1
|
Sandhu G, Adattini J, Gordon EA, O’Neill N, Bagnis C, Chambers P, Martin JH, Flynn A, Ibrahim K, Jardine MJ, Johnson DW, Jones GR, Karapetis CS, Kelly A, Kichenadasse G, Kliman DS, Liauw W, Lucas C, Mallett AJ, Malyszko J, Michael M, Pollock CA, Roberts DM, Rosner MH, Routledge DJ, Scuderi C, Shingleton J, Shortt J, Siderov J, Sprangers B, Stein BN, Tunnicliffe DJ, Webber K, Ward RL. Aligning kidney function assessment in patients with cancer to global practices in internal medicine. EClinicalMedicine 2025; 82:103102. [PMID: 40290845 PMCID: PMC12034077 DOI: 10.1016/j.eclinm.2025.103102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 04/30/2025] Open
Abstract
The kidney disease: Improving Global Outcomes (KDIGO) guideline recommends assessing kidney function using glomerular filtration rate (GFR) either through direct measurement or through estimation (eGFR) and describes a standardised classification of reduced kidney function. KDIGO guidelines have been adopted by most internal medicine specialities for the assessment and classification of kidney function, but not by cancer medicine. The development of the International Consensus Guideline on Anticancer Drug Dosing in Kidney Dysfunction (ADDIKD) aims to overcome the perceived challenges with KDIGO recommendations by describing their utility in patients with cancer. Two virtual, consensus building workshops were held consecutively, involving international, multidisciplinary participants (Part 1 of ADDIKD development). During these workshops, three consensus recommendations were agreed upon based on KDIGO's principles; to standardise kidney function assessment, classify kidney function, and determine a uniform approach to dose anticancer drugs in patients with reduced kidney function. Cancer clinicians attending the workshops identified issues regarding the adoption of KDIGO's recommendations. These issues were addressed by nephrologists, clinical pharmacologists, and other clinicians with extensive experience in the contemporary assessment of kidney function. The key concern for cancer specialists was a hesitancy to move away from the familiar and long-standing practice of using the Cockcroft-Gault equation to estimate creatinine clearance. The consensus building within the two multidisciplinary workshops allowed a thorough assessment of the evidence and clarified how directly measured GFR and eGFR, rather than creatinine clearance, could be optimally utilised in cancer care. The development of Part 1 of the ADDIKD guideline represents a standardised, contemporary approach to the assessment, classification, and utility of kidney function in the setting of cancer care and it harmonises with the approach used in other areas of medicine internationally. Funding Development of the ADDIKD guideline is funded by the Cancer Institute NSW as part of the NSW Government and received no funding from external commercial sources.
Collapse
Affiliation(s)
- Geeta Sandhu
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- eviQ, Cancer Institute NSW, St Leonards, NSW, Australia
- Pharmacy Department, St Vincent’s Hospital, Sydney, NSW, Australia
| | | | | | - Niamh O’Neill
- eviQ, Cancer Institute NSW, St Leonards, NSW, Australia
| | - Corrine Bagnis
- Nephrology Department, APHP Sorbonne University, Paris, France
| | - Pinkie Chambers
- University College London School of Pharmacy and University College London Hospital-University College London Centre for Medicines Optimisation Research and Education, London, United Kingdom
| | - Jennifer H. Martin
- Centre for Drug Repurposing, University of Newcastle, Newcastle, NSW, Australia
| | - Alex Flynn
- Centre for Drug Repurposing, University of Newcastle, Newcastle, NSW, Australia
| | - Karim Ibrahim
- Faculty of Medicine and Health, The University of New South Wales, Sydney, NSW, Australia
| | - Meg J. Jardine
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
- Concord Repatriation General Hospital, Concord, NSW, Australia
| | - David W. Johnson
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
| | - Graham R.D. Jones
- Faculty of Medicine and Health, The University of New South Wales, Sydney, NSW, Australia
- Department of Chemical Pathology, SydPath, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Christos S. Karapetis
- Flinders Centre for Innovation in Cancer, Flinders Medical Centre/Flinders University, Bedford Park, SA, Australia
| | - Aisling Kelly
- eviQ, Cancer Institute NSW, St Leonards, NSW, Australia
| | - Ganessan Kichenadasse
- Flinders Centre for Innovation in Cancer, Flinders Medical Centre/Flinders University, Bedford Park, SA, Australia
| | - David S. Kliman
- Department of Haematology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Winston Liauw
