1
|
Li W, Vazvaei-Smith F, Dear G, Boer J, Cuyckens F, Fraier D, Liang Y, Lu D, Mangus H, Moliner P, Pedersen ML, Romeo AA, Spracklin DK, Wagner DS, Winter S, Xu XS. Metabolite Bioanalysis in Drug Development: Recommendations from the IQ Consortium Metabolite Bioanalysis Working Group. Clin Pharmacol Ther 2024; 115:939-953. [PMID: 38073140 DOI: 10.1002/cpt.3144] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/05/2023] [Indexed: 03/13/2024]
Abstract
The intent of this perspective is to share the recommendations of the International Consortium for Innovation and Quality in Pharmaceutical Development Metabolite Bioanalysis Working Group on the fit-for-purpose metabolite bioanalysis in support of drug development and registration. This report summarizes the considerations for the trigger, timing, and rigor of bioanalysis in the various assessments to address unique challenges due to metabolites, with respect to efficacy and safety, which may arise during drug development from investigational new drug (IND) enabling studies, and phase I, phase II, and phase III clinical trials to regulatory submission. The recommended approaches ensure that important drug metabolites are identified in a timely manner and properly characterized for efficient drug development.
Collapse
Affiliation(s)
- Wenkui Li
- Pharmacokinetic Sciences, Novartis Biomedical Research, East Hanover, New Jersey, USA
| | - Faye Vazvaei-Smith
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Gordon Dear
- Drug Metabolism and Pharmacokinetics, GSK, Ware, UK
| | - Jason Boer
- Drug Metabolism and Pharmacokinetics, Incyte Corporation, Wilmington, Delaware, USA
| | - Filip Cuyckens
- Drug Metabolism and Pharmacokinetics, Janssen R & D, Beerse, Belgium
| | - Daniela Fraier
- Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yuexia Liang
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Ding Lu
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Inc., Boston, Massachusetts, USA
| | - Heidi Mangus
- Drug Metabolism and Pharmacokinetics, Agios Pharmaceuticals Inc., Cambridge, Massachusetts, USA
| | - Patricia Moliner
- Enzymology and Metabolism, Department of Translational Medicine and Early Development, Sanofi, Montpellier, Occitanie, France
| | - Mette Lund Pedersen
- DMPK, Research and Early Development, CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Andrea A Romeo
- Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Douglas K Spracklin
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., Groton, Connecticut, USA
| | - David S Wagner
- Drug Metabolism and Disposition, AbbVie, North Chicago, Illinois, USA
| | - Serge Winter
- Pharmacokinetic Sciences, Novartis Biomedical Research, Basel, Switzerland
| | - Xiaohui Sophia Xu
- Clinical Bioanalysis, Translation Medicine, Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| |
Collapse
|
2
|
Gao Q, Zhang W, Li T, Yang G, Zhu W, Chen N, Jin H. The efficacy and safety of glucokinase activators for the treatment of type-2 diabetes mellitus: A meta-analysis. Medicine (Baltimore) 2021; 100:e27476. [PMID: 34622877 PMCID: PMC8500571 DOI: 10.1097/md.0000000000027476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/22/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Glucokinase activators (GKAs) are a novel family of glucose-lowering agents used for the treatment of type-2 diabetes mellitus. Treatment with different GKAs has been shown to reduce blood glucose levels in these patients. We compared the efficacy/safety of GKAs in patients with type-2 diabetes mellitus through a meta-analysis. METHODS We searched the PubMed, Excerpt Medica Database, and Cochrane Central Register of Controlled Trials databases for articles published before December 30, 2020. We computed the weighted mean difference (WMD) and 95% confidence interval (CI) for the change from baseline to the study endpoint for GKA versus placebo treatments. RESULTS A total of 4 articles (5 studies) were included in the meta-analysis. GKAs were associated with reductions in glycated hemoglobin levels from baseline (WMD, -0.3%; 95% CI, -0.466% to -0.134%). No significant difference between GKA and placebo treatment was observed in the results of fasting plasma glucose levels from baseline (WMD 0.013 mmol/L; 95% CI, -0.304-0.33 mmol/L). A significantly higher change in 2-hour postprandial plasma glucose (2-h PPG) levels (WMD -2.434 mmol/L; 95% CI, -3.304 to -1.564 mmol/L) was observed following GKA than placebo treatment. GKAs were associated with a higher prevalence of causing hypoglycemic events than placebo treatment (risk difference [RD], 0.06; 95% CI 0.013-0.106). GKAs had no association with the risk of developing adverse effects (RD, 0.038; 95% CI, -0.03-0.106) and serious adverse events (RD, 0.01; 95% CI, -0.004-0.023). CONCLUSIONS GKAs were more effective for postprandial blood glucose control. However, these agents showed a significantly high risk of causing hypoglycemia. PROSPERO REGISTRATION NUMBER CRD42021220364.
