36 results on '"Eric D. Eisenmann"'
Search Results
2. Exploring pharmacokinetics of talazoparib in ABCB1/ABCG2-deficient mice using a novel UHPLC-MS/MS method
- Author
-
Zahra Talebi, Dominique A. Garrison, Eric D. Eisenmann, Kalindi Parmar, Geoffrey I. Shapiro, Michelle A. Rudek, Alex Sparreboom, and Yan Jin
- Subjects
ABC transporters ,Mouse plasma ,Pharmacokinetics ,Talazoparib ,UHPLC-MS/MS ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
A rapid, sensitive, and simple UHPLC-MS/MS method for the determination of the PARP inhibitor talazoparib in mouse plasma was developed and validated using [13C,2H4]-talazoparib as an internal standard (IS). The assay procedure involved extraction of talazoparib and the IS from plasma using a single-step deproteination and separation of the analytes was achieved on an ACQUITY UPLC RP18 HSS T3 column with a mobile phase gradient at a flow rate of 0.4 mL/min in a run time of 5 min. The calibration curve was linear (r2 > 0.99) over the concentration range of 0.5–100 ng/mL, and 10-fold dilution of samples could be accurately quantitated. The matrix effect and mean extraction recovery for talazoparib were between 93.7-109% and 87.7–105%, respectively. Precision and percent bias of quality control samples were always less than ±15%, indicating reproducibility and accuracy of the method. Talazoparib demonstrated bench-top stability at room temperature for 6 h, auto-sampler and reinjection stability at 4 °C for at least 24 h, and no significant degradation was observed after three freeze-thaw cycles. The developed method was successfully applied to pharmacokinetic studies involving serial blood sampling after oral administration of talazoparib to wild-type mice and animals with a genetic deficiency of the efflux transporters ABCB1 (P-gp) and ABCG2 (BCRP). Together, our results demonstrate the successful development of a suitable analytical method for talazoparib in mouse plasma and suggest that mice are a useful model to evaluate transporter-mediated drug-drug interactions involving therapy with talazoparib.
- Published
- 2023
- Full Text
- View/download PDF
3. Itraconazole-Induced Increases in Gilteritinib Exposure Are Mediated by CYP3A and OATP1B
- Author
-
Dominique A. Garrison, Yan Jin, Zahra Talebi, Shuiying Hu, Alex Sparreboom, Sharyn D. Baker, and Eric D. Eisenmann
- Subjects
gilteritinib ,OATP1B ,itraconazole ,pharmacokinetics ,drug–drug interactions ,CYP3A ,Organic chemistry ,QD241-441 - Abstract
Gilteritinib, an FDA-approved tyrosine kinase inhibitor approved for the treatment of relapsed/refractory FLT3-mutated acute myeloid leukemia, is primarily eliminated via CYP3A4-mediated metabolism, a pathway that is sensitive to the co-administration of known CYP3A4 inhibitors, such as itraconazole. However, the precise mechanism by which itraconazole and other CYP3A-modulating drugs affect the absorption and disposition of gilteritinib remains unclear. In the present investigation, we demonstrate that pretreatment with itraconazole is associated with a significant increase in the systemic exposure to gilteritinib in mice, recapitulating the observed clinical drug–drug interaction. However, the plasma levels of gilteritinib were only modestly increased in CYP3A-deficient mice and not further influenced by itraconazole. Ensuing in vitro and in vivo studies revealed that gilteritinib is a transported substrate of OATP1B-type transporters, that gilteritinib exposure is increased in mice with OATP1B2 deficiency, and that the ability of itraconazole to inhibit OATP1B-type transport in vivo is contingent on its metabolism by CYP3A isoforms. These findings provide new insight into the pharmacokinetic properties of gilteritinib and into the molecular mechanisms underlying drug–drug interactions with itraconazole.
- Published
- 2022
- Full Text
- View/download PDF
4. A Metabolomics Approach for Predicting OATP1B-Type Transporter-Mediated Drug–Drug Interaction Liabilities
- Author
-
Yang Li, Yan Jin, Hanieh Taheri, Keith T. Schmidt, Alice A. Gibson, Stefan A. J. Buck, Eric D. Eisenmann, Ron H. J. Mathijssen, William D. Figg, Sharyn D. Baker, Alex Sparreboom, and Shuiying Hu
- Subjects
endogenous biomarkers ,OATP1B ,taxane ,drug–drug interactions ,Pharmacy and materia medica ,RS1-441 - Abstract
In recent years, various endogenous compounds have been proposed as putative biomarkers for the hepatic uptake transporters OATP1B1 and OATP1B3 that have the potential to predict transporter-mediated drug–drug interactions (DDIs). However, these compounds have often been identified from top–down strategies and have not been fully utilized as a substitute for traditional DDI studies. In an attempt to eliminate observer bias in biomarker selection, we applied a bottom–up, untargeted metabolomics screening approach in mice and found that plasma levels of the conjugated bile acid chenodeoxycholate-24-glucuronide (CDCA-24G) are particularly sensitive to deletion of the orthologous murine transporter Oatp1b2 (31-fold increase vs. wild type) or the entire Oatp1a/1b(−/−)cluster (83-fold increased), whereas the humanized transgenic overexpression of hepatic OATP1B1 or OATP1B3 resulted in the partial restoration of transport function. Validation studies with the OATP1B1/OATP1B3 inhibitors rifampin and paclitaxel in vitro as well as in mice and human subjects confirmed that CDCA-24G is a sensitive and rapid response biomarker to dose-dependent transporter inhibition. Collectively, our study confirmed the ability of CDCA-24G to serve as a sensitive and selective endogenous biomarker of OATP1B-type transport function and suggests a template for the future development of biomarkers for other clinically important xenobiotic transporters.
- Published
- 2022
- Full Text
- View/download PDF
5. Influence of YES1 Kinase and Tyrosine Phosphorylation on the Activity of OCT1
- Author
-
Muhammad Erfan Uddin, Dominique A. Garrison, Kyeongmin Kim, Yan Jin, Eric D. Eisenmann, Kevin M. Huang, Alice A. Gibson, Zeping Hu, Alex Sparreboom, and Shuiying Hu
- Subjects
organic cation transporter 1 ,YES1 kinase ,tyrosine kinase inhibitors ,drug-transporter interactions ,post-translational modification ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Organic cation transporter 1 (OCT1) is a transporter that regulates the hepatic uptake and subsequent elimination of diverse cationic compounds. Although OCT1 has been involved in drug-drug interactions and causes pharmacokinetic variability of many prescription drugs, details of the molecular mechanisms that regulate the activity of OCT1 remain incompletely understood. Based on an unbiased phospho-proteomics screen, we identified OCT1 as a tyrosine-phosphorylated transporter, and functional validation studies using genetic and pharmacological approaches revealed that OCT1 is highly sensitive to small molecules that target the protein kinase YES1, such as dasatinib. In addition, we found that dasatinib can inhibit hepatic OCT1 function in mice as evidenced from its ability to modulate levels of isobutyryl L-carnitine, a hepatic OCT1 biomarker identified from a targeted metabolomics analysis. These findings provide novel insight into the post-translational regulation of OCT1 and suggest that caution is warranted with polypharmacy regimes involving the combined use of OCT1 substrates and kinase inhibitors that target YES1.
- Published
- 2021
- Full Text
- View/download PDF
6. TP-0903 is active in models of drug-resistant acute myeloid leukemia
- Author
-
Jae Yoon Jeon, Daelynn R. Buelow, Dominique A. Garrison, Mingshan Niu, Eric D. Eisenmann, Kevin M. Huang, Megan E. Zavorka Thomas, Robert H. Weber, Clifford J. Whatcott, Steve L. Warner, Shelley J. Orwick, Bridget Carmichael, Emily Stahl, Lindsey T. Brinton, Rosa Lapalombella, James S. Blachly, Erin Hertlein, John C. Byrd, Bhavana Bhatnagar, and Sharyn D. Baker
- Subjects
Hematology ,Oncology ,Medicine - Abstract
Effective treatment for AML is challenging due to the presence of clonal heterogeneity and the evolution of polyclonal drug resistance. Here, we report that TP-0903 has potent activity against protein kinases related to STAT, AKT, and ERK signaling, as well as cell cycle regulators in biochemical and cellular assays. In vitro and in vivo, TP-0903 was active in multiple models of drug-resistant FLT3 mutant AML, including those involving the F691L gatekeeper mutation and bone marrow microenvironment–mediated factors. Furthermore, TP-0903 demonstrated preclinical activity in AML models with FLT3-ITD and common co-occurring mutations in IDH2 and NRAS genes. We also showed that TP-0903 had ex vivo activity in primary AML cells with recurrent mutations including MLL-PTD, ASXL1, SRSF2, and WT1, which are associated with poor prognosis or promote clinical resistance to AML-directed therapies. Our preclinical studies demonstrate that TP-0903 is a multikinase inhibitor with potent activity against multiple drug-resistant models of AML that will have an immediate clinical impact in a heterogeneous disease like AML.
