1
|
Peng H, Yuan B, Mao J, Wang J, Luo S. Efficacy of Letrozole Combined with Urinary Gonadotropin for Ovulation Induction in Endocrine Abnormal Infertility Patients: A Retrospective Single-Center, Case-Control Study. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5363754. [PMID: 35860181 PMCID: PMC9293500 DOI: 10.1155/2022/5363754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 12/04/2022]
Abstract
Aims To ask lots of questions and try to find the truth about the medicine-based effectiveness of letrozole (LE) combined with human menopausal gonadotropin (HMG) in the treatment of inability to have children crops patients with endocrine (things that are different from what is usually expected and the effect on ovulation-related chemicals produced by the body). Materials and Methods A total of 160 unable to have children crops patients with endocrine things that are different from what is usually expected who were treated in our hospital from March 2019 to March 2022 were selected as the subjects of this look at how things were in the past study and were divided into instance of watching, making a statement group was treated with human menopausal gonadotropin on the basis of the control group. The differences in serum related to the process of making children, chemical produced by the body levels, ovarian function, and ovulation induction effect between the two groups were watched and compared. Results After treatment, LH, FSH, PRL, and E2 in the observation group were better than those in the control group. The ovarian volume, follicle size, follicle diameter, and endometrial thickness of the two groups of patients were significantly improved, and the observation group was better than the control group. Significance is P < 0.05. After treatment, the ovarian volume, follicle size, follicle diameter, and endometrial thickness in the two groups were significantly improved, and the observation group was better than the control group, and the difference was statistically significant (P < 0.05). The ovulation rate, pregnancy rate, singleton pregnancy rate, and multiple pregnancy rate of the observation group were higher than those of the control group, and the difference was statistically significant by the chi-square test (P < 0.05). Conclusion Letrozole can promote the improvement of sex hormones in infertile patients. After being combined with human menopausal gonadotropin treatment, the follicle development and ovulation of patients are significantly improved, and infertility is improved to a certain extent. It has a certain reference value in the clinical treatment of endocrine abnormal infertility.
Collapse
Affiliation(s)
- Hao Peng
- Department of Obstetrics and Gynecology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, 435000 Hubei Province, China
| | - Ben Yuan
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, 435000 Hubei Province, China
| | - Junbiao Mao
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, 430000 Hubei Province, China
| | - Junling Wang
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, 435000 Hubei Province, China
| | - Shuhong Luo
- Department of Reproductive Medicine, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, 435000 Hubei Province, China
| |
Collapse
|
2
|
Kell DB. The Transporter-Mediated Cellular Uptake and Efflux of Pharmaceutical Drugs and Biotechnology Products: How and Why Phospholipid Bilayer Transport Is Negligible in Real Biomembranes. Molecules 2021; 26:5629. [PMID: 34577099 PMCID: PMC8470029 DOI: 10.3390/molecules26185629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Over the years, my colleagues and I have come to realise that the likelihood of pharmaceutical drugs being able to diffuse through whatever unhindered phospholipid bilayer may exist in intact biological membranes in vivo is vanishingly low. This is because (i) most real biomembranes are mostly protein, not lipid, (ii) unlike purely lipid bilayers that can form transient aqueous channels, the high concentrations of proteins serve to stop such activity, (iii) natural evolution long ago selected against transport methods that just let any undesirable products enter a cell, (iv) transporters have now been identified for all kinds of molecules (even water) that were once thought not to require them, (v) many experiments show a massive variation in the uptake of drugs between different cells, tissues, and organisms, that cannot be explained if lipid bilayer transport is significant or if efflux were the only differentiator, and (vi) many experiments that manipulate the expression level of individual transporters as an independent variable demonstrate their role in drug and nutrient uptake (including in cytotoxicity or adverse drug reactions). This makes such transporters valuable both as a means of targeting drugs (not least anti-infectives) to selected cells or tissues and also as drug targets. The same considerations apply to the exploitation of substrate uptake and product efflux transporters in biotechnology. We are also beginning to recognise that transporters are more promiscuous, and antiporter activity is much more widespread, than had been realised, and that such processes are adaptive (i.e., were selected by natural evolution). The purpose of the present review is to summarise the above, and to rehearse and update readers on recent developments. These developments lead us to retain and indeed to strengthen our contention that for transmembrane pharmaceutical drug transport "phospholipid bilayer transport is negligible".
