Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Expert Opin Drug Discov ; 16(4): 463-473, 2021 04.
Article in English | MEDLINE | ID: mdl-33322954

ABSTRACT

Introduction: The ability to engineer mammalian genomes in a quick and cost-effective way has led to rapid adaptation of CRISPR technology in biomedical research. CRISPR-based engineering has the potential to accelerate drug discovery, to support the reduction of high attrition rate in drug development and to enhance development of cell and gene-based therapies.Areas covered: How CRISPR technology is transforming drug discovery is discussed in this review. From target identification to target validation in both in vitro and in vivo models, CRISPR technology is positively impacting the early stages of drug development by providing a straightforward way to genome engineering. This property also attracted attention for CRISPR application in the cell and gene therapy area.Expert opinion: CRISPR technology is rapidly becoming the preferred tool for genome engineering and nowadays it is hard to imagine the drug discovery pipeline without this technology. With the years to come, CRISPR technology will undoubtedly be further refined and will flourish into a mature technology that will play a key role in supporting genome engineering requirements in the drug discovery pipeline as well as in cell and gene therapy development.


Subject(s)
CRISPR-Cas Systems/genetics , Drug Discovery/methods , Genetic Therapy/methods , Animals , Biomedical Research/methods , Cell- and Tissue-Based Therapy , Cost-Benefit Analysis , Drug Development/methods , Genetic Engineering , Humans
2.
Oncogene ; 37(17): 2251-2269, 2018 04.
Article in English | MEDLINE | ID: mdl-29391599

ABSTRACT

Trastuzumab-refractory, HER2 (human epidermal growth factor receptor 2)-positive breast cancer is commonly treated with trastuzumab emtansine (T-DM1), an antibody-drug conjugate of trastuzumab and the microtubule-targeting agent, DM1. However, drug response reduces greatly over time due to acquisition of resistance whose molecular mechanisms are mostly unknown. Here, we uncovered a novel mechanism of resistance against T-DM1 by combining whole transcriptome sequencing (RNA-Seq), proteomics and a targeted small interfering RNA (siRNA) sensitization screen for molecular level analysis of acquired and de novo T-DM1-resistant models of HER2-overexpressing breast cancer. We identified Polo-like kinase 1 (PLK1), a mitotic kinase, as a resistance mediator whose genomic as well as pharmacological inhibition restored drug sensitivity. Both acquired and de novo resistant models exhibited synergistic growth inhibition upon combination of T-DM1 with a selective PLK1 inhibitor, volasertib, at a wide concentration range of the two drugs. Mechanistically, T-DM1 sensitization upon PLK1 inhibition with volasertib was initiated by a spindle assembly checkpoint (SAC)-dependent mitotic arrest, leading to caspase activation, followed by DNA damage through CDK1-dependent phosphorylation and inactivation of Bcl-2/xL. Furthermore, we showed that Ser70 phosphorylation of Bcl-2 directly regulates apoptosis by disrupting the binding to and sequestration of the pro-apoptotic protein Bim. Importantly, T-DM1 resistance signature or PLK1 expression correlated with cell cycle progression and DNA repair, and predicted a lower sensitivity to taxane/trastuzumab combination in HER2-positive breast cancer patients. Finally, volasertib in combination with T-DM1 greatly synergized in models of T-DM1 resistance in terms of growth inhibition both in three dimensional (3D) cell culture and in vivo. Altogether, our results provide promising pre-clinical evidence for potential testing of T-DM1/volasertib combination in T-DM1 refractory HER2-positive breast cancer patients for whom there is currently no treatment available.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Maytansine/analogs & derivatives , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Trastuzumab/therapeutic use , Ado-Trastuzumab Emtansine , Animals , Breast Neoplasms/metabolism , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Drug Synergism , Female , Humans , Maytansine/therapeutic use , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , bcl-X Protein/genetics , bcl-X Protein/metabolism , Polo-Like Kinase 1
3.
Drug Resist Updat ; 27: 72-88, 2016 07.
Article in English | MEDLINE | ID: mdl-27449599

ABSTRACT

It is now widely accepted that organic anion-transporting polypeptides (OATPs), especially members of the OATP1A/1B family, can have a major impact on the disposition and elimination of a variety of endogenous molecules and drugs. Owing to their prominent expression in the sinusoidal plasma membrane of hepatocytes, OATP1B1 and OATP1B3 play key roles in the hepatic uptake and plasma clearance of a multitude of structurally diverse anti-cancer and other drugs. Here, we present a thorough assessment of the currently available OATP1A and OATP1B knockout and transgenic mouse models as key tools to study OATP functions in vivo. We discuss recent studies using these models demonstrating the importance of OATPs, primarily in the plasma and hepatic clearance of anticancer drugs such as taxanes, irinotecan/SN-38, methotrexate, doxorubicin, and platinum compounds. We further discuss recent work on OATP-mediated drug-drug interactions in these mouse models, as well as on the role of OATP1A/1B proteins in the phenomenon of hepatocyte hopping, an efficient and flexible way of liver detoxification for both endogenous and exogenous substrates. Interestingly, glucuronide conjugates of both the heme breakdown product bilirubin and the protein tyrosine kinase-targeted anticancer drug sorafenib are strongly affected by this process. The clinical relevance of variation in OATP1A/1B activity in patients has been previously revealed by the effects of polymorphic variants and drug-drug interactions on drug toxicity. The development of in vivo tools to study OATP1A/1B functions has greatly advanced our mechanistic understanding of their functional role in drug pharmacokinetics, and their implications for therapeutic efficacy and toxic side effects of anticancer and other drug treatments.


