1
|
Fediuk DJ, Gorman DN, Stoddard SA, Zhang Y, Ogden AG, Winton JA, Saxena AR. Effect of Renal Impairment on the Pharmacokinetics of a Single Oral Dose of Danuglipron in Participants With Type 2 Diabetes. J Clin Pharmacol 2024; 64:449-460. [PMID: 37840155 DOI: 10.1002/jcph.2371] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Danuglipron (PF-06882961) is an oral, small-molecule glucagon-like peptide-1 receptor agonist in development for the treatment of type 2 diabetes (T2D) and obesity. Impaired renal function is prevalent in patients with T2D. This Phase 1, open-label study evaluated the effect of renal impairment on the pharmacokinetics, safety, and tolerability of danuglipron (20 mg) in healthy participants with normal renal function (estimated glomerular filtration rate [eGFR] unnormalized for body surface area: ≥90 mL/min), in participants with T2D and normal renal function (eGFR ≥90 mL/min), and in participants with T2D and mild (eGFR 60-89 mL/min), moderate (eGFR 30-59 mL/min), or severe (eGFR <30 mL/min) renal impairment (N = 39). Log-linear regression analyses and analyses of variance showed no evidence of a clinically significant effect of reduced renal function on danuglipron pharmacokinetics. Renal clearance of unchanged danuglipron was minimal (<1% across all renal function groups). Danuglipron pharmacokinetics were similar between healthy participants and participants with T2D and normal renal function. A single 20-mg oral dose of danuglipron was generally safe and well tolerated in all participant groups. In participants with T2D, renal impairment had no clinically meaningful effect on the pharmacokinetic, safety, and tolerability profiles of danuglipron, indicating that dose adjustment of danuglipron will not be required when administered to patients with T2D and reduced renal function.
Collapse
Affiliation(s)
- Daryl J Fediuk
- Clinical Pharmacology and Bioanalytics, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Donal N Gorman
- Global Biometrics and Data Management, Pfizer Global Product Development, Cambridge, UK
| | - Stephanie-An Stoddard
- Early Clinical Development, Clinical Sciences, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Yizhong Zhang
- Clinical Pharmacology and Bioanalytics, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Adam G Ogden
- Clinical Pharmacology and Bioanalytics, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Jennifer A Winton
- Clinical Pharmacology and Bioanalytics, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Aditi R Saxena
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| |
Collapse
|
2
|
Carriat L, Quaranta S, Solas C, Rony M, Ciccolini J. Renal impairment and DPD testing: watch out for false-positive results! Br J Clin Pharmacol 2022; 88:4928-4932. [PMID: 35939355 DOI: 10.1111/bcp.15482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Measuring uracil (U) levels in plasma is a convenient surrogate to establish DPD status in patients scheduled with 5-fluorouracil (5-FU) or capecitabine. To what extent renal impairment could impact on U levels and thus be a confounding factor is a rising concern. Here, we report the case of a cancer patient with severe renal impairment scheduled for 5-FU-based regimen. Determination of his DPD status was complicated because of his condition and the influence of intermittent hemodialysis when monitoring U levels. The patient was initially identified as markedly DPD-deficient upon U measurement (i.e., U = 40 ng/ml), but further monitoring between and immediately after dialysis showed mild deficiency only (i.e., U = 34 and U = 19 ng/ml, respectively). Despite this discrepancy, starting dose of 5-FU was cut by 50% upon treatment initiation. Tolerance was good and 5-FU dosing was next shifted to 25% reduction, then further shifted to normal dosing at the 5th course, with still no sign for drug-related toxicities. Further DPYD genotyping showed none of the 4 allelic variants usually associated with loss of DPD activity. Of note, the excellent tolerance upon standard dosing strongly suggests that this patient was actually not DPD-deficient, despite U values always above normal concentrations. This case report highlights how critical is the information regarding the renal function of patients with cancer when phenotyping DPD using U plasma as a surrogate, and that U accumulation in patients with such condition is likely to yield false-positive results.
Collapse
Affiliation(s)
- Laure Carriat
- Laboratoire de Pharmacocinétique et Toxicologie, CHU Timone, APHM, Marseille, France.,SMARTc unit, Centre de Recherche en Cancérologie de Marseille, Inserm, Marseille, France
| | - Sylvie Quaranta
- Laboratoire de Pharmacocinétique et Toxicologie, CHU Timone, APHM, Marseille, France
| | - Caroline Solas
- Laboratoire de Pharmacocinétique et Toxicologie, CHU Timone, APHM, Marseille, France
| | - Maelle Rony
- Oncologie Digestive, CHU Timone, APHM, Marseille, France
| | - Joseph Ciccolini
- Laboratoire de Pharmacocinétique et Toxicologie, CHU Timone, APHM, Marseille, France.,SMARTc unit, Centre de Recherche en Cancérologie de Marseille, Inserm, Marseille, France
| |
Collapse
|
3
|
Bonavia A, Stiles N. Renohepatic crosstalk: a review of the effects of acute kidney injury on the liver. Nephrol Dial Transplant 2021; 37:1218-1228. [PMID: 33527986 DOI: 10.1093/ndt/gfaa297] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Several theories regarding acute kidney injury (AKI)-related mortality have been entertained, although mounting evidence supports the paradigm that impaired kidney function directly and adversely affects the function of several remote organs. The kidneys and liver are fundamental to human metabolism and detoxification, and it is therefore hardly surprising that critical illness complicated by hepatorenal dysfunction portends a poor prognosis. Several diseases can simultaneously impact the proper functioning of the liver and kidneys, although this review will address the impact of AKI on liver function. While evidence for this relationship in humans remains sparse, we present supportive studies and then discuss the most likely mechanisms by which AKI can cause liver dysfunction. These include 'traditional' complications of AKI (uremia, volume overload and acute metabolic acidosis, among others) as well as systemic inflammation, hepatic leukocyte infiltration, cytokine-mediated liver injury and hepatic oxidative stress. We conclude by addressing the therapeutic implications of these findings to clinical medicine.
Collapse
Affiliation(s)
- Anthony Bonavia
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Nicholas Stiles
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
4
|
Lin YS, Thummel KE, Thompson BD, Totah RA, Cho CW. Sources of Interindividual Variability. Methods Mol Biol 2021; 2342:481-550. [PMID: 34272705 DOI: 10.1007/978-1-0716-1554-6_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The efficacy, safety, and tolerability of drugs are dependent on numerous factors that influence their disposition. A dose that is efficacious and safe for one individual may result in sub-therapeutic or toxic blood concentrations in others. A significant source of this variability in drug response is drug metabolism, where differences in presystemic and systemic biotransformation efficiency result in variable degrees of systemic exposure (e.g., AUC, Cmax, and/or Cmin) following administration of a fixed dose.Interindividual differences in drug biotransformation have been studied extensively. It is recognized that both intrinsic factors (e.g., genetics, age, sex, and disease states) and extrinsic factors (e.g., diet , chemical exposures from the environment, and the microbiome) play a significant role. For drug-metabolizing enzymes, genetic variation can result in the complete absence or enhanced expression of a functional enzyme. In addition, upregulation and downregulation of gene expression, in response to an altered cellular environment, can achieve the same range of metabolic function (phenotype), but often in a less predictable and time-dependent manner. Understanding the mechanistic basis for variability in drug disposition and response is essential if we are to move beyond the era of empirical, trial-and-error dose selection and into an age of personalized medicine that will improve outcomes in maintaining health and treating disease.
Collapse
Affiliation(s)
- Yvonne S Lin
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA.
| | - Kenneth E Thummel
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Brice D Thompson
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Christi W Cho
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Huang W, Nakano M, Sager J, Ragueneau-Majlessi I, Isoherranen N. Physiologically Based Pharmacokinetic Model of the CYP2D6 Probe Atomoxetine: Extrapolation to Special Populations and Drug-Drug Interactions. Drug Metab Dispos 2017; 45:1156-1165. [PMID: 28860113 PMCID: PMC5637815 DOI: 10.1124/dmd.117.076455] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 08/28/2017] [Indexed: 01/18/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling of drug disposition and drug-drug interactions (DDIs) has become a key component of drug development. PBPK modeling has also been considered as an approach to predict drug disposition in special populations. However, whether models developed and validated in healthy populations can be extrapolated to special populations is not well established. The goal of this study was to determine whether a drug-specific PBPK model validated using healthy populations could be used to predict drug disposition in specific populations and in organ impairment patients. A full PBPK model of atomoxetine was developed using a training set of pharmacokinetic (PK) data from CYP2D6 genotyped individuals. The model was validated using drug-specific acceptance criteria and a test set of 14 healthy subject PK studies. Population PBPK models were then challenged by simulating the effects of ethnicity, DDIs, pediatrics, and renal and hepatic impairment on atomoxetine PK. Atomoxetine disposition was successfully predicted in 100% of healthy subject studies, 88% of studies in Asians, 79% of DDI studies, and 100% of pediatric studies. However, the atomoxetine area under the plasma concentration versus time curve (AUC) was overpredicted by 3- to 4-fold in end stage renal disease and hepatic impairment. The results show that validated PBPK models can be extrapolated to different ethnicities, DDIs, and pediatrics but not to renal and hepatic impairment patients, likely due to incomplete understanding of the physiologic changes in these conditions. These results show that systematic modeling efforts can be used to further refine population models to improve the predictive value in this area.
Collapse
Affiliation(s)
- Weize Huang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Mariko Nakano
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Jennifer Sager
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | | | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
6
|
Hirai K, Ookawara S, Miyazawa H, Ito K, Ueda Y, Kaku Y, Hoshino T, Kimura SI, Yoshida I, Kakuta S, Morishita Y, Tabei K. Successful treatment of a hemodialyzed patient with pure red cell aplasia associated with epoetin beta pegol therapy with cyclosporine. CEN Case Rep 2017; 5:78-82. [PMID: 28509169 DOI: 10.1007/s13730-015-0196-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/03/2015] [Indexed: 11/30/2022] Open
Abstract
A 42-year-old man with end-stage renal failure had been receiving hemodialysis therapy since April 2009. Initially, darbepoetin alfa was administered to treat his renal anemia. After treatment was switched to epoetin beta pegol, the patient's hemoglobin levels rapidly decreased. He was diagnosed with pure red cell aplasia (PRCA) based on the results of a bone marrow examination. Epoetin beta pegol was strongly suspected to be the cause of the PRCA, and although he tested negative for anti-epoetin beta pegol antibodies, epoetin beta pegol was discontinued and cyclosporine therapy was initiated. Thereafter, his hemoglobin levels increased, and his anemia dramatically improved after 3 months.