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK;
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs Lyngby, Denmark
- Mellizyme Biotechnology Ltd., IC1, Liverpool Science Park, Mount Pleasant, Liverpool L3 5TF, UK
| |
Collapse
|
3
|
Rossi R, Ciofalo M. Current Advances in the Synthesis and Biological Evaluation of Pharmacologically Relevant 1,2,4,5-Tetrasubstituted-1H-Imidazole Derivatives. CURR ORG CHEM 2019. [DOI: 10.2174/1385272823666191014154129] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
:
In recent years, the synthesis and evaluation of the
biological properties of 1,2,4,5-tetrasubstituted-1H-imidazole
derivatives have been the subject of a large number of studies
by academia and industry. In these studies it has been shown
that this large and highly differentiated class of heteroarene
derivatives includes high valuable compounds having important
biological and pharmacological properties such as
antibacterial, antifungal, anthelmintic, anti-inflammatory, anticancer,
antiviral, antihypertensive, cholesterol-lowering, antifibrotic,
antiuricemic, antidiabetic, antileishmanial and antiulcer
activities.
:
The present review with 411 references, in which we focused on the literature data published mainly from 2011
to 2017, aims to update the readers on the recent developments on the synthesis and biological evaluation of
pharmacologically relevant 1,2,4,5-tetrasubstituted-1H-imidazole derivatives with an emphasis on their different
molecular targets and their potential use as drugs to treat various types of diseases. Reference was also
made to substantial literature data acquired before 2011 in this burgeoning research area.
Collapse
Affiliation(s)
- Renzo Rossi
- Dipartimento di Chimica e Chimica Industriale, University of Pisa - via Moruzzi, 3, I-56124 Pisa, Italy
| | - Maurizio Ciofalo
- Dipartimento di Scienze Agrarie, Alimentari e Forestali, University of Palermo - Viale delle Scienze, Edificio 4, I-90128 Palermo, Italy
| |
Collapse
|
4
|
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: 1.9] [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
|
5
|
Wu Y, Xiong FJ, Chen FE. Stereoselective synthesis of 3-hydroxy-3-methylglutaryl–coenzyme A reductase inhibitors. Tetrahedron 2015. [DOI: 10.1016/j.tet.2015.07.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
6
|
Cooper ME, Regnell SE. The hepatic cannabinoid 1 receptor as a modulator of hepatic energy state and food intake. Br J Clin Pharmacol 2015; 77:21-30. [PMID: 23452341 DOI: 10.1111/bcp.12102] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 02/12/2013] [Indexed: 12/11/2022] Open
Abstract
The cannabinoid 1 receptor (CB1R) has a well-established role in appetite regulation. Central CB1R antagonists, notably rimonabant, induced weight loss and improved the metabolic profile in obese individuals, but were discontinued due to psychiatric side-effects. The CB1R is also expressed peripherally, where its effects include promotion of liver fat accumulation, which consumes ATP. Type 2 diabetes in obese subjects is linked to excess liver fat, whilst there is a negative correlation between hepatic ATP content and insulin resistance. A decreased hepatic ATP/AMP ratio increases food intake by signals via the vagus nerve to the brain. The hepatic cannabinoid system is highly upregulated in obesity, and the effects of hepatic CB1R activation include increased activity of lipogenic and gluconeogenic transcription factors. Thus, blockade of hepatic CB1Rs could contribute significantly to the weight-reducing and insulin-sensitizing effects of CB1R antagonists. Additionally, upregulation of the hepatic CB1R may contribute to chronic liver inflammation, fibrosis and cirrhosis from causes including obesity, alcoholism and viral hepatitis. Peripheral CB1R antagonists induce weight loss and metabolic improvements in obese rodents; however, as there is evidence that hepatic CB1Rs are predominately intracellular, due to high intrinsic clearance, many drugs may not effectively block these receptors and therefore have limited efficacy. Hepatoselective CB1R antagonists may be effective at reducing hepatic steatosis, insulin resistance and bodyweight in obese, diabetic patients, with far fewer side-effects than first-generation CB1R antagonists. Additionally, such compounds may be effective in treating inflammatory liver disease, such as non-alcoholic steatohepatitis, reducing the likelihood of disease progression to cirrhosis or cancer.