- Published
- 2020
- Full Text
- View/download PDF
7. Interaction of Antifungal Drugs with CYP3A- and OATP1B-Mediated Venetoclax Elimination
- Author
-
Eric D. Eisenmann, Dominique A. Garrison, Zahra Talebi, Yan Jin, Josie A. Silvaroli, Jin-Gyu Kim, Alex Sparreboom, Michael R. Savona, Alice S. Mims, and Sharyn D. Baker
- Subjects
venetoclax ,OATP1B1 ,antifungal ,pharmacokinetics ,CYP3A ,Pharmacy and materia medica ,RS1-441 - Abstract
Venetoclax, a BCL-2 inhibitor used to treat certain hematological cancers, exhibits low oral bioavailability and high interpatient pharmacokinetic variability. Venetoclax is commonly administered with prophylactic antifungal drugs that may result in drug interactions, of which the underlying mechanisms remain poorly understood. We hypothesized that antifungal drugs may increase venetoclax exposure through inhibition of both CYP3A-mediated metabolism and OATP1B-mediated transport. Pharmacokinetic studies were performed in wild-type mice and mice genetically engineered to lack all CYP3A isoforms, or OATP1B2 that received venetoclax alone or in combination with ketoconazole or micafungin. In mice lacking all CYP3A isoforms, venetoclax AUC was increased by 1.8-fold, and pretreatment with the antifungal ketoconazole further increased venetoclax exposure by 1.6-fold, despite the absence of CYP3A. Ensuing experiments demonstrated that the deficiency of OATP1B-type transporters is also associated with increases in venetoclax exposure, and that many antifungal drugs, including micafungin, posaconazole, and isavuconazole, are inhibitors of this transport mechanism both in vitro and in vivo. These studies have identified OATP1B-mediated transport as a previously unrecognized contributor to the elimination of venetoclax that is sensitive to inhibition by various clinically-relevant antifungal drugs. Additional consideration is warranted when venetoclax is administered together with agents that inhibit both CYP3A-mediated metabolism and OATP1B-mediated transport.
- Published
- 2022
- Full Text
- View/download PDF
8. Role of OATP1B1 and OATP1B3 in Drug-Drug Interactions Mediated by Tyrosine Kinase Inhibitors
- Author
-
Dominique A. Garrison, Zahra Talebi, Eric D. Eisenmann, Alex Sparreboom, and Sharyn D. Baker
- Subjects
OATP1B1 ,OATP1B3 ,tyrosine kinase inhibitors ,drug-drug interactions ,Pharmacy and materia medica ,RS1-441 - Abstract
Failure to recognize important features of a drug’s pharmacokinetic characteristics is a key cause of inappropriate dose and schedule selection, and can lead to reduced efficacy and increased rate of adverse drug reactions requiring medical intervention. As oral chemotherapeutic agents, tyrosine kinase inhibitors (TKIs) are particularly prone to cause drug-drug interactions as many drugs in this class are known or suspected to potently inhibit the hepatic uptake transporters OATP1B1 and OATP1B3. In this article, we provide a comprehensive overview of the published literature and publicly-available regulatory documents in this rapidly emerging field. Our findings indicate that, while many TKIs can potentially inhibit the function of OATP1B1 and/or OATP1B3 and cause clinically-relevant drug-drug interactions, there are many inconsistencies between regulatory documents and the published literature. Potential explanations for these discrepant observations are provided in order to assist prescribing clinicians in designing safe and effective polypharmacy regimens, and to provide researchers with insights into refining experimental strategies to further predict and define the translational significance of TKI-mediated drug-drug interactions.
- Published
- 2020
- Full Text
- View/download PDF
9. Influence of Probenecid on the Pharmacokinetics and Pharmacodynamics of Sorafenib
- Author
-
Koen G. A. M. Hussaarts, Leni van Doorn, Karel Eechoute, Jeffrey Damman, Qiang Fu, Nadia van Doorn, Eric D. Eisenmann, Alice A. Gibson, Esther Oomen-de Hoop, Peter de Bruijn, Sharyn D. Baker, Stijn L. W. Koolen, Teun van Gelder, Roelof W. F. van Leeuwen, Ron H. J. Mathijssen, Alex Sparreboom, and Sander Bins
- Subjects
sorafenib ,probenecid ,hand-foot skin reaction (HFSR) ,pharmacokinetics ,OAT6 ,Pharmacy and materia medica ,RS1-441 - Abstract
Prior studies have demonstrated an organic anion transporter 6 (OAT6)-mediated accumulation of sorafenib in keratinocytes. The OAT6 inhibitor probenecid decreases sorafenib uptake in skin and might, therefore, decrease sorafenib-induced cutaneous adverse events. Here, the influence of probenecid on sorafenib pharmacokinetics and toxicity was investigated. Pharmacokinetic sampling was performed in 16 patients on steady-state sorafenib treatment at days 1 and 15 of the study. Patients received sorafenib (200–800 mg daily) in combination with probenecid (500 mg two times daily (b.i.d.)) on days 2–15. This study was designed to determine bioequivalence with geometric mean Area under the curve from zero to twelve hours (AUC0–12 h) as primary endpoint. During concomitant probenecid, sorafenib plasma AUC0–12 h decreased by 27% (90% CI: −38% to −14%; P < 0.01). Furthermore, peak and trough levels of sorafenib, as well as sorafenib concentrations in skin, decreased to a similar extent in the presence of probenecid. The metabolic ratio of sorafenib-glucuronide to parent drug increased (+29%) in the presence of probenecid. A decrease in systemic sorafenib concentrations during probenecid administration seems to have influenced cutaneous concentrations. Since sorafenib-glucuronide concentrations increased compared with sorafenib and sorafenib-N-oxide, probenecid may have interrupted enterohepatic circulation of sorafenib by inhibition of the organic anion transporting polypeptides 1B1 (OATP1B1). Sorafenib treatment with probenecid is, therefore, not bioequivalent to sorafenib monotherapy. A clear effect of probenecid on sorafenib toxicity could not be identified in this study.
- Published
- 2020
- Full Text
- View/download PDF
10. Supplementary Data from Sorafenib Population Pharmacokinetics and Skin Toxicities in Children and Adolescents with Refractory/Relapsed Leukemia or Solid Tumor Malignancies
- Author
-
Sharyn D. Baker, Jeffrey E. Rubnitz, Raul C. Ribeiro, Tanja A. Gruber, Ching-Hon Pui, Sheila Shurtleff, Yong-Dong Wang, Aksana Vasilyeva, Eric D. Eisenmann, Sara M. Federico, Fariba Navid, Lie Li, Lei Wang, Stanley B. Pounds, John C. Panetta, and Hiroto Inaba
- Abstract
Supplementary Figures and Tables
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Sorafenib Population Pharmacokinetics and Skin Toxicities in Children and Adolescents with Refractory/Relapsed Leukemia or Solid Tumor Malignancies
- Author
-
Sharyn D. Baker, Jeffrey E. Rubnitz, Raul C. Ribeiro, Tanja A. Gruber, Ching-Hon Pui, Sheila Shurtleff, Yong-Dong Wang, Aksana Vasilyeva, Eric D. Eisenmann, Sara M. Federico, Fariba Navid, Lie Li, Lei Wang, Stanley B. Pounds, John C. Panetta, and Hiroto Inaba
- Abstract
Purpose:To determine the pharmacokinetics and skin toxicity profile of sorafenib in children with refractory/relapsed malignancies.Patients and Methods:Sorafenib was administered concurrently or sequentially with clofarabine and cytarabine to patients with leukemia or with bevacizumab and cyclophosphamide to patients with solid tumor malignancies. The population pharmacokinetics (PPK) of sorafenib and its metabolites and skin toxicities were evaluated.Results:In PPK analysis, older age, bevacizumab and cyclophosphamide regimen, and higher creatinine were associated with decreased sorafenib apparent clearance (CL/f; P < 0.0001 for all), and concurrent clofarabine and cytarabine administration was associated with decreased sorafenib N-oxide CL/f (P = 7e−4). Higher bilirubin was associated with decreased sorafenib N-oxide and glucuronide CL/f (P = 1e−4). Concurrent use of organic anion-transporting polypeptide 1B1 inhibitors was associated with increased sorafenib and decreased sorafenib glucuronide CL/f (P < 0.003). In exposure–toxicity analysis, a shorter time to development of grade 2–3 hand–foot skin reaction (HFSR) was associated with concurrent (P = 0.0015) but not with sequential (P = 0.59) clofarabine and cytarabine administration, compared with bevacizumab and cyclophosphamide, and with higher steady-state concentrations of sorafenib (P = 0.0004) and sorafenib N-oxide (P = 0.0275). In the Bayes information criterion model selection, concurrent clofarabine and cytarabine administration, higher sorafenib steady-state concentrations, larger body surface area, and previous occurrence of rash appeared in the four best two-predictor models of HFSR. Pharmacokinetic simulations showed that once-daily and every-other-day sorafenib schedules would minimize exposure to sorafenib steady-state concentrations associated with HFSR.Conclusions:Sorafenib skin toxicities can be affected by concurrent medications and sorafenib steady-state concentrations. The described PPK model can be used to refine exposure–response relations for alternative dosing strategies to minimize skin toxicity.
