385 results on '"Kathleen M Giacomini"'
Search Results
2. Organic cation transporter 3 (Oct3) is a distinct catecholamines clearance route in adipocytes mediating the beiging of white adipose tissue.
- Author
-
Wenxin Song, Qi Luo, Yuping Zhang, Linkang Zhou, Ye Liu, Zhilong Ma, Jianan Guo, Yuedong Huang, Lili Cheng, Ziyi Meng, Zicheng Li, Bin Zhang, Siqi Li, Sook Wah Yee, Hao Fan, Peng Li, Kathleen M Giacomini, and Ligong Chen
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Beiging of white adipose tissue (WAT) is a particularly appealing target for therapeutics in the treatment of metabolic diseases through norepinephrine (NE)-mediated signaling pathways. Although previous studies report NE clearance mechanisms via SLC6A2 on sympathetic neurons or proinflammatory macrophages in adipose tissues (ATs), the low catecholamine clearance capacity of SLC6A2 may limit the cleaning efficiency. Here, we report that mouse organic cation transporter 3 (Oct3; Slc22a3) is highly expressed in WAT and displays the greatest uptake rate of NE as a selective non-neural route of NE clearance in white adipocytes, which differs from other known routes such as adjacent neurons or macrophages. We further show that adipocytes express high levels of NE degradation enzymes Maoa, Maob, and Comt, providing the molecular basis on NE clearance by adipocytes together with its reuptake transporter Oct3. Under NE administration, ablation of Oct3 induces higher body temperature, thermogenesis, and lipolysis compared with littermate controls. After prolonged cold challenge, inguinal WAT (ingWAT) in adipose-specific Oct3-deficient mice shows much stronger browning characteristics and significantly elevated expression of thermogenic and mitochondrial biogenesis genes than in littermate controls, and this response involves enhanced β-adrenergic receptor (β-AR)/protein kinase A (PKA)/cyclic adenosine monophosphate (cAMP)-responsive element binding protein (Creb) pathway activation. Glycolytic genes are reprogrammed to significantly higher levels to compensate for the loss of ATP production in adipose-specific Oct3 knockout (KO) mice, indicating the fundamental role of glucose metabolism during beiging. Inhibition of β-AR largely abolishes the higher lipolytic and thermogenic activities in Oct3-deficient ingWAT, indicating the NE overload in the vicinity of adipocytes in Oct3 KO adipocytes. Of note, reduced functional alleles in human OCT3 are also identified to be associated with increased basal metabolic rate (BMR). Collectively, our results demonstrate that Oct3 governs β-AR activity as a NE recycling transporter in white adipocytes, offering potential therapeutic applications for metabolic disorders.
- Published
- 2019
- Full Text
- View/download PDF
3. Organic cation transporter 1 (OCT1) modulates multiple cardiometabolic traits through effects on hepatic thiamine content.
- Author
-
Xiaomin Liang, Sook Wah Yee, Huan-Chieh Chien, Eugene C Chen, Qi Luo, Ling Zou, Meiling Piao, Arias Mifune, Ligong Chen, Meredith E Calvert, Sarah King, Frode Norheim, Janna Abad, Ronald M Krauss, and Kathleen M Giacomini
- Subjects
Biology (General) ,QH301-705.5 - Abstract
A constellation of metabolic disorders, including obesity, dysregulated lipids, and elevations in blood glucose levels, has been associated with cardiovascular disease and diabetes. Analysis of data from recently published genome-wide association studies (GWAS) demonstrated that reduced-function polymorphisms in the organic cation transporter, OCT1 (SLC22A1), are significantly associated with higher total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride (TG) levels and an increased risk for type 2 diabetes mellitus, yet the mechanism linking OCT1 to these metabolic traits remains puzzling. Here, we show that OCT1, widely characterized as a drug transporter, plays a key role in modulating hepatic glucose and lipid metabolism, potentially by mediating thiamine (vitamin B1) uptake and hence its levels in the liver. Deletion of Oct1 in mice resulted in reduced activity of thiamine-dependent enzymes, including pyruvate dehydrogenase (PDH), which disrupted the hepatic glucose-fatty acid cycle and shifted the source of energy production from glucose to fatty acids, leading to a reduction in glucose utilization, increased gluconeogenesis, and altered lipid metabolism. In turn, these effects resulted in increased total body adiposity and systemic levels of glucose and lipids. Importantly, wild-type mice on thiamine deficient diets (TDs) exhibited impaired glucose metabolism that phenocopied Oct1 deficient mice. Collectively, our study reveals a critical role of hepatic thiamine deficiency through OCT1 deficiency in promoting the metabolic inflexibility that leads to the pathogenesis of cardiometabolic disease.
- Published
- 2018
- Full Text
- View/download PDF
4. Genomic Characterization of Metformin Hepatic Response.
- Author
-
Marcelo R Luizon, Walter L Eckalbar, Yao Wang, Stacy L Jones, Robin P Smith, Megan Laurance, Lawrence Lin, Paul J Gallins, Amy S Etheridge, Fred Wright, Yihui Zhou, Cliona Molony, Federico Innocenti, Sook Wah Yee, Kathleen M Giacomini, and Nadav Ahituv
- Subjects
Genetics ,QH426-470 - Abstract
Metformin is used as a first-line therapy for type 2 diabetes (T2D) and prescribed for numerous other diseases. However, its mechanism of action in the liver has yet to be characterized in a systematic manner. To comprehensively identify genes and regulatory elements associated with metformin treatment, we carried out RNA-seq and ChIP-seq (H3K27ac, H3K27me3) on primary human hepatocytes from the same donor treated with vehicle control, metformin or metformin and compound C, an AMP-activated protein kinase (AMPK) inhibitor (allowing to identify AMPK-independent pathways). We identified thousands of metformin responsive AMPK-dependent and AMPK-independent differentially expressed genes and regulatory elements. We functionally validated several elements for metformin-induced promoter and enhancer activity. These include an enhancer in an ataxia telangiectasia mutated (ATM) intron that has SNPs in linkage disequilibrium with a metformin treatment response GWAS lead SNP (rs11212617) that showed increased enhancer activity for the associated haplotype. Expression quantitative trait locus (eQTL) liver analysis and CRISPR activation suggest that this enhancer could be regulating ATM, which has a known role in AMPK activation, and potentially also EXPH5 and DDX10, its neighboring genes. Using ChIP-seq and siRNA knockdown, we further show that activating transcription factor 3 (ATF3), our top metformin upregulated AMPK-dependent gene, could have an important role in gluconeogenesis repression. Our findings provide a genome-wide representation of metformin hepatic response, highlight important sequences that could be associated with interindividual variability in glycemic response to metformin and identify novel T2D treatment candidates.
- Published
- 2016
- Full Text
- View/download PDF
5. Illuminating the function of the orphan transporter, SLC22A10, in humans and other primates
- Author
-
Sook Wah Yee, Luis Ferrández-Peral, Pol Alentorn-Moron, Claudia Fontsere, Merve Ceylan, Megan L. Koleske, Niklas Handin, Virginia M. Artegoitia, Giovanni Lara, Huan-Chieh Chien, Xujia Zhou, Jacques Dainat, Arthur Zalevsky, Andrej Sali, Colin M. Brand, Finn D. Wolfreys, Jia Yang, Jason E. Gestwicki, John A. Capra, Per Artursson, John W. Newman, Tomàs Marquès-Bonet, and Kathleen M. Giacomini
- Subjects
Science - Abstract
Abstract SLC22A10 is an orphan transporter with unknown substrates and function. The goal of this study is to elucidate its substrate specificity and functional characteristics. In contrast to orthologs from great apes, human SLC22A10, tagged with green fluorescent protein, is not expressed on the plasma membrane. Cells expressing great ape SLC22A10 orthologs exhibit significant accumulation of estradiol-17β-glucuronide, unlike those expressing human SLC22A10. Sequence alignments reveal a proline at position 220 in humans, which is a leucine in great apes. Replacing proline with leucine in SLC22A10-P220L restores plasma membrane localization and uptake function. Neanderthal and Denisovan genomes show proline at position 220, akin to modern humans, indicating functional loss during hominin evolution. Human SLC22A10 is a unitary pseudogene due to a fixed missense mutation, P220, while in great apes, its orthologs transport sex steroid conjugates. Characterizing SLC22A10 across species sheds light on its biological role, influencing organism development and steroid homeostasis.