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
- School of Clinical Medicine, The University of New South Wales, Sydney, NSW, Australia
| | - Catherine Lucas
- Centre for Drug Repurposing, University of Newcastle, Newcastle, NSW, Australia
| | - Andrew J. Mallett
- Department of Renal Medicine, Townsville University Hospital, Townsville, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Townsville, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Medicine, Warsaw Medical University, Warsaw, Poland
| | - Michael Michael
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Carol A. Pollock
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Kolling Institute Medical Research, Sydney, NSW, Australia
| | - Darren M. Roberts
- Edith Collins Centre, Drug Health Services, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Mitchell H. Rosner
- Department of Medicine, University of Virginia Health, Charlottesville, VA, USA
| | - David J.M. Routledge
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Clinical Haematology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Carla Scuderi
- Pharmacy Department, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | | | - Jake Shortt
- Monash Haematology, Monash Health, Clayton, VIC, Australia
- Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Jim Siderov
- Pharmacy Department, Austin Health, Melbourne, VIC, Australia
| | - Ben Sprangers
- Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium
- Department of Nephrology, Ziekenhuis Oost Limburg, Genk, Belgium
| | - Brian N. Stein
- ICON Cancer Centre, Adelaide, SA, Australia
- The University of Adelaide, Adelaide, SA, Australia
| | - David J. Tunnicliffe
- Sydney School of Public Health, The University of Sydney, Sydney, NSW, Australia
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Kate Webber
- Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Robyn L. Ward
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- eviQ, Cancer Institute NSW, St Leonards, NSW, Australia
| | - ADDIKD Working Group
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- eviQ, Cancer Institute NSW, St Leonards, NSW, Australia
- Pharmacy Department, St Vincent’s Hospital, Sydney, NSW, Australia
- Nephrology Department, APHP Sorbonne University, Paris, France
- University College London School of Pharmacy and University College London Hospital-University College London Centre for Medicines Optimisation Research and Education, London, United Kingdom
- Centre for Drug Repurposing, University of Newcastle, Newcastle, NSW, Australia
- Faculty of Medicine and Health, The University of New South Wales, Sydney, NSW, Australia
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
- Concord Repatriation General Hospital, Concord, NSW, Australia
- Australasian Kidney Trials Network, The University of Queensland, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
- Department of Chemical Pathology, SydPath, St Vincent’s Hospital, Sydney, NSW, Australia
- Flinders Centre for Innovation in Cancer, Flinders Medical Centre/Flinders University, Bedford Park, SA, Australia
- Department of Haematology, Royal North Shore Hospital, Sydney, NSW, Australia
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
- School of Clinical Medicine, The University of New South Wales, Sydney, NSW, Australia
- Department of Renal Medicine, Townsville University Hospital, Townsville, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Townsville, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Department of Nephrology, Dialysis and Internal Medicine, Warsaw Medical University, Warsaw, Poland
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Kolling Institute Medical Research, Sydney, NSW, Australia
- Edith Collins Centre, Drug Health Services, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Department of Medicine, University of Virginia Health, Charlottesville, VA, USA
- Clinical Haematology, Royal Melbourne Hospital, Parkville, VIC, Australia
- Pharmacy Department, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
- Monash Haematology, Monash Health, Clayton, VIC, Australia
- Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Pharmacy Department, Austin Health, Melbourne, VIC, Australia
- Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium
- Department of Nephrology, Ziekenhuis Oost Limburg, Genk, Belgium
- ICON Cancer Centre, Adelaide, SA, Australia
- The University of Adelaide, Adelaide, SA, Australia
- Sydney School of Public Health, The University of Sydney, Sydney, NSW, Australia
- Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, NSW, Australia
- Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
2
|