- Published
- 2023
- Full Text
- View/download PDF
12. TP-0903 Is Active in Preclinical Models of Acute Myeloid Leukemia with TP53 Mutation/Deletion
- Author
-
Eric D. Eisenmann, Jack C. Stromatt, Sydney Fobare, Kevin M. Huang, Daelynn R. Buelow, Shelley Orwick, Jae Yoon Jeon, Robert H. Weber, Bill Larsen, Alice S. Mims, Erin Hertlein, John C. Byrd, and Sharyn D. Baker
- Subjects
Cancer Research ,Oncology ,TP53 ,AML ,TP-0903 ,decitabine ,multikinase ,aurora kinase - Abstract
Acute myeloid leukemia (AML) with mutations in the tumor suppressor gene TP53 confers a dismal prognosis with 3-year overall survival of
- Published
- 2022
- Full Text
- View/download PDF
13. Boosting the oral bioavailability of anticancer drugs through intentional drug–drug interactions
- Author
-
Alex Sparreboom, Sharyn D. Baker, Eric D. Eisenmann, and Zahra Talebi
- Subjects
Drug ,Cancer chemotherapy ,media_common.quotation_subject ,Administration, Oral ,Biological Availability ,Antineoplastic Agents ,Pharmacology ,Toxicology ,complex mixtures ,030226 pharmacology & pharmacy ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Humans ,Medicine ,Drug Interactions ,media_common ,Boosting (doping) ,business.industry ,Treatment options ,General Medicine ,Bioavailability ,Anticancer treatment ,Pharmacodynamics ,business ,030217 neurology & neurosurgery - Abstract
Oral anticancer drugs suffer from significant variability in pharmacokinetics and pharmacodynamics partially due to limited bioavailability. The limited bioavailability of anticancer drugs is due to both pharmaceutical limitations and physiological barriers. Pharmacokinetic boosting is a strategy to enhance the oral bioavailability of a therapeutic drug by inhibiting physiological barriers through an intentional drug-drug interaction (DDI). This type of strategy has proven effective across several therapeutic indications including anticancer treatment. Pharmacokinetic boosting could improve anticancer drugs lacking or with otherwise unacceptable oral formulations through logistic, economic, pharmacodynamic, and pharmacokinetic benefits. Despite these benefits, pharmacokinetic boosting strategies could result in unintended DDIs and are only likely to benefit a limited number of targets. Highlighting this concern, pharmacokinetic boosting has mixed results depending on the boosted drug. While pharmacokinetic boosting did not significantly improve certain drugs, it has resulted in the commercial approval of boosted oral formulations for other drugs. Pharmacokinetic boosting to improve oral anticancer therapy is an expanding area of research that is likely to improve treatment options for cancer patients.
- Published
- 2021
- Full Text
- View/download PDF
14. MATE1 Deficiency Exacerbates Dofetilide-Induced Proarrhythmia
- Author
-
Muhammad Erfan Uddin, Eric D. Eisenmann, Yang Li, Kevin M. Huang, Dominique A. Garrison, Zahra Talebi, Alice A. Gibson, Yan Jin, Mahesh Nepal, Ingrid M. Bonilla, Qiang Fu, Xinxin Sun, Alec Millar, Mikhail Tarasov, Christopher E. Jay, Xiaoming Cui, Heidi J. Einolf, Ryan M. Pelis, Sakima A. Smith, Przemysław B. Radwański, Douglas H. Sweet, Jörg König, Martin F. Fromm, Cynthia A. Carnes, Shuiying Hu, and Alex Sparreboom
- Subjects
Sulfonamides ,dofetilide ,organic cation transporters ,arrhythmia ,PBPK modeling ,Organic Chemistry ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Mice ,Atrial Fibrillation ,Phenethylamines ,Animals ,Humans ,ddc:610 ,Physical and Theoretical Chemistry ,Anti-Arrhythmia Agents ,Molecular Biology ,Spectroscopy - Abstract
Dofetilide is a rapid delayed rectifier potassium current inhibitor widely used to prevent the recurrence of atrial fibrillation and flutter. The clinical use of this drug is associated with increases in QTc interval, which predispose patients to ventricular cardiac arrhythmias. The mechanisms involved in the disposition of dofetilide, including its movement in and out of cardiomyocytes, remain unknown. Using a xenobiotic transporter screen, we identified MATE1 (SLC47A1) as a transporter of dofetilide and found that genetic knockout or pharmacological inhibition of MATE1 in mice was associated with enhanced retention of dofetilide in cardiomyocytes and increased QTc prolongation. The urinary excretion of dofetilide was also dependent on the MATE1 genotype, and we found that this transport mechanism provides a mechanistic basis for previously recorded drug-drug interactions of dofetilide with various contraindicated drugs, including bictegravir, cimetidine, ketoconazole, and verapamil. The translational significance of these observations was examined with a physiologically-based pharmacokinetic model that adequately predicted the drug-drug interaction liabilities in humans. These findings support the thesis that MATE1 serves a conserved cardioprotective role by restricting excessive cellular accumulation and warrant caution against the concurrent administration of potent MATE1 inhibitors and cardiotoxic substrates with a narrow therapeutic window.