- Published
- 2024
- Full Text
- View/download PDF
6. Maximum likelihood estimation of renal transporter ontogeny profiles for pediatric PBPK modeling
- Author
-
J. Porter Hunt, Samuel Dubinsky, Autumn M. McKnite, Kit Wun Kathy Cheung, Bianca D. vanGroen, Kathleen M. Giacomini, Saskia N. deWildt, Andrea N. Edginton, and Kevin M. Watt
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Optimal treatment of infants with many renally cleared drugs must account for maturational differences in renal transporter (RT) activity. Pediatric physiologically‐based pharmacokinetic (PBPK) models may incorporate RT activity, but this requires ontogeny profiles for RT activity in children, especially neonates, to predict drug disposition. Therefore, RT expression measurements from human kidney postmortem cortical tissue samples were normalized to represent a fraction of mature RT activity. Using these data, maximum likelihood estimated the distributions of RT activity across the pediatric age spectrum, including preterm and term neonates. PBPK models of four RT substrates (acyclovir, ciprofloxacin, furosemide, and meropenem) were evaluated with and without ontogeny profiles using average fold error (AFE), absolute average fold error (AAFE), and proportion of observations within the 5–95% prediction interval. Novel maximum likelihood profiles estimated ontogeny distributions for the following RT: OAT1, OAT3, OCT2, P‐gp, URAT1, BCRP, MATE1, MRP2, MRP4, and MATE‐2 K. Profiles for OAT3, P‐gp, and MATE1 improved infant furosemide and neonate meropenem PBPK model AFE from 0.08 to 0.70 and 0.53 to 1.34 and model AAFE from 12.08 to 1.44 and 2.09 to 1.36, respectively, and improved the percent of data within the 5–95% prediction interval from 48% to 98% for neonatal ciprofloxacin simulations, respectively. Even after accounting for other critical population‐specific maturational differences, novel RT ontogeny profiles substantially improved neonatal PBPK model performance, providing validated estimates of maturational differences in RT activity for optimal dosing in children.
- Published
- 2024
- Full Text
- View/download PDF
7. Genome-wide discovery of drug-dependent human liver regulatory elements.
- Author
-
Robin P Smith, Walter L Eckalbar, Kari M Morrissey, Marcelo R Luizon, Thomas J Hoffmann, Xuefeng Sun, Stacy L Jones, Shelley Force Aldred, Anuradha Ramamoorthy, Zeruesenay Desta, Yunlong Liu, Todd C Skaar, Nathan D Trinklein, Kathleen M Giacomini, and Nadav Ahituv
- Subjects
Genetics ,QH426-470 - Abstract
Inter-individual variation in gene regulatory elements is hypothesized to play a causative role in adverse drug reactions and reduced drug activity. However, relatively little is known about the location and function of drug-dependent elements. To uncover drug-associated elements in a genome-wide manner, we performed RNA-seq and ChIP-seq using antibodies against the pregnane X receptor (PXR) and three active regulatory marks (p300, H3K4me1, H3K27ac) on primary human hepatocytes treated with rifampin or vehicle control. Rifampin and PXR were chosen since they are part of the CYP3A4 pathway, which is known to account for the metabolism of more than 50% of all prescribed drugs. We selected 227 proximal promoters for genes with rifampin-dependent expression or nearby PXR/p300 occupancy sites and assayed their ability to induce luciferase in rifampin-treated HepG2 cells, finding only 10 (4.4%) that exhibited drug-dependent activity. As this result suggested a role for distal enhancer modules, we searched more broadly to identify 1,297 genomic regions bearing a conditional PXR occupancy as well as all three active regulatory marks. These regions are enriched near genes that function in the metabolism of xenobiotics, specifically members of the cytochrome P450 family. We performed enhancer assays in rifampin-treated HepG2 cells for 42 of these sequences as well as 7 sequences that overlap linkage-disequilibrium blocks defined by lead SNPs from pharmacogenomic GWAS studies, revealing 15/42 and 4/7 to be functional enhancers, respectively. A common African haplotype in one of these enhancers in the GSTA locus was found to exhibit potential rifampin hypersensitivity. Combined, our results further suggest that enhancers are the predominant targets of rifampin-induced PXR activation, provide a genome-wide catalog of PXR targets and serve as a model for the identification of drug-responsive regulatory elements.
- Published
- 2014
- Full Text
- View/download PDF
8. Genetic variation in the proximal promoter of ABC and SLC superfamilies: liver and kidney specific expression and promoter activity predict variation.
- Author
-
Stephanie E Hesselson, Pär Matsson, James E Shima, Hisayo Fukushima, Sook Wah Yee, Yuya Kobayashi, Jason M Gow, Connie Ha, Benjamin Ma, Annie Poon, Susan J Johns, Doug Stryke, Richard A Castro, Harunobu Tahara, Ji Ha Choi, Ligong Chen, Nicolas Picard, Elin Sjödin, Maarke J E Roelofs, Thomas E Ferrin, Richard Myers, Deanna L Kroetz, Pui-Yan Kwok, and Kathleen M Giacomini
- Subjects
Medicine ,Science - Abstract
Membrane transporters play crucial roles in the cellular uptake and efflux of an array of small molecules including nutrients, environmental toxins, and many clinically used drugs. We hypothesized that common genetic variation in the proximal promoter regions of transporter genes contribute to observed variation in drug response. A total of 579 polymorphisms were identified in the proximal promoters (-250 to +50 bp) and flanking 5' sequence of 107 transporters in the ATP Binding Cassette (ABC) and Solute Carrier (SLC) superfamilies in 272 DNA samples from ethnically diverse populations. Many transporter promoters contained multiple common polymorphisms. Using a sliding window analysis, we observed that, on average, nucleotide diversity (pi) was lowest at approximately 300 bp upstream of the transcription start site, suggesting that this region may harbor important functional elements. The proximal promoters of transporters that were highly expressed in the liver had greater nucleotide diversity than those that were highly expressed in the kidney consistent with greater negative selective pressure on the promoters of kidney transporters. Twenty-one promoters were evaluated for activity using reporter assays. Greater nucleotide diversity was observed in promoters with strong activity compared to promoters with weak activity, suggesting that weak promoters are under more negative selective pressure than promoters with high activity. Collectively, these results suggest that the proximal promoter region of membrane transporters is rich in variation and that variants in these regions may play a role in interindividual variation in drug disposition and response.
- Published
- 2009
- Full Text
- View/download PDF
9. Effect of Antioxidants in Medicinal Products on Intestinal Drug Transporters
- Author
-
Chetan P. Kulkarni, Jia Yang, Megan L. Koleske, Giovanni Lara, Khondoker Alam, Andre Raw, Bhagwant Rege, Liang Zhao, Dongmei Lu, Lei Zhang, Lawrence X. Yu, Robert A. Lionberger, Kathleen M. Giacomini, Deanna L. Kroetz, and Sook Wah Yee
- Subjects
nitrosamine ,antioxidants ,intestinal transporters ,OATP2B1 ,BCRP ,P-gp ,Pharmacy and materia medica ,RS1-441 - Abstract
The presence of mutagenic and carcinogenic N-nitrosamine impurities in medicinal products poses a safety risk. While incorporating antioxidants in formulations is a potential mitigation strategy, concerns arise regarding their interference with drug absorption by inhibiting intestinal drug transporters. Our study screened thirty antioxidants for inhibitory effects on key intestinal transporters—OATP2B1, P-gp, and BCRP in HEK-293 cells (OATP2B1) or membrane vesicles (P-gp, BCRP) using 3H-estrone sulfate, 3H-N-methyl quinidine, and 3H-CCK8 as substrates, respectively. The screen identified that butylated hydroxyanisole (BHA) and carnosic acid inhibited all three transporters (OATP2B1, P-gp, and BCRP), while ascorbyl palmitate (AP) inhibited OATP2B1 by more than 50%. BHA had IC50 values of 71 ± 20 µM, 206 ± 14 µM, and 182 ± 49 µM for OATP2B1, BCRP, and P-gp, respectively. AP exhibited IC50 values of 23 ± 10 µM for OATP2B1. The potency of AP and BHA was tested with valsartan, an OATP2B1 substrate, and revealed IC50 values of 26 ± 17 µM and 19 ± 11 µM, respectively, in HEK-293-OATP2B1 cells. Comparing IC50 values of AP and BHA with estimated intestinal concentrations suggests an unlikely inhibition of intestinal transporters at clinical concentrations of drugs formulated with antioxidants.
- Published
- 2024
- Full Text
- View/download PDF
10. GenEpi: gene-based epistasis discovery using machine learning
- Author
-
Yu-Chuan Chang, June-Tai Wu, Ming-Yi Hong, Yi-An Tung, Ping-Han Hsieh, Sook Wah Yee, Kathleen M. Giacomini, Yen-Jen Oyang, Chien-Yu Chen, and for the Alzheimer’s Disease Neuroimaging Initiative
- Subjects
GWAS ,Epistasis ,Machine learning ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background Genome-wide association studies (GWAS) provide a powerful means to identify associations between genetic variants and phenotypes. However, GWAS techniques for detecting epistasis, the interactions between genetic variants associated with phenotypes, are still limited. We believe that developing an efficient and effective GWAS method to detect epistasis will be a key for discovering sophisticated pathogenesis, which is especially important for complex diseases such as Alzheimer’s disease (AD). Results In this regard, this study presents GenEpi, a computational package to uncover epistasis associated with phenotypes by the proposed machine learning approach. GenEpi identifies both within-gene and cross-gene epistasis through a two-stage modeling workflow. In both stages, GenEpi adopts two-element combinatorial encoding when producing features and constructs the prediction models by L1-regularized regression with stability selection. The simulated data showed that GenEpi outperforms other widely-used methods on detecting the ground-truth epistasis. As real data is concerned, this study uses AD as an example to reveal the capability of GenEpi in finding disease-related variants and variant interactions that show both biological meanings and predictive power. Conclusions The results on simulation data and AD demonstrated that GenEpi has the ability to detect the epistasis associated with phenotypes effectively and efficiently. The released package can be generalized to largely facilitate the studies of many complex diseases in the near future.