Subject(s)
Antineoplastic Agents/toxicity , Inactivation, Metabolic/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Neoplasms/metabolism , Organic Cation Transport Proteins/genetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Camptothecin/toxicity , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/toxicity , Drug Interactions , Gene Expression , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Irinotecan , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver-Specific Organic Anion Transporter 1/metabolism , Methotrexate/pharmacokinetics , Methotrexate/pharmacology , Methotrexate/toxicity , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Niacinamide/analogs & derivatives , Niacinamide/pharmacokinetics , Niacinamide/pharmacology , Niacinamide/toxicity , Organic Cation Transport Proteins/deficiency , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Phenylurea Compounds/toxicity , Platinum Compounds/pharmacokinetics , Platinum Compounds/pharmacology , Platinum Compounds/toxicity , Sorafenib , Taxoids/pharmacokinetics , Taxoids/pharmacology , Taxoids/toxicity
4.
Mol Pharm ; 12(12): 4259-69, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26474710

ABSTRACT

The impact of OATP drug uptake transporters in drug-drug interactions (DDIs) is increasingly recognized. OATP1B1 and OATP1B3 are human hepatic uptake transporters that can mediate liver uptake of a wide variety of drugs. Recently, we generated transgenic mice with liver-specific expression of human OATP1B1 or OATP1B3 in a mouse Oatp1a/1b knockout background. Here, we investigated the applicability of these mice in OATP-mediated drug-drug interaction studies using the prototypic OATP inhibitor rifampicin and a good OATP substrate, the anticancer drug methotrexate (MTX). We next assessed the possibility of OATP-mediated interactions between telmisartan and MTX, a clinically relevant drug combination. Using HEK293 cells overexpressing OATP1B1 or OATP1B3, we estimated IC50 values for both rifampicin (0.9 or 0.3 µM) and telmisartan (6.7 or 7.9 µM) in inhibiting OATP-mediated MTX uptake in vitro. Using wild-type, Oatp1a/1b-/-, and OATP1B1- or OATP1B3-humanized transgenic mice, we found that rifampicin inhibits hepatic uptake of MTX mediated by the mouse Oatp1a/1b and human OATP1B1 and OATP1B3 transporters at clinically relevant concentrations. This highlights the applicability of these mouse models for DDI studies and may be exploited in the clinic to reduce the dose and thus methotrexate-mediated toxicity. On the other hand, telmisartan inhibited only human OATP1B1-mediated hepatic uptake of MTX at concentrations higher than those used in the clinic; therefore risks for OATP-mediated clinical DDIs for this drug combination are likely to be low. Overall, we show here that OATP1B1- and OATP1B3-humanized mice can be used as in vivo tools to assess and possibly predict clinically relevant DDIs.


Subject(s)
Drug Interactions/physiology , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Animals , Antineoplastic Agents/metabolism , Benzimidazoles/metabolism , Benzoates/metabolism , Biological Transport/physiology , HEK293 Cells , Humans , Liver/metabolism , Liver-Specific Organic Anion Transporter 1 , Male , Membrane Transport Proteins/metabolism , Methotrexate/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Solute Carrier Organic Anion Transporter Family Member 1B3 , Telmisartan
5.
J Inorg Biochem ; 152: 38-52, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26339715

