Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 16(4)2024 Mar 24.
Article in English | MEDLINE | ID: mdl-38675109

ABSTRACT

A large percentage (~60%) of prescription drugs and new molecular entities are designed for oral delivery, which requires passage through a semi-impervious membrane bilayer in the gastrointestinal wall. Passage through this bilayer can be dependent on membrane transporters that regulate the absorption of nutrients or endogenous substrates. Several investigations have provided links between nutrient, endogenous substrate, or drug absorption and the activity of certain membrane transporters. This knowledge has been key in the development of new therapeutics that can alleviate various symptoms of select diseases, such as cholestasis and diabetes. Despite this progress, recent studies revealed potential clinical dangers of unintended altered nutrient or endogenous substrate disposition due to the drug-mediated disruption of intestinal transport activity. This review outlines reports of glucose, folate, thiamine, lactate, and bile acid (re)absorption changes and consequent adverse events as examples. Finally, the need to comprehensively expand research on intestinal transporter-mediated drug interactions to avoid the unwanted disruption of homeostasis and diminish therapeutic adverse events is highlighted.

2.
Mol Pharmacol ; 101(6): 381-389, 2022 06.
Article in English | MEDLINE | ID: mdl-35383108

ABSTRACT

The organic anion transporting polypeptide family member (OATP) 1B3 is a hepatic uptake transporter that has a broad substrate recognition and plays a significant role in regulating elimination of endogenous biomolecules or xenobiotics. OATP1B3 works in tandem with OATP1B1, with which it shares approximately 80% sequence homology and a high degree of substrate overlap. Despite some substrates being recognized solely by OATP1B3, its ability to compensate for loss of OATP1B1-mediated elimination and recognition by regulatory agencies, little is known about OATP1B3 regulatory factors and how they are involved with drug-drug interaction. It was recently discovered that OATP1B1 function is mediated by the activity of a particular tyrosine kinase that is sensitive to a variety of tyrosine kinase inhibitors (TKIs). This study reports that OATP1B3 is similarly regulated, as at least 50% of its activity is reduced by 20 US Food and Drug Administration -approved TKIs. Nilotinib was assessed as the most potent OATP1B3 inhibitor among the investigated TKIs, which can occur at clinically relevant concentrations and acted predominantly through noncompetitive inhibition without impacting membrane expression. Finally, OATP1B3 function was determined to be sensitive to the knockdown of the Lck/Yes novel tyrosine kinase that is sensitive to nilotinib and has been previously implicated in mediating OATP1B1 activity. Collectively, our findings identify tyrosine kinase activity as a major regulator of OATP1B3 function which is sensitive to kinase inhibition. Given that OATP1B1 is similarly regulated, simultaneous disruption of these transporters can have drastic effects on systemic drug concentrations, which would promote adverse events. SIGNIFICANCE STATEMENT: The organic anion transporting polypeptide family member (OATP) 1B3 is a facilitator of hepatic drug elimination, although much is unknown of how OATP1B3 activity is mediated, or how such regulators contribute to drug-drug interactions. This study reports that OATP1B3 activity is dependent on the Lck/Yes novel tyrosine kinase, which is sensitive to numerous tyrosine kinase inhibitors. These findings provide insight into the occurrence of many clinical drug-drug interactions, and a rationale for future study of tyrosine kinases regulating drug disposition.


Subject(s)
Organic Anion Transporters , Protein-Tyrosine Kinases , Drug Interactions , Liver-Specific Organic Anion Transporter 1/metabolism , Membrane Transport Proteins/metabolism , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism
3.
Clin Pharmacol Ther ; 112(3): 461-484, 2022 09.
Article in English | MEDLINE | ID: mdl-35390174