- Published
- 2022
- Full Text
- View/download PDF
15. Determination of the endogenous OATP1B biomarkers glycochenodeoxycholate-3-sulfate and chenodeoxycholate-24-glucuronide in human and mouse plasma by a validated UHPLC-MS/MS method
- Author
-
Yan Jin, Yang Li, Eric D. Eisenmann, William D. Figg, Sharyn D. Baker, Alex Sparreboom, and Shuiying Hu
- Subjects
Methanol ,Clinical Biochemistry ,Reproducibility of Results ,Water ,Cell Biology ,General Medicine ,Chenodeoxycholic Acid ,Biochemistry ,Analytical Chemistry ,Bile Acids and Salts ,Mice ,Glucuronides ,Glycochenodeoxycholic Acid ,Tandem Mass Spectrometry ,Micafungin ,Animals ,Humans ,Rifampin ,Biomarkers ,Chromatography, High Pressure Liquid - Abstract
Glycochenodeoxycholate-3-sulfate (GCDCA-S) and chenodeoxycholate-24-glucuronide (CDCA-24G) are bile acid metabolites that potentially serve as endogenous biomarkers for drug-drug interactions mediated by the hepatic uptake transporters OATP1B1 and OATP1B3. We developed and validated a novel UHPLC-MS/MS method for the quantitative determination of GCDCA-S and CDCA-24G in mouse and human plasma with a lower limit of quantitation of 0.5 ng/mL. Chromatographic separation was achieved on an Accucore aQ column (50 mm × 2.1 mm, dp = 2.6 μm) maintained at 20 °C and a gradient mobile phase comprising 2 mM ammonium acetate in water and methanol. The extraction recoveries of GCDCA-S and CDCA-24G were80 %, and linear (r
- Published
- 2022
16. Neuronal uptake transporters contribute to oxaliplatin neurotoxicity in mice
- Author
-
Navjot Pabla, Maryam B. Lustberg, Alix F. Leblanc, Kevin M. Huang, Eric D. Eisenmann, Mingqing Chen, Jing Wang, Yang Li, Jiyoung Kim, Sherry H. Xia, Timothy C. Cope, Muhammad Erfan Uddin, Charles L. Loprinzi, Kristen W. Hong, Alice A. Gibson, Alex Sparreboom, Paola Alberti, Alessia Chiorazzi, Stephen N. Housley, Anne M. Noonan, Shuiying Hu, Duncan DiGiacomo, Guido Cavaletti, Jason A. Sprowl, Huang, K, Leblanc, A, Uddin, M, Kim, J, Chen, M, Eisenmann, E, Gibson, A, Li, Y, Hong, K, Digiacomo, D, Xia, S, Alberti, P, Chiorazzi, A, Housley, S, Cope, T, Sprowl, J, Wang, J, Loprinzi, C, Noonan, A, Lustberg, M, Cavaletti, G, Pabla, N, Hu, S, and Sparreboom, A
- Subjects
Male ,0301 basic medicine ,Colorectal cancer ,ved/biology.organism_classification_rank.species ,Pharmacology ,Oxaliplatin, neuropathy, neurotoxicity, OCT2, animal model, rodent model, neurophysiology ,Neuroprotection ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Medicine ,Model organism ,Mice, Knockout ,Neurons ,business.industry ,ved/biology ,Concise Communication ,Neurotoxicity ,Organic Cation Transporter 2 ,Cancer ,Transporter ,General Medicine ,medicine.disease ,Rats ,Oxaliplatin ,030104 developmental biology ,030220 oncology & carcinogenesis ,Genetically Engineered Mouse ,Female ,Neurotoxicity Syndromes ,business ,Neuroglia ,medicine.drug - Abstract
Peripheral neurotoxicity is a debilitating toxicity 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 to 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 preventative 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.
- Published
- 2020
- Full Text
- View/download PDF
17. BMX kinase mediates gilteritinib resistance in FLT3-mutated AML through microenvironmental factors
- Author
-
Daelynn R. Buelow, Bhavana Bhatnagar, Shelley J. Orwick, Jae Yoon Jeon, Eric D. Eisenmann, Jack C. Stromatt, Navjot Singh Pabla, James S. Blachly, Sharyn D. Baker, and Bradley W. Blaser
- Subjects
Leukemia, Myeloid, Acute ,Aniline Compounds ,fms-Like Tyrosine Kinase 3 ,Pyrazines ,Mutation ,Tumor Microenvironment ,Humans ,Hematology ,Protein-Tyrosine Kinases - Abstract
Despite the clinical benefit associated with gilteritinib in relapsed/refractory acute myeloid leukemia (AML), most patients eventually develop resistance through unknown mechanisms. To delineate the mechanistic basis of resistance to gilteritinib, we performed targeted sequencing and scRNASeq on primary FLT3-ITD-mutated AML samples. Co-occurring mutations in RAS pathway genes were the most common genetic abnormalities, and unresponsiveness to gilteritinib was associated with increased expression of bone marrow-derived hematopoietic cytokines and chemokines. In particular, we found elevated expression of the TEK-family kinase, BMX, in gilteritinib-unresponsive patients pre- and post-treatment. BMX contributed to gilteritinib resistance in FLT3-mutant cell lines in a hypoxia-dependent manner by promoting pSTAT5 signaling, and these phenotypes could be reversed with pharmacological inhibition and genetic knockout. We also observed that inhibition of BMX in primary FLT3-mutated AML samples decreased chemokine secretion and enhanced the activity of gilteritinib. Collectively, these findings indicate a crucial role for microenvironment-mediated factors modulated by BMX in the escape from targeted therapy and have implications for the development of novel therapeutic interventions to restore sensitivity to gilteritinib.
- Published
- 2022
18. Determination of the Endogenous Oatp1b Biomarkers Glycochenodeoxycholate-3-Sulfate and Chenodeoxycholate 24-Glucuronide in Human and Mouse Plasma
- Author
-
Yan Jin, Yang Li, Eric D. Eisenmann, William D. Figg, Sharyn D. Baker, Alex Sparreboom, and Shuiying Hu
- Published
- 2022
- Full Text
- View/download PDF
19. Intentional Modulation of Ibrutinib Pharmacokinetics through CYP3A Inhibition
- Author
-
Yan Jin, Qiang Fu, Alex Sparreboom, Robert H. Weber, John C. Byrd, Muhammad Erfan Uddin, Jennifer A. Woyach, Dominique Garrison, Eric D. Eisenmann, Sharyn D. Baker, and Elizabeth M. Muhowski
- Subjects
chemistry.chemical_compound ,Pharmacokinetics ,chemistry ,CYP3A ,business.industry ,Modulation ,Ibrutinib ,Medicine ,Pharmacology ,business ,Article - Abstract
Ibrutinib (Imbruvica; PCI-32765) is an orally administered inhibitor of Bruton's tyrosine kinase that has transformed the treatment of B-cell malignancies. However, ibrutinib has very low oral bioavailability that contributes to significant variability in systemic exposure between patients, and this has the potential to affect both efficacy and toxicity. We hypothesized that the oral bioavailability of ibrutinib is limited by CYP3A isoform–mediated metabolism, and that this pathway can be inhibited to improve the pharmacokinetic properties of ibrutinib. Pharmacokinetic studies were performed in wild-type mice and mice genetically engineered to lack all CYP3A isoforms (CYP3A−/−) that received ibrutinib alone or in combination with CYP3A inhibitors cobicistat or ketoconazole. Computational modeling was performed to derive doses of ibrutinib that, when given after a CYP3A inhibitor, results in therapeutically relevant drug levels. Deficiency of CYP3A in mice was associated with an approximately 10-fold increase in the AUC of ibrutinib. This result could be phenocopied by administration of cobicistat before ibrutinib in wild-type mice, but cobicistat did not influence levels of ibrutinib in CYP3A−/− mice. Population pharmacokinetic and prospectively validated physiologically based pharmacokinetic models established preclinical and clinical doses of ibrutinib that could be given safely in combination with cobicistat without negatively affecting antileukemic properties. These findings signify a dominant role for CYP3A-mediated metabolism in the elimination of ibrutinib, and suggest a role for pharmacologic inhibitors of this pathway to intentionally modulate the plasma levels and improve the therapeutic use of this clinically important agent. Significance: Ibrutinib has limited oral bioavailability, which contributes to significant interindividual pharmacokinetic variability. Using engineered mouse models, we here report a causal relationship between CYP3A-mediated metabolism and ibrutinib's bioavailability and drug–drug interaction with cobicistat. These results offer a mechanistic basis for reported pharmacokinetic interactions with ibrutinib, and in conjunction with a newly developed computational model, allow for the rational design of clinical trials aimed at improving the therapeutic use of ibrutinib.