- Published
- 2020
- Full Text
- View/download PDF
11. Organic Anion Transporter Polypeptide 1B1 Polymorphism Modulates the Extent of Drug–Drug Interaction and Associated Biomarker Levels in Healthy Volunteers
- Author
-
Sook Wah Yee, Marilyn M. Giacomini, Hong Shen, W. Griffith Humphreys, Howard Horng, William Brian, Yurong Lai, Deanna L. Kroetz, and Kathleen M. Giacomini
- Subjects
Therapeutics. Pharmacology ,RM1-950 ,Public aspects of medicine ,RA1-1270 - Abstract
Understanding transporter‐mediated drug–drug interactions is an integral part of risk assessment in drug development. Recent studies support the use of hexadecanedioate (HDA), tetradecanedioate (TDA), coproporphyrin (CP)‐I, and CP‐III as clinical biomarkers for evaluating organic anion‐transporting polypeptide (OATP)1B1 (SLCO1B1) inhibition. The current study investigated the effect of OATP1B1 genotype c.521T>C (OATP1B1‐Val174Ala) on the extent of interaction between cyclosporin A (CsA) and pravastatin, and associated endogenous biomarkers of the transporter (HDA, TDA, CP‐I, and CP‐III), in 20 healthy volunteers. The results show that the levels of each clinical biomarker and pravastatin were significantly increased in plasma samples of the volunteers following administration of pravastatin plus CsA compared with pravastatin plus placebo. The overall fold change in the area under the concentration–time curve (AUC) and maximum plasma concentration (Cmax) was similar among the four biomarkers (1.8–2.5‐fold, paired t‐test P value C genotype is significantly associated with CP‐I but not CP‐III levels. Overall, these results suggest that OATP1B1 genotype can modulate the effects of CsA on biomarker levels; the extent of modulation differs among the biomarkers.
- Published
- 2019
- Full Text
- View/download PDF
12. Diversity, Equity, and Inclusion: Translating Clinical Pharmacology forAll
- Author
-
Karthik Venkatakrishnan, Karen E. Brown, Kathleen M. Giacomini, and Piet H. van der Graaf
- Subjects
Pharmacology ,Pharmacology (medical) - Published
- 2023
13. Biomarkers Come of Age in Clinical Pharmacology
- Author
-
Kathleen M. Giacomini
- Subjects
Pharmacology ,Pharmacology (medical) - Published
- 2023
14. Characterization of P‐glycoprotein orthologs from human, sheep, pig, dog, and cat
- Author
-
Mina Azimi, Sook Wah Yee, Andrew Riselli, Dina Buitrago Silva, Craig P. Giacomini, Kathleen M. Giacomini, and Claire M. Brett
- Subjects
Pharmacology ,General Veterinary - Published
- 2023
15. Supplementary Figure 1 from Interactions of Tyrosine Kinase Inhibitors with Organic Cation Transporters and Multidrug and Toxic Compound Extrusion Proteins
- Author
-
Kathleen M. Giacomini and Tsuyoshi Minematsu
- Abstract
Supplementary Figure 1 from Interactions of Tyrosine Kinase Inhibitors with Organic Cation Transporters and Multidrug and Toxic Compound Extrusion Proteins
- Published
- 2023
16. Supplementary Tables 1-3 from Interactions of Tyrosine Kinase Inhibitors with Organic Cation Transporters and Multidrug and Toxic Compound Extrusion Proteins
- Author
-
Kathleen M. Giacomini and Tsuyoshi Minematsu
- Abstract
Supplementary Tables 1-3 from Interactions of Tyrosine Kinase Inhibitors with Organic Cation Transporters and Multidrug and Toxic Compound Extrusion Proteins
- Published
- 2023
17. Data from Interactions of Tyrosine Kinase Inhibitors with Organic Cation Transporters and Multidrug and Toxic Compound Extrusion Proteins
- Author
-
Kathleen M. Giacomini and Tsuyoshi Minematsu
- Abstract
The drug–drug interaction (DDI) potential of tyrosine kinase inhibitors (TKI) as interacting drugs via transporter inhibition has not been fully assessed. Here, we estimated the half maximal inhibitory concentration (IC50) values for 8 small-molecule TKIs (imatinib, dasatinib, nilotinib, gefitinib, erlotinib, sunitinib, lapatinib, and sorafenib) on [14C]metformin transport by human organic cation transporters (OCT), OCT1, OCT2, and OCT3, and multidrug and toxic compound extrusion (MATE) proteins, MATE1 and MATE2-K, using human embryonic kidney cells stably expressing these transporters. We then compared the estimated IC50 values to the maximum clinical concentrations of unbound TKIs in plasma (unbound Cmax,sys,p). Results showed that imatinib, nilotinib, gefitinib, and erlotinib exerted selectively potent inhibitory effects, with unbound Cmax,sys,p/IC50 values ≥0.1, on MATE1, OCT3, MATE2-K, and OCT1, respectively. In comparison to the common form of OCT1, the OCT1 polymorphism, M420del, was more sensitive to drug inhibition by erlotinib. Major metabolites of several TKIs showed IC50 values similar to those for unchanged TKIs. Taken together, these findings suggest the potential of clinical transporter-mediated DDIs between specific TKIs and OCTs and MATEs, which may affect the disposition, efficacy, and toxicity of metformin and other drugs that are substrates of these transporters. The study provides the basis for further clinical DDI studies with TKIs. Mol Cancer Ther; 10(3); 531–9. ©2011 AACR.
- Published
- 2023
18. Response to Comment on Dawed et al. Genome-Wide Meta-analysis Identifies Genetic Variants Associated With Glycemic Response to Sulfonylureas. Diabetes Care 2021;44:2673-2682
- Author
-
Adem Y, Dawed, Sook Wah, Yee, Kaixin, Zhou, Nienke, van Leeuwen, Yanfei, Zhang, Moneeza K, Siddiqui, Amy, Etheridge, Federico, Innocenti, Fei, Xu, Josephine H, Li, Joline W, Beulens, Amber A, van der Heijden, Roderick C, Slieker, Yu-Chuan, Chang, Josep M, Mercader, Varinderpal, Kaur, John S, Witte, Ming Ta Michael, Lee, Yoichiro, Kamatani, Yukihide, Momozawa, Michiaki, Kubo, Colin N A, Palmer, Jose C, Florez, Monique M, Hedderson, Leen M, 't Hart, Kathleen M, Giacomini, and Ewan R, Pearson
- Subjects
Advanced and Specialized Nursing ,Endocrinology, Diabetes and Metabolism ,Internal Medicine - Published
- 2023
19. Supplementary Figures S1-S6 from Vorinostat Increases Expression of Functional Norepinephrine Transporter in Neuroblastoma In Vitro and In Vivo Model Systems
- Author
-
Kathleen M. Giacomini, Katherine K. Matthay, Steven G. DuBois, Daphne A. Haas-Kogan, Sabine Mueller, William A. Weiss, Henry F. VanBrocklin, Youngho Seo, Michelle K. Tadano, Ethan G. Geier, Xiaodong Yang, Melissa Itsara, and Swati S. More
- Abstract
Supplementary Figures S1-S6.
- Published
- 2023
20. Data from A Genome-Wide Association Study of Overall Survival in Pancreatic Cancer Patients Treated with Gemcitabine in CALGB 80303
- Author
-
Mark J. Ratain, Yusuke Nakamura, Hedy L. Kindler, Richard L. Schilsky, Richard M. Goldberg, Howard L. McLeod, Kathleen M. Giacomini, Herbert Hurwitz, Dylan Glubb, Liewei Wang, Paula Friedman, Liang Li, Taisei Mushiroda, Donna Hollis, Chen Jiang, Hitoshi Zembutsu, Michiaki Kubo, Patrick Evans, Nancy L. Cox, Kouros Owzar, and Federico Innocenti
- Abstract
Background and Aims: Cancer and Leukemia Group B 80303 was a randomized, phase III study in patients with advanced pancreatic cancer treated with gemcitabine plus either bevacizumab or placebo. We prospectively collected germline DNA and conducted a genome-wide association study (GWAS) using overall survival (OS) as the endpoint.Experimental Design: DNA from 351 patients was genotyped for more than 550,000 single-nucleotide polymorphisms (SNP). Associations between OS and SNPs were investigated using the log-linear 2-way multiplicative Cox proportional hazards model. The subset of 294 genetically European patients was used for the primary analysis.Results: A nonsynonymous SNP in interleukin (IL)17F (rs763780, H161R) and an intronic SNP in strong linkage disequilibrium (rs7771466) were associated with OS using genome-wide criteria (P ≤ 10−7). Median OS was significantly shorter (P = 2.61 × 10−8) for the rs763780 heterozygotes [3.1 months; 95% confidence interval (CI), 2.3–4.3] than for the patients without this variant (6.8 months; 95% CI, 5.8–7.3). After adjustment by stratification factors, the P value for the association was 9.51 × 10−7.Conclusions: The variant 161R form of IL-17F is a natural antagonist of the antiangiogenic effects of wild-type 161H IL-17F, and angiogenesis may play an important role in the metastatic spread of pancreatic cancer. In this preliminary study, we hypothesize that the angiogenetic potential of pancreatic cancers in patients with variant IL-17F is higher than that of tumors in patients with wild-type IL-17F, conferring worse prognosis. This exploratory GWAS may provide the foundation for testing the biology and clinical effects of novel genes and their heritable variants through mechanistic and confirmatory studies in pancreatic cancer. Clin Cancer Res; 18(2); 577–84. ©2011 AACR.