ABSTRACT

Membrane transport proteins are involved in the absorption, disposition, efficacy, and/or toxicity of many drugs. Numerous mechanisms (e.g., nuclear receptors, epigenetic gene regulation, microRNAs, alternative splicing, post-translational modifications, and trafficking) regulate transport protein levels, localization, and function. Various factors associated with disease, medications, and dietary constituents, for example, may alter the regulation and activity of transport proteins in the intestine, liver, kidneys, brain, lungs, placenta, and other important sites, such as tumor tissue. This white paper reviews key mechanisms and regulatory factors that alter the function of clinically relevant transport proteins involved in drug disposition. Current considerations with in vitro and in vivo models that are used to investigate transporter regulation are discussed, including strengths, limitations, and the inherent challenges in predicting the impact of changes due to regulation of one transporter on compensatory pathways and overall drug disposition. In addition, translation and scaling of in vitro observations to in vivo outcomes are considered. The importance of incorporating altered transporter regulation in modeling and simulation approaches to predict the clinical impact on drug disposition is also discussed. Regulation of transporters is highly complex and, therefore, identification of knowledge gaps will aid in directing future research to expand our understanding of clinically relevant molecular mechanisms of transporter regulation. This information is critical to the development of tools and approaches to improve therapeutic outcomes by predicting more accurately the impact of regulation-mediated changes in transporter function on drug disposition and response.


Subject(s)
Carrier Proteins , Membrane Transport Proteins , Biological Transport , Carrier Proteins/metabolism , Gene Expression Regulation , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations , Receptors, Cytoplasmic and Nuclear/genetics
4.
Mol Pharmacol ; 100(4): 348-355, 2021 10.
Article in English | MEDLINE | ID: mdl-34330821

ABSTRACT

Cisplatin is a platinum-based drug, which remains among the most efficacious anticancer treatment options. Unfortunately, use of cisplatin is hindered by dose-limiting toxicities, including irreversible hearing loss, which can grossly affect patient quality of life. Cisplatin-induced ototoxicity is the result of cochlear hair cell damage through a mechanism that is poorly understood. However, cisplatin cytotoxicity is reliant on intracellular accumulation, a process that is largely dependent on the presence of particular membrane transporters. This review will provide an update on our current understanding of the various transporters known to be involved in the disposition and cytotoxicity of platinum drugs or their metabolites, as well as their role in mediating cisplatin-induced hearing loss. We also provide a summary of the successes and opportunities in therapeutically targeting membrane transporters to alleviate platinum-induced hearing loss. Moreover, we describe how this approach could be used to reduce the severity or onset of other adverse events associated with exposure to various forms of platinum drugs, without diminishing antitumor efficacy. SIGNIFICANCE STATEMENT: Cisplatin-induced hearing loss is a dose-limiting and irreversible adverse event with no current preventative or curative treatment measures. Pharmacological targeting of membrane transporters that regulate platinum uptake into cochlear hair cells, if conducted appropriately, may alleviate this devastating side effect and could be applied to alleviate other platinum-induced toxicities.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Drug Delivery Systems/trends , Hearing Loss/chemically induced , Hearing Loss/metabolism , Membrane Transport Proteins/metabolism , Hearing Loss/prevention & control , Humans , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/metabolism
5.
Clin Cancer Res ; 27(15): 4301-4310, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33664059

ABSTRACT

PURPOSE: OATP1B1 (SLCO1B1) is the most abundant and pharmacologically relevant uptake transporter in the liver and a key mediator of xenobiotic clearance. However, the regulatory mechanisms that determine OATP1B1 activity remain uncertain, and as a result, unexpected drug-drug interactions involving OATP1B1 substrates continue to be reported, including several involving tyrosine kinase inhibitors (TKI). EXPERIMENTAL DESIGN: OATP1B1-mediated activity in overexpressing HEK293 cells and hepatocytes was assessed in the presence of FDA-approved TKIs, while rosuvastatin pharmacokinetics in the presence of an OATP1B1 inhibiting TKI were measured in vivo. Tyrosine phosphorylation of OATP1B1 was determined by LC/MS-MS-based proteomics and transport function was measured following exposure to siRNAs targeting 779 different kinases. RESULTS: Twenty-nine of 46 FDA-approved TKIs studied significantly inhibit OATP1B1 function. Inhibition of OATP1B1 by TKIs, such as nilotinib, is predominantly noncompetitive, can increase systemic concentrations of rosuvastatin in vivo, and is associated with reduced phosphorylation of OATP1B1 at tyrosine residue 645. Using genetic screens and functional validation studies, the Src kinase LYN was identified as a potential regulator of OATP1B1 activity that is highly sensitive to inhibition by various TKIs at clinically relevant concentrations. CONCLUSIONS: A novel kinase-dependent posttranslational mechanism of OATP1B1 activation was identified and interference with this process by TKIs can influence the elimination of a broad range of xenobiotic substrates.