Collapse
|
7
|
Kell DB, Oliver SG. How drugs get into cells: tested and testable predictions to help discriminate between transporter-mediated uptake and lipoidal bilayer diffusion. Front Pharmacol 2014; 5:231. [PMID: 25400580 PMCID: PMC4215795 DOI: 10.3389/fphar.2014.00231] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/29/2014] [Indexed: 12/12/2022] Open
Abstract
One approach to experimental science involves creating hypotheses, then testing them by varying one or more independent variables, and assessing the effects of this variation on the processes of interest. We use this strategy to compare the intellectual status and available evidence for two models or views of mechanisms of transmembrane drug transport into intact biological cells. One (BDII) asserts that lipoidal phospholipid Bilayer Diffusion Is Important, while a second (PBIN) proposes that in normal intact cells Phospholipid Bilayer diffusion Is Negligible (i.e., may be neglected quantitatively), because evolution selected against it, and with transmembrane drug transport being effected by genetically encoded proteinaceous carriers or pores, whose “natural” biological roles, and substrates are based in intermediary metabolism. Despite a recent review elsewhere, we can find no evidence able to support BDII as we can find no experiments in intact cells in which phospholipid bilayer diffusion was either varied independently or measured directly (although there are many papers where it was inferred by seeing a covariation of other dependent variables). By contrast, we find an abundance of evidence showing cases in which changes in the activities of named and genetically identified transporters led to measurable changes in the rate or extent of drug uptake. PBIN also has considerable predictive power, and accounts readily for the large differences in drug uptake between tissues, cells and species, in accounting for the metabolite-likeness of marketed drugs, in pharmacogenomics, and in providing a straightforward explanation for the late-stage appearance of toxicity and of lack of efficacy during drug discovery programmes despite macroscopically adequate pharmacokinetics. Consequently, the view that Phospholipid Bilayer diffusion Is Negligible (PBIN) provides a starting hypothesis for assessing cellular drug uptake that is much better supported by the available evidence, and is both more productive and more predictive.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry, The University of Manchester Manchester, UK ; Manchester Institute of Biotechnology, The University of Manchester Manchester, UK
| | - Stephen G Oliver
- Department of Biochemistry, University of Cambridge Cambridge, UK ; Cambridge Systems Biology Centre, University of Cambridge Cambridge, UK
| |
Collapse
|
8
|
Wang Z, Cheng L, Kai Z, Wu F, Liu Z, Cai M. Molecular modeling studies of atorvastatin analogues as HMGR inhibitors using 3D-QSAR, molecular docking and molecular dynamics simulations. Bioorg Med Chem Lett 2014; 24:3869-76. [DOI: 10.1016/j.bmcl.2014.06.055] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/17/2014] [Accepted: 06/19/2014] [Indexed: 12/12/2022]
|
9
|
Ichikawa M, Ohtsuka M, Ohki H, Ota M, Haginoya N, Itoh M, Shibata Y, Sugita K, Ishigai Y, Terayama K, Kanda A, Usui H. Discovery of DF-461, a Potent Squalene Synthase Inhibitor. ACS Med Chem Lett 2013; 4:932-6. [PMID: 24900587 DOI: 10.1021/ml400151c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 09/04/2013] [Indexed: 11/30/2022] Open
Abstract
We report the development of a new trifluoromethyltriazolobenzoxazepine series of squalene synthase inhibitors. Structure-activity studies and pharmacokinetics optimization on this series led to the identification of compound 23 (DF-461), which exhibited potent squalene synthase inhibitory activity, high hepatic selectivity, excellent rat hepatic cholesterol synthesis inhibitory activity, and plasma lipid lowering efficacy in nonrodent repeated dose studies.