ABSTRACT

Four new cationic Pd(II) and Pt(II) 5,5-diethylbarbiturate (barb) complexes, [M(barb)(bpma)]X·H2O [M = Pd(II), X = Cl (1); M = Pt(II), X = NO3(-) (2)] and [M(barb)(terpy)]NO3·0.5H2O [M = Pd(II) (3); M = Pt(II) (4)], where bpma = bis(2-pyridylmethyl)amine and terpy = terpyridine, were synthesized and characterized by elemental analysis, IR, UV­vis, NMR, ESI-MS and X-ray crystallography. The DNA binding properties of the cationic complexes were investigated by spectroscopic titrations, displacement experiments, viscosity, DNA melting and electrophoresis measurements. The results revealed that the complexes effectively bind to FS-DNA (fish sperm DNA) via intercalative/minor groove binding modes with intrinsic binding constants (Kb) in the range of 0.50 × 10(4)­1.67 × 10(5) M(-1). Absorption, emission and synchronous fluorescence measurements showed strong association of the complexes with protein (BSA) through a static mechanism. The mode of interaction of complexes towards DNA and protein was also supported by molecular docking. Complexes 1 and 3 showed significant nuclear uptake in HT-29 cells. In addition, 1 and 3 showed higher inhibition than cisplatin on the growth of MCF-7 and HT-29 cells and induced apoptosis on these cells much more effectively than the rest of the complexes as evidenced by pyknotic nuclear morphology. The levels of caspase-cleaved cytokeratin 18 (M30 antigen) in HT-29 cells treated with 1 and 3 increased in a dose-dependent manner, suggesting apoptosis. Moreover, qRT-PCR experiments showed that 1 and 3 caused significant increases in the expression of TNFRSF10B in HT-29 cells, indicating the initiation of apoptosis via cell surface death receptors.


Subject(s)
Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Barbiturates/chemistry , DNA/metabolism , Organoplatinum Compounds/chemical synthesis , Palladium/chemistry , Serum Albumin, Bovine/metabolism , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Base Sequence , DNA/chemistry , HT29 Cells , Humans , MCF-7 Cells , Molecular Docking Simulation , Molecular Sequence Data , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/toxicity , Protein Binding , Pyridines/chemistry , Serum Albumin, Bovine/chemistry
6.
Mol Pharm ; 12(10): 3714-23, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26317243

ABSTRACT

We aimed to clarify the roles of the multidrug-detoxifying proteins ABCB1, ABCG2, ABCC2, and CYP3A in oral availability and brain accumulation of cabazitaxel, a taxane developed for improved therapy of docetaxel-resistant prostate cancer. Cabazitaxel pharmacokinetics were studied in Abcb1a/1b, Abcg2, Abcc2, Cyp3a, and combination knockout mice. We found that human ABCB1, but not ABCG2, transported cabazitaxel in vitro. Upon oral cabazitaxel administration, total plasma levels were greatly increased due to binding to plasma carboxylesterase Ces1c, which is highly upregulated in several knockout strains. Ces1c inhibition and in vivo hepatic Ces1c knockdown reversed these effects. Correcting for Ces1c effects, Abcb1a/1b, Abcg2, and Abcc2 did not restrict cabazitaxel oral availability, whereas Abcb1a/1b, but not Abcg2, dramatically reduced cabazitaxel brain accumulation (>10-fold). Coadministration of the ABCB1 inhibitor elacridar completely reversed this brain accumulation effect. After correction for Ces1c effects, Cyp3a knockout mice demonstrated a strong (six-fold) increase in cabazitaxel oral availability, which was completely reversed by transgenic human CYP3A4 in intestine and liver. Cabazitaxel markedly inhibited mouse Ces1c, but human CES1 and CES2 only weakly. Ces1c upregulation can thus complicate preclinical cabazitaxel studies. In summary, ABCB1 limits cabazitaxel brain accumulation and therefore potentially therapeutic efficacy against (micro)metastases or primary tumors positioned wholly or partly behind a functional blood-brain barrier. This can be reversed with elacridar coadministration, and similar effects may apply to ABCB1-expressing tumors. CYP3A4 profoundly reduces the oral availability of cabazitaxel. This may potentially be greatly improved by coadministering ritonavir or other CYP3A inhibitors, suggesting the option of patient-friendly oral cabazitaxel therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacokinetics , Brain Chemistry , Carboxylesterase/blood , Cytochrome P-450 CYP3A/metabolism , Taxoids/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/analysis , Carboxylic Ester Hydrolases/metabolism , Dogs , Madin Darby Canine Kidney Cells/metabolism , Male , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Taxoids/administration & dosage , Taxoids/analysis
7.
Cancer Res ; 75(13): 2729-36, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25952649

ABSTRACT

Recently, an efficient liver detoxification process dubbed "hepatocyte hopping" was proposed on the basis of findings with the endogenous compound, bilirubin glucuronide. According to this model, hepatocytic bilirubin glucuronide can follow a liver-to-blood shuttling loop via Abcc3 transporter-mediated efflux and subsequent Oatp1a/1b-mediated liver uptake. We hypothesized that glucuronide conjugates of xenobiotics, such as the anticancer drug sorafenib, can also undergo hepatocyte hopping. Using transporter-deficient mouse models, we show here that sorafenib-glucuronide can be extruded from hepatocytes into the bile by Abcc2 or back into the systemic circulation by Abcc3, and that it can be taken up efficiently again into neighboring hepatocytes by Oatp1a/1b. We further demonstrate that sorafenib-glucuronide excreted into the gut lumen can be cleaved by microbial enzymes to sorafenib, which is then reabsorbed, supporting its persistence in the systemic circulation. Our results suggest broad relevance of a hepatocyte shuttling process known as "hepatocyte hopping"-a novel concept in clinical pharmacology-for detoxification of targeted cancer drugs that undergo hepatic glucuronidation, such as sorafenib.