Collapse
|
3
|
Grewal AS, Lather V, Charaya N, Sharma N, Singh S, Kairys V. Recent Developments in Medicinal Chemistry of Allosteric Activators of Human Glucokinase for Type 2 Diabetes Mellitus Therapeutics. Curr Pharm Des 2020; 26:2510-2552. [PMID: 32286938 DOI: 10.2174/1381612826666200414163148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glucokinase (GK), a cytoplasmic enzyme catalyzes the metabolism of glucose to glucose- 6-phosphate with the help of ATP and aids in the controlling of blood glucose levels within the normal range in humans. In pancreatic β-cells, it plays a chief role by controlling the glucose-stimulated secretion of insulin and in liver hepatocyte cells, it controls the metabolism of carbohydrates. GK acts as a promising drug target for the pharmacological treatment of patients with type 2 diabetes mellitus (T2DM) as it plays an important role in the control of carbohydrate metabolism. METHODS Data used for this review was based on the search from several science databases as well as various patent databases. The main data search terms used were allosteric GK activators, diabetes mellitus, type 2 diabetes, glucokinase, glucokinase activators and human glucokinase. RESULTS This article discusses an overview of T2DM, the biology of GK, the role of GK in T2DM, recent updates in the development of small molecule GK activators reported in recent literature, mechanism of action of GK activators and their clinical status. CONCLUSION GK activators are the novel class of pharmacological agents that enhance the catalytic activity of GK enzyme and display their antihyperglycemic effects. Broad diversity of chemical entities including benzamide analogues, carboxamides, acrylamides, benzimidazoles, quinazolines, thiazoles, pyrimidines, pyridines, orotic acid amides, amino acid derivatives, amino phosphates and urea derivatives have been synthesized in past two decades as potent allosteric activators of GK. Presently, the pharmaceutical companies and researchers are focusing on the design and development of liver-selective GK activators for preventing the possible adverse effects associated with GK activators for the long-term treatment of T2DM.