- Published
- 2023
21. Supplementary Figures 1-3, Tables 1-3 from A Genome-Wide Association Study of Overall Survival in Pancreatic Cancer Patients Treated with Gemcitabine in CALGB 80303
- Author
-
Mark J. Ratain, Yusuke Nakamura, Hedy L. Kindler, Richard L. Schilsky, Richard M. Goldberg, Howard L. McLeod, Kathleen M. Giacomini, Herbert Hurwitz, Dylan Glubb, Liewei Wang, Paula Friedman, Liang Li, Taisei Mushiroda, Donna Hollis, Chen Jiang, Hitoshi Zembutsu, Michiaki Kubo, Patrick Evans, Nancy L. Cox, Kouros Owzar, and Federico Innocenti
- Abstract
PDF file - 155K
- Published
- 2023
22. Data from Organic Cation Transporters Modulate the Uptake and Cytotoxicity of Picoplatin, a Third-Generation Platinum Analogue
- Author
-
Kathleen M. Giacomini, David M. Jablons, Zhidong Xu, Ligong Chen, Sook Wah Yee, Shuanglian Li, and Swati S. More
- Abstract
Picoplatin, a third-generation platinum agent, is efficacious against lung cancers that are otherwise resistant or become refractory during platinum treatment. This effort was aimed at the determination of the influence of organic cation transporters 1, 2, and 3 (OCT1, OCT2, and OCT3) and their genetic variants on cellular uptake of picoplatin and on the individual components of the ensuing cytotoxicity such as DNA adduct formation. The effect of OCT1 on picoplatin pharmacokinetics and antitumor efficacy was determined using OCT knockout mice and HEK293 xenografts stably expressing OCT1. The uptake and DNA adduct formation of picoplatin were found to be significantly enhanced by the expression of the OCTs. Expression of OCT1 and OCT2, but not OCT3, significantly enhanced picoplatin cytotoxicity, which was reduced in the presence of an OCT inhibitor. Common reduced functional variants of OCT1 and OCT2 led to reduction in uptake and DNA adduct formation of picoplatin in comparison with the reference OCT1 and OCT2. Pharmacokinetic parameters of picoplatin in Oct1−/− and Oct1+/+ mice were not significantly different, suggesting that the transporters do not influence the disposition of the drug. In contrast, the volume of OCT1-expressing xenografts in mice was significantly reduced by picoplatin treatment, suggesting that OCT1 may enhance the antitumor efficacy of picoplatin. These studies provide a basis for follow-up clinical studies that would seek to examine the relationship between the anticancer efficacy of picoplatin and expression levels of OCTs and their genetic variants in tumors. Mol Cancer Ther; 9(4); 1058–69. ©2010 AACR.
- Published
- 2023
23. Data from Vorinostat Increases Expression of Functional Norepinephrine Transporter in Neuroblastoma In Vitro and In Vivo Model Systems
- Author
-
Kathleen M. Giacomini, Katherine K. Matthay, Steven G. DuBois, Daphne A. Haas-Kogan, Sabine Mueller, William A. Weiss, Henry F. VanBrocklin, Youngho Seo, Michelle K. Tadano, Ethan G. Geier, Xiaodong Yang, Melissa Itsara, and Swati S. More
- Abstract
Purpose: Histone deacetylase (HDAC) inhibition causes transcriptional activation or repression of several genes that in turn can influence the biodistribution of other chemotherapeutic agents. Here, we hypothesize that the combination of vorinostat, a HDAC inhibitor, with 131I-meta-iodobenzylguanidine (MIBG) would lead to preferential accumulation of the latter in neuroblastoma (NB) tumors via increased expression of the human norepinephrine transporter (NET).Experimental Design:In vitro and in vivo experiments examined the effect of vorinostat on the expression of NET, an uptake transporter for 131I-MIBG. Human NB cell lines (Kelly and SH-SY-5Y) and NB1691-luc mouse xenografts were employed. The upregulated NET protein was characterized for its effect on 123I-MIBG biodistribution.Results: Preincubation of NB cell lines, Kelly, and SH-SY-5Y, with vorinostat caused dose-dependent increases in NET mRNA and protein levels. Accompanying this was a corresponding dose-dependent increase in MIBG uptake in NB cell lines. Four- and 2.5-fold increases were observed in Kelly and SH-SY-5Y cells, respectively, pretreated with vorinostat in comparison to untreated cells. Similarly, NB xenografts, created by intravenous tail vein injection of NB1691-luc, and harvested from nude mice livers treated with vorinostat (150 mg/kg i.p.) showed substantial increases in NET protein expression. Maximal effect of vorinostat pretreatment in NB xenografts on 123I-MIBG biodistribution was observed in tumors that exhibited enhanced uptake in vorinostat-treated [0.062 ± 0.011 μCi/(mg tissue-dose injected)] vs. -untreated mice [0.022 ± 0.003 μCi/(mg tissue-dose injected); P < 0.05].Conclusions: The results of our study provide preclinical evidence that vorinostat treatment can enhance NB therapy with 131I-MIBG. Clin Cancer Res; 17(8); 2339–49. ©2011 AACR.
- Published
- 2023
24. Data from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Author
-
Kathleen M. Giacomini, Stephen J. Lippard, Xin Chen, Joe W. Gray, Takafumi Komori, Ying Chen, Anna Lapuk, Yan Shu, Leah L. Lagpacan, James E. Shima, Katherine S. Lovejoy, and Shuzhong Zhang
- Abstract
Although the platinum-based anticancer drugs cisplatin, carboplatin, and oxaliplatin have similar DNA-binding properties, only oxaliplatin is active against colorectal tumors. The mechanisms for this tumor specificity of platinum-based compounds are poorly understood but could be related to differences in uptake. This study shows that the human organic cation transporters (OCT) 1 and 2 (SLC22A1 and SLC22A2) markedly increase oxaliplatin, but not cisplatin or carboplatin, accumulation and cytotoxicity in transfected cells, indicating that oxaliplatin is an excellent substrate of these transporters. The cytotoxicity of oxaliplatin was greater than that of cisplatin in six colon cancer cell lines [mean ± SE of IC50 in the six cell lines, 3.9 ± 1.4 μmol/L (oxaliplatin) versus 11 ± 2.0 μmol/L (cisplatin)] but was reduced by an OCT inhibitor, cimetidine, to a level similar to, or even lower than that of, cisplatin (29 ± 11 μmol/L for oxaliplatin versus 19 ± 4.3 μmol/L for cisplatin). Structure-activity studies indicated that organic functionalities on nonleaving groups coordinated to platinum are critical for selective uptake by OCTs. These results indicate that OCT1 and OCT2 are major determinants of the anticancer activity of oxaliplatin and may contribute to its antitumor specificity. They also strongly suggest that expression of OCTs in tumors should be investigated as markers for selecting specific platinum-based therapies in individual patients. The development of new anticancer drugs, specifically targeted to OCTs, represents a novel strategy for targeted drug therapy. The results of the present structure-activity studies indicate specific tactics for realizing this goal. (Cancer Res 2006; 66(17): 8847-57)
- Published
- 2023
25. Supplementary Table 1 from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Author
-
Kathleen M. Giacomini, Stephen J. Lippard, Xin Chen, Joe W. Gray, Takafumi Komori, Ying Chen, Anna Lapuk, Yan Shu, Leah L. Lagpacan, James E. Shima, Katherine S. Lovejoy, and Shuzhong Zhang
- Abstract
Supplementary Table 1 from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Published
- 2023
26. Supplementary Figure 1 Legend from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Author
-
Kathleen M. Giacomini, Stephen J. Lippard, Xin Chen, Joe W. Gray, Takafumi Komori, Ying Chen, Anna Lapuk, Yan Shu, Leah L. Lagpacan, James E. Shima, Katherine S. Lovejoy, and Shuzhong Zhang
- Abstract
Supplementary Figure 1 Legend from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Published
- 2023
27. Supplementary Table 2 from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Author
-
Kathleen M. Giacomini, Stephen J. Lippard, Xin Chen, Joe W. Gray, Takafumi Komori, Ying Chen, Anna Lapuk, Yan Shu, Leah L. Lagpacan, James E. Shima, Katherine S. Lovejoy, and Shuzhong Zhang
- Abstract
Supplementary Table 2 from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Published
- 2023
28. Supplementary Figure 1 from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Author
-
Kathleen M. Giacomini, Stephen J. Lippard, Xin Chen, Joe W. Gray, Takafumi Komori, Ying Chen, Anna Lapuk, Yan Shu, Leah L. Lagpacan, James E. Shima, Katherine S. Lovejoy, and Shuzhong Zhang