Collapse
Affiliation(s)
- Keiji Hirai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | - Susumu Ookawara
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan.
| | - Haruhisa Miyazawa
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | - Kiyonori Ito
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | - Yuichiro Ueda
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | - Yoshio Kaku
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | - Taro Hoshino
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | - Shun-Ichi Kimura
- Division of Hematology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Izumi Yoshida
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | | | - Yoshiyuki Morishita
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| | - Kaoru Tabei
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama, Saitama, 330-8503, Japan
| |
Collapse
|
7
|
Kropeit D, Scheuenpflug J, Erb-Zohar K, Halabi A, Stobernack HP, Hulskotte EGJ, van Schanke A, Zimmermann H, Rübsamen-Schaeff H. Pharmacokinetics and safety of letermovir, a novel anti-human cytomegalovirus drug, in patients with renal impairment. Br J Clin Pharmacol 2017; 83:1944-1953. [PMID: 28345163 DOI: 10.1111/bcp.13292] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 12/18/2022] Open
Abstract
AIMS Human cytomegalovirus remains a significant issue for immunocompromised patients and existing viral polymerase targeting therapies are associated with significant toxicity. Accordingly, the viral terminase complex inhibitor, letermovir, is in development. We assessed letermovir pharmacokinetics in renal impairment. METHODS This was a Phase 1, open-label, nonrandomised trial. Estimated glomerular filtration rate based on the Modification of Diet Renal Disease equation was used to create three groups of eight subjects: healthy function (estimated glomerular filtration rate ≥ 90 ml min-1 1.73m-2 ), moderate (30-59 ml min-1 1.73m-2 ) and severe (<30 ml min-1 1.73m-2 ) impairment. Oral letermovir 120 mg was dosed once-daily for 8 days and blood collected for pharmacokinetic analyses. RESULTS All 24 subjects enrolled completed the trial. Moderate and severe renal impairment increased mean unbound letermovir fractions by 11% and 26%, respectively, vs. healthy subjects. Exposure (AUCτ,ss and Css,max ) was increased with renal impairment [least square mean ratios (90% confidence intervals) total letermovir vs. healthy subjects, AUCτ,ss 192% (143-258%) and 142% (83-243%) for moderate and severe impairment, respectively; Css,max 125% (87-182%) and 106% (75-151%), respectively]. Clearance was decreased vs. healthy subjects. Correlation analyses indicated a correlation between decreasing renal function and increased unbound letermovir concentration (R2 = 0.5076, P < 0.0001). Correlations were identified between decreased clearance with both decreased renal function (R2 = 0.0662, P = 0.2249 and R2 = 0.1861, P = 0.0353 total and unbound clearance, respectively) and increased age (R2 = 0.3548, P = 0.0021 and R2 = 0.3166, P = 0.0042 total and unbound clearance, respectively). Multiple-dose letermovir 120 mg was well tolerated across groups. CONCLUSIONS Renal impairment increased exposure to letermovir, although age was a confounding factor.
Collapse
Affiliation(s)
- Dirk Kropeit
- AiCuris Anti-infective Cures GmbH, Wuppertal, Germany
| | | | | | - Atef Halabi
- CRS Clinical Research Services Kiel GmbH, Kiel, Germany
| | | | | | | | | | | |
Collapse
|
8
|
Hamamura K, Matsunaga N, Ikeda E, Kondo H, Ikeyama H, Tokushige K, Itcho K, Furuichi Y, Yoshida Y, Matsuda M, Yasuda K, Doi A, Yokota Y, Amamoto T, Aramaki H, Irino Y, Koyanagi S, Ohdo S. Alterations of Hepatic Metabolism in Chronic Kidney Disease via D-box-binding Protein Aggravate the Renal Dysfunction. J Biol Chem 2016; 291:4913-27. [PMID: 26728457 DOI: 10.1074/jbc.m115.696930] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 11/06/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with an increase in serum retinol; however, the underlying mechanisms of this disorder are poorly characterized. Here, we found that the alteration of hepatic metabolism induced the accumulation of serum retinol in 5/6 nephrectomy (5/6Nx) mice. The liver is the major organ responsible for retinol metabolism; accordingly, microarray analysis revealed that the hepatic expression of most CYP genes was changed in 5/6Nx mice. In addition, D-box-binding protein (DBP), which controls the expression of several CYP genes, was significantly decreased in these mice. Cyp3a11 and Cyp26a1, encoding key proteins in retinol metabolism, showed the greatest decrease in expression in 5/6Nx mice, a process mediated by the decreased expression of DBP. Furthermore, an increase of plasma transforming growth factor-β1 (TGF-β1) in 5/6Nx mice led to the decreased expression of the Dbp gene. Consistent with these findings, the alterations of retinol metabolism and renal dysfunction in 5/6Nx mice were ameliorated by administration of an anti-TGF-β1 antibody. We also show that the accumulation of serum retinol induced renal apoptosis in 5/6Nx mice fed a normal diet, whereas renal dysfunction was reduced in mice fed a retinol-free diet. These findings indicate that constitutive Dbp expression plays an important role in mediating hepatic dysfunction under CKD. Thus, the aggravation of renal dysfunction in patients with CKD might be prevented by a recovery of hepatic function, potentially through therapies targeting DBP and retinol.
Collapse
Affiliation(s)
- Kengo Hamamura
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan, the Drug Innovation Research Center, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Naoya Matsunaga
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Eriko Ikeda
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan, the Drug Innovation Research Center, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Hideaki Kondo
- the Center for Sleep Medicine, Saiseikai Nagasaki Hospital, Katafuchi, Nagasaki 850-0003, Japan
| | - Hisako Ikeyama
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Kazutaka Tokushige
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Kazufumi Itcho
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Yoko Furuichi
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Yuya Yoshida
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Masaki Matsuda
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Kaori Yasuda
- Cell-Innovator Inc., EC Building, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Atsushi Doi
- Cell-Innovator Inc., EC Building, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Yoshifumi Yokota
- the Department of Molecular Genetics, School of Medicine, Fukui University, Matsuoka, Fukui 910-1193, Japan
| | - Toshiaki Amamoto
- Neues Corporation, Tenyamachi-cho, Hakata-ku, Fukuoka 812-0025, Japan, and
| | - Hironori Aramaki
- the Drug Innovation Research Center, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan, the Department of Molecular Biology, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Yasuhiro Irino
- the Integrated Center for Mass Spectrometry, Division of Membrane Biology, and Department of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Satoru Koyanagi
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan
| | - Shigehiro Ohdo
- From the Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8512, Japan,
| |
Collapse
|
9
|
Droppa M, Tschernow D, Müller KAL, Tavlaki E, Karathanos A, Stimpfle F, Schaeffeler E, Schwab M, Tolios A, Siller-Matula JM, Gawaz M, Geisler T. Evaluation of clinical risk factors to predict high on-treatment platelet reactivity and outcome in patients with stable coronary artery disease (PREDICT-STABLE). PLoS One 2015; 10:e0121620. [PMID: 25799149 PMCID: PMC4370634 DOI: 10.1371/journal.pone.0121620] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/02/2015] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES This study was designed to identify the multivariate effect of clinical risk factors on high on-treatment platelet reactivity (HPR) and 12 months major adverse events (MACE) under treatment with aspirin and clopidogrel in patients undergoing non-urgent percutaneous coronary intervention (PCI). METHODS 739 consecutive patients with stable coronary artery disease (CAD) undergoing PCI were recruited. On-treatment platelet aggregation was tested by light transmittance aggregometry. Clinical risk factors and MACE during one-year follow-up were recorded. An independent population of 591 patients served as validation cohort. RESULTS Degree of on-treatment platelet aggregation was influenced by different clinical risk factors. In multivariate regression analysis older age, diabetes mellitus, elevated BMI, renal function and left ventricular ejection fraction were independent predictors of HPR. After weighing these variables according to their estimates in multivariate regression model, we developed a score to predict HPR in stable CAD patients undergoing elective PCI (PREDICT-STABLE Score, ranging 0-9). Patients with a high score were significantly more likely to develop MACE within one year of follow-up, 3.4% (score 0-3), 6.3% (score 4-6) and 10.3% (score 7-9); odds ratio 3.23, P=0.02 for score 7-9 vs. 0-3. This association was confirmed in the validation cohort. CONCLUSIONS Variability of on-treatment platelet function and associated outcome is mainly influenced by clinical risk variables. Identification of high risk patients (e.g. with high PREDICT-STABLE score) might help to identify risk groups that benefit from more intensified antiplatelet regimen. Additional clinical risk factor assessment rather than isolated platelet function-guided approaches should be investigated in future to evaluate personalized antiplatelet therapy in stable CAD-patients.
Collapse
Affiliation(s)
- Michal Droppa
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Dimitri Tschernow
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Karin A. L. Müller
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Elli Tavlaki
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Athanasios Karathanos
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Fabian Stimpfle
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- Department of Clinical Pharmacology, University Hospital of Tübingen, Tübingen, Germany
| | - Alexander Tolios
- Laboratory Medicine Institute, Medical Center of the University of Munich, Munich, Germany
| | | | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Cardiovascular Medicine, University Hospital of Tübingen, Tübingen, Germany
- * E-mail:
| |
Collapse
|
10
|
Mita S, Meyers D, Pal P, Lin T, Majumdar T, Rebello S, Sunkara G, Chen J. Effect of Renal Impairment on the Pharmacokinetics of Pradigastat, a Novel Diacylglycerol Acyltransferase1 (DGAT1) Inhibitor. Clin Pharmacokinet 2015; 54:751-60. [DOI: 10.1007/s40262-015-0234-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
11
|
K Gupta S, Kantesaria B, Glue P. Exploring the influence of renal dysfunction on the pharmacokinetics of ribavirin after oral and intravenous dosing. Drug Discov Ther 2014; 8:89-95. [PMID: 24815584 DOI: 10.5582/ddt.8.89] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Although ribavirin is minimally cleared by renal elimination, its pharmacokinetics are substantially altered in patients with chronic renal impairment. This open-label study assessed the pharmacokinetics of single 400-mg oral and intravenous (IV) doses of ribavirin in two healthy volunteers and 12 patients with varying degrees of chronic renal impairment. Blood and urine samples were collected pre-dose and up to 168 h post-dose for pharmacokinetic analyses. Ribavirin area under the plasma concentration-time curve from time zero to time of final quantifiable sample and maximum plasma concentration values were increased, and total plasma clearance (CL), renal clearance (CLr), non-renal clearance (CLnr), volume of distribution at steady state (Vdss), and amount excreted values were reduced in patients with renal dysfunction compared with those who had normal renal function. Following IV administration, mean CLr was 54%, 23%, and 10% in patients with mild, moderate, and severe renal dysfunction, respectively, relative to control subjects, and was 56%, 28%, and 9% of control values after oral dosing. After IV dosing, mean CLnr was 94%, 76%, and 75% of control values in patients with mild, moderate, and severe renal dysfunction, respectively, and was 54%, 48%, and 27% of control values after oral dosing. Mean oral bioavailability of ribavirin was 35%, 60%, 57%, and 71% in control subjects and patients with mild, moderate, and severe renal dysfunction, respectively. These data indicate that there are multiple mechanisms (increased oral bioavailability, reduced CLr and CLnr, reduced Vd) contributing to altered ribavirin pharmacokinetics in chronic renal impairment.
Collapse
Affiliation(s)
- Samir K Gupta
- Departments of Drug Metabolism/Pharmacokinetics and Clinical Pharmacology, Merck Research Lab
| | | | | |
Collapse
|
12
|
Areberg J, Petersen KB, Chen G, Naik H. Population pharmacokinetic meta-analysis of vortioxetine in healthy individuals. Basic Clin Pharmacol Toxicol 2014; 115:552-9. [PMID: 24766668 PMCID: PMC4257570 DOI: 10.1111/bcpt.12256] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/10/2014] [Indexed: 11/27/2022]
Abstract
The objective was to describe the pharmacokinetics of vortioxetine and evaluate the effect of intrinsic and extrinsic factors in the healthy population. Data from 26 clinical pharmacology studies were pooled. A total of 21,758 vortioxetine quantifiable plasma concentrations were collected from 887 subjects with corresponding demography. The doses ranged from 2.5 to 75 mg (single dose) and 2.5-60 mg (multiple QD doses). The pharmacokinetics of vortioxetine was best characterised by a two-compartment model with first-order absorption, lag-time and linear elimination, with interindividual error terms for absorption rate constant, oral clearance and central volume of distribution. The population mean was 32.7 L/hr for oral clearance and 1.97∙10(3) L for the central volume of distribution. The average elimination half-life was 65.8 hr. CYP2D6 inferred metabolic status (ultra, extensive, intermediate or poor metabolisers) and age on oral clearance and height on central volume of distribution were identified as statistically significant covariate-parameter relationships. For CYP2D6 poor metabolisers, CL/F was approximately 50% to that seen in CYP2D6 extensive metabolisers. The impact of height on V2/F and age on CL/F was low and not considered to be clinically relevant. The final model was found to be reliable, stable and predictive. A reliable, stable and predictive pharmacokinetic model was developed to characterise pharmacokinetics of vortioxetine in the healthy population.
Collapse
Affiliation(s)
- Johan Areberg
- Division of Clinical and Quantitative Pharmacology, H. Lundbeck A/S, Valby, Denmark
| | | | | | | |
Collapse
|
13
|
Effects of renal impairment on the pharmacokinetics of morinidazole: uptake transporter-mediated renal clearance of the conjugated metabolites. Antimicrob Agents Chemother 2014; 58:4153-61. [PMID: 24820074 DOI: 10.1128/aac.02414-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Morinidazole is a novel 5-nitroimidazole antimicrobial drug that undergoes extensive metabolism in humans via N(+)-glucuronidation (N(+)-glucuronide of S-morinidazole [M8-1] and N(+)-glucuronide of R-morinidazole [M8-2]) and sulfation (sulfate conjugate of morinidazole [M7]). Our objectives were to assess the effects of renal impairment on the pharmacokinetics (PK) of morinidazole and to elucidate the potential mechanisms. In this parallel-group study, healthy subjects and patients with severe renal impairment received an intravenous infusion of 500 mg of morinidazole. Plasma and urine samples were collected and analyzed. The areas under the plasma concentration-time curves (AUC) for M7, M8-1, and M8-2 were 15.1, 20.4, and 17.4 times higher, respectively, in patients with severe renal impairment than in healthy subjects, while the AUC for morinidazole was 1.5 times higher. The urinary recovery of the major metabolites was not significantly different between the two groups over 0 to 48 h, but the renal clearances of M7, M8-1, and M8-2 in patients were 85.3%, 92.5%, and 92.2% lower, respectively. In vitro transporter studies revealed that M7 is a substrate for organic anion transporter 1 (OAT1) and OAT3 (Km = 28.6 and 54.0 μM, respectively). Only OAT3 transported M8-1 and M8-2. Morinidazole was not a substrate for the transporter-transfected cells examined. These results revealed that the function or activity of renal uptake transporters might be impaired in patients with severe renal impairment, which accounted for dramatically increased plasma exposure and reduced renal clearance of the conjugated metabolites of morinidazole, the substrates of renal transporters in patients. It will help clinicians to adjust the dose in patients with severe renal impairment and to predict possible transporter-based drug-drug interactions.
Collapse
|
14
|
Maruyama H, Fukuoka T, Deguchi I, Ohe Y, Kato Y, Horiuchi Y, Hayashi T, Nagamine Y, Sano H, Tanahashi N. Response to clopidogrel and its association with chronic kidney disease in noncardiogenic ischemic stroke patients. Intern Med 2014; 53:215-9. [PMID: 24492689 DOI: 10.2169/internalmedicine.53.1316] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Noncardiogenic ischemic stroke patients with chronic kidney disease (CKD) are known to have a greater rate of ischemic stroke recurrence than those without. Although clopidogrel is often used to prevent the recurrence of noncardiogenic ischemic stroke, the relationship between the response to clopidogrel and CKD is unclear. In the present study, the relationship between the response to clopidogrel and the presence of CKD was investigated in noncardiogenic ischemic stroke patients. METHODS A total of 129 noncardiogenic ischemic stroke patients receiving 75 mg/day of clopidogrel for ≥1 week were evaluated. The VerifyNow P2Y12 Assay was used to measure the level of platelet aggregation induced by 20 μM of adenosine diphosphate, and the degree of platelet aggregation and frequency of clopidogrel resistance were compared between 34 patients with CKD and 95 patients without CKD. Clopidogrel resistance was defined as a P2Y12 Reaction Units (PRU) value of >230 and/or % inhibition <20%. RESULTS The PRU value was 201.9±91.3 in the patients with CKD and 163.3±86.4 in the patients without CKD (p=0.035). The frequency of a PRU value of >230 was 44.1% (15 patients) among the patients with CKD and 17.9% (17 patients) among those without CKD (p=0.002). The percent inhibition was 29.9%±28.1% among the patients with CKD and 41.1%±28.0% among the patients without CKD (p=0.030). The frequency of % inhibition <20% was 47.1% (16 patients) among the patients with CKD and 26.3% (25 patients) among those without CKD (p=0.026). CONCLUSION The present study showed that noncardiogenic ischemic stroke patients with CKD have a greater frequency of clopidogrel resistance, thus suggesting that the response to clopidogrel is diminished in these patients.
Collapse
Affiliation(s)
- Hajime Maruyama
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Chen X, Shi JG, Emm T, Scherle PA, McGee RF, Lo Y, Landman RR, Punwani NG, Williams WV, Yeleswaram S. Pharmacokinetics and pharmacodynamics of orally administered ruxolitinib (INCB018424 phosphate) in renal and hepatic impairment patients. Clin Pharmacol Drug Dev 2013; 3:34-42. [DOI: 10.1002/cpdd.77] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 09/13/2013] [Indexed: 01/01/2023]
|
16
|
Effects of chronic kidney disease and uremia on hepatic drug metabolism and transport. Kidney Int 2013; 85:522-8. [PMID: 24132209 PMCID: PMC4276411 DOI: 10.1038/ki.2013.399] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/24/2013] [Accepted: 07/17/2013] [Indexed: 01/15/2023]
Abstract
The pharmacokinetics of non-renally cleared drugs in patients with chronic kidney disease is often unpredictable. Some of this variability may be due to alterations in the expression and activity of extra-renal drug metabolizing enzymes and transporters, primarily localized in the liver and intestine. Studies conducted in rodent models of renal failure have shown decreased mRNA and protein expression of many members of the cytochrome P450 enzyme (CYP) gene family and the ATP-Binding Cassette (ABC) and Solute Carrier (SLC) gene families of drug transporters. Uremic toxins interfere with transcriptional activation, cause down-regulation of gene expression mediated by proinflammatory cytokines, and directly inhibit the activity of the cytochrome P450s and drug transporters. While much has been learned about the effects of kidney disease on non-renal drug disposition, important questions remain regarding the mechanisms of these effects, as well as the interplay between drug metabolizing enzymes and drug transporters in the uremic milieu. In this review, we have highlighted the existing gaps in our knowledge and understanding of the impact of chronic kidney disease on non-renal drug clearance, and identified areas of opportunity for future research.
Collapse
|
17
|
Vestergaard B, Agersø H, Lykkesfeldt J. Nephrectomized and Hepatectomized Animal Models as Tools in Preclinical Pharmacokinetics. Basic Clin Pharmacol Toxicol 2013; 113:75-86. [DOI: 10.1111/bcpt.12077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 04/03/2013] [Indexed: 12/13/2022]
Affiliation(s)
| | - Henrik Agersø
- Biopharm Research Unit; Novo Nordisk A/S; Måløv; Denmark
| | - Jens Lykkesfeldt
- Department of Veterinary Disease Biology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen; Denmark
| |
Collapse
|
18
|
Jhaveri KD, Flombaum C, Shah M, Latcha S. A retrospective observational study on the use of capecitabine in patients with severe renal impairment (GFR <30 mL/min) and end stage renal disease on hemodialysis. J Oncol Pharm Pract 2012; 18:140-7. [PMID: 22392964 DOI: 10.1177/1078155210390255] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Capecitabine (Xeloda) is an orally administered precursor of 5'deoxy-5-fluorouridine, which is a preferentially activated to 5-fluorouracil in tumors. It is used in the treatment of colorectal, gastric, and breast cancers. Based on a single Phase II trial, which included a total of 4 patients with severe renal impairment (GFR <30 mL/min), the manufacturer issued a 'Dear Doctor' letter contraindicating the use of capecitabine in these patients since a high rate of grade 3 and 4 adverse events were observed and because these patients tolerated shorter treatment durations.(1) We retrospectively studied 12 patients with a GFR <30 mL/min, including 2 patients with end stage renal disease on hemodialysis, who received capecitabine for mean duration of 7.1 months (1-26 months). The mean serum creatinine at the time of initiation of the drug was 2.63 mg/dL (1.8-6.4 mg/dL) and mean GFR was 20.9 mL/min (8-29 mL/min). Two patients remained on capecitabine after they progressed to end stage renal disease (ESRD) requiring hemodialysis (HD) for an additional 17 and 6 months, respectively. Most patients reported grade 1 and 2 adverse effects (AE), 2 patients reported grade 3 diarrhea and one patient died while on treatment with capecitabine. The starting dose ranged from 250 to 1000 mg/m(2), given twice daily at variable intervals. Dose modifications, with reductions of up to 50% of the starting dose, were made following reports of AEs. Serum tumor marker levels and/or follow up imaging studies were available on 9 patients. Response to capecitabine was documented in 4 patients, stable disease in 2, and disease progression in 3. We conclude that, with close monitoring of their clinical and chemical data, and with dose modification based on reported AEs, capecitabine can be safely administered to patients with severe renal impairment, including patients on hemodialysis.