Collapse
Affiliation(s)
- Ajmer S Grewal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Neha Charaya
- Jan Nayak Ch. Devi Lal Memorial College of Pharmacy, Haryana, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Visvaldas Kairys
- Department of Bioinformatics, Institute of Biotechnology, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
4
|
Bergman A, Bi Y, Mathialagan S, Litchfield J, Kazierad DJ, Pfefferkorn JA, Varma MV. Effect of Hepatic Organic Anion‐Transporting Polypeptide 1B Inhibition and Chronic Kidney Disease on the Pharmacokinetics of a Liver‐Targeted Glucokinase Activator: A Model‐Based Evaluation. Clin Pharmacol Ther 2019; 106:792-802. [DOI: 10.1002/cpt.1419] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/22/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Arthur Bergman
- Clinical PharmacologyWorldwide Research and DevelopmentPfizer Inc. Groton Connecticut USA
| | - Yi‐an Bi
- Medicine DesignWorldwide Research and DevelopmentPfizer Inc. Groton Connecticut USA
| | - Sumathy Mathialagan
- Medicine DesignWorldwide Research and DevelopmentPfizer Inc. Groton Connecticut USA
| | - John Litchfield
- Worldwide Research and DevelopmentPfizer Inc. Cambridge Massachusetts USA
| | - David J. Kazierad
- Worldwide Research and DevelopmentPfizer Inc. Cambridge Massachusetts USA
| | | | - Manthena V.S. Varma
- Medicine DesignWorldwide Research and DevelopmentPfizer Inc. Groton Connecticut USA
| |
Collapse
|
5
|
Li Z, Fisher C, Gardner I, Ghosh A, Litchfield J, Maurer TS. Modeling Exposure to Understand and Predict Kidney Injury. Semin Nephrol 2019; 39:176-189. [DOI: 10.1016/j.semnephrol.2018.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
6
|
Sharma R, Bergman A, Litchfield J, Atkinson K, Kazierad DJ, Kalgutkar AS. Metabolism and excretion of ( S)-6-(3-cyclopentyl-2-(4-trifluoromethyl)-1 H-imidazol-1-yl)propanamido)nicotinic acid (PF-04991532), a hepatoselective glucokinase activator, in humans: confirmation of the MIST potential noted in first-in-Human metabolite scouting studies. Xenobiotica 2019; 49:1447-1457. [PMID: 30747552 DOI: 10.1080/00498254.2019.1581960] [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: 10/27/2022]
Abstract
1. The absorption, metabolism, and excretion of a single oral 450-mg dose of [14C]-(S)-6-(3-cyclopentyl-2-(4-trifluoromethyl)-1H-imidazol-1-yl)propanamido)nicotinic acid (PF-04991532), a hepatoselective glucokinase activator, was investigated in humans. Mass balance was achieved with ∼94.6% of the administered dose recovered in urine and feces. The total administered radioactivity excreted in feces and urine was 70.6% and 24.1%, respectively. Unchanged PF-04991532 collectively accounted for ∼47.2% of the dose excreted in feces and urine, suggestive of moderate metabolic elimination in humans. 2. The biotransformation pathways involved acyl glucuronidation (M1), amide bond hydrolysis (M3), and CYP3A4-mediated oxidative metabolism on the cyclopentyl ring in PF-04991532 yielding monohydroxylated isomers (M2a-d). Unchanged PF-04991532 was the major circulating component (64.4% of total radioactivity) whereas M2a-d collectively represented 28.9% of the total plasma radioactivity. 3. Metabolites M2a-d were not detected systemically in rats and dogs, the preclinical species for the toxicological evaluation of PF-04991532. In contrast, cynomologus monkeys dosed orally with unlabeled PF-04991532 revealed M2a-d in circulation, whose UV abundance was comparable to the profile in humans. This observation suggested that monkeys could potentially serve as a non-rodent alternative for studying the toxicity of PF-04991532 and its metabolites M2a-d. 4. The present results are in excellent agreement with our previously generated metabolite scouting data, which provided preliminary evidence for the disproportionate metabolism of PF-04991532 in humans.
