Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sook Wah Yee is active.

Publication


Featured researches published by Sook Wah Yee.


Nature Reviews Drug Discovery | 2010

Membrane transporters in drug development

Kathleen M. Giacomini; Shiew Mei Huang; Donald J. Tweedie; Leslie Z. Benet; Kim L. R. Brouwer; Xiaoyan Chu; Amber Dahlin; Raymond Evers; Volker Fischer; Kathleen M. Hillgren; Keith Hoffmaster; Toshihisa Ishikawa; Dietrich Keppler; Richard B. Kim; Caroline A. Lee; Mikko Niemi; Joseph W. Polli; Yuicchi Sugiyama; Peter W. Swaan; Joseph A. Ware; Stephen H. Wright; Sook Wah Yee; Lei Zhang

Membrane transporters can be major determinants of the pharmacokinetic, safety and efficacy profiles of drugs. This presents several key questions for drug development, including which transporters are clinically important in drug absorption and disposition, and which in vitro methods are suitable for studying drug interactions with these transporters. In addition, what criteria should trigger follow-up clinical studies, and which clinical studies should be conducted if needed. In this article, we provide the recommendations of the International Transporter Consortium on these issues, and present decision trees that are intended to help guide clinical studies on the currently recognized most important drug transporter interactions. The recommendations are generally intended to support clinical development and filing of a new drug application. Overall, it is advised that the timing of transporter investigations should be driven by efficacy, safety and clinical trial enrolment questions (for example, exclusion and inclusion criteria), as well as a need for further understanding of the absorption, distribution, metabolism and excretion properties of the drug molecule, and information required for drug labelling.


Clinical Pharmacology & Therapeutics | 2011

Clinical Pharmacogenetics Implementation Consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing.

Mary V. Relling; Gardner Ee; William J. Sandborn; Kjeld Schmiegelow; Ching-Hon Pui; Sook Wah Yee; C.M. Stein; Michelle Whirl Carrillo; William E. Evans; Teri E. Klein

Thiopurine methyltransferase (TPMT) activity exhibits monogenic co‐dominant inheritance, with ethnic differences in the frequency of occurrence of variant alleles. With conventional thiopurine doses, homozygous TPMT‐deficient patients (~1 in 178 to 1 in 3,736 individuals with two nonfunctional TPMT alleles) experience severe myelosuppression, 30–60% of individuals who are heterozygotes (~3–14% of the population) show moderate toxicity, and homozygous wild‐type individuals (~86–97% of the population) show lower active thioguanine nucleolides and less myelosuppression. We provide dosing recommendations (updates at http://www.pharmgkb.org) for azathioprine, mercaptopurine (MP), and thioguanine based on TPMT genotype.


Clinical Pharmacology & Therapeutics | 2013

Clinical Pharmacogenetics Implementation Consortium Guidelines for Thiopurine Methyltransferase Genotype and Thiopurine Dosing: 2013 Update

Mary V. Relling; Gardner Ee; William J. Sandborn; Kjeld Schmiegelow; Ching-Hon Pui; Sook Wah Yee; C.M. Stein; Michelle Whirl Carrillo; William E. Evans; J K Hicks; Matthias Schwab; Teri E. Klein

The Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Thiopurine Methyltransferase Genotype and Thiopurine Dosing was originally published in March 2011. We reviewed recent literature and concluded that although relevant new evidence has been generated, none of the evidence would change the primary dosing recommendations in the original guideline; therefore, the original publication remains clinically current. Up‐to‐date information on thiopurine methyltransferase (TPMT) gene alleles and nomenclature can be found at PharmGKB (http://www.pharmgkb.org).


Pharmacogenetics and Genomics | 2009

Effect of genetic variation in the organic cation transporter 2 on the renal elimination of metformin

Ying Chen; Shuanglian Li; Chaline Brown; Stephen Cheatham; Richard A. Castro; Maya K. Leabman; Thomas J. Urban; Ligong Chen; Sook Wah Yee; Ji Ha Choi; Yong Huang; Claire M. Brett; Esteban G. Burchard; Kathleen M. Giacomini

Objective The goal of this study was to determine the effect of a genetic variant in the organic cation transporter 2 (OCT2), OCT2-808G/T, which results in an amino acid change, A270S, on the pharmacokinetics of the antidiabetic drug, metformin. Methods The uptake of metformin was performed in stably transfected HEK-293 cells expressing the empty vector (MOCK), the reference OCT2-808G, and the variant OCT2-808T. Healthy individuals with known OCT2 genotypes [14 homozygous for the OCT2 reference allele (808G/G) and nine heterozygous for the variant allele (808G/T, *3D)] were recruited to this study. Metformin concentrations in plasma and urine were measured by liquid chromatography–tandem mass spectrometry method. Creatinine levels were also measured in plasma and urine. Pharmacokinetic parameters were evaluated for both the groups. Results We observed that in HEK-293 stably transfected cells, OCT2-808T had a greater capacity to transport metformin than did the reference OCT2. Metformin pharmacokinetics was characterized in 23 healthy volunteers of Caucasian and African-American ancestries. We observed that the renal clearance (CLR) and the net secretion (SrCLR) of metformin were significantly different between the volunteers heterozygous for the variant allele (808G/T), and the volunteers homozygous for the reference allele (808G/G) (P<0.005). Multivariate analysis revealed that OCT2 genotype was a significant predictor of CLR and SrCLR of metformin (P<0.01). Conclusion We conclude that genetic variation in OCT2 plays an important role in the CLR and SrCLR of metformin in healthy volunteers.