Subject(s)
HEK293 Cells/metabolism , Hepatocytes/metabolism , Liver-Specific Organic Anion Transporter 1/physiology , Protein-Tyrosine Kinases/physiology , Animals , Humans , Mice , Phosphorylation
6.
J Clin Invest ; 130(9): 4601-4606, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32484793

ABSTRACT

Peripheral neurotoxicity is a debilitating condition that afflicts up to 90% of patients with colorectal cancer receiving oxaliplatin-containing therapy. Although emerging evidence has highlighted the importance of various solute carriers to the toxicity of anticancer drugs, the contribution of these proteins to oxaliplatin-induced peripheral neurotoxicity remains controversial. Among candidate transporters investigated in genetically engineered mouse models, we provide evidence for a critical role of the organic cation transporter 2 (OCT2) in satellite glial cells in oxaliplatin-induced neurotoxicity, and demonstrate that targeting OCT2 using genetic and pharmacological approaches ameliorates acute and chronic forms of neurotoxicity. The relevance of this transport system was verified in transporter-deficient rats as a secondary model organism, and translational significance of preventive strategies was demonstrated in preclinical models of colorectal cancer. These studies suggest that pharmacological targeting of OCT2 could be exploited to afford neuroprotection in cancer patients requiring treatment with oxaliplatin.


Subject(s)
Neuroglia/metabolism , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Organic Cation Transporter 2/metabolism , Oxaliplatin , Animals , Female , Male , Mice , Mice, Knockout , Neuroglia/pathology , Neurons/pathology , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , Organic Cation Transporter 2/genetics , Oxaliplatin/adverse effects , Oxaliplatin/pharmacokinetics , Oxaliplatin/pharmacology , Rats
7.
Clin Pharmacol Ther ; 104(5): 799-802, 2018 11.
Article in English | MEDLINE | ID: mdl-30028006

ABSTRACT

Toxicity is among the greatest concerns during clinical use of drugs, and it has been well documented that transporter proteins facilitate such events by regulating drug accumulation. However, an important question is whether strategies could be adopted to diminish such toxicities. Although the field is currently in its infancy, some examples exist demonstrating how mechanistic understanding of the role of transporters in drug disposition can aid in alleviating transporter-mediated toxic effects.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/prevention & control , Membrane Transport Modulators/pharmacology , Membrane Transport Proteins/drug effects , Pharmaceutical Preparations/metabolism , Animals , Biological Transport , Drug Interactions , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/genetics , Drug-Related Side Effects and Adverse Reactions/metabolism , Genotype , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Pharmacogenomic Variants , Phenotype , Risk Assessment
8.
Clin Pharmacol Ther ; 104(5): 900-915, 2018 11.
Article in English | MEDLINE | ID: mdl-29756222

ABSTRACT

Drug transporters are critically important for the absorption, distribution, metabolism, and excretion (ADME) of many drugs and endogenous compounds. Therefore, disruption of these pathways by inhibition, induction, genetic polymorphisms, or disease can have profound effects on overall physiology, drug pharmacokinetics, drug efficacy, and toxicity. This white paper provides a review of changes in transporter function associated with acute and chronic disease states, describes regulatory pathways affecting transporter expression, and identifies opportunities to advance the field.