Collapse
Affiliation(s)
- Masanori Ichikawa
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masami Ohtsuka
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hitoshi Ohki
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masahiro Ota
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Noriyasu Haginoya
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masao Itoh
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yoshihiro Shibata
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Kazuyuki Sugita
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yutaka Ishigai
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Koji Terayama
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Akira Kanda
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Hiroyuki Usui
- R&D Division, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| |
Collapse
|
10
|
Pfefferkorn JA. Strategies for the design of hepatoselective glucokinase activators to treat type 2 diabetes. Expert Opin Drug Discov 2013; 8:319-30. [PMID: 23289965 DOI: 10.1517/17460441.2013.748744] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Type 2 diabetes mellitus (T2DM) represents a rapidly expanding healthcare challenge. There is a significant need for novel therapies to help patients achieve and maintain glycemic control in order to avoid the long-term microvascular and macrovascular complications associated with the disease. Small molecule allosteric activators of the glucokinase enzyme, an important regulator of glucose homeostasis, have emerged as a potential new class of therapeutics. Glucokinase activators have been shown to effectively lower fasting and postprandial glucose in T2DM patients; however, hypoglycemia emerged as a potential risk limiting their therapeutic potential. To mitigate this risk, recent efforts have focused on the design of liver-specific activators that seek to normalize hepatic glucose uptake and production without potentiating glucose-stimulated insulin secretion. AREAS COVERED The article reviews the various drug discovery strategies that have emerged for the development of candidates that selectively activate glucokinase in the liver. Literature from 2000 to 2012 is surveyed including scientific publications, patent applications, conferences and clinical trials. EXPERT OPINION Liver selective agents have proven to be an effective strategy for mitigating the hypoglycemia risk that has been historically associated with this mechanism. The ultimate therapeutic potential of this approach will depend on the results of longer patient studies which are currently being conducted with several clinical candidates. The discovery of these liver-specific activators has highlighted several challenges in the design of tissue-selective therapeutics, which will need to be overcome in the future.
Collapse
Affiliation(s)
- Jeffrey A Pfefferkorn
- Cardiovascular, Metabolic & Endocrine Disease Research Unit, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, MA 02139, USA.
| |
Collapse
|
11
|
Joharapurkar AA, Dhote VV, Jain MR. Selective Thyromimetics Using Receptor and Tissue Selectivity Approaches: Prospects for Dyslipidemia. J Med Chem 2012; 55:5649-75. [DOI: 10.1021/jm2004706] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Amit A. Joharapurkar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Sarkhej Bavla NH 8A, Moraiya,
Ahmedabad 382210, India
| | - Vipin V. Dhote
- Department of Pharmacology and Toxicology, Zydus Research Centre, Sarkhej Bavla NH 8A, Moraiya,
Ahmedabad 382210, India
| | - Mukul R. Jain
- Department of Pharmacology and Toxicology, Zydus Research Centre, Sarkhej Bavla NH 8A, Moraiya,
Ahmedabad 382210, India
| |
Collapse
|
12
|
Lai Y, Varma M, Feng B, Stephens JC, Kimoto E, El-Kattan A, Ichikawa K, Kikkawa H, Ono C, Suzuki A, Suzuki M, Yamamoto Y, Tremaine L. Impact of drug transporter pharmacogenomics on pharmacokinetic and pharmacodynamic variability - considerations for drug development. Expert Opin Drug Metab Toxicol 2012; 8:723-43. [PMID: 22509796 DOI: 10.1517/17425255.2012.678048] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Drug transporter proteins are expressed on the cell membrane, regulating substrate exposure in systemic circulation and/or peripheral tissues. Genetic polymorphism of drug transporter genes encoding these proteins could alter the functional activity and/or protein expression, having effects on absorption, distribution, metabolism and excretion (ADME), efficacy and adverse effects. AREAS COVERED The authors provide the reader with an overview of the pharmacogenetics (PGx) of 12 membrane transporters. The clinical literature is summarized as to the quantitative significance on pharmacokinetics (PK) and implications on pharmacodynamics (PD) and adverse effects, due to transporter influence on intracellular drug concentrations. EXPERT OPINION Unlike polymorphisms for cytochrome P450s (CYPs) resulting in large magnitude of PK variation, genetic mutations for membrane transporters are typically less than threefold alteration in systemic PK for drugs with a few exceptions. However, substantially greater changes in intracellular drug levels may result. We are aware of 1880 exome variants in 12 of the best-studied transporters to date, and nearly 40% of these change the amino acid. However, the functional consequences of most of these variants remain to be determined, and have only been empirically evaluated for a handful. To the extent that genetic polymorphisms impact ADME, it is a variable that will contribute to ethnic differences due to substantial frequency differences for the known variants.