Clinical Pharmacology & Therapeutics | 2010

The Role of Organic Anion–Transporting Polypeptides and Their Common Genetic Variants in Mycophenolic Acid Pharmacokinetics

Nicolas Picard; Sook Wah Yee; Jean-Baptiste Woillard; Yvon Lebranchu; Y. Le Meur; Kathleen M. Giacomini; Pierre Marquet

The goal of this study was to determine the roles of the organic anion–transporting polypeptides (OATPs) OATP1A2, OATP1B1, and OATP1B3 and their genetic variants in the pharmacokinetics of the immunosuppressive drug mycophenolate mofetil (MMF). Using OATP‐transfected human embryonic kidney (HEK) cells, we measured the uptake of mycophenolic acid (MPA) and its glucuronide (MPAG). MPAG, but not MPA, significantly accumulated in cells expressing OATP1B3 or OATP1B1 (P < 0.05). The pharmacokinetics of both MPA and MPAG were significantly influenced by the OATP1B3 polymorphism 334T>G/699G>A in 70 renal transplant patients receiving combination treatment of MMF with either tacrolimus or sirolimus, but not in 115 patients receiving MMF and cyclosporine. The decrease in dose‐normalized (dn) MPA exposure and the concomitant increase in the MPAG/MPA metabolic ratio are consistent with reduced enterohepatic cycling in patients carrying the OATP1B3 334G–699A haplotype. Further studies demonstrated that this variant of OATP1B3 exhibited a reduced maximal velocity (Vmax) in transfected HEK cells, thereby providing functional evidence to support our clinical findings.


Nature Reviews Drug Discovery | 2015

SLC transporters as therapeutic targets: emerging opportunities

Lawrence Lin; Sook Wah Yee; Richard B. Kim; Kathleen M. Giacomini

Solute carrier (SLC) transporters — a family of more than 300 membrane-bound proteins that facilitate the transport of a wide array of substrates across biological membranes — have important roles in physiological processes ranging from the cellular uptake of nutrients to the absorption of drugs and other xenobiotics. Several classes of marketed drugs target well-known SLC transporters, such as neurotransmitter transporters, and human genetic studies have provided powerful insight into the roles of more-recently characterized SLC transporters in both rare and common diseases, indicating a wealth of new therapeutic opportunities. This Review summarizes knowledge on the roles of SLC transporters in human disease, describes strategies to target such transporters, and highlights current and investigational drugs that modulate SLC transporters, as well as promising drug targets.


Clinical Pharmacology & Therapeutics | 2013

The effect of novel promoter variants in MATE1 and MATE2 on the pharmacokinetics and pharmacodynamics of metformin.

Sophie L. Stocker; Kari M. Morrissey; Sook Wah Yee; Richard A. Castro; Lu Xu; Amber Dahlin; Andrea H. Ramirez; Dan M. Roden; Russ A. Wilke; Catherine A. McCarty; Robert L. Davis; Claire M. Brett; Kathleen M. Giacomini

Interindividual variation in response to metformin, first‐line therapy for type 2 diabetes, is substantial. Given that transporters are determinants of metformin pharmacokinetics, we examined the effects of promoter variants in both multidrug and toxin extrusion protein 1 (MATE1) (g.–66T→C, rs2252281) and MATE2 (g.–130G→A, rs12943590) on variation in metformin disposition and response. The pharmacokinetics and glucose‐lowering effects of metformin were assessed in healthy volunteers (n = 57) receiving metformin. The renal and secretory clearances of metformin were higher (22% and 26%, respectively) in carriers of variant MATE2 who were also MATE1 reference (P < 0.05). Both MATE genotypes were associated with altered post‐metformin glucose tolerance, with variant carriers of MATE1 and MATE2 having an enhanced (P < 0.01) and reduced (P < 0.05) response, respectively. Consistent with these results, patients with diabetes (n = 145) carrying the MATE1 variant showed enhanced metformin response. These findings suggest that promoter variants of MATE1 and MATE2 are important determinants of metformin disposition and response in healthy volunteers and diabetic patients.