Subject(s)
Glucuronides/metabolism , Hepatocytes/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Niacinamide/analogs & derivatives , Organic Cation Transport Proteins/metabolism , Phenylurea Compounds/pharmacokinetics , Animals , Female , Glucuronidase/metabolism , Glucuronides/blood , Humans , Intestines/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Niacinamide/blood , Niacinamide/pharmacokinetics , Phenylurea Compounds/blood , Rats , Sf9 Cells , Sorafenib
8.
Adv Cancer Res ; 125: 1-41, 2015.
Article in English | MEDLINE | ID: mdl-25640265

ABSTRACT

ATP-binding cassette (ABC) transporters are transmembrane efflux transporters that mediate cellular extrusion of a broad range of substrates ranging from amino acids, lipids, and ions to xenobiotics including many anticancer drugs. ABCB1 (P-GP) and ABCG2 (BCRP) are the most extensively studied apical ABC drug efflux transporters. They are highly expressed in apical membranes of many pharmacokinetically relevant tissues such as epithelial cells of the small intestine and endothelial cells of the blood capillaries in brain and testis, and in the placental maternal-fetal barrier. In these tissues, they have a protective function as they efflux their substrates back to the intestinal lumen or blood and thus restrict the intestinal uptake and tissue disposition of many compounds. This presents a major challenge for the use of many (anticancer) drugs, as most currently used anticancer drugs are substrates of these transporters. Herein, we review the latest findings on the role of apical ABC transporters in the disposition of anticancer drugs. We discuss that many new, rationally designed anticancer drugs are substrates of these transporters and that their oral availability and/or brain disposition are affected by this interaction. We also summarize studies that investigate the improvement of oral availability and brain disposition of many cytotoxic (e.g., taxanes) and rationally designed (e.g., tyrosine kinase inhibitor) anticancer drugs, using chemical inhibitors of these transporters. These findings provide a better understanding of the importance of apical ABC transporters in chemotherapy and may therefore advance translation of promising preclinical insights and approaches to clinical studies.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/pharmacokinetics , Drug Resistance, Neoplasm/physiology , Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Biological Availability , Biological Transport/physiology , Blood-Brain Barrier/physiology , Bridged-Ring Compounds/metabolism , Bridged-Ring Compounds/pharmacokinetics , Bridged-Ring Compounds/therapeutic use , Humans , Intestinal Absorption/physiology , Mice , Neoplasms/metabolism , Taxoids/metabolism , Taxoids/pharmacokinetics , Taxoids/therapeutic use
9.
Pharm Res ; 32(7): 2205-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25563977

ABSTRACT

PURPOSE: Regorafenib is a novel multikinase inhibitor, currently approved for the treatment of metastasized colorectal cancer and advanced gastrointestinal stromal tumors. We investigated whether regorafenib is a substrate for the multidrug efflux transporters ABCG2 and ABCB1 and whether oral availability, brain and testis accumulation of regorafenib and its active metabolites are influenced by these transporters. METHODS: We used in vitro transport assays to assess human (h)ABCB1- or hABCG2- or murine (m)Abcg2-mediated active transport at high and low concentrations of regorafenib. To study the single and combined roles of Abcg2 and Abcb1a/1b in oral regorafenib disposition and the impact of Cyp3a-mediated metabolism, we used appropriate knockout mouse strains. RESULTS: Regorafenib was transported well by mAbcg2 and hABCG2 and modestly by hABCB1 in vitro. Abcg2 and to a lesser extent Abcb1a/1b limited brain and testis accumulation of regorafenib and metabolite M2 (brain only) in mice. Regorafenib oral availability was not increased in Abcg2(-/-);Abcb1a/1b(-/-) mice. Up till 2 h, metabolite M5 was undetectable in plasma and organs. CONCLUSIONS: Brain and testis accumulation of regorafenib and brain accumulation of metabolite M2 are restricted by Abcg2 and Abcb1a/1b. Inhibition of these transporters may be of clinical relevance for patients with brain (micro)metastases positioned behind an intact blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/pharmacokinetics , Brain/metabolism , Neoplasm Proteins/metabolism , Phenylurea Compounds/pharmacokinetics , Pyridines/pharmacokinetics , Testis/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Administration, Oral , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Biological Transport , Dogs , Humans , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Phenylurea Compounds/blood , Phenylurea Compounds/metabolism , Pyridines/blood , Pyridines/metabolism , Tissue Distribution , Transfection
10.
Pharm Res ; 32(1): 37-46, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24962512