Collapse
Affiliation(s)
- Kenar D Jhaveri
- Weill Cornell Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | | | | | | |
Collapse
|
19
|
Awad AS, Kamel R, Sherief MAE. Effect of thymoquinone on hepatorenal dysfunction and alteration of CYP3A1 and spermidine/spermine N-1-acetyl-transferase gene expression induced by renal ischaemia-reperfusion in rats. ACTA ACUST UNITED AC 2011; 63:1037-42. [PMID: 21718287 DOI: 10.1111/j.2042-7158.2011.01303.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Renal ischaemia-reperfusion (I/R) is a well-characterised model of acute renal failure that causes both local and remote organ injury. The aim of this work was to investigate the effect of thymoquinone, the main constituent of the volatile oil extracted from Nigella sativa seeds, on renal and hepatic changes after renal ischaemia-reperfusion. METHODS Male Sprague-Dawley rats were divided into sham I/R vehicle-treated groups, and I/R thymoquinone-treated groups. Thymoquinone (10 mg/kg, p.o.) was administered for ten consecutive days to the I/R thymoquinone group before injury. I/R and I/R thymoquinone groups were subjected to 30-min ischaemia followed by 4-h reperfusion. KEY FINDINGS I/R resulted in a significant increase in malondialdehyde (MDA) level and decreases in glutathione-S-transferase (GST) and superoxide dismutase (SOD) activity in liver and kidney tissues. Thymoquinone treatment caused the reversal of I/R-induced changes in MDA as well as GST and SOD activity. Moreover, I/R caused a significant rise in creatinine and alanine aminotransferase serum levels. CYP3A1 mRNA expression was induced significantly by I/R in both liver and kidney tissues compared with sham group. Thymoquinone reduced significantly this increase. I/R caused induction of mRNA expression of spermidine/spermine N-1-acetyl-transferase (SSAT), a catabolic enzyme that participates in polyamine metabolism, in liver and kidney tissues. Thymoquinone reduced SSAT mRNA expression significantly in liver and markedly in kidney. CONCLUSIONS These findings suggested that thymoquinone protected against renal I/R-induced damage through an antioxidant mechanism as well as the decrease of CYP3A1 and SSAT gene expression.
Collapse
Affiliation(s)
- Azza S Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University (Girls), Nasr City, Egypt.
| | | | | |
Collapse
|
20
|
Vlčková H, Solichová D, Bláha M, Solich P, Nováková L. Microextraction by packed sorbent as sample preparation step for atorvastatin and its metabolites in biological samples—Critical evaluation. J Pharm Biomed Anal 2011; 55:301-8. [DOI: 10.1016/j.jpba.2011.01.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 01/20/2011] [Accepted: 01/22/2011] [Indexed: 11/27/2022]
|
21
|
Htun P, Fateh-Moghadam S, Bischofs C, Banya W, Müller K, Bigalke B, Stellos K, May AE, Flather M, Gawaz M, Geisler T. Low responsiveness to clopidogrel increases risk among CKD patients undergoing coronary intervention. J Am Soc Nephrol 2011; 22:627-33. [PMID: 21273381 DOI: 10.1681/asn.2010020220] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Patients with CKD are at higher risk for major events after percutaneous coronary intervention (PCI) compared with subjects with normal renal function. The aims of this study were to evaluate responsiveness to clopidogrel in patients with CKD and to examine the effect of antiplatelet drug response on post-PCI outcome. We retrospectively evaluated a consecutive cohort of 1567 patients with symptomatic coronary artery disease undergoing PCI, 648 (41%) of whom had stage 3 to 5 CKD. We assessed responsiveness to clopidogrel by ADP-induced platelet aggregation after oral administration of a 600-mg clopidogrel loading dose and 100 mg of aspirin. In a multivariate survival analysis that included 1335 (85%) of the cohort, stage 3 to 5 CKD and low response to clopidogrel were independent predictors of the primary end point (composite of myocardial infarction, ischemic stroke, and death within 1 year). In summary, a low response to clopidogrel might be an additional risk factor for the poorer outcomes in patients with stage 3 to 5 CKD compared with patients with better renal function.
Collapse
Affiliation(s)
- Patrik Htun
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universitätsklinikum der Eberhard-Karls-Universität Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sun H, Frassetto LA, Huang Y, Benet LZ. Hepatic clearance, but not gut availability, of erythromycin is altered in patients with end-stage renal disease. Clin Pharmacol Ther 2010; 87:465-72. [PMID: 20090676 DOI: 10.1038/clpt.2009.247] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Nonrenal clearance of drugs can be significantly lower in patients with end-stage renal disease (ESRD) than in those with normal renal function. Using erythromycin (ER) as a probe compound, we investigated whether this decrease in nonrenal clearance is due to reduced hepatic clearance (CL(H)) and/or gut metabolism. We also examined the potential effects of the uremic toxins 3-carboxy-4-methyl-5-propyl-2-furan propanoic acid (CMPF) and indoxyl sulfate (Indox) on ER disposition. Route-randomized, two-way crossover pharmacokinetic studies of ER were conducted in 12 ESRD patients and 12 healthy controls after oral (250 mg) and intravenous (125 mg) dosing with ER. In patients with ESRD, CL(H) decreased 31% relative to baseline values (0.35 +/- 0.14 l/h/kg vs. 0.51 +/- 0.13 l/h/kg, P = 0.01), with no change in steady-state volume of distribution. With oral dosing, the bioavailability of ER increased 36% in patients with ESRD, and this increase was not related to changes in gut availability. As expected, plasma levels of CMPF and Indox were significantly higher in the patients than in the healthy controls. However, no correlation was observed between CL(H) of ER and the levels of uremic toxins.
Collapse
Affiliation(s)
- H Sun
- Department of Biopharmaceutical Sciences, School of Pharmacy, University of California, San Francisco, San Francisco, California, USA
| | | | | | | |
Collapse
|
23
|
Albarellos GA, Montoya L, Landoni MF. Pharmacokinetics of erythromycin after intravenous, intramuscular and oral administration to cats. Vet J 2009; 187:129-32. [PMID: 19854664 DOI: 10.1016/j.tvjl.2009.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 08/13/2009] [Accepted: 09/20/2009] [Indexed: 10/20/2022]
Abstract
The aim of this study was to characterise the pharmacokinetic properties of different formulations of erythromycin in cats. Erythromycin was administered as lactobionate (4 mg/kg intravenously (IV)), base (10mg/kg, intramuscularly (IM)) and ethylsuccinate tablets or suspension (15 mg/kg orally (PO)). After IV administration, the major pharmacokinetic parameters were (mean ± SD): area under the curve (AUC)((0-∞)) 2.61 ± 1.52 microgh/mL; volume of distribution (V(z)) 2.34 ± 1.76L/kg; total body clearance (Cl(t)) 2.1 0 ± 1.37 L/hkg; elimination half-life (t(½)(λ)) 0.75 ± 0.09 h and mean residence time (MRT) 0.88 ± 0.13 h. After IM administration, the principal pharmacokinetic parameters were (mean ± DS): peak concentration (C(max)), 3.54 ± 2.16 microg/mL; time of peak (T(max)), 1.22 ± 0.67 h; t(½)(λ), 1.94 ± 0.21 h and MRT, 3.50 ± 0.82 h. The administration of erythromycin ethylsuccinate (tablets and suspension) did not result in measurable serum concentrations. After IM and IV administrations, erythromycin serum concentrations were above minimum inhibitory concentration (MIC)(90)=0.5 microg/mL for 7 and 1.5h, respectively. However, these results should be interpreted cautiously since tissue erythromycin concentrations have not been measured and can reach much higher concentrations than in blood, which may be associated with enhanced clinical efficacy.
Collapse
Affiliation(s)
- G A Albarellos
- Cátedra de Farmacología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Chorroarín 280 (1427), Buenos Aires, Argentina.
| | | | | |
Collapse
|
24
|
Nováková L, Vlcková H, Satínský D, Sadílek P, Solichová D, Bláha M, Bláha V, Solich P. Ultra high performance liquid chromatography tandem mass spectrometric detection in clinical analysis of simvastatin and atorvastatin. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 877:2093-103. [PMID: 19540175 DOI: 10.1016/j.jchromb.2009.05.052] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Revised: 04/24/2009] [Accepted: 05/26/2009] [Indexed: 11/26/2022]
Abstract
Simvastatin and atorvastatin belong to the group of hypolipidemic drugs, more exactly to the second generation of inhibitors of microsomal 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase. They induce a significant reduction in total cholesterol, low-density lipoprotein cholesterol and plasma triglycerides, therefore they are widely used in the treatment of hypercholesterolemia even of its severe form-familiar hypercholesterolemia. Simvastatin and atorvastatin as the most widely used statins in clinical treatment and their hydroxy-acid/lactone forms were determined by means of UPLC in connection with triple quadrupole mass spectrometer. Deuterium labeled reference standard compounds were used as internal standards for the quantitation. Separation was performed on Acquity BEH C18 (100 mm x 2.1 mm, 1.7 microm) using gradient elution by mobile phase containing acetonitrile and ammonium acetate pH 4.0, which is convenient in order to prevent interconversion of analytes. ESI in positive mode was used for the ionization of all compounds. Two SRM (selected reaction monitoring) transitions were carefully optimized for each analyte in order to get high sensitivity and selectivity. SPE on Discovery DSC-18 was used as a sample preparation step. Intra-day precision was generally within 10% RSD, while inter-day precision within 15% RSD. Method accuracy expressed as recovery ranged from 75 to 100%. The method was validated with the sensitivity reaching LOQ 0.08-5.46 nmol/l and LOD 0.01-1.80 nmol/l in biological samples. Atorvastatin, simvastatin, its metabolites and hydroxy-acid/lactone forms were monitored in human serum and in lipoprotein fractions (LDL, HDL and VLDL) at patients with end stage renal diseases.
Collapse
Affiliation(s)
- Lucie Nováková
- Department of Analytical Chemistry, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Dasgupta A, Steinhubl SR, Bhatt DL, Berger PB, Shao M, Mak KH, Fox KAA, Montalescot G, Weber MA, Haffner SM, Dimas AP, Steg PG, Topol EJ. Clinical outcomes of patients with diabetic nephropathy randomized to clopidogrel plus aspirin versus aspirin alone (a post hoc analysis of the clopidogrel for high atherothrombotic risk and ischemic stabilization, management, and avoidance [CHARISMA] trial). Am J Cardiol 2009; 103:1359-63. [PMID: 19427428 DOI: 10.1016/j.amjcard.2009.01.342] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 01/23/2009] [Accepted: 01/23/2009] [Indexed: 11/16/2022]
Abstract
No prospective randomized trial has specifically examined the long-term outcomes of clopidogrel use in patients with chronic kidney disease. This study aimed to determine the risks and benefits of long-term clopidogrel administration in patients with diabetic nephropathy, the most common form of chronic kidney disease. We performed a post hoc analysis of the CHARISMA trial, which randomly assigned patients without active acute coronary syndrome, but with established atherosclerotic disease (symptomatic) or multiple risk factors for atherosclerotic disease (asymptomatic), to clopidogrel plus aspirin versus placebo plus aspirin. All CHARISMA patients (n = 15,603) were separated into the 3 groups: nondiabetic patients, diabetic patients without nephropathy, and diabetic patients with nephropathy. Within each group, outcomes of patients randomly assigned to clopidogrel were compared with those of patients randomly assigned to placebo. Outcomes in the prespecified CHARISMA subgroups of asymptomatic and symptomatic patients were also compared with respect to study drug assignment and nephropathy status. Patients with nephropathy who received clopidogrel had no difference in bleeding, but experienced significantly increased cardiovascular (CV) and overall mortality compared with those randomly assigned to placebo. There were no differences in bleeding, overall mortality, or CV mortality for nondiabetic or diabetic patients without nephropathy who received clopidogrel versus placebo. In the asymptomatic cohort, patients with nephropathy randomly assigned to clopidogrel had significantly increased overall and CV mortality compared with placebo, whereas asymptomatic patients without nephropathy randomly assigned to clopidogrel had no significant mortality difference compared with placebo. In conclusion, this post hoc analysis suggested that clopidogrel may be harmful in patients with diabetic nephropathy. Additional studies are needed to investigate this possible interaction.