Clinical Pharmacology & Therapeutics | 2013

International Transporter Consortium Commentary on Clinically Important Transporter Polymorphisms

Kathleen M. Giacomini; P. V. Balimane; Sk Cho; M. Eadon; Timi Edeki; Kathleen M. Hillgren; Huang Sm; Yuichi Sugiyama; Dietmar Weitz; Yujia Wen; Cindy Xia; Sook Wah Yee; H. Zimdahl; Mikko Niemi

This Commentary focuses on genetic polymorphisms in membrane transporters. We present two polymorphisms for which there is a compelling body of literature supporting their clinical relevance: OATP1B1 (c.521T>C, p.V174A, rs4149056) and BCRP (c.421C>A, p.Q141K, rs2231142). The clinical evidence demonstrating their role in variation in pharmacokinetics and pharmacodynamics is described along with their allele frequencies in ethnic populations. Recommendations for incorporating studies of transporter polymorphisms in drug development are provided, along with the regulatory implications.


Proceedings of the National Academy of Sciences of the United States of America | 2014

OCT1 is a high-capacity thiamine transporter that regulates hepatic steatosis and is a target of metformin

Ligong Chen; Yan Shu; Xiaomin Liang; Eugene C. Chen; Sook Wah Yee; Arik A. Zur; Shuanglian Li; Lu Xu; Kayvan R. Keshari; Michael J. Lin; Huan-Chieh Chien; Youcai Zhang; Kari M. Morrissey; Jason Liu; Jonathan M. Ostrem; Noah S. Younger; John Kurhanewicz; Kevan M. Shokat; Kaveh Ashrafi; Kathleen M. Giacomini

Significance This manuscript describes a previously unidentified mechanism for organic cation transporter 1 (OCT1), the major hepatic metformin transporter, in hepatic steatosis. Here we show that OCT1, long thought to function primarily as a transporter for drugs, functions as a major thiamine transporter in the liver, which has profound implications in cellular metabolism. Collectively, our results point to an important role of thiamine (through OCT1) in hepatic steatosis and suggest that the modulation of thiamine disposition by metformin may contribute to its pharmacologic effects. Organic cation transporter 1, OCT1 (SLC22A1), is the major hepatic uptake transporter for metformin, the most prescribed antidiabetic drug. However, its endogenous role is poorly understood. Here we show that similar to metformin treatment, loss of Oct1 caused an increase in the ratio of AMP to ATP, activated the energy sensor AMP-activated kinase (AMPK), and substantially reduced triglyceride (TG) levels in livers from healthy and leptin-deficient mice. Conversely, livers of human OCT1 transgenic mice fed high-fat diets were enlarged with high TG levels. Metabolomic and isotopic uptake methods identified thiamine as a principal endogenous substrate of OCT1. Thiamine deficiency enhanced the phosphorylation of AMPK and its downstream target, acetyl-CoA carboxylase. Metformin and the biguanide analog, phenformin, competitively inhibited OCT1-mediated thiamine uptake. Acute administration of metformin to wild-type mice reduced intestinal accumulation of thiamine. These findings suggest that OCT1 plays a role in hepatic steatosis through modulation of energy status. The studies implicate OCT1 as well as metformin in thiamine disposition, suggesting an intriguing and parallel mechanism for metformin and its major hepatic transporter in metabolic function.


Clinical Pharmacology & Therapeutics | 2011

A common 5′-UTR variant in MATE2-K is associated with poor response to metformin

Choi Jh; Sook Wah Yee; Andrea H. Ramirez; Kari M. Morrissey; Jang Gh; Joski Pj; Joel Mefford; Stephanie Hesselson; Avner Schlessinger; Gerard Jenkins; Richard A. Castro; Susan J. Johns; Douglas Stryke; Andrej Sali; Thomas E. Ferrin; John S. Witte; Pui-Yan Kwok; Dan M. Roden; Russell A. Wilke; Catherine A. McCarty; Robert L. Davis; Kathleen M. Giacomini

Multidrug and toxin extrusion 2 (MATE2‐K (SLC47A2)), a polyspecific organic cation exporter, facilitates the renal elimination of the antidiabetes drug metformin. In this study, we characterized genetic variants of MATE2‐K, determined their association with metformin response, and elucidated their impact by means of a comparative protein structure model. Four nonsynonymous variants and four variants in the MATE2‐K basal promoter region were identified from ethnically diverse populations. Two nonsynonymous variants—c.485C>T and c.1177G>A—were shown to be associated with significantly lower metformin uptake and reduction in protein expression levels. MATE2‐K basal promoter haplotypes containing the most common variant, g.−130G>A (>26% allele frequency), were associated with a significant increase in luciferase activities and reduced binding to the transcriptional repressor myeloid zinc finger 1 (MZF‐1). Patients with diabetes who were homozygous for g.−130A had a significantly poorer response to metformin treatment, assessed as relative change in glycated hemoglobin (HbA1c) (−0.027 (−0.076, 0.033)), as compared with carriers of the reference allele, g.−130G (−0.15 (−0.17, −0.13)) (P = 0.002). Our study showed that MATE2‐K plays a role in the antidiabetes response to metformin.

Collaboration


Dive into the Sook Wah Yee's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ligong Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaomin Liang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susan J. Johns

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eugene C. Chen

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge