
Affiliation(s)
- Mariana Babayeva
- Department of Biomedical and Pharmaceutical Sciences, Touro College of Pharmacy, New York, NY 10027, USA
| | - Zvi G Loewy
- Department of Biomedical and Pharmaceutical Sciences, Touro College of Pharmacy, New York, NY 10027, USA
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
5
|
Woerdenbag HJ, Olinga P, Kok EA, Brugman DAP, van Ark UF, Ramcharan AS, Lebbink PW, Hoogwater FJH, Knapen DG, de Groot DJA, Nijkamp MW. Potential, Limitations and Risks of Cannabis-Derived Products in Cancer Treatment. Cancers (Basel) 2023; 15:cancers15072119. [PMID: 37046779 PMCID: PMC10093248 DOI: 10.3390/cancers15072119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
The application of cannabis products in oncology receives interest, especially from patients. Despite the plethora of research data available, the added value in curative or palliative cancer care and the possible risks involved are insufficiently proven and therefore a matter of debate. We aim to give a recommendation on the position of cannabis products in clinical oncology by assessing recent literature. Various types of cannabis products, characteristics, quality and pharmacology are discussed. Standardisation is essential for reliable and reproducible quality. The oromucosal/sublingual route of administration is preferred over inhalation and drinking tea. Cannabinoids may inhibit efflux transporters and drug-metabolising enzymes, possibly inducing pharmacokinetic interactions with anticancer drugs being substrates for these proteins. This may enhance the cytostatic effect and/or drug-related adverse effects. Reversely, it may enable dose reduction. Similar interactions are likely with drugs used for symptom management treating pain, nausea, vomiting and anorexia. Cannabis products are usually well tolerated and may improve the quality of life of patients with cancer (although not unambiguously proven). The combination with immunotherapy seems undesirable because of the immunosuppressive action of cannabinoids. Further clinical research is warranted to scientifically support (refraining from) using cannabis products in patients with cancer.
Collapse
Affiliation(s)
- Herman J. Woerdenbag
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Ellen A. Kok
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Donald A. P. Brugman
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Ulrike F. van Ark
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | - Paul W. Lebbink
- Transvaal Apotheek, Kempstraat 113, 2572 GC Den Haag, The Netherlands
| | - Frederik J. H. Hoogwater
- Department of Surgery, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Daan G. Knapen
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Derk Jan A. de Groot
- Department of Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Maarten W. Nijkamp
- Department of Surgery, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
6
|
Tagawa K, Maruo Y, Mimura Y, Ikushiro S. Effects of common genetic variants of human uridine diphosphate glucuronosyltransferase subfamilies on irinotecan glucuronidation. Toxicol Mech Methods 2023; 33:197-205. [PMID: 35930428 DOI: 10.1080/15376516.2022.2109229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The adverse effects (diarrhea and neutropenia) of irinotecan (7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) are associated with genetic variants of uridine diphosphate glucuronosyltransferase 1A subfamilies (UGT1As). UGT1As are enzymes that metabolize the active form of irinotecan, 7-ethyl-10 hydroxycamptothecin (SN-38), by glucuronidation in the liver. They are widely known as predictive factors of severe adverse effects, such as neutropenia and diarrhea. Some studies have suggested that variants of UGT1As affect SN-38 glucuronidation activities, thus exerting severe adverse effects. We aimed to identify UGT1A isoforms that show SN-38 glucuronidation activity and determine the relationship between UGT1A variants and SN-38 glucuronidation in vitro. We found that UGT1A1 and UGT1A6-UGT1A10 displayed SN-38 glucuronidation activity. Among these, UGT1A1 was the most active. Furthermore, the variants of these isoforms showed decreased SN-38 glucuronidation activity. In our study, we compared the different variants of UGT1As, such as UGT1A1.6, UGT1A1.7, UGT1A1.27, UGT1A1.35, UGT1A7.3, UGT1A8.4, UGT1A10M59I, and UGT1A10T202I, to determine the differences in the reduction of glucuronidation. Our study elucidates the relationship between UGT1A variants and the level of glucuronidation associated with each variant. Therefore, testing can be done before the initiation of irinotecan treatment to predict potential toxicities and adverse effects.