Subject(s)
Acute Disease , Chronic Disease/drug therapy , Membrane Transport Modulators/pharmacology , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Animals , Drug Interactions , Drug-Related Side Effects and Adverse Reactions/etiology , Drug-Related Side Effects and Adverse Reactions/metabolism , Humans , Membrane Transport Modulators/metabolism , Membrane Transport Proteins/genetics , Models, Biological , Risk Assessment
9.
J Clin Invest ; 128(2): 816-825, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29337310

ABSTRACT

Paclitaxel is among the most widely used anticancer drugs and is known to cause a dose-limiting peripheral neurotoxicity, the initiating mechanisms of which remain unknown. Here, we identified the murine solute carrier organic anion-transporting polypeptide B2 (OATP1B2) as a mediator of paclitaxel-induced neurotoxicity. Additionally, using established tests to assess acute and chronic paclitaxel-induced neurotoxicity, we found that genetic or pharmacologic knockout of OATP1B2 protected mice from mechanically induced allodynia, thermal hyperalgesia, and changes in digital maximal action potential amplitudes. The function of this transport system was inhibited by the tyrosine kinase inhibitor nilotinib through a noncompetitive mechanism, without compromising the anticancer properties of paclitaxel. Collectively, our findings reveal a pathway that explains the fundamental basis of paclitaxel-induced neurotoxicity, with potential implications for its therapeutic management.


Subject(s)
Hyperalgesia/chemically induced , Liver-Specific Organic Anion Transporter 1/deficiency , Liver-Specific Organic Anion Transporter 1/genetics , Paclitaxel/toxicity , Peripheral Nervous System Diseases/chemically induced , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/toxicity , Biomarkers/metabolism , Cell Line, Tumor , Genotype , HEK293 Cells , Humans , Hyperalgesia/prevention & control , Inhibitory Concentration 50 , MCF-7 Cells , Mice , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Organic Anion Transporters/genetics , Peripheral Nervous System Diseases/prevention & control , Phenotype
10.
Nat Commun ; 7: 10880, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26979622

ABSTRACT

Membrane transporters are key determinants of therapeutic outcomes. They regulate systemic and cellular drug levels influencing efficacy as well as toxicities. Here we report a unique phosphorylation-dependent interaction between drug transporters and tyrosine kinase inhibitors (TKIs), which has uncovered widespread phosphotyrosine-mediated regulation of drug transporters. We initially found that organic cation transporters (OCTs), uptake carriers of metformin and oxaliplatin, were inhibited by several clinically used TKIs. Mechanistic studies showed that these TKIs inhibit the Src family kinase Yes1, which was found to be essential for OCT2 tyrosine phosphorylation and function. Yes1 inhibition in vivo diminished OCT2 activity, significantly mitigating oxaliplatin-induced acute sensory neuropathy. Along with OCT2, other SLC-family drug transporters are potentially part of an extensive 'transporter-phosphoproteome' with unique susceptibility to TKIs. On the basis of these findings we propose that TKIs, an important and rapidly expanding class of therapeutics, can functionally modulate pharmacologically important proteins by inhibiting protein kinases essential for their post-translational regulation.


Subject(s)
Organic Cation Transport Proteins/drug effects , Phosphotyrosine/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-yes/drug effects , Animals , Antineoplastic Agents/pharmacology , Ganglia, Spinal/drug effects , HEK293 Cells , HeLa Cells , Humans , Liver-Specific Organic Anion Transporter 1 , Mice , Models, Molecular , Organic Anion Transporters/drug effects , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/drug effects , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2 , Organoplatinum Compounds/pharmacology , Oxaliplatin , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-yes/metabolism
11.
Proc Natl Acad Sci U S A ; 112(16): 5231-6, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25848011