- Published
- 2021
20. Sorafenib Population Pharmacokinetics and Skin Toxicities in Children and Adolescents with Refractory/Relapsed Leukemia or Solid Tumor Malignancies
- Author
-
Lei Wang, Raul C. Ribeiro, Stanley Pounds, Sara M. Federico, Lie Li, John C. Panetta, Ching-Hon Pui, Jeffrey E. Rubnitz, Aksana Vasilyeva, Tanja A. Gruber, Sheila A. Shurtleff, Yong-Dong Wang, Sharyn D. Baker, Fariba Navid, Eric D. Eisenmann, and Hiroto Inaba
- Subjects
Adult ,0301 basic medicine ,Sorafenib ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Bevacizumab ,Cyclophosphamide ,urologic and male genital diseases ,Skin Diseases ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Clofarabine ,Tissue Distribution ,heterocyclic compounds ,Child ,neoplasms ,Leukemia ,business.industry ,Cytarabine ,medicine.disease ,female genital diseases and pregnancy complications ,digestive system diseases ,Regimen ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Neoplasm Recurrence, Local ,business ,medicine.drug - Abstract
Purpose: To determine the pharmacokinetics and skin toxicity profile of sorafenib in children with refractory/relapsed malignancies. Patients and Methods: Sorafenib was administered concurrently or sequentially with clofarabine and cytarabine to patients with leukemia or with bevacizumab and cyclophosphamide to patients with solid tumor malignancies. The population pharmacokinetics (PPK) of sorafenib and its metabolites and skin toxicities were evaluated. Results: In PPK analysis, older age, bevacizumab and cyclophosphamide regimen, and higher creatinine were associated with decreased sorafenib apparent clearance (CL/f; P < 0.0001 for all), and concurrent clofarabine and cytarabine administration was associated with decreased sorafenib N-oxide CL/f (P = 7e−4). Higher bilirubin was associated with decreased sorafenib N-oxide and glucuronide CL/f (P = 1e−4). Concurrent use of organic anion-transporting polypeptide 1B1 inhibitors was associated with increased sorafenib and decreased sorafenib glucuronide CL/f (P < 0.003). In exposure–toxicity analysis, a shorter time to development of grade 2–3 hand–foot skin reaction (HFSR) was associated with concurrent (P = 0.0015) but not with sequential (P = 0.59) clofarabine and cytarabine administration, compared with bevacizumab and cyclophosphamide, and with higher steady-state concentrations of sorafenib (P = 0.0004) and sorafenib N-oxide (P = 0.0275). In the Bayes information criterion model selection, concurrent clofarabine and cytarabine administration, higher sorafenib steady-state concentrations, larger body surface area, and previous occurrence of rash appeared in the four best two-predictor models of HFSR. Pharmacokinetic simulations showed that once-daily and every-other-day sorafenib schedules would minimize exposure to sorafenib steady-state concentrations associated with HFSR. Conclusions: Sorafenib skin toxicities can be affected by concurrent medications and sorafenib steady-state concentrations. The described PPK model can be used to refine exposure–response relations for alternative dosing strategies to minimize skin toxicity.
- Published
- 2019
- Full Text
- View/download PDF
21. Myocardial hypersensitivity to ischemic injury is not reversed by clonidine or propranolol in a predator-based rat model of posttraumatic stress disorder
- Author
-
Brandon L. Johnson, Sarah L. Seeley, Boyd R. Rorabaugh, Albert D. Bui, Megan E. Heikkila, Phillip R. Zoladz, Madelaine R. Huntley, Eric D. Eisenmann, and Robert M. Rose
- Subjects
Male ,medicine.medical_specialty ,Elevated plus maze ,Sympathetic Nervous System ,medicine.medical_treatment ,Myocardial Ischemia ,Ischemia ,Adrenergic ,Propranolol ,Disease ,Anxiety ,Article ,Clonidine ,Rats, Sprague-Dawley ,Stress Disorders, Post-Traumatic ,Random Allocation ,03 medical and health sciences ,Adrenergic Agents ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Treatment Failure ,Saline ,Biological Psychiatry ,Pharmacology ,business.industry ,Myocardium ,Cardiovascular Agents ,Heart ,medicine.disease ,030227 psychiatry ,Disease Models, Animal ,Cardiology ,Disease Susceptibility ,medicine.symptom ,business ,medicine.drug - Abstract
• Individuals with posttraumatic stress disorder (PTSD) are at increased risk for cardiovascular disease. We previously reported that a predator-based model of PTSD increases myocardial sensitivity to ischemic injury. Heightened sympathetic signaling has a well-established role in the formation of anxiety associated with PTSD and may also contribute to worsening of myocardial injury in the ischemic heart. Thus, we examined whether suppression of sympathetic tone protects the ischemic heart in rats subjected to this model of PTSD. Rats were treated with saline or clonidine throughout the 31-day stress paradigm. Behavior on the elevated plus maze (EPM) was assessed on Day 32, and hearts were subjected to an ischemic insult on day 33. Stressed rats exhibited increased anxiety on the EPM and significantly larger myocardial infarcts following ischemia. Clonidine reversed the anxiety-like behavior but had no impact on infarct size. In a subsequent experiment, rats were treated with propranolol in their drinking water throughout the stress paradigm. Propranolol had no effect on either anxiety or myocardial sensitivity to ischemic injury. These findings suggest that the myocardial hypersensitivity to ischemic injury observed in this model is not caused by increased sympathetic tone or chronic β-adrenergic receptor signaling.
- Published
- 2019
- Full Text
- View/download PDF
22. The Use, Validity, and Translational Utility of Animal Models of Posttraumatic Stress Disorder
- Author
-
Phillip R. Zoladz, Eric D. Eisenmann, and Chelsea E. Cadle
- Subjects
Posttraumatic stress ,business.industry ,Medicine ,business ,Clinical psychology - Published
- 2021
- Full Text
- View/download PDF
23. Preclinical Characterization of TP‐0903, a Novel Multikinase Inhibitor, in TP53 Mutant Acute Myeloid Leukemia
- Author
-
Kevin M. Huang, Erin Hertlein, Bill Larsen, Robert J. Weber, Daelynn R. Buelow, John C. Byrd, Josie A. Silvaroli, Sharyn D. Baker, Shelley Orwick, Eric D. Eisenmann, Sydney Fobare, Jae Yoon Jeon, and Jack Stromatt
- Subjects
Multikinase inhibitor ,Chemistry ,Mutant ,Genetics ,Cancer research ,Myeloid leukemia ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2021
- Full Text
- View/download PDF
24. Targeting OCT3 attenuates doxorubicin-induced cardiac injury
- Author
-
Megan E. Zavorka Thomas, Eric D. Eisenmann, Muhammad Erfan Uddin, Sherry H. Xia, Vincenzo Coppola, Markus Keiser, Yan Jin, Pearlly S. Yan, Paul W. Burridge, Sharyn D. Baker, Duncan DiGiacomo, Kevin M. Huang, Ralf Bundschuh, Tarek Magdy, Sakima A. Smith, Anne T. Nies, Taosheng Chen, Alexander Pan, Kara N. Corps, Daniel Addison, Moray J. Campbell, Qiang Fu, Alice A. Gibson, Alex Sparreboom, Stefan Oswald, Joanne Wang, Cynthia A. Carnes, Shuiying Hu, Maryam B. Lustberg, Yang Li, Marcus Otter, and Ingrid M. Bonilla
- Subjects
Side effect ,Organic Anion Transporters, Sodium-Independent ,SLC22A3 ,Mice ,Neoplasms ,Medicine ,Animals ,Humans ,Doxorubicin ,Myocytes, Cardiac ,Molecular Targeted Therapy ,Child ,Cardiotoxicity ,Multidisciplinary ,Organic cation transport proteins ,biology ,business.industry ,Sequence Analysis, RNA ,Transporter ,Biological Sciences ,medicine.disease ,Leukemia ,Pyrimidines ,Nilotinib ,Gene Expression Regulation ,Heart Injuries ,Cancer research ,biology.protein ,business ,medicine.drug - Abstract
Doxorubicin is a commonly used anticancer agent that can cause debilitating and irreversible cardiac injury. The initiating mechanisms contributing to this side effect remain unknown, and current preventative strategies offer only modest protection. Using stem-cell–derived cardiomyocytes from patients receiving doxorubicin, we probed the transcriptomic landscape of solute carriers and identified organic cation transporter 3 (OCT3) (SLC22A3) as a critical transporter regulating the cardiac accumulation of doxorubicin. Functional validation studies in heterologous overexpression models confirmed that doxorubicin is transported into cardiomyocytes by OCT3 and that deficiency of OCT3 protected mice from acute and chronic doxorubicin-related changes in cardiovascular function and genetic pathways associated with cardiac damage. To provide proof-of-principle and demonstrate translational relevance of this transport mechanism, we identified several pharmacological inhibitors of OCT3, including nilotinib, and found that pharmacological targeting of OCT3 can also preserve cardiovascular function following treatment with doxorubicin without affecting its plasma levels or antitumor effects in multiple models of leukemia and breast cancer. Finally, we identified a previously unrecognized, OCT3-dependent pathway of doxorubicin-induced cardiotoxicity that results in a downstream signaling cascade involving the calcium-binding proteins S100A8 and S100A9. These collective findings not only shed light on the etiology of doxorubicin-induced cardiotoxicity, but also are of potential translational relevance and provide a rationale for the implementation of a targeted intervention strategy to prevent this debilitating side effect.