Collapse
Affiliation(s)
- Arijit Dasgupta
- Gill Heart Institute, University of Kentucky, Lexington, Kentucky, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhang Y, Zhang L, Abraham S, Apparaju S, Wu TC, Strong JM, Xiao S, Atkinson Jr AJ, Thummel KE, Leeder JS, Lee C, Burckart GJ, Lesko LJ, Huang SM. Assessment of the Impact of Renal Impairment on Systemic Exposure of New Molecular Entities: Evaluation of Recent New Drug Applications. Clin Pharmacol Ther 2008; 85:305-11. [DOI: 10.1038/clpt.2008.208] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
27
|
Vilay AM, Churchwell MD, Mueller BA. Clinical review: Drug metabolism and nonrenal clearance in acute kidney injury. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 12:235. [PMID: 19040780 PMCID: PMC2646335 DOI: 10.1186/cc7093] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Decreased renal drug clearance is an obvious consequence of acute kidney injury (AKI). However, there is growing evidence to suggest that nonrenal drug clearance is also affected. Data derived from human and animal studies suggest that hepatic drug metabolism and transporter function are components of nonrenal clearance affected by AKI. Acute kidney injury may also impair the clearance of formed metabolites. The fact that AKI does not solely influence kidney function may have important implications for drug dosing, not only of renally eliminated drugs but also of those that are hepatically cleared. A review of the literature addressing the topic of drug metabolism and clearance alterations in AKI reveals that changes in nonrenal clearance are highly complicated and poorly studied, but they may be quite common. At present, our understanding of how AKI affects drug metabolism and nonrenal clearance is limited. However, based on the available evidence, clinicians should be cognizant that even hepatically eliminated drugs and formed drug metabolites may accumulate during AKI, and renal replacement therapy may affect nonrenal clearance as well as drug metabolite clearance.
Collapse
Affiliation(s)
- A Mary Vilay
- Department of Clinical, Social and Administrative Sciences, University of Michigan College of Pharmacy, 428 Church Street, Ann Arbor, MI 48109-1065, USA.
| | | | | |
Collapse
|
28
|
Egorin M, Ramanathan RK, Gibbons J, Petros W, Ivy SP, Remick SC. In Reply. J Clin Oncol 2008. [DOI: 10.1200/jco.2008.18.5470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Merrill Egorin
- University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Ramesh K. Ramanathan
- Scottsdale Clinical Research Institute/Translational Genomics Institute, Scottsdale, AZ
| | | | | | | | | |
Collapse
|
29
|
Dreisbach AW, Lertora JJL. The effect of chronic renal failure on drug metabolism and transport. Expert Opin Drug Metab Toxicol 2008; 4:1065-74. [PMID: 18680441 PMCID: PMC2745294 DOI: 10.1517/17425255.4.8.1065] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Chronic renal failure (CRF) has been shown to significantly reduce the nonrenal clearance and alter bioavailability of drugs predominantly metabolized by the liver and intestine. OBJECTIVES The purpose of this article is to review all significant animal and clinical studies dealing with the effect of CRF on drug metabolism and transport. METHODS A search of the National Library of Medicine PubMed was done with terms such as chronic renal failure, cytochrome P450 [CYP], liver metabolism, efflux drug transport and uptake transport, including relevant articles back to 1969. RESULTS Animal studies in CRF have shown a significant downregulation (40-85%) of hepatic and intestinal CYP metabolism. High levels of parathyroid hormone, cytokines and uremic toxins have been shown to reduce CYP activity. Phase II reactions and drug transporters such as P-glycoprotein and organic anion transporting polypeptide are also affected. CONCLUSION CRF alters intestinal, renal and hepatic drug metabolism and transport producing a clinically significant impact on drug disposition and increasing the risk for adverse drug reactions.
Collapse
Affiliation(s)
- Albert W Dreisbach
- University of Mississippi Medical Center, Division of Nephrology, Department of Medicine, 2500 North State Street, Jackson, MS 39216, USA.
| | | |
Collapse
|
30
|
Simard E, Naud J, Michaud J, Leblond FA, Bonnardeaux A, Guillemette C, Sim E, Pichette V. Downregulation of hepatic acetylation of drugs in chronic renal failure. J Am Soc Nephrol 2008; 19:1352-9. [PMID: 18417721 PMCID: PMC2440299 DOI: 10.1681/asn.2007090974] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 02/02/2008] [Indexed: 02/05/2023] Open
Abstract
Drug metabolism can be affected by chronic renal failure (CRF). Although it is known that several drugs that are known to be acetylated accumulate in CRF, the effect of CRF on N-acetyltransferase (NAT), the enzyme responsible for this acetylation, is unknown. Herein is reported that protein and gene expression of both Nat isoforms in the liver was reduced by >30% and Nat2 activity was reduced by 50% in rats with CRF compared with control rats. Incubation of hepatocytes with serum from rats with CRF suggested that a circulating factor is responsible for the decrease in protein and gene expression. For testing the hypothesis that parathyroid hormone may be this factor, CRF was induced in parathyroidectomized rats; downregulation of Nat expression and activity was not observed in these rats. Furthermore, addition of parathyroid hormone to cultured hepatocytes induced a decrease in Nat2 protein and gene expression. In conclusion, liver acetylation of drugs in a rat model of CRF is reduced by a downregulation of Nat1 and Nat2 isoforms, secondary to decreased gene expression. Parathyroid hormone seems to be an important mediator of this phenomenon.
Collapse
Affiliation(s)
- Emilie Simard
- Service de néphrologie et Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Canada
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Judson IR. Imatinib for Patients With Liver or Kidney Dysfunction: No Need to Modify the Dose. J Clin Oncol 2008; 26:521-2. [DOI: 10.1200/jco.2007.14.5110] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Ian R. Judson
- Sarcoma Unit, Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
32
|
Gibbons J, Egorin MJ, Ramanathan RK, Fu P, Mulkerin DL, Shibata S, Takimoto CH, Mani S, LoRusso PA, Grem JL, Pavlick A, Lenz HJ, Flick SM, Reynolds S, Lagattuta TF, Parise RA, Wang Y, Murgo AJ, Ivy SP, Remick SC. Phase I and Pharmacokinetic Study of Imatinib Mesylate in Patients With Advanced Malignancies and Varying Degrees of Renal Dysfunction: A Study by the National Cancer Institute Organ Dysfunction Working Group. J Clin Oncol 2008; 26:570-6. [PMID: 18235116 DOI: 10.1200/jco.2007.13.3819] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PurposeThis study was undertaken to determine the safety, dose-limiting toxicities (DLT), maximum-tolerated dose (MTD), and pharmacokinetics of imatinib in cancer patients with renal impairment and to develop dosing guidelines for imatinib in such patients.Patients and MethodsSixty adult patients with advanced solid tumors and varying renal function (normal, creatinine clearance [CrCL] ≥ 60 mL/min; mild dysfunction, CrCL 40 to 59 mL/min; moderate dysfunction, CrCL 20 to 39 mL/min; and severe dysfunction, CrCL < 20 mL/min) received daily imatinib doses of 100 to 800 mg. Treatment cycles were 28 days long.ResultsThe MTD was not reached for any group. DLTs occurred in two mild group patients (600 and 800 mg) and two moderate group patients (200 and 600 mg). Serious adverse events (SAEs) were more common in the renal dysfunction groups than in the normal group (P = .0096). There was no correlation between dose and SAEs in any group. No responses were observed. Several patients had prolonged stable disease. Imatinib exposure, expressed as dose-normalized imatinib area under the curve, was significantly greater in the mild and moderate groups than in the normal group. There was a positive correlation between serum alpha-1 acid glycoprotein (AGP) concentration and plasma imatinib, and an inverse correlation between plasma AGP concentration and imatinib clearance. Urinary excretion accounted for 3% to 5% of the daily imatinib dose.ConclusionDaily imatinib doses up to 800 or 600 mg were well tolerated by patients with mild and moderate renal dysfunction, respectively, despite their having increased imatinib exposure.
Collapse
Affiliation(s)
- Joseph Gibbons
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Merrill J. Egorin
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Ramesh K. Ramanathan
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Pingfu Fu
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Daniel L. Mulkerin
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Stephen Shibata
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Chris H.M. Takimoto
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Sridhar Mani
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Patricia A. LoRusso
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Jean L. Grem
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Anna Pavlick
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Heinz-Josef Lenz
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Susan M. Flick
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Sherrie Reynolds
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Theodore F. Lagattuta
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Robert A. Parise
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Yanfeng Wang
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Anthony J. Murgo
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - S. Percy Ivy
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| | - Scot C. Remick
- From the Developmental Therapeutics Program, Case Comprehensive Cancer Center, Ireland Cancer Center at University Hospitals of Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH; University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Wisconsin Comprehensive Cancer Center, Madison, WI; City of Hope National Medical Center, Duarte; University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA; University of Texas Health
| |
Collapse
|
33
|
Naud J, Michaud J, Leblond FA, Lefrancois S, Bonnardeaux A, Pichette V. Effects of chronic renal failure on liver drug transporters. Drug Metab Dispos 2008; 36:124-8. [PMID: 17940133 DOI: 10.1124/dmd.107.018192] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Chronic renal failure (CRF) is associated with a decrease in liver drug metabolism, particularly mediated by the cytochrome P450. CRF also impedes intestinal drug transporters [mainly P-glycoprotein (P-gp) and multidrug resistance protein (MRP)]. However, very few studies have evaluated the effects of CRF on liver drug transport. The present study aimed to investigate the repercussions of CRF on liver drug transporters involved in hepatic uptake [organic anion transporting polypeptide (Oatp) 2] and in drug extrusion (P-gp and MRP2). Two groups of rats were studied: control and CRF. Oatp2, P-gp, and MRP2 protein expressions and mRNA levels, as well as some of their metabolic activity, were assessed. The effects of CRF serum on drug transporters were also evaluated in cultured hepatocytes. Compared with control, creatinine clearance was reduced by 70% (p < 0.01) in rats with CRF. Protein expression and mRNA levels of P-gp were increased by 25 and 40% (p < 0.01), respectively, in liver from rats with CRF. MRP2 protein expression was identical in both groups, whereas its mRNA levels were increased by 35% (p < 0.01) in CRF rats. Finally, Oatp2 protein expression was reduced by 35%, whereas its mRNA levels remained unchanged. Similar results were obtained when hepatocytes were incubated with uremic serum. In conclusion, CRF is associated with a decrease in liver transporters involved in drug absorption and an increase in those involved in drug extrusion. Uremic mediators appear to be responsible for these modifications.