Collapse
Affiliation(s)
- Raman Sharma
- Medicine Design Pfizer Worldwide Research and Development , Groton , CT , USA
| | - Arthur Bergman
- Clinical Pharmacology/Pharmacometrics Pfizer Worldwide Research and Development , Groton , CT , USA
| | - John Litchfield
- Medicine Design Pfizer Worldwide Research and Development , Cambridge , MA , USA
| | - Karen Atkinson
- Medicine Design Pfizer Worldwide Research and Development , Groton , CT , USA
| | - David J Kazierad
- Clinical Sciences Pfizer Worldwide Research and Development , Cambridge , MA , USA
| | - Amit S Kalgutkar
- Medicine Design Pfizer Worldwide Research and Development , Cambridge , MA , USA
| |
Collapse
|
7
|
Xi XN, Liu N, Wang QQ, Wu HT, He HB, Wang LL, Zhang TJ, Sun L, Yin Z, Chen Y, Lu YX. Pharmacokinetics, tissue distribution and excretion of ACT001 in Sprague-Dawley rats and metabolism of ACT001. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1104:29-39. [DOI: 10.1016/j.jchromb.2018.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 11/15/2022]
|
8
|
Zhu XX, Zhu DL, Li XY, Li YL, Jin XW, Hu TX, Zhao Y, Li YG, Zhao GY, Ren S, Zhang Y, Ding YH, Chen L. Dorzagliatin (HMS5552), a novel dual-acting glucokinase activator, improves glycaemic control and pancreatic β-cell function in patients with type 2 diabetes: A 28-day treatment study using biomarker-guided patient selection. Diabetes Obes Metab 2018; 20:2113-2120. [PMID: 29707866 DOI: 10.1111/dom.13338] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/19/2018] [Accepted: 04/24/2018] [Indexed: 01/04/2023]
Abstract
AIMS To investigate the pharmacokinetics and pharmacodynamics of a dual-acting glucokinase activator, dorzagliatin, and its safety, tolerability and effect on pancreatic β-cell function in Chinese patients with type 2 diabetes (T2D). MATERIALS AND METHODS A total of 24 T2D patients were selected, utilizing a set of predefined clinical biomarkers, and were randomized to receive dorzagliatin 75 mg twice or once daily (BID, QD respectively) for 28 days. Changes in HbA1c and glycaemic parameters from baseline to Day 28 were assessed. In addition, changes in β-cell function from baseline to Day 32 were evaluated. RESULTS Significant reductions in HbA1c were observed in both regimens on Day 28 (-0.79%, 75 mg BID; -1.22%, 75 mg QD). Similar trends were found in the following parameters, including reductions from baseline in fasting plasma glucose by 1.20 mmol/L and 1.51 mmol/L, in 2-hour postprandial glucose by 2.48 mmol/L and 5.03 mmol/L, and in glucose AUC0-24 by 18.59% and 20.98%, for the BID and QD groups, respectively. Both regimens resulted in improvement in β-cell function as measured by steady state HOMA 2 parameter, %B, which increased by 36.31% and 40.59%, and by dynamic state parameter, ΔC30 /ΔG30 , which increased by 24.66% and 167.67%, for the BID and QD groups, respectively. Dorzagliatin was well tolerated in both regimens, with good pharmacokinetic profiles. CONCLUSIONS Dorzagliatin treatment for 28 days in Chinese T2D patients, selected according to predefined biomarkers, resulted in significant improvement in β-cell function and glycaemic control. The safety and pharmacokinetic profile of dorzagliatin supports a subsequent Phase II trial design and continued clinical development.
Collapse
Affiliation(s)
- Xiao-Xue Zhu
- Phase I Clinical Trial Unit, The First Hospital of Jilin University, Changchun, China
| | - Da-Long Zhu
- Department of Endocrinology and Metabolism, Nanjing Drum Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiao-Ying Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ya-Lin Li
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Xiao-Wei Jin
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Tian-Xin Hu
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Yu Zhao
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Yong-Guo Li
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Gui-Yu Zhao
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Shuang Ren
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Yi Zhang
- Hua Medicine (Shanghai) Limited, Shanghai, China
| | - Yan-Hua Ding
- Phase I Clinical Trial Unit, The First Hospital of Jilin University, Changchun, China
| | - Li Chen
- Hua Medicine (Shanghai) Limited, Shanghai, China
| |
Collapse
|
9
|
Tong Z, Atsriku C, Yerramilli U, Wang X, Li Y, Reyes J, Fan B, Yang H, Hoffmann M, Surapaneni S. Absorption, distribution, metabolism and excretion of an isocitrate dehydrogenase-2 inhibitor enasidenib in rats and humans. Xenobiotica 2018; 49:200-210. [PMID: 29320949 DOI: 10.1080/00498254.2018.1425511] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
1. The absorption, distribution, metabolism and excretion of enasidenib were studied following a single oral dose of [14C]enasidenib to rats (10 mg/kg; 100 μCi/kg) and healthy volunteers (100 mg; 318 nCi). 2. Enasidenib was readily absorbed, extensively metabolized and primarily eliminated via the hepatobiliary pathway. Enasidenib-derived radioactivity was widely distributed in rats. Excretion of radioactivity was approximately 95-99% of the dose from rats in 168 h post-dose and 82.4% from human volunteers in 504 h post-dose. In rat bile, approximately 35-42% of the administered dose was recovered, with less than 5% of the dose excreted as the parent drug. Renal elimination was a minor pathway, with <12% of the dose excreted in rat urine and <10% of the dose excreted in human urine. 3. Enasidenib was the prominent radioactive component in rat and human systemic circulation. Enasidenib was extensively metabolized in rats and human volunteers through N-dealkylation, oxidation, direct glucuronidation and combinations of these pathways. Glucuronidation was the major metabolic pathway in rats while N-dealkylation was the prominent metabolic pathway in human volunteers. All human metabolites were detected in rats.