ABSTRACT

BACKGROUND: Rucaparib is a potent, orally available, small-molecule inhibitor of poly ADP-ribose polymerase (PARP) 1 and 2. Ongoing clinical trials are assessing the efficacy of rucaparib alone or in combination with other cytotoxic drugs, mainly in breast and ovarian cancer patients with mutations in the breast cancer associated (BRCA) genes. PURPOSE: We aimed to establish whether the multidrug efflux transporters ABCG2 (BCRP) and ABCB1 (P-gp, MDR1) affect the oral availability and brain penetration of rucaparib in mice. RESULTS: In vitro, rucaparib was efficiently transported by both human ABCB1 and ABCG2, and very efficiently by mouse Abcg2. Transport could be inhibited by the small-molecule ABCB1 and ABCG2 inhibitors zosuquidar and Ko143, respectively. In vivo, oral availability (plasma AUC0-1 and AUC0-24) and brain levels of rucaparib at 1 and 24 h were increased by the absence of both Abcg2 and Abcb1a/1b after oral administration of rucaparib at 10 mg/kg. CONCLUSIONS: Our data show to our knowledge for the first time that oral availability and brain accumulation of a PARP inhibitor are markedly and additively restricted by Abcg2 and Abcb1a/1b. This may have clinical relevance for improvement of rucaparib therapy in PARP inhibitor-resistant tumors with ABCB1 and/or ABCG2 expression and in patients with brain (micro)metastases positioned behind a functional blood-brain barrier.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Brain/metabolism , Indoles/pharmacokinetics , Neoplasm Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Administration, Oral , Animals , Biological Availability , Biological Transport , Cell Culture Techniques , Dogs , Female , Humans , Indoles/administration & dosage , Indoles/blood , Madin Darby Canine Kidney Cells , Mice, Knockout , Neoplasm Proteins/genetics , Substrate Specificity , Tissue Distribution
11.
Clin Cancer Res ; 20(12): 3133-45, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24727322

ABSTRACT

PURPOSE: To clarify the role of ABCB1, ABCG2, and CYP3A in blood and brain exposure of everolimus using knockout mouse models. EXPERIMENTAL DESIGN: We used wild-type, Abcb1a/1b(-/-), Abcg2(-/-), Abcb1a/1b;Abcg2(-/-), and Cyp3a(-/-) mice to study everolimus oral bioavailability and brain accumulation. RESULTS: Following everolimus administration, brain concentrations and brain-to-liver ratios were substantially increased in Abcb1a/1b(-/-)and Abcb1a/1b;Abcg2(-/-), but not Abcg2(-/-)mice. The fraction of everolimus located in the plasma compartment was highly increased in all knockout strains. In vitro, everolimus was rapidly degraded in wild-type but not knockout plasma. Carboxylesterase 1c (Ces1c), a plasma carboxylesterase gene, was highly upregulated (∼80-fold) in the liver of knockout mice relative to wild-type mice, and plasma Ces1c likely protected everolimus from degradation by binding and stabilizing it. This binding was prevented by preincubation with the carboxylesterase inhibitor BNPP. In vivo knockdown experiments confirmed the involvement of Ces1c in everolimus stabilization. Everolimus also markedly inhibited the hydrolysis of irinotecan and p-nitrophenyl acetate by mouse plasma carboxylesterase and recombinant human CES2, respectively. After correcting for carboxylesterase binding, Cyp3a(-/-), but not Abcb1a/1b(-/-), Abcg2(-/-), or Abcb1a/1b;Abcg2(-/-)mice, displayed highly (>5-fold) increased oral availability of everolimus. CONCLUSIONS: Brain accumulation of everolimus was restricted by Abcb1, but not Abcg2, suggesting the use of coadministered ABCB1 inhibitors to improve brain tumor treatment. Cyp3a, but not Abcb1a/1b, restricted everolimus oral availability, underscoring drug-drug interaction risks via CYP3A. Upregulated Ces1c likely mediated the tight binding and stabilization of everolimus, causing higher plasma retention in knockout strains. This Ces upregulation might confound other pharmacologic studies.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/physiology , ATP-Binding Cassette Transporters/physiology , Brain Neoplasms/drug therapy , Carboxylic Ester Hydrolases/blood , Cytochrome P-450 Enzyme System/physiology , Immunosuppressive Agents/pharmacology , Sirolimus/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Biological Availability , Biological Transport , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cytochrome P-450 CYP3A , Everolimus , Humans , Immunosuppressive Agents/pharmacokinetics , Mice , Mice, Knockout , Sirolimus/pharmacokinetics , Sirolimus/pharmacology , Tissue Distribution
12.
Biomed Chromatogr ; 28(10): 1366-70, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24619951

ABSTRACT

Regorafenib has recently been approved for the treatment of colorectal cancer. A bioanalytical liquid chromatography-tandem mass spectrometric assay for this multikinase inhibitor was developed and validated in plasma. The concentration range of the assay was 25-25,000 ng/mL. Protein precipitation with acetonitrile was used as sample pre-treatment with sorafenib as internal standard. The extract was diluted with methanol (25%, v/v) and then injected onto the sub-2 µm particle, bridged ethylsilicia hybrid trifunctional bonded C18 column. Isocratic elution using 0.02% (v/v) formic acid in a methanol-water mixture was used. Compounds were monitored by a triple quadrupole mass spectrometer in the selected reaction monitoring mode after positive electrospray ionization. Double logarithmic calibration was used; within-day precisions, between-day precisions, and accuracies were 3.2-9.2, 4.1-12.3 and 94.8-103.0%, respectively. High drug stability was observed under all relevant storage conditions. The assay was used to measure drug concentrations in a pharmacokinetic study in wild-type FVB mice.