Collapse
Affiliation(s)
- Kouji Tagawa
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Yu Mimura
- Department of Pediatrics, Toyosato Hospital, Shiga, Japan
| | - Shinichi Ikushiro
- Department of Biotechnology, Toyama Prefectural University, Toyama, Japan
| |
Collapse
|
7
|
Liu C, Wu K, Li J, Mu X, Gao H, Xu X. Nanoparticle-mediated therapeutic management in cholangiocarcinoma drug targeting: Current progress and future prospects. Biomed Pharmacother 2023; 158:114135. [PMID: 36535198 DOI: 10.1016/j.biopha.2022.114135] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with cholangiocarcinoma (CCA) often have an unfavorable prognosis because of its insidious nature, low resectability rate, and poor response to anticancer drugs and radiotherapy, which makes early detection and treatment difficult. At present, CCA has a five-year overall survival rate (OS) of only 5%, despite advances in therapies. New an increasing number of evidence suggests that nanoplatforms may play a crucial role in enhancing the pharmacological effects and in reducing both short- and long-term side effects of cancer treatment. This document reviews the advantages and shortcomings of nanoparticles such as liposomes, polymeric nanoparticle,inorganic nanoparticle, nano-metals and nano-alloys, carbon dots, nano-micelles, dendrimer, nano-capsule, bio-Nanomaterials in the diagnosis and treatment of CCA and discuss the current challenges in of nanoplatforms for CCA.
Collapse
Affiliation(s)
- Chunkang Liu
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianyang Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xupeng Mu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Huan Gao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Patel P, Xue Z, King KS, Parham L, Ford S, Lou Y, Bakshi KK, Sutton K, Margolis D, Hughes AR, Spreen WR. Evaluation of the effect of UGT1A1 polymorphisms on the pharmacokinetics of oral and long-acting injectable cabotegravir. J Antimicrob Chemother 2021; 75:2240-2248. [PMID: 32361755 PMCID: PMC7366207 DOI: 10.1093/jac/dkaa147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/28/2020] [Accepted: 03/18/2020] [Indexed: 12/31/2022] Open
Abstract
Background Cabotegravir is an HIV integrase inhibitor in clinical development with both oral and long-acting (LA) injectable formulations. Cabotegravir is primarily metabolized by uridine 5′-diphospho-glucuronosyltransferase (UGT) 1A1, a known polymorphic enzyme with functional variants that can affect drug metabolism and exposure. Objectives To investigate the pharmacogenetic effects of the reduced-function alleles UGT1A1*6, UGT1A1*28 and/or UGT1A1*37 on steady-state pharmacokinetics (PK) and safety of oral cabotegravir (30 mg/day) and intramuscular cabotegravir LA (400 mg every 4 weeks or 600 mg every 8 weeks). Methods Plasma cabotegravir PK was assessed in 346 UGT-genotyped participants with and without UGT1A1 functional variants across six studies (four Phase I and two Phase II) of oral cabotegravir, including 215 HIV-infected participants who received oral cabotegravir followed by cabotegravir LA. Changes from baseline in total bilirubin and ALT were assessed in one study (LATTE; NCT01641809). Results Statistically significant (P < 0.05) associations were observed between UGT1A1 genotype and plasma cabotegravir PK parameters, with 28%–50% increases following oral cabotegravir [plasma cabotegravir concentration at the end of the dosing interval (Ctau), 1.50-fold; AUCtau, 1.41-fold; and Cmax, 1.28-fold] and 16%–24% increases following cabotegravir LA administration (48 week Ctau, 1.24-fold; AUCtau, 1.16-fold; and Cmax, 1.18-fold) among those with low-versus-normal genetically predicted UGT1A1 activity. A statistically significant (P < 10−5) association between predicted UGT1A1 activity and maximum change in total bilirubin was also observed (2.45-fold asymptomatic increase for low versus normal) without a corresponding change in ALT. Conclusions This modest increase in oral and parenteral cabotegravir exposure associated with a reduced function of UGT1A1 is not considered clinically relevant based on accumulated safety data; no dose adjustment is required.