Collapse
Affiliation(s)
- Judith Naud
- Service de Néphrologie et Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Université de Montréal, Québec, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Naud J, Michaud J, Boisvert C, Desbiens K, Leblond FA, Mitchell A, Jones C, Bonnardeaux A, Pichette V. Down-regulation of intestinal drug transporters in chronic renal failure in rats. J Pharmacol Exp Ther 2007; 320:978-85. [PMID: 17135344 DOI: 10.1124/jpet.106.112631] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic renal failure (CRF) is associated with an increased bioavailability of drugs by a poorly understood mechanism. One hypothesis is a reduction in the elimination of drugs by the intestine, i.e., drug elimination mediated by protein membrane transporters such as P-glycoprotein (Pgp) and multidrug-resistance-related protein (MRP) 2. The present study aimed to investigate the repercussions of CRF on intestinal transporters involved in drug absorption [organic anion-transportingpolypeptide (Oatp)] and those implicated in drug extrusion (Pgp and MRP2). Pgp, MRP2, MRP3, Oatp2, and Oatp3 protein expression and Pgp, MRP2, and Oatp3 mRNA expression were assessed in the intestine of CRF (induced by five-sixth nephrectomy) and control rats. Pgp and MRP2 activities were measured using the everted gut technique. Rat enterocytes and Caco-2 cells were incubated with sera from control and CRF rats to characterize the mechanism of transporters' down-regulation. Protein expression of Pgp, MRP2, and MRP3 were reduced by more than 40% (p < 0.01) in CRF rats, whereas Oatp2 and Oatp3 expression remained unchanged. There was no difference in the mRNA levels assessed by real-time polymerase chain reaction. Pgp and MRP2 activities were decreased by 30 and 25%, respectively, in CRF rats compared with control (p < 0.05). Uremic sera induced a reduction in protein expression and in activity of drug transporters compared with control sera. Our results demonstrate that CRF in rats is associated with a decrease in intestinal Pgp and MRP2 protein expression and function secondarily to serum uremic factors. This reduction could explain the increased bioavailability of drugs in CRF.
Collapse
Affiliation(s)
- Judith Naud
- Centre de Recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, 5415 Boulevard de l'Assomption, Montreal, Quebec, Canada H1T 2M4
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tanabe H, Taira S, Taguchi M, Hashimoto Y. Pharmacokinetics and Hepatic Extraction of Metoprolol in Rats with Glycerol-Induced Acute Renal Failure. Biol Pharm Bull 2007; 30:552-5. [PMID: 17329855 DOI: 10.1248/bpb.30.552] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to evaluate the effect of glycerol-induced acute renal failure (ARF) on the pharmacokinetics and hepatic extraction of metoprolol in rats. Experimental ARF in rats was induced by injections of 50% glycerol into the leg muscle (10 ml/kg). Pharmacokinetics and hepatic extraction of metoprolol was evaluated by means of intravenous, intra-intestinal, and intra-portal administration of the drug. The blood metoprolol concentration following intravenous infusion in ARF rats was similar to that in control rats. On the other hand, the blood metoprolol concentration at 5--10 min after intra-intestinal administration in ARF rats was significantly higher than that in control rats, and the oral clearance (CL/F) of the drug was significantly decreased in ARF rats. Hepatic extraction following intra-portal infusion was not altered by glycerol-induced ARF; however, hepatic first-pass extraction of metoprolol was dose-dependent and saturable in both ARF and control rats. These results suggested that the decreased CL/F of metoprolol in rats with glycerol-induced ARF is mainly a result of the increased initial absorption rate in the intestine followed by partial saturation of hepatic first-pass metabolism.
Collapse
Affiliation(s)
- Hiroko Tanabe
- Graduate School of Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | | | | | | |
Collapse
|
36
|
Michaud J, Naud J, Chouinard J, Désy F, Leblond FA, Desbiens K, Bonnardeaux A, Pichette V. Role of Parathyroid Hormone in the Downregulation of Liver Cytochrome P450 in Chronic Renal Failure. J Am Soc Nephrol 2006; 17:3041-8. [PMID: 17021269 DOI: 10.1681/asn.2006010035] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Chronic renal failure (CRF) is associated with a decrease in drug metabolism secondary to a decrease in liver cytochrome P450 (P450). The predominant theory to explain this decrease is the presence of factors in the blood of uremic patients. This study tested the hypothesis that parathyroid hormone (PTH) could be this factor. The objectives of this study were to determine (1) the role of PTH in the downregulation of hepatocyte P450 induced by rat uremic serum, (2) the role of PTH in the downregulation of liver P450 in rats with CRF, and (3) the effects of PTH on P450 in hepatocytes. For this purpose, (1) hepatocytes were incubated with serum from rat with CRF that was depleted with anti-PTH antibodies or with serum from parathyroidectomized (CRF-PTX) rat with CRF, (2) the effect of PTX on liver P450 was evaluated in rats with CRF, and (3) the effects of PTH on P450 in hepatocytes were determined. The depletion of PTH from CRF serum completely reversed the downregulating effect of CRF serum on P450 in hepatocytes. Addition of PTH (10(-9) M) to depleted CRF serum induced a decrease in P450 similar to nondepleted CRF serum. The serum of CRF-PTX rats had no effect on P450 in hepatocytes compared with CRF serum. Adding PTH to CRF-PTX serum induced a similar decrease in P450 as obtained with CRF serum. Finally, PTX prevented the decrease of liver P450 in rats with CRF. In summary, PTH is the major mediator implicated in the downregulation of liver P450 in rats with CRF.
Collapse
Affiliation(s)
- Josée Michaud
- Service de néphrologie et Centre de Recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Unfortunately, there is no endogenous marker for hepatic clearance that can be used as a guide for drug dosing. In order to predict the kinetic behaviour of drugs in cirrhotic patients, agents can be grouped according to their extent of hepatic extraction. For drugs with a high hepatic extraction (low bioavailability in healthy subjects), bioavailability increases and hepatic clearance decreases in cirrhotic patients. If such drugs are administered orally to cirrhotic patients, their initial dose has to be reduced according to hepatic extraction. Furthermore, their maintenance dose has to be adapted irrespective of the route of administration, if possible, according to kinetic studies in cirrhotic patients. For drugs with a low hepatic extraction, bioavailability is not affected by liver disease, but hepatic clearance may be affected. For such drugs, only the maintenance dose has to be reduced, according to the estimated decrease in hepatic drug metabolism. For drugs with an intermediate hepatic extraction, initial oral doses should be chosen in the low range of normal in cirrhotic patients and maintenance doses should be reduced as for high extraction drugs. In cholestatic patients, the clearance of drugs with predominant biliary elimination may be impaired. Guidelines for dose reduction in cholestasis exist for many antineoplastic drugs, but are mostly lacking for other drugs with biliary elimination. Dose adaptation of such drugs in cholestatic patients is, therefore, difficult and has to be performed according to pharmacological effect and/or toxicity. Importantly, the dose of drugs with predominant renal elimination may also have to be adapted in patients with liver disease. Cirrhotic patients often have impaired renal function, despite a normal serum creatinine level. In cirrhotic patients, creatinine clearance should, therefore, be measured or estimated to gain a guideline for the dosing of drugs with predominant renal elimination. Since the creatinine clearance tends to overestimate glomerular filtration in cirrhotic patients, the dose of a given drug may still be too high after adaptation to creatinine clearance. Therefore, the clinical monitoring of pharmacological effects and toxicity of such drugs is important. Besides the mentioned kinetic changes, the dynamics of some drugs is also altered in cirrhotic patients. Examples include opiates, benzodiazepines, NSAIDs and diuretics. Such drugs may exhibit unusual adverse effects that clinicians should be aware of for their safe use. However, it is important to realise that the recommendations for dose adaptation remain general and cannot replace accurate clinical monitoring of patients with liver disease treated with critical drugs.
Collapse
Affiliation(s)
- Fabiola Delcò
- Division of Gastroenterology, University Hospital of Basel, Switzerland
| | | | | | | | | |
Collapse
|
38
|
Sun H, Frassetto L, Benet LZ. Effects of renal failure on drug transport and metabolism. Pharmacol Ther 2006; 109:1-11. [PMID: 16085315 DOI: 10.1016/j.pharmthera.2005.05.010] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2005] [Indexed: 01/11/2023]
Abstract
Renal failure not only alters the renal elimination, but also the non-renal disposition of drugs that are extensively metabolized by the liver. Although reduced metabolic enzyme activity in some cases can be responsible for the reduced drug clearance, alterations in the transporter systems may also be involved in the process. With the development of renal failure, the renal secretion of organic ions mediated by organic anion transporters (OATs) and organic cation transporters (OCTs) is decreased. 3-Carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF) and other organic anionic uremic toxins may directly inhibit the renal excretion of various drugs and endogenous organic acids by competitively inhibiting OATs. In addition, the expression of OAT1 and OCT2 was reduced in chronic renal failure (CRF) rats. Renal failure also impairs the liver uptake of drugs and organic anions, such as bromosulphophthalein (BSP), indocyanine green (ICG), and thyroxine, where organic anion transport polypeptides (OATPs) are the major transporters. Most previous studies have been done in animals or cell culture, very often in rat models, but these are presumed to reflect the presentation of advanced renal disease in humans as well. Recent studies demonstrate that the uremic toxins CMPF and indoxyl sulfate (IS) can directly inhibit rOatp2 and hOATP-C in hepatocytes. The protein content of the liver uptake transporters Oatp1, 2, and 4 were significantly decreased in CRF rats. Decreased activity of the intestinal efflux transporter, P-glycoprotein (P-gp), was also observed in CRF rats, with no significant change of protein content, suggesting that uremic toxins may suppress P-gp function. However, increased protein levels of multidrug resistance-associated protein (MRP) 2 in the kidney and MRP3 in the liver were found in CRF rats, suggesting an adaptive response that may serve as a protective mechanism. Increases in drug areas under the curve (AUCs) in subjects with advanced renal disease for drugs that are not renally excreted are consistent with uremic toxin effects on either intestinal or hepatic cell transporters, metabolizing enzymes, or both. In conclusion, alterations of drug transporters, as well as metabolic enzymes, in patients with renal failure can be responsible for reduced drug clearance.