Subject(s)
Chromatography, Liquid/methods , Phenylurea Compounds/blood , Pyridines/blood , Tandem Mass Spectrometry/methods , Animals , Humans , Linear Models , Mice , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
13.
Int J Cancer ; 135(7): 1700-10, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24554572

ABSTRACT

Organic anion-transporting polypeptides (OATPs) are important drug uptake transporters, mediating distribution of substrates to several pharmacokinetically relevant organs. Doxorubicin is a widely used anti-cancer drug extensively studied for its interactions with various drug transporters, but not OATPs. Here, we investigated the role of OATP1A/1B proteins in the distribution of doxorubicin. In vitro, we observed ∼ 2-fold increased doxorubicin uptake in HEK293 cells overexpressing human OATP1A2, but not OATP1B1 or OATP1B3. In mice, absence of Oatp1a/1b transporters led to up to 2-fold higher doxorubicin plasma concentrations and 1.3-fold higher plasma AUC. Conversely, liver AUC and liver-to-plasma ratios of Oatp1a/1b(-/-) mice were 1.4-fold and up to 4.1-fold lower than in wild-type mice, respectively. Decreased doxorubicin levels in the small intestinal content reflected those in the liver, indicating a reduced biliary excretion of doxorubicin in Oatp1a/1b(-/-) mice. These results demonstrate important control of doxorubicin plasma clearance and hepatic uptake by mouse Oatp1a/1b transporters. This is unexpected, as the fairly hydrophobic weak base doxorubicin is an atypical OATP1A/1B substrate. Interestingly, transgenic liver-specific expression of human OATP1A2, OATP1B1 or OATP1B3 could partially rescue the increased doxorubicin plasma levels of Oatp1a/1b(-/-) mice. Hepatic uptake and bile-derived intestinal excretion of doxorubicin were completely reverted to wild-type levels by OATP1A2, and partially by OATP1B1 and OATP1B3. Thus, doxorubicin is transported by hepatocyte-expressed OATP1A2, -1B1 and -1B3 in vivo, illustrating an unexpectedly wide substrate specificity. These findings have possible implications for the uptake, disposition, therapy response and toxicity of doxorubicin, also in human tumors and tissues expressing these transporters.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Doxorubicin/pharmacokinetics , Liver/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/physiology , Animals , Antineoplastic Agents/administration & dosage , Biological Transport , Blotting, Western , Chromatography, High Pressure Liquid , Doxorubicin/administration & dosage , Female , HEK293 Cells , Humans , Liver/drug effects , Liver-Specific Organic Anion Transporter 1 , Mice , Mice, Transgenic , Solute Carrier Organic Anion Transporter Family Member 1B3 , Tissue Distribution
14.
J Pharm Biomed Anal ; 88: 626-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24216281

ABSTRACT

A quantitative bioanalytical liquid chromatography-tandem mass spectrometric (LC-MS/MS) assay for the poly(ADP-ribose) polymerase-1 inhibitor rucaparib was developed and validated. Plasma samples were pre-treated using protein precipitation with acetonitrile containing gefitinib as internal standard. Diluted extract was directly injected into the reversed-phase chromatographic system. The eluate was transferred into the electrospray interface with positive ionization and the analyte was detected in the selected reaction monitoring mode of a triple quadrupole mass spectrometer. The assay was validated in a 1.25-2000ng/ml calibration range with r(2)=0.9958±0.0012 for linear regression with quadratic weighting (n=6). Within day precisions (n=18) were 2.0-5.4%, between day (3 days; n=18) precisions 3.2-8.0% and accuracies (n=18) were 89.7-93.2%. At the lower limit of quantification (1.25ng/ml) these parameters were 9.6%, 13.7% and 85.3%, respectively. The drug was sufficiently stable under all relevant analytical conditions. Finally, the assay was successfully used to determine drug pharmacokinetics in female FVB wild type mice.