Collapse
Affiliation(s)
- Parul Patel
- ViiV Healthcare, Research Triangle Park, NC, USA
| | | | | | | | - Susan Ford
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | - Yu Lou
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | | | | | | | | | | |
Collapse
|
9
|
Effect of UGT1A1, CYP3A and CES Activities on the Pharmacokinetics of Irinotecan and its Metabolites in Patients with UGT1A1 Gene Polymorphisms. Eur J Drug Metab Pharmacokinet 2021; 46:317-324. [PMID: 33619631 DOI: 10.1007/s13318-021-00675-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Irinotecan (CPT-11) is metabolized to an active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase (CES). SN-38 is then converted to the inactive metabolite SN-38 glucuronide (SN-38G) by glucuronosyltransferase 1A1 (UGT1A1). Genetic polymorphisms in UGT1A1 have been associated with altered SN-38 pharmacokinetics, which increase the risk of toxicity in patients. CPT-11 is also converted to 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin (APC) and 7-ethyl-10-(4-amino-1-piperidino) carbonyloxycamptothecin (NPC) by cytochrome P450 3A (CYP3A), and this route also affects the plasma concentration of SN-38. We evaluated the activities of UGT1A1, CYP3A, and CES and the factors affecting the pharmacokinetics of plasma SN-38 in patients with UGT1A1 gene polymorphisms. METHODS Three male patients aged 56, 65, and 49 years were recruited for the analysis. All patients had pancreatic cancer, received FOLFIRINOX, and had UGT1A1*6/*6 (patients 1 and 3) or *6/*28 (patient 2) genetic polymorphisms. The rate constants for evaluating the enzyme activity were determined from the measured plasma concentration of CPT-11 and its metabolites using a two-compartment model by WinNonlin. RESULTS The area under the plasma concentration-time curve (AUC) of SN-38 was patient 1 > patient 2 > patient 3. The rate constants obtained from the model analysis indicated the respective enzyme activities of UGT1A1 (k57), CYP3A (k13 + k19), and CES (k15). The order of values for UGT1A1 activity was patient 2 > patient 3 > patient 1. Since UGT1A1 activity was low in patient 1 with a high AUC of SN-38, it can be said that the increase in plasma concentration was due to a decrease in UGT1A1 activity. Conversely, the order of values for CYP3A and CES activities was patient 3 > patient 1 > patient 2 and patient 2 > patient 1 > patient 3, respectively. Patient 3 had the lowest AUC of SN-38, caused by a lower level of CES activity and increased CYP3A activity. CONCLUSION In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Using pharmacokinetic analysis, it is possible to directly evaluate enzyme activity and consider what kind of enzyme variation causes the increase in the AUC of SN-38.
Collapse
|
10
|
Ingram CJE, Ekong R, Ansari-Pour N, Bradman N, Swallow DM. Group-based pharmacogenetic prediction: is it feasible and do current NHS England ethnic classifications provide appropriate data? THE PHARMACOGENOMICS JOURNAL 2020; 21:47-59. [PMID: 32683419 DOI: 10.1038/s41397-020-0175-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
Inter-individual variation of drug metabolising enzymes (DMEs) leads to variable efficacy of many drugs and even adverse drug responses. Consequently, it would be desirable to test variants of many DMEs before drug treatment. Inter-ethnic differences in frequency mean that the choice of SNPs to test may vary across population groups. Here we examine the utility of testing representative groups as a way of assessing what variants might be tested. We show that publicly available population information is potentially useful for determining loci for pre-treatment genetic testing, and for determining the most prevalent risk haplotypes in defined groups. However, we also show that the NHS England classifications have limitations for grouping for these purposes, in particular for people of African descent. We conclude: (1) genotyping of hospital patients and people from the hospital catchment area confers no advantage over using samples from appropriate existing ethnic group collections or publicly available data, (2) given the current NHS England Black African grouping, a decision as to whether to test, would have to apply to all patients of recent Black African ancestry to cover reported risk alleles and (3) the current scarcity of available genome and drug effect data from Africans is a problem for both testing and treatment decisions.