- Abstract
Supplementary Figure 1 from Organic Cation Transporters Are Determinants of Oxaliplatin Cytotoxicity
- Published
- 2023
29. ORGANIC CATION AND ZWITTERION TRANSPORTERS
- Author
-
Megan L. Koleske, Xiaomin Liang, Osatohanmwen Jessica Enogieru, Dina Buitrago, and Kathleen M. Giacomini
- Published
- 2022
30. The Clinical Pharmacogenetics Implementation Consortium Guideline for SLCO1B1 , ABCG2 , and CYP2C9 genotypes and Statin‐Associated Musculoskeletal Symptoms
- Author
-
Rhonda M. Cooper‐DeHoff, Mikko Niemi, Laura B. Ramsey, Jasmine A. Luzum, E. Katriina Tarkiainen, Robert J. Straka, Li Gong, Sony Tuteja, Russell A. Wilke, Mia Wadelius, Eric A. Larson, Dan M. Roden, Teri E. Klein, Sook Wah Yee, Ronald M. Krauss, Richard M. Turner, Latha Palaniappan, Andrea Gaedigk, Kathleen M. Giacomini, Kelly E. Caudle, Deepak Voora, HUSLAB, Department of Clinical Pharmacology, Medicum, Department of Diagnostics and Therapeutics, INDIVIDRUG - Individualized Drug Therapy, and HUS Diagnostic Center
- Subjects
Member 2 ,PHARMACOKINETICS ,Simvastatin ,Genotype ,IMPACT ,ATP Binding Cassette Transporter ,VARIANTS ,Subfamily G ,Cardiovascular ,INDUCED MYOPATHY ,Clinical Research ,Genetics ,Humans ,Pharmacology (medical) ,Pharmacology & Pharmacy ,Rosuvastatin Calcium ,Cytochrome P-450 CYP2C9 ,RISK ,Pharmacology ,Liver-Specific Organic Anion Transporter 1 ,Evaluation of treatments and therapeutic interventions ,Pharmacology and Pharmaceutical Sciences ,ALLELES ,RHABDOMYOLYSIS ,Neoplasm Proteins ,Good Health and Well Being ,Pharmacogenetics ,3121 General medicine, internal medicine and other clinical medicine ,6.1 Pharmaceuticals ,3111 Biomedicine ,Patient Safety ,Hydroxymethylglutaryl-CoA Reductase Inhibitors - Abstract
Statins reduce cholesterol, prevent cardiovascular disease, and are among the most commonly prescribed medications in the world. Statin-associated musculoskeletal symptoms (SAMS) impact statin adherence and ultimately can impede the long-term effectiveness of statin therapy. There are several identified pharmacogenetic variants that impact statin disposition and adverse events during statin therapy. SLCO1B1 encodes a transporter (SLCO1B1; alternative names include OATP1B1 or OATP-C) that facilitates the hepatic uptake of all statins. ABCG2 encodes an efflux transporter (BCRP) that modulates the absorption and disposition of rosuvastatin. CYP2C9 encodes a phase I drug metabolizing enzyme responsible for the oxidation of some statins. Genetic variation in each of these genes alters systemic exposure to statins (i.e., simvastatin, rosuvastatin, pravastatin, pitavastatin, atorvastatin, fluvastatin, lovastatin), which can increase the risk for SAMS. We summarize the literature supporting these associations and provide therapeutic recommendations for statins based on SLCO1B1, ABCG2, and CYP2C9 genotype with the goal of improving the overall safety, adherence, and effectiveness of statin therapy. This document replaces the 2012 and 2014 Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for SLCO1B1 and simvastatin-induced myopathy.
- Published
- 2022
31. High Throughput Screening of a Prescription Drug Library for Inhibitors of Organic Cation Transporter 3, OCT3
- Author
-
Eugene C. Chen, Pär Matsson, Mina Azimi, Xujia Zhou, Niklas Handin, Sook Wah Yee, Per Artursson, and Kathleen M. Giacomini
- Subjects
Pharmacology ,Prescription Drugs ,Organic Cation Transport Proteins ,Organic Chemistry ,EMT ,Pharmaceutical Science ,Pharmacology and Toxicology ,Farmaceutiska vetenskaper ,Farmakologi och toxikologi ,High-Throughput Screening Assays ,Pharmaceutical Sciences ,HEK293 Cells ,Cations ,Humans ,Molecular Medicine ,Pharmacology (medical) ,Solute carrier superfamily ,extraneuronal monoamine transporter ,Biotechnology - Abstract
Introduction The organic cation transporter 3 (OCT3, SLC22A3) is ubiquitously expressed and interacts with a wide array of compounds including endogenous molecules, environmental toxins and prescription drugs. Understudied as a determinant of pharmacokinetics and pharmacodynamics, OCT3 has the potential to be a major determinant of drug absorption and disposition and to be a target for drug-drug interactions (DDIs). Goal The goal of the current study was to identify prescription drug inhibitors of OCT3. Methods We screened a compound library consisting of 2556 prescription drugs, bioactive molecules, and natural products using a high throughput assay in HEK-293 cells stably expressing OCT3. Results We identified 210 compounds that at 20 μM inhibit 50% or more of OCT3-mediated uptake of 4-Di-1-ASP (2 μM). Of these, nine were predicted to inhibit the transporter at clinically relevant unbound plasma concentrations. A Structure-Activity Relationship (SAR) model included molecular descriptors that could discriminate between inhibitors and non-inhibitors of OCT3 and was used to identify additional OCT3 inhibitors. Proteomics of human brain microvessels (BMVs) indicated that OCT3 is the highest expressed OCT in the human blood-brain barrier (BBB). Conclusions This study represents the largest screen to identify prescription drug inhibitors of OCT3. Several are sufficiently potent to inhibit the transporter at therapeutic unbound plasma levels, potentially leading to DDIs or off-target pharmacologic effects.
- Published
- 2022
32. Identification of genetic variation influencing metformin response in a multi-ancestry genome-wide association study in the Diabetes Prevention Program (DPP)
- Author
-
Diabetes Prevention Program Research Group, Jose C. Florez, Toni I. Pollin, William C. Knowler, Steven E. Kahn, Robert L. Hanson, Paul W. Franks, L. Keoki Williams, Shujie Xiao, Adriana M. Hung, Ayush Giri, Kathleen M. Giacomini, Ewan R. Pearson, Sook Wah Yee, Adem Y. Dawed, Qing Pan, Josep M. Mercader, Maegan Harden, Jennifer N. Todd, Ling Chen, Shylaja Srinivasan, Kathleen A. Jablonski, James A. Perry, and Josephine Li
- Abstract
Genome-wide significant loci for metformin response in type 2 diabetes reported elsewhere have not replicated in the Diabetes Prevention Program (DPP). To assess pharmacogenetic interactions in pre-diabetes, we conducted a genome-wide association study (GWAS) in the DPP. Cox proportional hazards models tested associations with diabetes incidence in metformin (MET, n=876) and placebo (PBO, n=887) arms. Multiple linear regression assessed association with one-year change in metformin-related quantitative traits, adjusted for baseline trait, age, sex, and 10 ancestry principal components. We tested for gene-by-treatment interaction. No significant associations emerged for diabetes incidence. We identified four genome-wide significant variants after correcting for correlated traits (pENOSF1 (minor allele frequency [MAF]AFR=0.07, MAFEUR=0.002) was associated with an increase in % glycated hemoglobin (per minor allele β=0.39 [95% CI 0.28, 0.50], p=2.8×10-12). Rs145591055 near OMSR (MAF=0.10 in American Indians), was associated with weight loss (kg) (per G allele β=-7.55 [95% CI -9.88, -5.22], p=3.2×10-10) in MET. Neither variant was significant in PBO; gene-by-treatment interaction was significant for both variants (p(G×T)
- Published
- 2023
33. Emerging Roles of the Human Solute Carrier 22 Family
- Author
-
Sook Wah Yee and Kathleen M. Giacomini
- Subjects
Pharmacology ,Pharmaceutical Science - Published
- 2021
34. Identification of Genetic Variation Influencing Metformin Response in a Multi-Ancestry Genome-Wide Association Study in the Diabetes Prevention Program (DPP)
- Author
-
Josephine H, Li, James A, Perry, Kathleen A, Jablonski, Shylaja, Srinivasan, Ling, Chen, Jennifer N, Todd, Maegan, Harden, Josep M, Mercader, Qing, Pan, Adem Y, Dawed, Sook Wah, Yee, Ewan R, Pearson, Kathleen M, Giacomini, Ayush, Giri, Adriana M, Hung, Shujie, Xiao, L Keoki, Williams, Paul W, Franks, Robert L, Hanson, Steven E, Kahn, William C, Knowler, Toni I, Pollin, and Jose C, Florez
- Subjects
Endocrinology, Diabetes and Metabolism ,Internal Medicine - Abstract