Subject(s)
Indoles/blood , Poly(ADP-ribose) Polymerase Inhibitors , Acetonitriles/chemistry , Animals , Area Under Curve , Calibration , Chromatography, Liquid , Edetic Acid/chemistry , Female , Gefitinib , Indoles/pharmacokinetics , Mice , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Quality Control , Quinazolines/chemistry , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
15.
Article in English | MEDLINE | ID: mdl-23892825

ABSTRACT

A bioanalytical liquid chromatography-tandem mass spectrometry assay for the tyrosine kinase inhibitor pelitinib was developed and validated in plasma. Acetonitrile containing the internal standard erlotinib was used to precipitate proteins. The extract was diluted with water and then directly injected onto the sub-2µm particle, bridged ethylsilica hybrid trifunctional bonded C18 column. A gradient consisting of 0.02% (v/v) formic acid in a methanol-water mixture was used. The ionization mode of the electrospray interface was positive and the analyte was detected by a triple quadrupole mass spectrometer in the selected reaction monitoring mode. The calibration range of the assay was 1-200ng/ml. The within day precision, the between day precision, and the accuracy were 3.5-7.4%, 4.5-8.6%, and 94.0-104.8%, respectively. The stability of the drug was sufficient under all relevant conditions. The validated assay was used to measure drug levels in wild-type FVB mice and pharmacokinetic parameters were assessed.


Subject(s)
Aminoquinolines/blood , Aniline Compounds/blood , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/blood , Aminoquinolines/pharmacokinetics , Aniline Compounds/pharmacokinetics , Animals , Drug Stability , ErbB Receptors/metabolism , Humans , Male , Mice , Protein Kinase Inhibitors/pharmacokinetics , Tandem Mass Spectrometry
16.
Article in English | MEDLINE | ID: mdl-23537694

ABSTRACT

A quantitative bioanalytical liquid chromatography-tandem mass spectrometric (LC-MS/MS) assay for the mutated BRAF inhibitor dabrafenib was developed and validated. Plasma samples were pre-treated using protein precipitation with acetonitrile containing PLX4720 as internal standard. The extract was directly injected into the reversed-phase chromatographic system after dilution with water. The eluate was transferred into the electrospray interface with positive ionization and the analyte was detected in the selected reaction monitoring mode of a triple quadrupole mass spectrometer. The assay was validated in a 2-2000 ng/ml calibration range with r(2)=0.993±0.002 for linear regression with quadratic weighting (n=5). Within day precisions (n=6) were 3.3-5.2%, between day (3 days; n=18) precisions 4.7-8.2%. Accuracies were between 95-104% for the whole calibration range. The drug was sufficiently stable under all relevant analytical conditions. Finally, the assay was successfully used to determine drug pharmacokinetics in mice.


Subject(s)
Chromatography, Liquid/methods , Imidazoles/blood , Oximes/blood , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Tandem Mass Spectrometry/methods , Animals , Female , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Linear Models , Mice , Oximes/chemistry , Oximes/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/methods
17.
Mol Pharm ; 9(11): 3236-45, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23020847

ABSTRACT

Vemurafenib (PLX4032) is a novel tyrosine kinase inhibitor that has clinical efficacy against metastatic melanoma harboring a BRAF(V600E) mutation. We aimed to establish whether oral availability and brain penetration of vemurafenib could be restricted by the multidrug efflux transporters P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2), as these might limit therapeutic efficacy, especially against brain metastases. In vitro, vemurafenib was efficiently transported by both human ABCB1 and ABCG2, and very efficiently by mouse Abcg2, but not by mouse Abcc2. Upon oral administration of vemurafenib (5 mg/kg), Abcb1a/1b(-/-) mice had a 1.6-fold increased, Abcg2(-/-) mice a 2.3-fold increased, and Abcb1a/1b(-/-);Abcg2(-/-) mice a 6.6-fold increased plasma AUC, respectively, compared to wild-type (WT) mice, indicating a marked and additive role of these transporters in limiting vemurafenib oral availability. Brain-to-plasma ratios of vemurafenib (oral, 25 mg/kg) were not increased in Abcg2(-/-) mice, only 1.7-fold in Abcb1a/1b(-/-) mice, but 21.4-fold in Abcb1a/1b(-/-);Abcg2(-/-) mice, indicating pronounced overlapping functions of these transporters in reducing vemurafenib brain accumulation. Oral coadministration of the dual ABCB1 and ABCG2 inhibitor elacridar almost completely eliminated the roles of Abcb1 and Abcg2 in restricting oral availability and brain accumulation of vemurafenib. As predicted by previously described pharmacokinetic modeling, halving the amount of active efflux transport at the WT blood-brain barrier by testing heterozygous Abcb1a/1b(+/-);Abcg2(+/-) mice had little impact on vemurafenib brain accumulation. Our data suggest that elacridar coadministration may be considered to improve the therapeutic efficacy of vemurafenib, especially for brain metastases located behind a functional blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP-Binding Cassette Transporters/antagonists & inhibitors , Acridines/pharmacology , Blood-Brain Barrier/drug effects , Indoles/pharmacokinetics , Sulfonamides/pharmacokinetics , Tetrahydroisoquinolines/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/physiology , Administration, Oral , Animals , Biological Availability , Biological Transport , Drug Combinations , Female , Humans , Indoles/administration & dosage , Indoles/pharmacology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Tissue Distribution , Vemurafenib
18.
Article in English | MEDLINE | ID: mdl-22476054