Collapse
Affiliation(s)
- Catherine J E Ingram
- Research Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Rosemary Ekong
- Research Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Naser Ansari-Pour
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7LF, UK
| | - Neil Bradman
- Henry Stewart Group, 40-41 Museum Street, London, WC1A 1LT, UK
| | - Dallas M Swallow
- Research Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
11
|
Yang L, Jiang X, Yan H, Li Y, Zhen H, Chang B, Kariminia S, Li Q. Irinotecan-containing doublet treatment versus irinotecan monotherapy as second-line choice for advanced gastric cancer. BMC Gastroenterol 2018; 18:43. [PMID: 29609559 PMCID: PMC5879920 DOI: 10.1186/s12876-018-0772-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/26/2018] [Indexed: 12/25/2022] Open
Abstract
Background For patients with advanced gastric cancer (AGC), second-line chemotherapy regimen remains controversial. The efficacy and safety of irinotecan-containing doublet treatment and irinotecan monotherapy were compared in this systematic analysis. Methods A search was conducted on EMBASE and Medline databases. All articles compared irinotecan-containing doublet to irinotecan as second-line chemotherapy for AGC. STATA statistical software (Version 12.0) was used to analyze the data. Results Seven studies, including 905 cases, were included in the analysis. Irinotecan-containing doublet treatment significantly prolonged progression-free survival compared to irinotecan monotherapy (HR = 0.82, 95% CI: 0.70–0.95). However, doublet treatment neither significantly prolong overall survival compared to monotherapy (HR = 0.94, 95% CI: 0.81–1.10), nor did it significantly increase the overall response rates and disease control rates, when compared to monotherapy. In addition, the irinotecan-containing doublet group had an increase in incidences of ≥ Grade 3 neutropenia (RR = 1.23, 95% CI: 1.01–1.51) and anemia (RR = 2.00, 95% CI: 1.37–2.92). Conclusions When compared to irinotecan monotherapy, irinotecan-containing doublet treatment increased progression free survival and was tolerable as a second- line chemotherapy for AGC.
Collapse
|
12
|
Schneider JS, Gasse A, Schürenkamp M, Sibbing U, Banken S, Pfeiffer H, Schürenkamp J, Vennemann M. Multiplex analysis of genetic polymorphisms within UGT1A9, a gene involved in phase II of Δ 9-THC metabolism. Int J Legal Med 2018; 133:365-372. [PMID: 30191314 DOI: 10.1007/s00414-018-1919-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 08/16/2018] [Indexed: 11/30/2022]
Abstract
We present a novel multiplex assay for the simultaneous detection of 12 polymorphisms within the UGT1A9 sequence, which codes for enzymes involved in phase II biotransformation. The assay combines a multiplexed amplification step with single-base extension sequencing. The method described here is fast, cost-effective, and easy-to-use, combining the relevant features of screening methods for research and diagnostics in pharmacogenetics. To validate the assay, we tested reproducibility and sensitivity and analysed allele frequencies of 110 Caucasian individuals. Furthermore, we describe combining genetic information of individuals consuming Cannabis sativa products with respective plasma concentrations of a metabolite.
Collapse
Affiliation(s)
- Julia Sophie Schneider
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany
| | - Angela Gasse
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany
| | - Marianne Schürenkamp
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany
| | - Ursula Sibbing
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany
| | - Sabrina Banken
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany
| | - Heidi Pfeiffer
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany
| | - Jennifer Schürenkamp
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany.
| | - Marielle Vennemann
- Institute of Legal Medicine, University of Münster, Röntgenstraße 23, 48149, Münster, Germany
| |
Collapse
|
13
|
Chen DW, Du Z, Zhang CZ, Zhang WH, Cao YF, Sun HZ, Zhu ZT, Yang K, Liu YZ, Zhao ZW, Fu ZW, Gu WQ, Yu Y, Fang ZZ. The inhibition of UDP-glucuronosyltransferases (UGTs) by tetraiodothyronine (T4) and triiodothyronine (T3). Xenobiotica 2017; 48:250-257. [DOI: 10.1080/00498254.2017.1304593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Da-Wei Chen
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
| | - Zuo Du
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
| | - Chun-Ze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China, and
| | - Wei-Hua Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China, and
| | - Yun-Feng Cao
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China
| | - Hong-Zhi Sun
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China
| | - Zhi-Tu Zhu
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China
| | - Kun Yang
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
| | - Yong-Zhe Liu
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
| | - Ze-Wei Zhao
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
| | - Zhi-Wei Fu
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
| | - Wen-Qing Gu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,
| | - Yang Yu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,
| | - Zhong-Ze Fang
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China,
| |
Collapse
|