ABSTRACT

Acute kidney injury (AKI) is a potentially fatal syndrome characterized by a rapid decline in kidney function caused by ischemic or toxic injury to renal tubular cells. The widely used chemotherapy drug cisplatin accumulates preferentially in the renal tubular cells and is a frequent cause of drug-induced AKI. During the development of AKI the quiescent tubular cells reenter the cell cycle. Strategies that block cell-cycle progression ameliorate kidney injury, possibly by averting cell division in the presence of extensive DNA damage. However, the early signaling events that lead to cell-cycle activation during AKI are not known. In the current study, using mouse models of cisplatin nephrotoxicity, we show that the G1/S-regulating cyclin-dependent kinase 4/6 (CDK4/6) pathway is activated in parallel with renal cell-cycle entry but before the development of AKI. Targeted inhibition of CDK4/6 pathway by small-molecule inhibitors palbociclib (PD-0332991) and ribociclib (LEE011) resulted in inhibition of cell-cycle progression, amelioration of kidney injury, and improved overall survival. Of additional significance, these compounds were found to be potent inhibitors of organic cation transporter 2 (OCT2), which contributes to the cellular accumulation of cisplatin and subsequent kidney injury. The unique cell-cycle and OCT2-targeting activities of palbociclib and LEE011, combined with their potential for clinical translation, support their further exploration as therapeutic candidates for prevention of AKI.


Subject(s)
Acute Kidney Injury/drug therapy , Cell Cycle Checkpoints , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Acute Kidney Injury/pathology , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Cell Cycle Checkpoints/drug effects , Cisplatin , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Humans , Kidney Tubules/drug effects , Kidney Tubules/enzymology , Kidney Tubules/pathology , Mice , Organic Cation Transport Proteins/deficiency , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2 , Piperazines/pharmacology , Piperazines/therapeutic use , Protective Agents/pharmacology , Protective Agents/therapeutic use , Purines/pharmacology , Purines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
12.
Pharmacogenomics ; 16(4): 323-32, 2015.
Article in English | MEDLINE | ID: mdl-25823781

ABSTRACT

AIM: Assuming that genetic variants of the SLC22A2 and SLC31A1 transporter affect patients' susceptibility to cisplatin-induced ototoxicity, we compared the distribution of 11 SLC22A2 variants and the SLC31A1 variant rs10981694 between patients with and without cisplatin-induced ototoxicity. PATIENTS & METHODS: Genotyping was performed in 64 pediatric patients and significant findings were re-evaluated in 66 adults. RESULTS: The SLC22A2 polymorphism rs316019 (c.808G>T; Ser270Ala) was significantly associated with protection from cisplatin-induced ototoxicity in the pediatric (p = 0.022) and the adult cohort (p = 0.048; both: Fisher's exact test). This result was confirmed by multiple logistic regression analysis accounting for age which was identified as a relevant factor for ototoxicity as well (rs316019: OR [G/T vs G/G] = 0.12, p = 0.009; age: OR [per year]: 0.84, p = 0.02). CONCLUSION: These results identified rs316019 as potential pharmacogenomic marker for cisplatin-induced ototoxicity and point to a critical role of SLC22A2 for cisplatin transport in humans and its contribution to the organ specific side effects of this drug. Original submitted 17 September 2014; Revision submitted 19 December 2014.


Subject(s)
Cation Transport Proteins/genetics , Cisplatin/adverse effects , Neoplasms/drug therapy , Organic Cation Transport Proteins/genetics , Adolescent , Adult , Child , Child, Preschool , Cisplatin/administration & dosage , Copper Transporter 1 , Drug-Related Side Effects and Adverse Reactions/genetics , Female , Genetic Association Studies , Genotype , Haplotypes , Humans , Male , Neoplasms/genetics , Neoplasms/pathology , Organic Cation Transporter 2 , Polymorphism, Single Nucleotide
13.
Clin Cancer Res ; 20(15): 4026-35, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24916697