Collapse
Affiliation(s)
- Zeen Tong
- a Nonclinical Development , Celgene Corporation , Summit , NJ , USA
| | | | - Usha Yerramilli
- a Nonclinical Development , Celgene Corporation , Summit , NJ , USA
| | - Xiaomin Wang
- a Nonclinical Development , Celgene Corporation , Summit , NJ , USA
| | - Yan Li
- b Clinical Pharmacology , Celgene Corporation , Summit , NJ , USA , and
| | - Josephine Reyes
- b Clinical Pharmacology , Celgene Corporation , Summit , NJ , USA , and
| | - Bin Fan
- c DMPK/Clinical Pharmacology , Agios Pharmaceutical , Cambridge , MA , USA
| | - Hua Yang
- c DMPK/Clinical Pharmacology , Agios Pharmaceutical , Cambridge , MA , USA
| | - Matthew Hoffmann
- a Nonclinical Development , Celgene Corporation , Summit , NJ , USA
| | | |
Collapse
|
10
|
Tong Z, Atsriku C, Yerramilli U, Wang X, Nissel J, Li Y, Surapaneni S. Absorption, distribution, metabolism, and excretion of mTOR kinase inhibitor CC-223 in rats, dogs, and humans. Xenobiotica 2017; 49:43-53. [DOI: 10.1080/00498254.2017.1413718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Zeen Tong
- Nonclinical Development, Celgene Corporation, Summit, NJ, USA and
| | | | - Usha Yerramilli
- Nonclinical Development, Celgene Corporation, Summit, NJ, USA and
| | - Xiaomin Wang
- Nonclinical Development, Celgene Corporation, Summit, NJ, USA and
| | - Jim Nissel
- Clinical Pharmacology, Celgene Corporation, Summit, NJ, USA
| | - Yan Li
- Clinical Pharmacology, Celgene Corporation, Summit, NJ, USA
| | | |
Collapse
|
11
|
Luffer-Atlas D, Atrakchi A. A decade of drug metabolite safety testing: industry and regulatory shared learning. Expert Opin Drug Metab Toxicol 2017; 13:897-900. [DOI: 10.1080/17425255.2017.1364362] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Debra Luffer-Atlas
- Drug Disposition and Toxicology, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Aisar Atrakchi
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
12
|
Grixti JM, O'Hagan S, Day PJ, Kell DB. Enhancing Drug Efficacy and Therapeutic Index through Cheminformatics-Based Selection of Small Molecule Binary Weapons That Improve Transporter-Mediated Targeting: A Cytotoxicity System Based on Gemcitabine. Front Pharmacol 2017; 8:155. [PMID: 28396636 PMCID: PMC5366350 DOI: 10.3389/fphar.2017.00155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/10/2017] [Indexed: 12/23/2022] Open
Abstract
The transport of drug molecules is mainly determined by the distribution of influx and efflux transporters for which they are substrates. To enable tissue targeting, we sought to develop the idea that we might affect the transporter-mediated disposition of small-molecule drugs via the addition of a second small molecule that of itself had no inhibitory pharmacological effect but that influenced the expression of transporters for the primary drug. We refer to this as a “binary weapon” strategy. The experimental system tested the ability of a molecule that on its own had no cytotoxic effect to increase the toxicity of the nucleoside analog gemcitabine to Panc1 pancreatic cancer cells. An initial phenotypic screen of a 500-member polar drug (fragment) library yielded three “hits.” The structures of 20 of the other 2,000 members of this library suite had a Tanimoto similarity greater than 0.7 to those of the initial hits, and each was itself a hit (the cheminformatics thus providing for a massive enrichment). We chose the top six representatives for further study. They fell into three clusters whose members bore reasonable structural similarities to each other (two were in fact isomers), lending strength to the self-consistency of both our conceptual and experimental strategies. Existing literature had suggested that indole-3-carbinol