Collapse
Affiliation(s)
- Yurong Lai
- Pfizer Worldwide Research and Development, Department of Pharmacokinetics, Dynamics and Metabolism, Groton, CT 06340, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Explication of interactions between HMGCR isoform 2 and various statins through In silico modeling and docking. Comput Biol Med 2012; 42:156-63. [DOI: 10.1016/j.compbiomed.2011.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 10/28/2011] [Accepted: 11/07/2011] [Indexed: 11/24/2022]
|
14
|
Pfefferkorn JA, Guzman-Perez A, Litchfield J, Aiello R, Treadway JL, Pettersen J, Minich ML, Filipski KJ, Jones CS, Tu M, Aspnes G, Risley H, Bian J, Stevens BD, Bourassa P, D’Aquila T, Baker L, Barucci N, Robertson AS, Bourbonais F, Derksen DR, MacDougall M, Cabrera O, Chen J, Lapworth AL, Landro JA, Zavadoski WJ, Atkinson K, Haddish-Berhane N, Tan B, Yao L, Kosa RE, Varma MV, Feng B, Duignan DB, El-Kattan A, Murdande S, Liu S, Ammirati M, Knafels J, DaSilva-Jardine P, Sweet L, Liras S, Rolph TP. Discovery of (S)-6-(3-Cyclopentyl-2-(4-(trifluoromethyl)-1H-imidazol-1-yl)propanamido)nicotinic Acid as a Hepatoselective Glucokinase Activator Clinical Candidate for Treating Type 2 Diabetes Mellitus. J Med Chem 2012; 55:1318-33. [DOI: 10.1021/jm2014887] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Jeffrey A. Pfefferkorn
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Angel Guzman-Perez
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - John Litchfield
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Robert Aiello
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Judith L. Treadway
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - John Pettersen
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Martha L. Minich
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin J. Filipski
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Christopher S. Jones
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Meihua Tu
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Gary Aspnes
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Hud Risley
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jianwei Bian
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Benjamin D. Stevens
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Patricia Bourassa
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Theresa D’Aquila
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Levenia Baker
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Nicole Barucci
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Alan S. Robertson
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Francis Bourbonais
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - David R. Derksen
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Margit MacDougall
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Over Cabrera
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Jing Chen
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amanda Lee Lapworth
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - James A. Landro
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - William J. Zavadoski
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Karen Atkinson
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Nahor Haddish-Berhane
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Beijing Tan
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Lili Yao
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Rachel E. Kosa
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Manthena V. Varma
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Bo Feng
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - David B. Duignan
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ayman El-Kattan
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sharad Murdande
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Shenping Liu
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Mark Ammirati
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - John Knafels
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Paul DaSilva-Jardine
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Laurel Sweet
- Groton Laboratories, Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Spiros Liras
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Timothy P. Rolph
- Cambridge Laboratories, Pfizer Worldwide Research & Development, 620 Memorial Drive, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Fenner KS, Jones HM, Ullah M, Kempshall S, Dickins M, Lai Y, Morgan P, Barton HA. The evolution of the OATP hepatic uptake transport protein family in DMPK sciences: from obscure liver transporters to key determinants of hepatobiliary clearance. Xenobiotica 2011; 42:28-45. [PMID: 22077101 DOI: 10.3109/00498254.2011.626464] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Over the last two decades the impact on drug pharmacokinetics of the organic anion transporting polypeptides (OATPs: OATP-1B1, 1B3 and 2B1), expressed on the sinusoidal membrane of the hepatocyte, has been increasingly recognized. OATP-mediated uptake into the hepatocyte coupled with subsequent excretion into bile via efflux proteins, such as MRP2, is often referred to as hepatobiliary excretion. OATP transporter proteins can impact some drugs in several ways including pharmacokinetic variability, pharmacodynamic response and drug-drug interactions (DDIs). The impact of transporter mediated hepatic clearance is illustrated with case examples, from the literature and also from the Pfizer portfolio. The currently available in vitro techniques to study the hepatic transporter proteins involved in the hepatobiliary clearance of drugs are reviewed herein along with recent advances in using these in vitro data to predict the human clearance of compounds recognized by hepatic uptake transporters.
Collapse
Affiliation(s)
- Katherine S Fenner
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Sandwich, Kent, UK.
| | | | | | | | | | | | | | | |
Collapse
|