Genome-wide significant loci for metformin response in type 2 diabetes reported elsewhere have not replicated in the Diabetes Prevention Program (DPP). To assess pharmacogenetic interactions in pre-diabetes, we conducted a genome-wide association study (GWAS) in the DPP. Cox proportional hazards models tested associations with diabetes incidence in metformin (MET, n=876) and placebo (PBO, n=887) arms. Multiple linear regression assessed association with one-year change in metformin-related quantitative traits, adjusted for baseline trait, age, sex, and 10 ancestry principal components. We tested for gene-by-treatment interaction. No significant associations emerged for diabetes incidence. We identified four genome-wide significant variants after correcting for correlated traits (p
- Published
- 2022
35. Functional genomics of OCTN2 variants informs protein-specific variant effect predictor for Carnitine Transporter Deficiency
- Author
-
Megan L. Koleske, Gregory McInnes, Julia E. H. Brown, Neil Thomas, Keino Hutchinson, Marcus Y. Chin, Antoine Koehl, Michelle R. Arkin, Avner Schlessinger, Renata C. Gallagher, Yun S. Song, Russ B. Altman, and Kathleen M. Giacomini
- Subjects
Multidisciplinary ,Organic Cation Transport Proteins ,variant interpretation ,Prevention ,Human Genome ,rare disease ,Genomics ,HEK293 Cells ,machine learning ,Carnitine ,transporter ,Genetics ,Humans ,2.1 Biological and endogenous factors ,Generic health relevance ,Aetiology ,Solute Carrier Family 22 Member 5 - Abstract
Genetic variants in SLC22A5 , encoding the membrane carnitine transporter OCTN2, cause the rare metabolic disorder Carnitine Transporter Deficiency (CTD). CTD is potentially lethal but actionable if detected early, with confirmatory diagnosis involving sequencing of SLC22A5 . Interpretation of missense variants of uncertain significance (VUSs) is a major challenge. In this study, we sought to characterize the largest set to date ( n = 150) of OCTN2 variants identified in diverse ancestral populations, with the goals of furthering our understanding of the mechanisms leading to OCTN2 loss-of-function (LOF) and creating a protein-specific variant effect prediction model for OCTN2 function. Uptake assays with 14 C-carnitine revealed that 105 variants (70%) significantly reduced transport of carnitine compared to wild-type OCTN2, and 37 variants (25%) severely reduced function to less than 20%. All ancestral populations harbored LOF variants; 62% of green fluorescent protein (GFP)–tagged variants impaired OCTN2 localization to the plasma membrane of human embryonic kidney (HEK293T) cells, and subcellular localization significantly associated with function, revealing a major LOF mechanism of interest for CTD. With these data, we trained a model to classify variants as functional (>20% function) or LOF (
- Published
- 2022
36. Advancing Precision Medicine Through the New Pharmacogenomics Global Research Network
- Author
-
Akinyemi Oni-Orisan, William A. Murphy, Michelle Whirl-Carrillo, Kathleen M. Giacomini, Kristine R. Crews, Laura B. Ramsey, Jason H. Karnes, Andrew A. Monte, and Jun J. Yang
- Subjects
Pharmacology ,medicine.medical_specialty ,business.industry ,Research ,MEDLINE ,Precision medicine ,Article ,Pharmacogenetics ,Pharmacogenomics ,Humans ,Medicine ,Pharmacology (medical) ,Medical physics ,Precision Medicine ,business - Abstract
The new Pharmacogenomics Global Research Network (PGRN) is an independent society that builds on the National Institutes of Health (NIH)–funded Pharmacogenomics Research Network that was established in 2000. Leveraging the original PGRN’s previous success, the new network continues to be a leader in the field of personalized medicine focusing on research, discovery, and translation of genomic variation influencing drug efficacy and adverse events, while simultaneously increasing inclusion in the field and expanding globally.
- Published
- 2021
37. Oxypurinol pharmacokinetics and pharmacodynamics in healthy volunteers: Influence of BCRP Q141K polymorphism and patient characteristics
- Author
-
Marina Sirota, Radojka M. Savic, Deanna J. Brackman, Kathleen M. Giacomini, Maria Garcia-Cremades, Ling Zou, and Bianca Vora
- Subjects
Male ,Pharmacogenomic Variants ,Pharmacology ,Cardiorespiratory Medicine and Haematology ,Subfamily G ,chemistry.chemical_compound ,Models ,Medicine ,ATP Binding Cassette Transporter, Subfamily G, Member 2 ,Prospective Studies ,General Pharmacology, Toxicology and Pharmaceutics ,General Clinical Medicine ,Volume of distribution ,education.field_of_study ,General Neuroscience ,General Medicine ,Single Nucleotide ,Articles ,Middle Aged ,Healthy Volunteers ,Neoplasm Proteins ,6.1 Pharmaceuticals ,Creatinine ,Female ,Public aspects of medicine ,RA1-1270 ,medicine.drug ,Glomerular Filtration Rate ,Half-Life ,Member 2 ,Adult ,Adolescent ,ATP Binding Cassette Transporter ,Population ,Oncology and Carcinogenesis ,Mutation, Missense ,Allopurinol ,Oxypurinol ,RM1-950 ,Models, Biological ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Article ,Young Adult ,Sex Factors ,Pharmacokinetics ,Clinical Research ,Genetics ,Humans ,Polymorphism ,education ,Other Medical and Health Sciences ,business.industry ,Research ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Biological ,Gout ,Uric Acid ,Renal Elimination ,chemistry ,Pharmacodynamics ,Mutation ,Uric acid ,Therapeutics. Pharmacology ,Missense ,business ,Blood sampling - Abstract
The missense variant, breast cancer resistance protein (BCRP) p.Q141K, which encodes a reduced function BCRP, has been linked to poor response to allopurinol. Using a multifaceted approach, we aimed to characterize the relationship(s) between BCRP p.Q141K, the pharmacokinetics (PK) and pharmacodynamics (PD) of oxypurinol (the active metabolite of allopurinol), and serum uric acid (SUA) levels. A prospective clinical study (NCT02956278) was conducted in which healthy volunteers were given a single oral dose of 300mg allopurinol followed by intensive blood sampling. Data were analyzed using noncompartmental analysis and population PK/PD modeling. Additionally, electronic health records were analyzed to investigate whether clinical inhibitors of BCRP phenocopied the effects of the p.Q141K variant with respect to SUA. Subjects homozygous for p.Q141K had a longer half-life (34.2±12.2h vs. 19.1±1.42h) of oxypurinol. The PK/PD model showed that women had a 24.8% lower volume of distribution. Baseline SUA was affected by p.Q141K genotype and renal function; that is, it changed by 48.8% for every 1mg/dl difference in serum creatinine. Real-world data analyses showed that patients prescribed clinical inhibitors of BCRP have higher SUA levels than those that have not been prescribed inhibitors of BCRP, consistent with the idea that BCRP inhibitors phenocopy the effects of p.Q141K on uric acid levels. This study identified important covariates of oxypurinol PK/PD that could affect its efficacy for the treatment of gout as well as a potential side effect of BCRP inhibitors on increasing uric acid levels, which has not been described previously.
- Published
- 2021
38. Cardiac contraction and relaxation are regulated by beta 1 adrenergic receptor-generated cAMP pools at distinct membrane locations
- Author
-
Ting-Yu Lin, Quynh N. Mai, Hao Zhang, Emily Wilson, Huan-Chieh Chien, Sook Wah Yee, Kathleen M. Giacomini, Jeffrey E. Olgin, and Roshanak Irannejad
- Abstract
Cells interpret a variety of signals through G protein-coupled receptors (GPCRs) and stimulate the generation of second messengers such as cyclic adenosine monophosphate (cAMP). A long-standing puzzle is deciphering how GPCRs elicit different physiological responses despite generating similar levels of cAMP. We previously showed that some GPCRs generate cAMP from both the plasma membrane and the Golgi apparatus. Here, we demonstrate that cardiomyocytes distinguish between subcellular cAMP inputs to elicit different physiological outputs. We show that generating cAMP from the Golgi leads to regulation of a specific PKA target that increases the rate of cardiomyocyte relaxation. In contrast, cAMP generation from the plasma membrane activates a different PKA target that increases contractile force. We further validated the physiological consequences of these observations in intact zebrafish and mice. Thus, we demonstrate that the same GPCR acting through the same second messenger regulates cardiac contraction and relaxation dependent on its subcellular location.