ABSTRACT

PURPOSE: Tubular secretion of cisplatin is abolished in mice deficient for the organic cation transporters Oct1 and Oct2 (Oct1/2(-/-)mice), and these animals are protected from severe cisplatin-induced kidney damage. Since tubular necrosis is not completely absent in Oct1/2(-/-)mice, we hypothesized that alternate pathways are involved in the observed injury. EXPERIMENTAL DESIGN: Studies were done in wild-type, Oct1/2(-/-), or p53-deficient animals, all on an FVB background, receiving cisplatin intraperitoneally at 15 mg/kg. Cisplatin metabolites were analyzed using mass spectrometry, and gene expression was assessed using Affymetrix microarrays and RT-PCR arrays. RESULTS: KEGG pathway analyses on kidneys from mice exposed to cisplatin revealed that the most significantly altered genes were associated with the p53 signaling network, including Cdnk1a and Mdm2, in both wild-type (P = 2.40 × 10(-11)) and Oct1/2(-/-)mice (P = 1.92 × 10(-8)). This was confirmed by demonstrating that homozygosity for a p53-null allele partially reduced renal tubular damage, whereas loss of p53 in Oct1/2(-/-)mice (p53(-/-)/Oct1/2(-/-)) completely abolished nephrotoxicity. We found that pifithrin-α, an inhibitor of p53-dependent transcriptional activation, inhibits Oct2 and can mimic the lack of nephrotoxicity observed in p53(-/-)/Oct1/2(-/-)mice. CONCLUSIONS: These findings indicate that (i) the p53 pathway plays a crucial role in the kidney in response to cisplatin treatment and (ii) clinical exploration of OCT2 inhibitors may not lead to complete nephroprotection unless the p53 pathway is simultaneously antagonized.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Octamer Transcription Factor-1/physiology , Organic Cation Transport Proteins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Antineoplastic Agents/metabolism , Benzothiazoles/pharmacology , Biomarkers/metabolism , Cisplatin/metabolism , Female , Gene Expression Profiling , Homeodomain Proteins/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Knockout , Octamer Transcription Factor-1/antagonists & inhibitors , Oligonucleotide Array Sequence Analysis , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transporter 2 , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors
14.
Drug Metab Dispos ; 42(4): 611-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24378324

ABSTRACT

Members of the solute carrier (SLC) family of transporters are responsible for the cellular influx of a broad range of endogenous compounds and xenobiotics in multiple tissues. Many of these transporters are highly expressed in the gastrointestinal tract, liver, and kidney and are considered to be of particular importance in governing drug absorption, elimination, and cellular sensitivity of specific organs to a wide variety of oncology drugs. Although the majority of studies on the interaction of oncology drugs with SLC have been restricted to the use of exploratory in vitro model systems, emerging evidence suggests that several SLCs, including OCT2 and OATP1B1, contribute to clinically important phenotypes associated with those agents. Recent literature has indicated that modulation of SLC activity may result in drug-drug interactions, and genetic polymorphisms in SLC genes have been described that can affect the handling of substrates. Alteration of SLC function by either of these mechanisms has been demonstrated to contribute to interindividual variability in the pharmacokinetics and toxicity associated with several oncology drugs. In this report, we provide an update on this rapidly emerging field.


Subject(s)
Antineoplastic Agents , Drug-Related Side Effects and Adverse Reactions/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Drug-Related Side Effects and Adverse Reactions/etiology , Humans , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Tissue Distribution
16.
Proc Natl Acad Sci U S A ; 110(27): 11199-204, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776246

ABSTRACT

Oxaliplatin is an integral component of colorectal cancer therapy, but its clinical use is associated with a dose-limiting peripheral neurotoxicity. We found that the organic cation transporter 2 (OCT2) is expressed on dorsal root ganglia cells within the nervous system where oxaliplatin is known to accumulate. Cellular uptake of oxaliplatin was increased by 16- to 35-fold in cells overexpressing mouse Oct2 or human OCT2, and this process was associated with increased DNA platination and oxaliplatin-induced cytotoxicity. Furthermore, genetic or pharmacologic knockout of Oct2 protected mice from hypersensitivity to cold or mechanical-induced allodynia, which are established tests to assess acute oxaliplatin-induced neurotoxicity. These findings provide a rationale for the development of targeted approaches to mitigate this debilitating toxicity.