Collapse
Affiliation(s)
- Hong Sun
- Department of Biopharmaceutical Sciences, University of California, San Francisco, CA 94143-0446, United States
| | | | | |
Collapse
|
39
|
Higashi T, Urai M, Taguchi M, Hashimoto Y. Mechanism responsible for the decreased hepatic NADPH generation rate in rats with bilateral ureter ligation-induced renal failure. Biol Pharm Bull 2005; 28:1809-12. [PMID: 16141568 DOI: 10.1248/bpb.28.1809] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that a decrease in hepatic NADPH generation results in reduced hepatic first-pass clearance of propranolol in rats with bilateral ureteral ligation (BUL)-induced renal failure. The aim of the present study was to evaluate the mechanisms responsible for the reduced NADPH generation in the supernatant of liver homogenates (SUP) obtained from rats with BUL. The NADPH generation in the SUP in the presence of NADP(+) was decreased in BUL rats as compared with control rats. After the addition of glucose-6-phosphate or 6-phosphogluconic acid, the increase in NADPH in the SUP of BUL rats was similar to that in control rats. The NADPH generation in the SUP after the addition of the ultrafiltrate of BUL rat SUP was smaller than that after the addition of the ultrafiltrate of control rat SUP. These findings suggest that the enzymatic activities in the pentose phosphate pathway were not decreased significantly in BUL rats, and that the decrease in the generation of NADPH in BUL rats was mainly caused by the decreased concentration of endogenous substrate(s) and/or the increased concentration of endogenous inhibitor(s) for the pentose phosphate pathway.
Collapse
Affiliation(s)
- Takashi Higashi
- Graduate School of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, Toyama, Japan
| | | | | | | |
Collapse
|
40
|
Michaud J, Dubé P, Naud J, Leblond FA, Desbiens K, Bonnardeaux A, Pichette V. Effects of serum from patients with chronic renal failure on rat hepatic cytochrome P450. Br J Pharmacol 2005; 144:1067-77. [PMID: 15700027 PMCID: PMC1576090 DOI: 10.1038/sj.bjp.0706138] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. In humans, chronic renal failure (CRF) is associated with decreased hepatic drug metabolism, particularly that mediated by the cytochrome P450 (P450). The mechanisms remain poorly understood. The present study aimed to investigate the effects of the serum of patients with CRF on liver P450, and to evaluate whether renal replacement therapies (dialysis or transplantation) impede the inhibition of CRF serum on P450. 2. Rat hepatocytes were incubated for 24 h with serum from patients with severe CRF and from controls to measure (1) P450 level, (2) protein expression and mRNA levels of P450 isoforms and (3) metabolic activities of CYP3A and CYP1A. Similar experiments were performed with serum of patients once on chronic hemodialysis and after kidney transplantation. 3. In rat hepatocytes incubated for 24 h with serum from patients with CRF, P450 level and protein expression, as well as mRNA levels of P450 isoforms (CYP1A2, 2C6, 2C11, 2D1/2D2, 3A2 and 4A1/4A3), were decreased by more than 45% (P<0.001) compared to control serum, while the levels of CYP2E1 were not modified. CYP3A and CYP1A activities were decreased by 51 and 59% (P<0.001), respectively. The inhibitory effect of serum obtained from patients before first dialysis was similar after 1 or 6 months on chronic hemodialysis but was lost after successful kidney transplantation. In CRF serum, the fraction containing proteins between 10 and 15 kDa decreases P450. 4. Human uremic serum contains mediator(s) that decreases rat hepatic P450 activity and expression secondary to reduced gene expression. The inhibitory effect of serum persists even after initiation of dialysis, but disappears after normalization of renal function following kidney transplantation.
Collapse
Affiliation(s)
- Josée Michaud
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, 5415 boul.de l'Assomption, Montreal, Québec, Canada H1T 2H4
| | - Pierre Dubé
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, 5415 boul.de l'Assomption, Montreal, Québec, Canada H1T 2H4
| | - Judith Naud
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, 5415 boul.de l'Assomption, Montreal, Québec, Canada H1T 2H4
| | - Francois A Leblond
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, 5415 boul.de l'Assomption, Montreal, Québec, Canada H1T 2H4
| | - Karine Desbiens
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, 5415 boul.de l'Assomption, Montreal, Québec, Canada H1T 2H4
| | - Alain Bonnardeaux
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, 5415 boul.de l'Assomption, Montreal, Québec, Canada H1T 2H4
| | - Vincent Pichette
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, 5415 boul.de l'Assomption, Montreal, Québec, Canada H1T 2H4
- Author for correspondence:
| |
Collapse
|
41
|
Sun H, Huang Y, Frassetto L, Benet LZ. Effects of uremic toxins on hepatic uptake and metabolism of erythromycin. Drug Metab Dispos 2004; 32:1239-46. [PMID: 15286055 DOI: 10.1124/dmd.104.000521] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hepatic clearance of erythromycin (Ery) is significantly reduced in patients with end stage renal disease. Since Ery is primarily eliminated via excretion of unchanged drug in the bile, we suspect that this change could be due to the effect of uremic toxins on hepatic uptake and/or efflux transporters. Using rat hepatocytes and microsomes as model proof of concept systems, we examined six uremic toxins, 3-carboxy-4-methyl-5-propyl-2-furan-propanoic acid (CMPF), indoxyl sulfate (IS), hippuric acid (HA), indole acetic acid (IA), guanidinosuccinic acid (GSA), and indoxyl-beta-D-glucuronide (IG), for their effects on Ery uptake and metabolism. Ery and the metabolite N-demethyl-Ery were measured by liquid chromatography/tandem mass spectrometry. The uptake of Ery by rat hepatocytes was markedly inhibited by rifampin and digoxin, but not by quinidine, suggesting that Oatp2 plays a major role in the uptake of Ery. At 50 microM, CMPF significantly (p < 0.05) reduced hepatocyte accumulation of Ery and N-demethyl-Ery. At higher concentrations (>200 microM), CMPF appears to also inhibit the enzymatic metabolism of Ery. In contrast, IS did not significantly inhibit the hepatocyte uptake of Ery, even at the highest concentration (800 microM) tested, but reduced metabolite generation (p < 0.001). The other uremic toxins, HA, IA, IG, and GSA, did not affect either hepatic uptake or microsomal metabolism of Ery. CMPF, IS, and HA were shown not to inhibit differential P-glycoprotein transport of Ery in cellular systems. Our results suggest that CMPF can directly inhibit the uptake of Ery by inhibiting Oatp2, whereas IS is more likely to inhibit the enzymatic metabolism of Ery.
Collapse
Affiliation(s)
- Hong Sun
- Department of Biopharmaceutical Sciences, University of California, San Francisco, California 94143-0446, USA
| | | | | | | |
Collapse
|
42
|
Okabe H, Hasunuma M, Hashimoto Y. The hepatic and intestinal metabolic activities of P450 in rats with surgery- and drug-induced renal dysfunction. Pharm Res 2004; 20:1591-4. [PMID: 14620512 DOI: 10.1023/a:1026131216669] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE The hepatic and intestinal metabolic activities of P450 were evaluated in rats with surgery- and drug-induced renal dysfunction. METHODS Renal failure was induced by five-sixths nephrectomy (NR), bilateral ureter ligation (BUL), the intramuscular injection of glycerol (GL), and the intraperitoneal injection of cisplatin (CDDP). Phenytoin 4-hydroxylation, debrisoquine 4-hydroxylation, and testosterone 6beta-hydroxylation were estimated to evaluate the metabolic activities of cytochrome P450 (CYP) 2C, 2D, and 3A, respectively. RESULTS The hepatic CYP3A metabolic activities were decreased by 65.9% and 60.2% in NR and GL rats, respectively. The hepatic CYP2C metabolic activity was decreased by 48.8% in CDDP rats. No alteration in hepatic drug-metabolizing activities was observed in BUL rats. On the other hand, the intestinal CYP3A metabolic activity was weakly increased in GL rats but not significantly altered in NR, CDDP, and BUL rats. CONCLUSIONS This study suggested (a) that only selected P450 metabolic activity in the liver is decreased in renal failure, (b) that extent of the decrease in hepatic metabolic activities of P450 is dependent on the etiology of renal failure, and (c) that alteration of CYP3A metabolic activity in the intestine is not always correlated with that in the liver.
Collapse
Affiliation(s)
- Hiromi Okabe
- Graduate School of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, 2630 Sugitani, Toyama 930-0194, Japan
| | | | | |
Collapse
|
43
|
Rege B, Krieg R, Gao N, Sarkar MA. Down-regulation of hepatic CYP3A in chronic renal insufficiency. Pharm Res 2004; 20:1600-6. [PMID: 14620514 DOI: 10.1023/a:1026135317578] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE The objective of this study was to investigate the mechanisms underlying the decrease in hepatic clearance of some drugs metabolized by CYP450 enzymes in chronic renal insufficiency (CRI). METHODS CRI was induced in male Sprague-Dawley rats (n = 7) by the remnant kidney model (RKM); control animals (C) (n = 12) underwent sham surgery, of which n = 6 rats were pair-fed (CPF) with CRI rats and others (n = 6) had free access to food. Serum creatinine (Scr) and urea nitrogen (SUN) were monitored every 2 weeks. On day 36, livers were isolated, and microsomes were prepared. Catalytic activities were measured through O-demethylation (CYP2D) and N-demethylation of dextromethorphan (CYP3A) and O-deethylation of 7-ethoxyresorufin (CYP1A2). CYP450 protein and mRNA levels were also measured. RESULTS Compared with CPF, Scr and SUN levels in CRI rats were increased twofold (p < 0.01) and 2.5-fold (p < 0.01), respectively. No effect on CYP1A2 and CYP2D activities, mRNA, or protein levels was observed between the groups. There was a reduction (41.8 +/- 20%, p < 0.01) in CYP3A activity, mRNA (p < 0.05), and protein levels (p < 0.05) in CRI rats compared to CPF. CONCLUSIONS CRI induced by RKM does not have an effect on hepatic CYP1A2 and CYP2D enzymes but does reduce CYP3A activity, probably through down-regulation of CYP3A2.
Collapse
Affiliation(s)
- Bhaskar Rege
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
44
|
|
45
|
Okabe H, Higashi T, Ohta T, Hashimoto Y. Intestinal Absorption and Hepatic Extraction of Propranolol and Metoprolol in Rats with Bilateral Ureteral Ligation. Biol Pharm Bull 2004; 27:1422-7. [PMID: 15340230 DOI: 10.1248/bpb.27.1422] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate the mechanism responsible for the increased bioavailability of propranolol in bilateral ureter-ligated (BUL) rats, the intestinal absorption and hepatic extraction of propranolol and metoprolol were evaluated. The initial absorption rate of these drugs after intra-intestinal administration was only slightly increased in the BUL rats, whereas the blood drug concentration in these rats was higher than that in control rats. The blood propranolol and metoprolol concentrations during intra-portal infusion in the BUL rat were significantly higher than that in the control rat. In the presence of NADPH, the intrinsic metabolic activity of metoprolol in hepatic microsomes was not altered by BUL. On the other hand, the NADPH generation rate in the hepatic cytosol in the BUL group was lower than that in the control group. These results indicate that the absorption rate-dependent decrease in hepatic first-pass clearance of propranolol and metoprolol due to saturation kinetics is marginal, and that the hepatic metabolic activity and extraction of the drugs is significantly decreased in BUL rats probably due to the reduced NADPH generation rate in the liver.