- Published
- 2021
25. Influence of YES1 Kinase and Tyrosine Phosphorylation on the Activity of OCT1
- Author
-
Yan Jin, Muhammad Erfan Uddin, Dominique Garrison, Kyeongmin Kim, Shuiying Hu, Eric D. Eisenmann, Zeping Hu, Alice A. Gibson, Alex Sparreboom, and Kevin M. Huang
- Subjects
0301 basic medicine ,YES1 ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,tyrosine kinase inhibitors ,medicine ,Pharmacology (medical) ,organic cation transporter 1 ,Protein kinase A ,Original Research ,Organic cation transport proteins ,biology ,Kinase ,Chemistry ,lcsh:RM1-950 ,Transporter ,Tyrosine phosphorylation ,Small molecule ,Dasatinib ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,post-translational modification ,030220 oncology & carcinogenesis ,drug-transporter interactions ,YES1 kinase ,biology.protein ,medicine.drug - Abstract
Organic cation transporter 1 (OCT1) is a transporter that regulates the hepatic uptake and subsequent elimination of diverse cationic compounds. Although OCT1 has been involved in drug-drug interactions and causes pharmacokinetic variability of many prescription drugs, details of the molecular mechanisms that regulate the activity of OCT1 remain incompletely understood. Based on an unbiased phospho-proteomics screen, we identified OCT1 as a tyrosine-phosphorylated transporter, and functional validation studies using genetic and pharmacological approaches revealed that OCT1 is highly sensitive to small molecules that target the protein kinase YES1, such as dasatinib. In addition, we found that dasatinib can inhibit hepatic OCT1 function in mice as evidenced from its ability to modulate levels of isobutyryl L-carnitine, a hepatic OCT1 biomarker identified from a targeted metabolomics analysis. These findings provide novel insight into the post-translational regulation of OCT1 and suggest that caution is warranted with polypharmacy regimes involving the combined use of OCT1 substrates and kinase inhibitors that target YES1.
- Published
- 2020
26. TP-0903 is active in models of drug-resistant acute myeloid leukemia
- Author
-
Robert H. Weber, Dominique Garrison, James S. Blachly, Mingshan Niu, Daelynn R. Buelow, Megan E. Zavorka Thomas, Shelley Orwick, Steve L. Warner, Eric D. Eisenmann, Lindsey T. Brinton, Emily Stahl, Jae Yoon Jeon, Rosa Lapalombella, Erin Hertlein, John C. Byrd, Clifford J. Whatcott, Kevin M. Huang, Sharyn D. Baker, Bhavana Bhatnagar, and Bridget Carmichael
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Male ,medicine.medical_specialty ,Mice, Nude ,Drug resistance ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Internal medicine ,Cell Line, Tumor ,Gene Duplication ,hemic and lymphatic diseases ,Leukemias ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Protein Kinase Inhibitors ,neoplasms ,Sulfonamides ,Hematology ,business.industry ,Myeloid leukemia ,General Medicine ,Cell cycle ,Xenograft Model Antitumor Assays ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,Pyrimidines ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Medicine ,Female ,Bone marrow ,Drug therapy ,business ,Protein Kinases ,Ex vivo ,Research Article - Abstract
Effective treatment for AML is challenging due to the presence of clonal heterogeneity and the evolution of polyclonal drug resistance. Here, we report that TP-0903 has potent activity against protein kinases related to STAT, AKT, and ERK signaling, as well as cell cycle regulators in biochemical and cellular assays. In vitro and in vivo, TP-0903 was active in multiple models of drug-resistant FLT3 mutant AML, including those involving the F691L gatekeeper mutation and bone marrow microenvironment–mediated factors. Furthermore, TP-0903 demonstrated preclinical activity in AML models with FLT3-ITD and common co-occurring mutations in IDH2 and NRAS genes. We also showed that TP-0903 had ex vivo activity in primary AML cells with recurrent mutations including MLL-PTD, ASXL1, SRSF2, and WT1, which are associated with poor prognosis or promote clinical resistance to AML-directed therapies. Our preclinical studies demonstrate that TP-0903 is a multikinase inhibitor with potent activity against multiple drug-resistant models of AML that will have an immediate clinical impact in a heterogeneous disease like AML., TP-0903, a multikinase inhibitor, demonstrates preclinical activity in models of drug-resistant AML, including those involving FLT3 mutations, bone marrow microenvironment-mediated factors and recurrent mutations.
- Published
- 2020
27. Influence of Probenecid on the Pharmacokinetics and Pharmacodynamics of Sorafenib
- Author
-
Peter de Bruijn, Ron H.J. Mathijssen, Esther Oomen-de Hoop, Teun van Gelder, Alice A. Gibson, Nadia van Doorn, Alex Sparreboom, Eric D. Eisenmann, Roelof W F van Leeuwen, Karel Eechoute, Koen G A M Hussaarts, Qiang Fu, Jeffrey Damman, Sharyn D. Baker, Sander Bins, Leni van Doorn, Stijn L.W. Koolen, Medical Oncology, Pathology, and Pharmacy
- Subjects
Sorafenib ,Organic anion transporter 1 ,lcsh:RS1-441 ,Pharmaceutical Science ,Bioequivalence ,Pharmacology ,OAT6 ,urologic and male genital diseases ,Article ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,medicine ,heterocyclic compounds ,Enterohepatic circulation ,neoplasms ,030304 developmental biology ,0303 health sciences ,hand-foot skin reaction (HFSR) ,biology ,business.industry ,Area under the curve ,digestive system diseases ,female genital diseases and pregnancy complications ,Probenecid ,probenecid ,030220 oncology & carcinogenesis ,Toxicity ,biology.protein ,sorafenib ,business ,pharmacokinetics ,medicine.drug - Abstract
Prior studies have demonstrated an organic anion transporter 6 (OAT6)-mediated accumulation of sorafenib in keratinocytes. The OAT6 inhibitor probenecid decreases sorafenib uptake in skin and might, therefore, decrease sorafenib-induced cutaneous adverse events. Here, the influence of probenecid on sorafenib pharmacokinetics and toxicity was investigated. Pharmacokinetic sampling was performed in 16 patients on steady-state sorafenib treatment at days 1 and 15 of the study. Patients received sorafenib (200&ndash, 800 mg daily) in combination with probenecid (500 mg two times daily (b.i.d.)) on days 2&ndash, 15. This study was designed to determine bioequivalence with geometric mean Area under the curve from zero to twelve hours (AUC0&ndash, 12 h) as primary endpoint. During concomitant probenecid, sorafenib plasma AUC0&ndash, 12 h decreased by 27% (90% CI: &minus, 38% to &minus, 14%, P <, 0.01). Furthermore, peak and trough levels of sorafenib, as well as sorafenib concentrations in skin, decreased to a similar extent in the presence of probenecid. The metabolic ratio of sorafenib-glucuronide to parent drug increased (+29%) in the presence of probenecid. A decrease in systemic sorafenib concentrations during probenecid administration seems to have influenced cutaneous concentrations. Since sorafenib-glucuronide concentrations increased compared with sorafenib and sorafenib-N-oxide, probenecid may have interrupted enterohepatic circulation of sorafenib by inhibition of the organic anion transporting polypeptides 1B1 (OATP1B1). Sorafenib treatment with probenecid is, therefore, not bioequivalent to sorafenib monotherapy. A clear effect of probenecid on sorafenib toxicity could not be identified in this study.