might play a similar role to that of our fragments, but in our hands it was without effect; nor was it structurally similar to any of our hits. As there was no evidence that the fragments could affect toxicity directly, we looked for effects on transporter transcript levels. In our hands, only the ENT1-3 uptake and ABCC2,3,4,5, and 10 efflux transporters displayed measurable transcripts in Panc1 cultures, along with a ribonucleoside reductase RRM1 known to affect gemcitabine toxicity. Very strikingly, the addition of gemcitabine alone increased the expression of the transcript for ABCC2 (MRP2) by more than 12-fold, and that of RRM1 by more than fourfold, and each of the fragment “hits” served to reverse this. However, an inhibitor of ABCC2 was without significant effect, implying that RRM1 was possibly the more significant player. These effects were somewhat selective for Panc cells. It seems, therefore, that while the effects we measured were here mediated more by efflux than influx transporters, and potentially by other means, the binary weapon idea is hereby fully confirmed: it is indeed possible to find molecules that manipulate the expression of transporters that are involved in the bioactivity of a pharmaceutical drug. This opens up an entirely new area, that of chemical genomics-based drug targeting.
Collapse
Affiliation(s)
- Justine M Grixti
- Faculty of Biology, Medicine and Health, University of ManchesterManchester, UK; Manchester Institute of Biotechnology, University of ManchesterManchester, UK
| | - Steve O'Hagan
- Manchester Institute of Biotechnology, University of ManchesterManchester, UK; School of Chemistry, University of ManchesterManchester, UK; Centre for Synthetic Biology of Fine and Speciality Chemicals, University of ManchesterManchester, UK
| | - Philip J Day
- Faculty of Biology, Medicine and Health, University of ManchesterManchester, UK; Manchester Institute of Biotechnology, University of ManchesterManchester, UK
| | - Douglas B Kell
- Manchester Institute of Biotechnology, University of ManchesterManchester, UK; School of Chemistry, University of ManchesterManchester, UK; Centre for Synthetic Biology of Fine and Speciality Chemicals, University of ManchesterManchester, UK
| |
Collapse
|
13
|
Rines AK, Sharabi K, Tavares CDJ, Puigserver P. Targeting hepatic glucose metabolism in the treatment of type 2 diabetes. Nat Rev Drug Discov 2016; 15:786-804. [PMID: 27516169 DOI: 10.1038/nrd.2016.151] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is characterized by the dysregulation of glucose homeostasis, resulting in hyperglycaemia. Although current diabetes treatments have exhibited some success in lowering blood glucose levels, their effect is not always sustained and their use may be associated with undesirable side effects, such as hypoglycaemia. Novel antidiabetic drugs, which may be used in combination with existing therapies, are therefore needed. The potential of specifically targeting the liver to normalize blood glucose levels has not been fully exploited. Here, we review the molecular mechanisms controlling hepatic gluconeogenesis and glycogen storage, and assess the prospect of therapeutically targeting associated pathways to treat type 2 diabetes.
Collapse
Affiliation(s)
- Amy K Rines
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Clint D J Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
14
|
Kamimura H, Ito S. Assessment of chimeric mice with humanized livers in new drug development: generation of pharmacokinetics, metabolism and toxicity data for selecting the final candidate compound. Xenobiotica 2015; 46:557-69. [DOI: 10.3109/00498254.2015.1091113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|