- Published
- 2022
39. More Than Pharmacokinetics: Transporters in Clinical Pharmacology
- Author
-
Kathleen M. Giacomini and Shiew Mei Huang
- Subjects
Pharmacology ,Pharmacology, Clinical ,Humans ,Membrane Transport Proteins ,Pharmacology (medical) ,Pharmacokinetics - Published
- 2022
40. Drugs in COVID‐19 Clinical Trials: Predicting Transporter‐Mediated Drug‐Drug Interactions Using In Vitro Assays and Real‐World Data
- Author
-
Ling Zou, Idit Kosti, Marina Sirota, Tomiko Oskotsky, Sebastian Jakobsen, Osatohanmwen J. Enogieru, Megan L. Koleske, Sook Wah Yee, Mattias Rödin, Bianca Vora, and Kathleen M. Giacomini
- Subjects
Drug ,Drug-Related Side Effects and Adverse Reactions ,media_common.quotation_subject ,Virus Replication ,Bioinformatics ,Antiviral Agents ,Risk Assessment ,030226 pharmacology & pharmacy ,Article ,03 medical and health sciences ,0302 clinical medicine ,Chloroquine ,Electronic Health Records ,Humans ,Medicine ,Potency ,Drug Interactions ,Pharmacology (medical) ,media_common ,Pharmacology ,Clinical Trials as Topic ,SARS-CoV-2 ,business.industry ,Research ,In vitro toxicology ,COVID-19 ,Membrane Transport Proteins ,Hydroxychloroquine ,Articles ,Virus Internalization ,COVID-19 Drug Treatment ,Clinical trial ,HEK293 Cells ,030220 oncology & carcinogenesis ,Concomitant ,Drug Monitoring ,business ,Risk assessment ,medicine.drug - Abstract
Numerous drugs are currently under accelerated clinical investigation for the treatment of coronavirus disease 2019 (COVID‐19); however, well‐established safety and efficacy data for these drugs are limited. The goal of this study was to predict the potential of 25 small molecule drugs in clinical trials for COVID‐19 to cause clinically relevant drug‐drug interactions (DDIs), which could lead to potential adverse drug reactions (ADRs) with the use of concomitant medications. We focused on 11 transporters, which are targets for DDIs. In vitro potency studies in membrane vesicles or HEK293 cells expressing the transporters coupled with DDI risk assessment methods revealed that 20 of the 25 drugs met the criteria from regulatory authorities to trigger consideration of a DDI clinical trial. Analyses of real‐world data from electronic health records, including a database representing nearly 120,000 patients with COVID‐19, were consistent with several of the drugs causing transporter‐mediated DDIs (e.g., sildenafil, chloroquine, and hydroxychloroquine). This study suggests that patients with COVID‐19, who are often older and on various concomitant medications, should be carefully monitored for ADRs. Future clinical studies are needed to determine whether the drugs that are predicted to inhibit transporters at clinically relevant concentrations, actually result in DDIs.
- Published
- 2021
41. SNP Analysis and Presentation in the Pharmacogenetics of Membrane Transporters Project.
- Author
-
Doug Stryke, Conrad C. Huang, Michiko Kawamoto, Susan J. Johns, Elaine J. Carlson, Joseph A. Deyoung, Maya K. Leabman, Ira Herskowitz, Kathleen M. Giacomini, and Thomas E. Ferrin
- Published
- 2003
42. Characterization of cytochrome P450 (CYP) 2D6 drugs as substrates of human organic cation transporters and multidrug and toxin extrusion proteins
- Author
-
Anne T. Nies, Elke Schaeffeler, Kathrin Klein, Kathleen M. Giacomini, Michel Eichelbaum, Matthias Schwab, Ute Hofmann, Claudia Neul, and Stefan Winter
- Subjects
0301 basic medicine ,CYP2D6 ,SLC47A1 ,Organic Cation Transport Proteins ,Pharmacology ,Hydroxylation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cations ,medicine ,Humans ,Organic cation transport proteins ,biology ,Chemistry ,Sparteine ,Organic Cation Transporter 2 ,Cytochrome P450 ,Phenotype ,030104 developmental biology ,Cytochrome P-450 CYP2D6 ,Pharmaceutical Preparations ,Debrisoquine ,Perhexiline ,biology.protein ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background and purpose The metabolic activity of cytochrome P450 (CYP) 2D6 is highly variable and CYP2D6 genotypes insufficiently explain the extensive and intermediate metabolic phenotypes, limiting the prediction of drug response plus adverse drug reactions. Since CYP2D6 prototypic substrates are positively charged, the aim of this study was to evaluate the organic cation transporters (OCTs) and multidrug and toxin extrusion proteins (MATEs) as potential contributors to the variability of CYP2D6 hydroxylation of debrisoquine, dextromethorphan, diphenhydramine, perhexiline and sparteine. Experimental approach OCT1/SLC22A1-, OCT2/SLC22A2-, OCT3/SLC22A3-, MATE1/SLC47A1-, and MATE2K/SLC47A2-overexpressing cell lines were used to investigate the transport of the selected drugs. Individuals from a study cohort, well defined with respect to CYP2D6 genotype and sparteine pharmacokinetics, were genotyped for the common OCT1 variants rs12208357 (OCT1-R61C), rs34130495 (OCT1-G401S), rs202220802 (OCT1-Met420del), rs34059508 (OCT1-G465R), OCT2 variant rs316019 (OCT2-A270S) and MATE1 variant rs2289669. Sparteine pharmacokinetics was stratified according to CYP2D6 and OCT1, OCT2 or MATE1 genotype. Key results OCTs and MATE1 transport sparteine and debrisoquine with high affinity in vitro, but OCT- and MATE1-dependent transport of dextromethorphan, diphenhydramine and perhexiline was not detected. Sparteine and debrisoquine transport depends on OCT1 genotype; however, sparteine pharmacokinetics is independent from OCT1 genotype. Conclusions and implications Some drugs that are substrates of CYP2D6 are also substrates of OCTs and MATE1, suggesting overlapping specificities. Variability in sparteine hydroxylation in extensive and intermediate metabolizers cannot be explained by OCT1 genetic variants indicating presence of other factors. Dose-dependent toxicities of dextromethorphan, diphenhydramine and perhexiline appear to be independent from OCTs and MATEs.
- Published
- 2021
43. A New Era in Pharmacovigilance: Toward Real‐World Data and Digital Monitoring
- Author
-
Adam Lavertu, Stefano E. Rensi, Kathleen M. Giacomini, Bianca Vora, and Russ B. Altman
- Subjects
Databases, Pharmaceutical ,MEDLINE ,Reviews ,030226 pharmacology & pharmacy ,Mini‐Reviews ,03 medical and health sciences ,Pharmacovigilance ,0302 clinical medicine ,Product lifecycle ,medicine ,Adverse Drug Reaction Reporting Systems ,Electronic Health Records ,Humans ,Pharmacology (medical) ,Social media ,Safety monitoring ,Pharmacology ,United States Food and Drug Administration ,Mini‐Review ,medicine.disease ,United States ,Clinical trial ,Drug development ,030220 oncology & carcinogenesis ,Business ,Medical emergency ,Real world data ,Social Media - Abstract
Adverse drug reactions (ADRs) are a major concern for patients, clinicians, and regulatory agencies. The discovery of serious ADRs leading to substantial morbidity and mortality has resulted in mandatory phase IV clinical trials, black box warnings, and withdrawal of drugs from the market. Real-world data, data collected during routine clinical care, is being adopted by innovators, regulators, payors, and providers to inform decision making throughout the product life cycle. We outline several different approaches to modern pharmacovigilance, including spontaneous reporting databases, electronic health record monitoring and research frameworks, social media surveillance, and the use of digital devices. Some of these platforms are well-established while others are still emerging or experimental. We highlight both the potential opportunity, as well as the existing challenges within these pharmacovigilance systems that have already begun to impact the drug development process, as well as the landscape of postmarket drug safety monitoring. Further research and investment into different and complementary pharmacovigilance systems is needed to ensure the continued safety of pharmacotherapy.
- Published
- 2021
44. A Tribute to Professor Per Artursson - Scientist, Explorer, Mentor, Innovator, and Giant in Pharmaceutical Research
- Author
-
Katrin Palm, Christel A. S. Bergström, Tommy B. Andersson, Pär Matsson, Pawel Baranczewski, Kathleen M. Giacomini, William N. Charman, and Johan E. Palm
- Subjects
Field (Bourdieu) ,Mentors ,Pharmaceutical Research ,Pharmaceutical Science ,Tribute ,Pharmacy ,02 engineering and technology ,History, 20th Century ,021001 nanoscience & nanotechnology ,030226 pharmacology & pharmacy ,Intestinal absorption ,03 medical and health sciences ,0302 clinical medicine ,Portrait ,Innovator ,Honor ,Humans ,Engineering ethics ,Sociology ,Pharmaceutical sciences ,0210 nano-technology - Abstract
This issue of the Journal of Pharmaceutical Sciences is dedicated to Professor Per Artursson and the groundbreaking contributions he has made and continues to make in the Pharmaceutical Sciences. Per is one of the most cited researchers in his field, with more than 30,000 citations and an h-index of 95 as of September 2020. Importantly, these citations are distributed over the numerous fields he has explored, clearly showing the high impact the research has had on the discipline. We provide a short portrait of Per, with emphasis on his personality, driving forces and the inspirational sources that shaped his career as a world-leading scientist in the field. He is a curious scientist who deftly moves between disciplines and has continued to innovate, expand boundaries, and profoundly impact the pharmaceutical sciences throughout his career. He has developed new tools and provided insights that have significantly contributed to today’s molecular and mechanistic approaches to research in the fields of intestinal absorption, cellular disposition, and exposure-efficacy relationships of pharmaceutical drugs. We want to celebrate these important contributions in this special issue of the Journal of Pharmaceutical Sciences in Per’s honor.