- Published
- 2020
28. Development and validation of a sensitive UHPLC-MS/MS analytical method for venetoclax in mouse plasma, and its application to pharmacokinetic studies
- Author
-
Robert H. Weber, Yan Jin, Sharyn D. Baker, Eric D. Eisenmann, and Alex Sparreboom
- Subjects
Male ,Analyte ,Calibration curve ,Electrospray ionization ,Clinical Biochemistry ,Administration, Oral ,030226 pharmacology & pharmacy ,01 natural sciences ,Biochemistry ,Uhplc ms ms ,Sensitivity and Specificity ,Analytical Chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Pharmacokinetics ,Tandem Mass Spectrometry ,Animals ,Drug Interactions ,Chromatography, High Pressure Liquid ,Sulfonamides ,Chromatography ,Chemistry ,Venetoclax ,010401 analytical chemistry ,Reproducibility of Results ,Cell Biology ,General Medicine ,Plasma ,Bridged Bicyclo Compounds, Heterocyclic ,Blood proteins ,0104 chemical sciences ,Ketoconazole ,Linear Models ,Female - Abstract
A rapid and sensitive analytical method was developed to quantify venetoclax, an oral BH3-mimetic that blocks the anti-apoptotic protein BCL-2, in mouse plasma using ultra-high-performance liquid chromatography with electrospray ionization tandem mass spectrometric detection. Plasma protein precipitation was performed on 5 µL samples, and separation of the analytes was accomplished on an Accucore aQ column using gradient elution at a flow rate of 0.4 mL/min. The calibration curve was linear (R2 ≥ 0.99) over the concentration range of 5–1,000 ng/mL, and the lower limit of quantitation was 5 ng/mL. The intra-day and inter-day precisions (RSD%) were
- Published
- 2020
29. Predator-based psychosocial stress model of PTSD differentially influences voluntary ethanol consumption depending on methodology
- Author
-
Kasey E. Mucher, Kiera L. Robinson, Brandon L. Johnson, Robert M. Rose, Madelaine R. Huntley, Eric D. Eisenmann, Phillip R. Zoladz, Megan E. Heikkila, and Brooke A. Kohls
- Subjects
Male ,Health (social science) ,Alcohol Drinking ,Self Administration ,Alcohol ,Toxicology ,Choice Behavior ,Biochemistry ,Stress Disorders, Post-Traumatic ,03 medical and health sciences ,Behavioral Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Stress, Physiological ,Negative cognitions ,Animals ,Medicine ,Predator ,Ethanol ,business.industry ,General Medicine ,Rats ,030227 psychiatry ,Disease Models, Animal ,Mood ,Neurology ,chemistry ,Turnover ,Predatory Behavior ,Psychosocial stress ,Ethanol intake ,business ,030217 neurology & neurosurgery ,Clinical psychology - Abstract
Post-traumatic stress disorder (PTSD) is a debilitating psychological disorder typified by diagnostic symptom clusters including hyperarousal, avoidance, negative cognitions and mood, and intrusive re-experiencing of the traumatic event. Patients with PTSD have been reported to self-medicate with alcohol to ameliorate hyperarousal symptoms associated with the disorder. Research utilizing rodent models of PTSD to emulate this behavioral phenomenon has thus far yielded inconsistent results. In the present study, we examined the effects of a predator-based psychosocial stress model of PTSD on voluntary ethanol consumption. In the first of two experiments, following exposure to a 31-day stress or control paradigm, rats were singly housed during the dark cycle with free access to 1% sucrose solution or 10% ethanol, which was also sweetened with 1% sucrose. Over the course of a 20-day period of ethanol access, stressed rats consumed significantly less ethanol than non-stressed rats. These counterintuitive results prompted the completion of a second experiment which was identical to the first, except rats were also exposed to the two-bottle paradigm for 20 days before the stress or control paradigm. In the second experiment, after the stress manipulation, stressed rats exhibited significantly greater ethanol preference than non-stressed rats. These findings suggest that prior exposure to ethanol influences the subsequent effect of stress on ethanol intake. They also validate the use of the present model of PTSD to examine potential mechanisms underlying stress-related changes in ethanol-seeking behavior.
- Published
- 2018
- Full Text
- View/download PDF
30. High-Dimensional Analysis Identifies Mechanisms of Gilteritinib Resistance in FLT3-Mutated AML
- Author
-
Jae Yoon Jeon, James S. Blachly, Shelley Orwick, Eric D. Eisenmann, Navjot Pabla, Bhavana Bhatnagar, Daelynn R. Buelow, Sharyn D. Baker, Jack Stromatt, Bradley W. Blaser, and Megan E. Zavorka Thomas
- Subjects
Resistance (ecology) ,Chemistry ,Immunology ,Gilteritinib ,Cell Biology ,Hematology ,Computational biology ,High dimensional ,Biochemistry - Abstract
While clinical benefit has been observed with gilteritinib in patients with FLT3 mutated relapsed/refractory acute myeloid leukemia (AML), most patients relapse through mechanisms that are incompletely understood. In this study, to investigate mechanisms of gilteritinib sensitivity and resistance, we performed targeted sequencing (21 patients) and scRNASeq analysis (8 patients) of FLT3-ITD-positive AML samples obtained before and during treatment. Before treatment, co-occurring mutations were observed in 33 genes among 21 patients. Mutations in RAS pathway genes (PTPN11, KRAS, NRAS, CBL) were the most common and observed in 57% (12/21) of patients. Seven patients pretreatment already contained RAS pathway mutations, of which 6 of these mutations were maintained over the course of treatment. During treatment, 9 patients showed emerging RAS mutations, 4 of which initially presented with a different RAS pathway mutation pre-treatment. Other mutations that arose during treatment were observed in CEBPA, IDH1, SF1 and WT1; as well as CSF3R, CUX1, PLEKHG5, and XPO1, not previously identified in gilteritinib-treated patients. Mutational clonality was generally maintained over treatment in both responders and non-responders. scRNASeq revealed global gene expression differences in myeloblast populations between gilteritinib-responsive and -unresponsive patients. Previous studies in vitro have shown that bone marrow-derived hematopoietic and inflammatory cytokines/chemokines confer resistance to FLT3 inhibitors. In the unresponsive group, we observed an increase in expression of CCL5, CXCL1, CXCL2, CXCL8, FLT3, IL6R, IL3RA, and CSF2RA during gilteritinib treatment, supporting the concept from preclinical studies that AML microenvironment-mediated factors play a critical role in drug resistance. Baseline expression of the Tec kinase BMX was significantly higher in unresponsive patients (Log2FoldChange, 6.65; adjusted P value, 0.00186), and this was maintained in the expanding myeloblast populations during treatment. Previously, upregulated BMX was shown to contribute to sorafenib resistance in patients with FLT3-ITD-positive AML, through cell-nonautonomous microenvironment hypoxia-dependent effects. Further in vitro investigation confirmed gilteritinib resistance could be reversed through genetic and pharmacological manipulation of BMX. Gene module analysis showed associations between gilteritinib responsive and upregulation of genes and pathways involved in lymphocyte differentiation and myeloid leukocyte activation, including TBX21, GATA3, CD33, and LYZ. By contrast, there was association between unresponsiveness to gilteritinib and upregulation of cell-cycle, DNA, and RNA metabolic processes, including pathways involving METTL1 and DNMT3A, as well as pre-treatment expression of pathways associated with protein translation. Together, these data provide support for microenvironment-dependent escape from targeted therapy and suggest that BMX may contribute to gilteritinib resistance. High-dimensional analysis with scRNA-seq provides a deeper understanding of targets and pathways for potential therapeutic intervention to restore gilteritinib sensitivity. Disclosures Blachly: INNATE: Consultancy, Honoraria; KITE: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; AstraZeneca: Consultancy, Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
31. Sex-dependent effects of sleep deprivation on myocardial sensitivity to ischemic injury
- Author
-
Brandon L. Johnson, Sarah L. Seeley, Megan E Fry, Albert D. Bui, Phillip R. Zoladz, Eric D. Eisenmann, Boyd R. Rorabaugh, and Anna Krivenko
- Subjects
Male ,Physiology ,Myocardial Infarction ,Ischemia ,Diastole ,Blood Pressure ,030204 cardiovascular system & hematology ,Rats, Sprague-Dawley ,03 medical and health sciences ,Behavioral Neuroscience ,Sex Factors ,0302 clinical medicine ,medicine ,Animals ,Myocardial infarction ,Endocrine and Autonomic Systems ,business.industry ,Myocardium ,Heart ,Recovery of Function ,medicine.disease ,Sleep in non-human animals ,Rats ,Psychiatry and Mental health ,Sleep deprivation ,Neuropsychology and Physiological Psychology ,Blood pressure ,Rate pressure product ,Anesthesia ,Sleep Deprivation ,End-diastolic volume ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Sleep deprivation is associated with increased risk of myocardial infarction. However, it is unknown whether the effects of sleep deprivation are limited to increasing the likelihood of experiencing a myocardial infarction or if sleep deprivation also increases the extent of myocardial injury. In this study, rats were deprived of paradoxical sleep for 96 h using the platform-over-water method. Control rats were subjected to the same condition except the control platform was large enough for the rats to sleep. Hearts from sleep deprived and control rats were subjected to 20 min ischemia on a Langendorff isolated heart system. Infarct size and post ischemic recovery of contractile function were unaffected by sleep deprivation in male hearts. In contrast, hearts from sleep-deprived females exhibited significantly larger infarcts than hearts from control females. Post ischemic recovery of rate pressure product and + dP/dT were significantly attenuated by sleep deprivation in female hearts, and post ischemic recovery of end diastolic pressure was significantly elevated in hearts from sleep deprived females compared to control females, indicating that post ischemic recovery of both systolic and diastolic function were worsened by sleep deprivation. These data provide evidence that sleep deprivation increases the extent of ischemia-induced injury in a sex-dependent manner.