ABSTRACT

A quantitative bioanalytical liquid chromatography-tandem mass spectrometric (LC-MS/MS) assay for the JAK2 inhibitor CYT387 was developed and validated. Plasma samples were pre-treated using protein precipitation with acetonitrile containing cediranib as internal standard. The extract was directly injected into the chromatographic system after dilution with water. This system consisted of a sub-2 µm particle, trifunctional bonded octadecyl silica column with a gradient using 0.005% (v/v) of formic acid in a mixture of water and methanol. The eluate was transferred into the electrospray interface with positive ionization and the analyte was detected in the selected reaction monitoring mode of a triple quadrupole mass spectrometer. The assay was validated in a 0.25-1000 ng/ml calibration range. Within day precisions were 3.0-13.5%, between day precisions 5.7% and 14.5%. Accuracies were between 96% and 113% for the whole calibration range. The drug was stable under all relevant analytical conditions. Finally, the assay was successfully used to assess drug levels in mice.


Subject(s)
Benzamides/blood , Chromatography, Liquid/methods , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/blood , Pyrimidines/blood , Tandem Mass Spectrometry/methods , Animals , Benzamides/pharmacokinetics , Benzamides/pharmacology , Calibration , Drug Stability , Humans , Linear Models , Mice , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization
19.
J Chromatogr B Analyt Technol Biomed Life Sci ; 895-896: 169-73, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22476055

ABSTRACT

A quantitative bioanalytical assay for cediranib and its N(+)-glucuronide metabolite was developed and validated. Human plasma samples were pre-treated using protein precipitation with acetonitrile containing erlotinib and CYT-387 as internal standards for the glucuronide metabolite and parent compound, respectively. The extract was diluted with water and injected into the chromatographic system. This system consisted of sub-2 µm particles, a trifunctional bonded octadecyl silica column with gradient elution using 0.005% (v/v) of formic acid in a mixture of water and methanol. The eluate was transferred into the electrospray interface with positive ionization and the analytes were detected in the selected reaction monitoring mode of a triple quadrupole mass spectrometer. The assay was validated in a 1-290 ng/ml calibration range for cediranib and 0.2-52 ng/ml for its glucuronide metabolite. The lowest levels of these ranges corresponded to the lower limits of quantification for both compounds. Within day precisions were 4.0-6.7% for cediranib and 4.1-11.9% for its glucuronide, between day precisions were 4.2-10.2 and 4.8-14.4% and accuracies were between 99 and 106 and 84 and 94% for cediranib and its metabolite, respectively. Stabilities of both compounds were sufficient under all relevant conditions. Finally, the assay was successfully used to assess drug levels in a pharmacokinetic mouse study.


Subject(s)
Chromatography, Liquid/methods , Glucuronides/blood , Quinazolines/blood , Tandem Mass Spectrometry/methods , Animals , Benzamides/chemistry , Glucuronides/metabolism , Glucuronides/pharmacokinetics , Humans , Linear Models , Mice , Pyrimidines/chemistry , Quinazolines/metabolism , Quinazolines/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Reproducibility of Results , Sensitivity and Specificity
20.
Article in English | MEDLINE | ID: mdl-22386128

ABSTRACT

A bioanalytical assay for the mutated BRAF inhibitor vemurafenib was developed and validated. For the quantitative assay, human plasma samples were pre-treated using protein precipitation with water-acetonitrile (1/3, v/v) containing sorafenib as internal standard. The extract was directly injected into the chromatographic system. This system consisted of a sub-2 µm particle, trifunctional bonded octadecyl silica column with isocratic elution using 0.01% (v/v) of formic acid in a mixture of water and methanol. The eluate was transferred into the electrospray interface with positive ionization and the analyte was detected in the selected reaction monitoring mode of a triple quadrupole mass spectrometer. The assay was validated in a 0.1-100 µg/ml calibration range. Within day precisions were 1.6-3.2%, between day precisions 2.7% and 8.2% and accuracies were between 99% and 106% for the whole calibration range. The drug was stable under all relevant conditions. Finally, the assay was successfully used to assess drug levels in a pharmacokinetic mouse study.


Subject(s)
Chromatography, Liquid/methods , Indoles/blood , Protein Kinase Inhibitors/blood , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/blood , Tandem Mass Spectrometry/methods , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Drug Stability , Humans , Indoles/pharmacokinetics , Mice , Protein Kinase Inhibitors/pharmacokinetics , Regression Analysis , Reproducibility of Results , Sulfonamides/pharmacokinetics , Vemurafenib
SELECTION OF CITATIONS
SEARCH DETAIL
...