Subject(s)
Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Organic Cation Transport Proteins/physiology , Organoplatinum Compounds/toxicity , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Male , Mice , Mice, Knockout , Neurotoxicity Syndromes/genetics , Octamer Transcription Factor-1/deficiency , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-1/physiology , Organic Cation Transport Proteins/deficiency , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2 , Organoplatinum Compounds/pharmacokinetics , Oxaliplatin
17.
Mol Cancer Ther ; 12(8): 1537-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23757163

ABSTRACT

Expression of the human organic anion transporting polypeptides OATP1B1 and OATP1B3 has been previously believed to be restricted to hepatocytes. Here we show that the gene encoding OATP1B3, but not OATP1B1, is abundantly expressed in multiple human solid tumors that include hepatocellular, lung, and ovarian carcinomas. Surprisingly, OATP1B3 gene expression in a panel of 60 human tumor cell lines was linked with sensitivity to multiple cytotoxic agents, including the platinum anticancer drugs cisplatin, carboplatin, and oxaliplatin. In addition, overexpression of OATP1B3 in mammalian cells increased cellular accumulation of platinum agents and decreased cell survival. In mice with a targeted disruption of the ortholog transporter Oatp1b2, the liver-to-plasma ratio of cisplatin was significantly reduced compared with wild-type mice, without concurrent changes in expression profiles of other transporter genes. Our findings indicate an unexpected role for tumoral and host OATP1B-type carriers in the toxicity and disposition of platinum anticancer drugs, and may provide a foundation for understanding the extensive interindividual pharmacodynamic variability seen with these drugs in patients.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Organic Anion Transporters/metabolism , Platinum/metabolism , Platinum/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Biological Transport , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression , Humans , Liver-Specific Organic Anion Transporter 1 , Male , Mice , Neoplasms/genetics , Neoplasms/metabolism , Organic Anion Transporters/deficiency , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Platinum/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3
18.
Clin Cancer Res ; 19(12): 3316-24, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23640974

ABSTRACT

PURPOSE: Paclitaxel is used for the treatment of several solid tumors and displays a high interindividual variation in exposure and toxicity. Neurotoxicity is one of the most prominent side effects of paclitaxel. This study explores potential predictive pharmacokinetic and pharmacogenetic determinants for the onset and severity of neurotoxicity. EXPERIMENTAL DESIGN: In an exploratory cohort of patients (n = 261) treated with paclitaxel, neurotoxicity incidence, and severity, pharmacokinetic parameters and pharmacogenetic variants were determined. Paclitaxel plasma concentrations were measured by high-performance liquid chromatography or liquid chromatography/tandem mass spectrometry, and individual pharmacokinetic parameters were estimated from previously developed population pharmacokinetic models by nonlinear mixed effects modeling. Genetic variants of paclitaxel pharmacokinetics tested were CYP3A4*22, CYP2C8*3, CYP2C8*4, and ABCB1 3435 C>T. The association between CYP3A4*22 and neurotoxicity observed in the exploratory cohort was validated in an independent patient cohort (n = 239). RESULTS: Exposure to paclitaxel (logAUC) was correlated with severity of neurotoxicity (P < 0.00001). Female CYP3A4*22 carriers were at increased risk of developing neurotoxicity (P = 0.043) in the exploratory cohort. CYP3A4*22 carrier status itself was not associated with pharmacokinetic parameters (CL, AUC, Cmax, or T>0.05) of paclitaxel in males or females. Other genetic variants displayed no association with neurotoxicity. In the subsequent independent validation cohort, CYP3A4*22 carriers were at risk of developing grade 3 neurotoxicity (OR = 19.1; P = 0.001). CONCLUSIONS: Paclitaxel exposure showed a relationship with the severity of paclitaxel-induced neurotoxicity. In this study, female CYP3A4*22 carriers had increased risk of developing severe neurotoxicity during paclitaxel therapy. These observations may guide future individualization of paclitaxel treatment.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Genetic Association Studies , Neoplasms/complications , Neurotoxicity Syndromes/genetics , Paclitaxel/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Drug-Related Side Effects and Adverse Reactions/genetics , Female , Genotype , Humans , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/genetics , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Paclitaxel/adverse effects , Polymorphism, Single Nucleotide , Sex Characteristics
19.
Drug Metab Pharmacokinet ; 28(1): 19-27, 2013.
Article in English | MEDLINE | ID: mdl-22986709