Collapse
Affiliation(s)
- Hiromi Okabe
- Graduate School of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University; 2630 Sugitani, Toyama 930-0194, Japan
| | | | | | | |
Collapse
|
46
|
Nolin TD, Frye RF, Matzke GR. Hepatic drug metabolism and transport in patients with kidney disease. Am J Kidney Dis 2003; 42:906-25. [PMID: 14582035 DOI: 10.1016/j.ajkd.2003.07.019] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The disposition of many drugs is altered in patients with acute (AKD) and chronic kidney disease (CKD). A decline in renal clearance of several drugs has been correlated significantly with residual renal function (ie, creatinine clearance) of subjects. Reductions in nonrenal clearance of some compounds also have been reported and associated with clearance of markers of oxidative and/or conjugative metabolism or P-glycoprotein-mediated transport. Although initial accounts of reduced hepatic microsomal cytochrome P-450 (CYP) content and activity in animal models of AKD and CKD were published almost 25 years ago, it is only in the last decade that technical advances in molecular biology and clinical pharmacology have enabled researchers to begin to characterize the phenotypic expression of individual enzymes and, importantly, distinguish the molecular and/or genetic basis for these changes. The selective modulation of hepatic CYP enzyme activity observed in kidney disease is caused, at least in part, by differentially altered expression of several CYP isoforms. This review summarizes data available through June 2003 regarding the effect of AKD and CKD on drug metabolism. Knowledge of the impact and nature of these alterations associated with kidney disease may facilitate the individualization of medication management in this patient population.
Collapse
Affiliation(s)
- Thomas D Nolin
- Department of Pharmacy Services and Division of Nephrology and Renal Transplantation, Maine Medical Center, Portland, ME, USA
| | | | | |
Collapse
|
47
|
Budde K, Neumayer HH, Fritsche L, Sulowicz W, Stompôr T, Eckland D. The pharmacokinetics of pioglitazone in patients with impaired renal function. Br J Clin Pharmacol 2003; 55:368-74. [PMID: 12680885 PMCID: PMC1884238 DOI: 10.1046/j.1365-2125.2003.01785.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS To evaluate the effect of renal impairment on the pharmacokinetics and safety of pioglitazone and its metabolites M-III and M-IV with impaired renal function and normal renal function. METHODS In a phase-I, open-label, parallel-group study, six healthy subjects with normal renal function (creatinine clearance> 80 ml min-1), nine patients with moderate renal impairment (creatinine clearance 30-60 ml min-1) and 12 patients with severe renal impairment (creatinine clearance < 30 ml min-1) received single and multiple oral doses of pioglitazone 45 mg. The serum pharmacokinetic profiles of pioglitazone and its metabolites M-III and M-IV were assessed for the first and last dose administered (day 1 and day 12, respectively). RESULTS Pharmacokinetic data revealed no significant accumulation of pioglitazone or its metabolites M-III and M-IV in patients with renal impairment. There was no significant difference in the pharmacokinetic profile of pioglitazone in subjects with normal and with moderately impaired renal function. After single oral doses, mean area under the concentration-time curve (AUC) values were decreased in patients with severe renal impairment compared with healthy subjects with normal renal function for pioglitazone (13 476 vs 17 387, P = 0.371; -23%; confidence interval (CI) -57, 38), M-III metabolite (13 394 vs 15 071, P = 0.841; -11%; CI -74, 194) and M-IV metabolite (27 991 vs 49 856, P = 0.006; -44%; CI -62, -17). After repeated oral doses of pioglitazone, mean AUC values (microg.h l-1) were decreased in patients with severe renal impairment compared with healthy subjects with normal renal function for pioglitazone (8744 vs 14,565, P = 0.004; -40%; CI -57, -16), M-III (3991 vs 7,289, P = 0.0009; -45%; CI -60, -25) and M-IV (21 080 vs 25 706, P = 0.181; -18%; CI 39, 10). The tolerability and safety profile of pioglitazone was comparable between groups. CONCLUSIONS Pioglitazone was well tolerated in patients with varying degrees of renal impairment. Although mean serum concentrations of pioglitazone and its metabolites are increased in patients with severe renal impairment, adjustment of starting and maintenance doses in these patients is probably unwarranted.
Collapse
Affiliation(s)
- Klemens Budde
- Department of Nephrology, University Hospital Charité, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Alteration in liver function are not typically present in patients with uremic syndrome, but varying degrees of liver dysfunction were observed in animals with experimental uremia and, to a lesser degree, in patients with chronic renal failure. This article summarizes the data obtained during the last 2 decades on protein, carbohydrate, and lipid metabolism by the liver in uremia and molecular aspects of regulation of lipids and protein synthesis. Particular attention is given to the role of cytosolic calcium ([Ca(2+)](i)) regulation and calcium signal transduction in hepatocytes in chronic renal failure. It is proposed that the parathyroid hormone (PTH)-mediated increase in the [Ca(2+)](i) of hepatocytes in chronic renal failure is a major signal for the downregulation of hepatic receptors for PTH-PTHrP, vasopressin and angiotensin II as well as as hepatic lipase. It is possible that the mRNA of other hormone receptors and various proteins of the liver cells are affected similarly by the elevated basal levels of [Ca(2+)](i) in CRF.
Collapse
Affiliation(s)
- Miroslaw J Smogorzewski
- Division of Nephrology and the Department of Medicine, University of Southern California, Los Angeles, USA.
| | | |
Collapse
|
49
|
Guévin C, Michaud J, Naud J, Leblond FA, Pichette V. Down-regulation of hepatic cytochrome p450 in chronic renal failure: role of uremic mediators. Br J Pharmacol 2002; 137:1039-46. [PMID: 12429576 PMCID: PMC1573574 DOI: 10.1038/sj.bjp.0704951] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. Chronic renal failure (CRF) is associated with a decrease in liver cytochrome p450 (p450). The mechanism remains poorly understood. The present study aimed to investigate the effects of the serum of rats with CRF on liver p450. 2. Normal rat hepatocytes were incubated for 24 h with serum (concentration of 10%) from rats with CRF and from control animals in order to measure (1). total p450 level, (2). protein expression and mRNA levels of major p450 isoforms, and (3). some of their specific metabolic activities (N-demethylation of erythromycin). Time-course experiments (incubation time from 12 to 48 h) and dose-response curves (concentration of serum ranging from 1 to 30%) have been conducted. 3. In normal hepatocytes incubated for 24 h with serum (concentration of 10%) from rats with CRF, total p450 level, protein expression and mRNA levels of several p450 isoforms (CYP2C6, 2C11, 3A1 and 3A2) were decreased by more than 35% (P<0.001) compared to serum from control animals. The protein expression as well as the mRNA levels of CYP2D were similar in hepatocytes incubated with serum from either control or CRF rats. The N-demethylation of erythromycin was decreased by more than 35% (P<0.001) in hepatocytes incubated with serum from rats with CRF. The inhibitory effect of serum from rats with CRF tended to peak at 48 h of incubation and was maximum at a concentration of 20%. 4. In conclusion, uremic serum contains mediator(s) that down-regulate the cytochrome p450 of normal hepatocytes secondary to reduced gene expression.
Collapse
MESH Headings
- Animals
- Blood Proteins/chemistry
- Blood Proteins/pharmacology
- Blotting, Western
- Body Weight
- Chemical Fractionation
- Culture Media/chemistry
- Culture Media/pharmacology
- Cytochrome P-450 Enzyme System/genetics
- Cytochrome P-450 Enzyme System/metabolism
- Dose-Response Relationship, Drug
- Down-Regulation
- Erythromycin/metabolism
- Gene Expression Regulation, Enzymologic/drug effects
- Hepatocytes/cytology
- Hepatocytes/drug effects
- Hepatocytes/enzymology
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Kidney Failure, Chronic/blood
- Kidney Failure, Chronic/enzymology
- Liver/cytology
- Liver/enzymology
- Male
- Microsomes, Liver/drug effects
- Microsomes, Liver/metabolism
- Molecular Weight
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Time Factors
- Uremia/blood
- Uremia/enzymology
Collapse
Affiliation(s)
- Carl Guévin
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, Québec, Canada
| | - Josée Michaud
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, Québec, Canada
| | - Judith Naud
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, Québec, Canada
| | - Francois A Leblond
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, Québec, Canada
| | - Vincent Pichette
- Service de néphrologie et Centre de recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Université de Montréal, Québec, Canada
- Author for correspondence:
| |
Collapse
|
50
|
Leblond F, Guévin C, Demers C, Pellerin I, Gascon-Barré M, Pichette V. Downregulation of hepatic cytochrome P450 in chronic renal failure. J Am Soc Nephrol 2001; 12:326-332. [PMID: 11158222 DOI: 10.1681/asn.v122326] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Chronic renal failure (CRF) is associated with a decrease in drug metabolism. The mechanism remains poorly understood. The present study investigated the repercussions of CRF on liver cytochrome P450 (CYP450). Three groups of rats were defined: control, control paired-fed, and CRF. Total CYP450 activity, protein expression of several CYP450 isoforms as well as their mRNA, and the in vitro N-demethylation of erythromycin were assessed in liver microsomes. The regulation of liver CYP450 by dexamethasone and phenobarbital was assessed in CRF rats. Compared with control and control paired-fed rats, creatinine clearance was reduced by 60% (P: < 0.01) in CRF rats. Weight was reduced by 30% (P: < 0.01) in control paired-fed and CRF rats, compared with control animals. There was no difference in the CYP450 parameters between control and control paired-fed. Compared with control paired-fed rats, total CYP450 was reduced by 47% (P: < 0.001) in CRF rats. Protein expression of CYP2C11, CYP3A1, and CYP3A2 were considerably reduced (>40%, P: < 0.001) in rats with CRF. The levels of CYP1A2, CYP2C6, CYP2D, and CYP2E1 were the same in the three groups. Northern blot analysis revealed a marked downregulation in gene expression of CYP2C11, 3A1, and 3A2 in CRF rats. Although liver CYP450 was reduced in CRF, its induction by dexamethasone and phenobarbital was present. N-demethylation of erythromycin was decreased by 50% in CRF rats compared with control (P: < 0.001). In conclusion, CRF in rats is associated with a decrease in liver cytochrome P450 activity (mainly in CYP2C11, CYP3A1, and 3A2), secondary to reduced gene expression.
Collapse
Affiliation(s)
- Francois Leblond
- Service de Néphrologie et Centre de Recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Université de Montréal, Québec, Canada
| | - Carl Guévin
- Service de Néphrologie et Centre de Recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Université de Montréal, Québec, Canada
| | - Christian Demers
- Centre Hospitalier Universitaire de Montréal, Faculté de Médecine, Université de Montréal, Québec, Canada
| | - Isabelle Pellerin
- Service de Néphrologie et Centre de Recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Université de Montréal, Québec, Canada
| | - Marielle Gascon-Barré
- Centre Hospitalier Universitaire de Montréal, Faculté de Médecine, Université de Montréal, Québec, Canada
| | - Vincent Pichette
- Service de Néphrologie et Centre de Recherche Guy-Bernier, Hôpital Maisonneuve-Rosemont, Université de Montréal, Québec, Canada
| |
Collapse
|