- Published
- 2016
- Full Text
- View/download PDF
32. Modulation of CYP3A Activity to Increase the Oral Bioavailability of Ibrutinib
- Author
-
Bob Leroy Weber, John C. Byrd, Jennifer A. Woyach, Sharyn D. Baker, Shuiying Hu, Alex Sparreboom, Qiang Fu, Eric D. Eisenmann, and Dominique Garrison
- Subjects
chemistry.chemical_compound ,Chemistry ,CYP3A ,Modulation ,Ibrutinib ,Genetics ,Pharmacology ,Molecular Biology ,Biochemistry ,Biotechnology ,Bioavailability - Published
- 2020
- Full Text
- View/download PDF
33. Activity of the multikinase inhibitor TP‐0903 in RAS mutant acute myeloid leukemia
- Author
-
Mingshan Niu, Erin Hertlein, Megan E. Zavorka Thomas, Daelynn R. Buelow, Shelley Orwick, John C. Byrd, Sharyn D. Baker, Dominique Garrison, Clifford J. Whatcott, Bhavana Bhatnagar, Eric D. Eisenmann, Jae Yoon Jeon, and Steven L. Warner
- Subjects
Multikinase inhibitor ,Chemistry ,Mutant ,Genetics ,Cancer research ,Myeloid leukemia ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2020
- Full Text
- View/download PDF
34. Acute Stress Decreases but Chronic Stress Increases Myocardial Sensitivity to Ischemic Injury in Rodents
- Author
-
Boyd R. Rorabaugh, Phillip R. Zoladz, and Eric D. Eisenmann
- Subjects
medicine.medical_specialty ,lcsh:RC435-571 ,Rat model ,Ischemia ,Disease ,Review ,ischemia ,030204 cardiovascular system & hematology ,03 medical and health sciences ,stress ,0302 clinical medicine ,lcsh:Psychiatry ,Internal medicine ,Medicine ,In patient ,Chronic stress ,cardiovascular diseases ,Acute stress ,Psychiatry ,business.industry ,cardiovascular ,rodent ,Ischemic injury ,PTSD ,medicine.disease ,anxiety ,rodent models ,Psychiatry and Mental health ,Anesthesia ,Cardiology ,Anxiety ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Cardiovascular disease is the largest cause of mortality worldwide, and stress is a significant contributor to the development of cardiovascular disease. The relationship between acute and chronic stress and cardiovascular disease is well-evidenced. Acute stress can lead to arrhythmias and ischemic injury. However, recent evidence in rodent models suggests that acute stress can decrease sensitivity to myocardial ischemia-reperfusion injury. Conversely, chronic stress is arrythmogenic and increases sensitivity to myocardial ischemia-reperfusion injury. Few studies have examined the impact of validated animal models of stress-related psychological disorders on the ischemic heart. This review examines the work that has been completed using rat models to study the effects of stress on myocardial sensitivity to ischemic injury. Utilization of animal models of stress-related psychological disorders is critical in the prevention and treatment of cardiovascular disorders in patients experiencing stress-related psychiatric conditions.
- Published
- 2016
- Full Text
- View/download PDF
35. Sex-dependent effects of chronic psychosocial stress on myocardial sensitivity to ischemic injury
- Author
-
Brandon L. Johnson, Boyd R. Rorabaugh, Megan E Fry, Sarah L. Seeley, Phillip R. Zoladz, Lauren E. Stoner, Anna Krivenko, Albert D. Bui, Eric D. Eisenmann, and Joseph D. Lawson
- Subjects
Male ,Elevated plus maze ,Physiology ,Ischemia ,Myocardial Ischemia ,Disease ,Anxiety ,Rats, Sprague-Dawley ,Stress Disorders, Post-Traumatic ,Behavioral Neuroscience ,medicine ,Animals ,Sex Characteristics ,Endocrine and Autonomic Systems ,Ischemic injury ,Heart ,medicine.disease ,Rats ,Psychiatry and Mental health ,Disease Models, Animal ,Neuropsychology and Physiological Psychology ,Anesthesia ,Psychosocial stress ,Female ,medicine.symptom ,Cues ,Psychology ,Psychosocial ,Stress, Psychological ,Sex characteristics - Abstract
Individuals with post-traumatic stress disorder (PTSD) experience many debilitating symptoms, including intrusive memories, persistent anxiety and avoidance of trauma-related cues. PTSD also results in numerous physiological complications, including increased risk for cardiovascular disease (CVD). However, characterization of PTSD-induced cardiovascular alterations is lacking, especially in preclinical models of the disorder. Thus, we examined the impact of a psychosocial predator-based animal model of PTSD on myocardial sensitivity to ischemic injury. Male and female Sprague-Dawley rats were exposed to psychosocial stress or control conditions for 31 days. Stressed rats were given two cat exposures, separated by a period of 10 days, and were subjected to daily social instability throughout the paradigm. Control rats were handled daily for the duration of the experiment. Rats were tested on the elevated plus maze (EPM) on day 32, and hearts were isolated on day 33 and subjected to 20 min ischemia and 2 h reperfusion on a Langendorff isolated heart system. Stressed male and female rats gained less body weight relative to controls, but only stressed males exhibited increased anxiety on the EPM. Male, but not female, rats exposed to psychosocial stress exhibited significantly larger infarcts and attenuated post-ischemic recovery of contractile function compared to controls. Our data demonstrate that predator stress combined with daily social instability sex-dependently increases myocardial sensitivity to ischemic injury. Thus, this manipulation may be useful for studying potential mechanisms underlying cardiovascular alterations in PTSD, as well as sex differences in the cardiovascular stress response.
- Published
- 2015
36. Abstract 105: Increased Myocardial Sensitivity to Ischemic Injury in an Animal Model of Posttraumatic Stress Disorder
- Author
-
Boyd R Rorabaugh, Albert Bui, Sarah L Seeley, Anna Krivenko, Eric D Eisenmann, Megan E Fry, Joseph D Lawson, Lauren E Stoner, and Phillip R Zoladz
- Subjects
Physiology ,Cardiology and Cardiovascular Medicine - Abstract
Background: Posttraumatic stress disorder (PTSD) is a psychological disorder characterized by the formation of traumatic memories following exposure to a life threatening event. In addition to psychological manifestations, PTSD promotes atherosclerosis and increases the incidence of myocardial infarction. However, it is unknown whether the effects of PTSD are limited to increasing the incidence of myocardial infarction or if PTSD also increases infarct severity. Therefore, we used an animal model of PTSD to determine whether posttraumatic stress influences infarct size and postischemic recovery of cardiac contractile function. Methods: Rats were subjected to a well-established animal model of PTSD that is based on predator exposure and psychosocial stress (Zoladz et al., Stress 11:259-281). Rats subjected to this model exhibit many PTSD-like characteristics including the formation of traumatic memories, increased anxiety, increased startle reflex, hypertension, and alterations in the hypothalamic-pituitary adrenal axis. Male rats (7 weeks of age) were either subjected to psychosocial stress (n = 9) or continuously housed in their home cages (n = 8) for 31 days. Hearts were subsequently isolated and subjected to 20 minutes of ischemia and 2 hours reperfusion on a Langendorff isolated heart system. Results: Stressed rats exhibited significantly elevated corticosterone concentrations and anxiety-like behavior in the elevated plus maze. Infarct sizes were significantly larger in hearts from stressed rats (44.7 ± 1.7 % of area at risk) compared to nonstressed rats (31.0 ± 5.4 % of area at risk). Consistent with increased myocardial injury, postischemic recovery of rate pressure product (stressed = 16,922 ± 1,554 mmHg*bpm; nonstressed = 26,407 ± 2,977 mmHg*bpm) and +dP/dT (stressed = 1,901 ± 189 mmHg/sec; nonstressed =3,259 ± 498 mmHg/sec) were significantly attenuated in hearts from stressed rats. Furthermore, postischemic end diastolic pressure was significantly elevated in hearts from stressed (57 ± 6 mmHg) compared to nonstressed (32 ± 7 mmHg) rats. Conclusion: This animal model suggests that PTSD may make the myocardium more sensitive to ischemic injury through a mechanism that is independent from its ability to promote atherosclerosis.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.