ABSTRACT

Several solute carriers and ATP-binding cassette transporters have been implicated in the influx or efflux of platinum-based chemotherapeutic agents such as cisplatin, carboplatin, and oxaliplatin. Given that many of these proteins are highly polymorphic, the genetic status of these proteins could be an important contributor to the extensive interindividual pharmacokinetic variability associated with the clinical use of these agents. In this review article, we provide an updated overview of the various transporters that have shown promise in animal models or patient populations in facilitating the movement of platinum-based agents across cell membranes, and how their function is associated with drug disposition or pharmacodynamic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Membrane Transport Proteins/genetics , Platinum Compounds/pharmacology , Platinum Compounds/pharmacokinetics , Animals , Cation Transport Proteins/genetics , Copper Transporter 1 , Humans , Multidrug Resistance-Associated Proteins/genetics , Organic Cation Transport Proteins/genetics , Polymorphism, Single Nucleotide
20.
BMC Cancer ; 12: 381, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22938713

ABSTRACT

BACKGROUND: Since proteins involved in chemotherapy drug pharmacokinetics and pharmacodynamics have a strong impact on the uptake, metabolism, and efflux of such drugs, they likely play critical roles in resistance to chemotherapy drugs in cancer patients. METHODS: To investigate this hypothesis, we conducted a whole genome microarray study to identify difference in the expression of genes between isogenic doxorubicin-sensitive and doxorubicin-resistant MCF-7 breast tumour cells. We then assessed the degree of over-representation of doxorubicin pharmacokinetic and pharmacodynamic genes in the dataset of doxorubicin resistance genes. RESULTS: Of 27,958 Entrez genes on the array, 7.4 per cent or 2,063 genes were differentially expressed by ≥ 2-fold between wildtype and doxorubicin-resistant cells. The false discovery rate was set at 0.01 and the minimum p value for significance for any gene within the "hit list" was 0.01. Seventeen and 43 per cent of doxorubicin pharmacokinetic genes were over-represented in the hit list, depending upon whether the gene name was identical or within the same gene family, respectively. The most over-represented genes were within the 1C and 1B families of aldo-keto reductases (AKRs), which convert doxorubicin to doxorubicinol. Other genes convert doxorubicin to other metabolites or affect the influx, efflux, or cytotoxicity of the drug. In further support of the role of AKRs in doxorubicin resistance, we observed that, in comparison to doxorubicin, doxorubincol exhibited dramatically reduced cytotoxicity, reduced DNA-binding activity, and strong localization to extra nuclear lysosomes. Pharmacologic inhibition of the above AKRs in doxorubicin-resistant cells increased cellular doxorubicin levels, restored doxorubicin cytotoxicity and re-established doxorubicin localization to the nucleus. The properties of doxorubicinol were unaffected. CONCLUSIONS: These findings demonstrate the utility of using curated pharmacokinetic and pharmacodynamic knowledge bases to identify highly relevant genes associated with doxorubicin resistance. The induction of one or more of these genes was found to be correlated with changes in the drug's properties, while inhibiting one specific class of these genes (the AKRs) increased cellular doxorubicin content and restored drug DNA binding, cytotoxicity, and subcellular localization.


Subject(s)
Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , 20-Hydroxysteroid Dehydrogenases/biosynthesis , 20-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxysteroid Dehydrogenases/biosynthesis , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Aldehyde Reductase/biosynthesis , Aldo-Keto Reductase Family 1 Member C3 , Aldo-Keto Reductases , Breast Neoplasms/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cholic Acids/pharmacology , Cyclosporine/pharmacology , DNA, Neoplasm/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Gene Expression Profiling , Humans , Hydroxyprostaglandin Dehydrogenases/biosynthesis , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , MCF-7 Cells , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...