- Published
- 2021
45. Neural production of kynurenic acid in Caenorhabditis elegans requires the AAT-1 transporter
- Author
-
George A. Lemieux, Lin Lin, Kathleen M. Giacomini, Osatohanmwen J. Enogieru, and Kaveh Ashrafi
- Subjects
Nervous system ,Kynurenic Acid ,Medical and Health Sciences ,Large Neutral Amino Acid-Transporter 1 ,Eating ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Kynurenic acid ,Behavioral and Social Science ,Genetics ,medicine ,Animals ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Kynurenine ,Cognitive deficit ,030304 developmental biology ,Therapeutic strategy ,Neurons ,0303 health sciences ,learning ,biology ,Psychology and Cognitive Sciences ,Neurodegeneration ,Neurosciences ,Transporter ,Biological Sciences ,medicine.disease ,biology.organism_classification ,medicine.anatomical_structure ,chemistry ,transporter ,030220 oncology & carcinogenesis ,Mutation ,Neurological ,C. elegans ,medicine.symptom ,metabolism ,Neuroscience ,Developmental Biology - Abstract
Kynurenic acid (KynA) levels link peripheral metabolic status to neural functions including learning and memory. Since neural KynA levels dampen learning capacity, KynA reduction has been proposed as a therapeutic strategy for conditions of cognitive deficit such as neurodegeneration. While KynA is generated locally within the nervous system, its precursor, kynurenine (Kyn), is largely derived from peripheral resources. The mechanisms that import Kyn into the nervous system are poorly understood. Here, we provide genetic, anatomical, biochemical, and behavioral evidence showing that in C. elegans an ortholog of the human LAT1 transporter, AAT-1, imports Kyn into sites of KynA production.
- Published
- 2020
46. COVID‐19: A Defining Moment for Clinical Pharmacology?
- Author
-
Piet H. van der Graaf and Kathleen M. Giacomini
- Subjects
2019-20 coronavirus outbreak ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Antiviral Agents ,law.invention ,Betacoronavirus ,Pharmacovigilance ,Drug Development ,law ,Drug Discovery ,Humans ,Medicine ,Pharmacology (medical) ,Medical physics ,Pandemics ,Pharmacology ,Clinical pharmacology ,SARS-CoV-2 ,business.industry ,Research ,Drug Repositioning ,COVID-19 ,Clinical method ,Moment (mathematics) ,Pharmacology, Clinical ,Coronavirus Infections ,business - Published
- 2020
47. Oral Anticoagulants and Precision Medicine: Something Old, Something New
- Author
-
Kathleen M. Giacomini and Mohamed H. Shahin
- Subjects
Pharmacology ,medicine.medical_specialty ,business.industry ,MEDLINE ,Administration, Oral ,Anticoagulants ,Precision medicine ,Humans ,Medicine ,Pharmacology (medical) ,Medical physics ,Precision Medicine ,business ,Factor Xa Inhibitors - Published
- 2020
48. A Critical Overview of the Biological Effects of Excipients (Part I): Impact on Gastrointestinal Absorption
- Author
-
Marilyn N. Martinez, Balint Sinko, Fang Wu, Talia Flanagan, Enikő Borbás, Eleftheria Tsakalozou, and Kathleen M. Giacomini
- Subjects
Excipients ,Intestinal Absorption ,Solubility ,Pharmaceutical Science ,Permeability - Abstract
Traditionally, excipients have been considered in drug development from the perspective of their influence on drug solubility, manufacturability, and ability to control in vitro and in vivo drug release. These effects have been largely evaluated through studies involving in vitro dissolution methods. However, there is a growing awareness that what had previously been considered biologically inert excipients can exert numerous in vivo effects. This includes the potential to change gastrointestinal (GI) transit time, enterocyte passive transcellular or paracellular permeability, active transport activity, or presystemic drug metabolism. In this critical overview of the biological effects of excipients (Part I), we provide a summary of select published studies that explore these various in vivo factors. We also include a table that points readers to published reviews that list a range of excipients known to have biological activity. A subsequent discussion on in vitro, in vivo, and in silico methods that can be used to explore these excipient effects is provided in a separate (Part 2) continuation of this critical overview.
- Published
- 2022
49. New and Emerging Research on Solute Carrier and ATP Binding Cassette Transporters in Drug Discovery and Development: Outlook From the International Transporter Consortium
- Author
-
Kathleen M. Giacomini, Sook W. Yee, Megan L. Koleske, Ling Zou, Pär Matsson, Eugene C. Chen, Deanna L. Kroetz, Miles A. Miller, Elnaz Gozalpour, and Xiaoyan Chu
- Subjects
Pharmacology ,Proteomics ,1.1 Normal biological development and functioning ,Membrane Transport Proteins ,Pharmacology and Pharmaceutical Sciences ,Underpinning research ,5.1 Pharmaceuticals ,Drug Discovery ,Genetics ,Humans ,Pharmacology (medical) ,ATP-Binding Cassette Transporters ,Patient Safety ,Generic health relevance ,Pharmacology & Pharmacy ,Development of treatments and therapeutic interventions ,Biotechnology - Abstract
Enabled by a plethora of new technologies, research in membrane transporters has exploded in the past decade. The goal of this state-of-the-art article is to describe recent advances in research on membrane transporters that are particularly relevant to drug discovery and development. This review covers advances in basic, translational, and clinical research that has led to an increased understanding of membrane transporters at all levels. At the basic level, we describe the available crystal structures of membrane transporters in both the solute carrier (SLC) and ATP binding cassette superfamilies, which has been enabled by the development of cryogenic electron microscopy methods. Next, we describe new research on lysosomal and mitochondrial transporters as well as recently deorphaned transporters in the SLC superfamily. The translational section includes a summary of proteomic research, which has led to a quantitative understanding of transporter levels in various cell types and tissues and new methods to modulate transporter function, such as allosteric modulators and targeted protein degraders of transporters. The section ends with a review of the effect of the gut microbiome on modulation of transporter function followed by a presentation of 3D cell cultures, which may enable in vivo predictions of transporter function. In the clinical section, we describe new genomic and pharmacogenomic research, highlighting important polymorphisms in transporters that are clinically relevant to many drugs. Finally, we describe new clinical tools, which are becoming increasingly available to enable precision medicine, with the application of tissue-derived small extracellular vesicles and real-world biomarkers.
- Published
- 2022
50. Contributors
- Author
-
Darrell R. Abernethy, Balaji Agoram, John M. Allen, Mark E. Arnold, Arthur J. Atkinson, Thomas J. Bateman, Kimberly Bergman, Brian Booth, David W. Boulton, Robert A. Branch, Gilbert J. Burckart, Mary Buschmann, Owen Carmichael, Christine Chamberlain, Ligong Chen, Charles E. Daniels, Promi Das, Jana G. Delfino, John N. Van Den Anker, Albert W. Dreisbach, Michael Dyszel, Justin C. Earp, M. Khair ElZarrad, Osatohanmwen J. Enogieru, Elimika Pfuma Fletcher, David M. Foster, Marilynn C. Frederiksen, Aleksandra Galetin, Pamela D. Garzone, Kathleen M. Giacomini, Megan A. Gibbs, Jack A Gilbert, Danijela Gnjidic, Charles T. Gombar, Denis M. Grant, Charles Grudzinskas, Bengt Hamren, Nicholas H.G. Holford, Shiew-Mei Huang, Renee Iacona, Nina Isoherranen, Denise Jin, Bridgette L. Jones, Gregory L. Kearns, Cindy Kortepeter, Elizabeth Kunkoski, S.W. Johnny Lau, Christopher Leptak, Juan J.L. Lertora, Lawrence J. Lesko, Jiang Liu, Qi Liu, Rajanikanth Madabushi, Raymond Miller, Diane R. Mould, Monica Muñoz, Thomas D. Nolin, Robert Joseph Noveck, R. Scott Obach, Michael Pacanowski, Mary F. Paine, Carl C. Peck, Anuradha Ramamoorthy, A. David Rodrigues, Malcolm Rowland, Chandrahas G. Sahajwalla, Martina Dagmar Sahre, Robert N. Schuck, Khushboo Sharma, Tristan Sissung, Catherine S. Stika, Chris H. Takimoto, Helen Tomkinson, Jack Uetrecht, Paolo Vicini, Karen D. Vo, John A. Wagner, Yaning Wang, Yow-Ming C. Wang, Peter G. Wells, Michael J. Wick, Sook Wah Yee, Ophelia Yin, Nathalie K. Zgheib, Lei Zhang, and Hao Zhu
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.