Huang W, Bowman C, Yin M, Han LW, Wen YW, Ahn SK, Chen Y. A review of physiologically based pharmacokinetic modeling of renal drug disposition. Drug Metab Dispos 2025; 53:100042. [PMID: 40048993 DOI: 10.1016/j.dmd.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/14/2025] [Indexed: 03/30/2025] Open
Abstract
The human kidney is a critical organ for the elimination of numerous drugs and metabolites. The mechanisms of renal drug handling are manifold including unbound filtration, transporter-mediated active secretion, bidirectional passive diffusion, and occasionally active reabsorption and renal metabolism. These mechanisms collectively dictate the fate of drugs at various spatiotemporal points as drug molecules travel through the renal vasculature, tubules, and cells, posing a significant challenge in accurately describing and predicting renal drug disposition. Toward this end, a physiologically based kidney model serves as a promising tool to combine the anatomical and physiological features of the kidney (eg, tubular flow rate, pH, and transporter expression) with the unique properties of drugs (eg, protein binding, lipophilicity, ionization, and transporter substrate) to capture the dynamic system-drug interactions. Despite the exciting progress over the past several decades, physiologically based pharmacokinetic modeling has overall been predominantly used to predict intestinal absorption and hepatic drug-drug interaction. In comparison, pharmacokinetic modeling of renal drug handling has been underappreciated. In this review, we first provide an overview of kidney function and physiology, renal clearance mechanisms, and the evolutionary history of the physiologically based mechanistic kidney model. We then summarize the recent efforts spent in different areas of kidney model application, particularly: (1) renal transporter-mediated drug-drug interaction, (2) disease effect from both renal and hepatic impairment, and (3) model applications across the lifespan (eg, pediatrics and geriatrics). Finally, we identify remaining knowledge gaps, future directions, and potential model utilities. SIGNIFICANCE STATEMENT: This review summarizes pharmacokinetic model case studies that are related to renal drug disposition, illustrating the current framework of modeling renal drug handling, highlighting knowledge gaps in predicting renal transporter-mediated drug-drug interactions, and modeling the effects of disease and age on renal drug handling. A discussion on robust model validation and areas for future directions is also provided.
Collapse
Affiliation(s)
- Weize Huang
- Genentech Inc., South San Francisco, California.
| | | | - Mengyue Yin
- University of Washington, Seattle, Washington
| | | | | | | | - Yuan Chen
- Genentech Inc., South San Francisco, California
| |
Collapse
|
3
|
Thakur A, Singh DK, Hart KD, Kis E, Gáborik Z, Denton TT, Clarke JD, Paine MF, Prasad B. From discovery to translation: Endogenous substrates of OAT1 and OAT3 as clinical biomarkers for renal secretory function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636675. [PMID: 39975069 PMCID: PMC11838602 DOI: 10.1101/2025.02.05.636675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The recent ICH M12 guidance on Drug Interaction Studies encourages the use of alternate approaches for predicting drug-drug interaction (DDI) potential of new chemical entities. One approach involves biomarkers, which are endogenous substrates of drug metabolizing enzymes and transporters (DMET) and can be used to assess the inhibitory potential of new chemical entities during Phase 1 clinical studies. Thus, biomarkers could potentially eliminate the need for dedicated DDI studies with exogenous probe substrates. Metabolomics, in conjunction with in vitro and/or in vivo preclinical models or clinical studies, can be used for biomarker discovery. We developed and applied a novel metabolomics-based DMET biomarker discovery (MDBD) approach to identify and qualify biomarkers of renal organic anion transporter 1 (OAT1) and OAT3. Untargeted metabolomics of pooled plasma and urine samples from a pharmacokinetic DDI study using the OAT1/3 inhibitor, probenecid, yielded 153 features identified as putative OAT1/3 biomarkers. Subsequently, in vitro transporter uptake assays using processed urine samples confirmed 57 of these features as OAT1 and/or OAT3 substrates. Finally, 23 features were clinically validated as OAT1/3 biomarkers through a detailed pharmacokinetic analysis (0-24 h) of plasma and urine samples. These biomarkers, either alone or as part of a panel, can predict OAT1/3-mediated DDIs and interindividual variability in the renal secretory clearance of organic anions across different populations, thereby enabling translational utility in clinical settings. The novel MDBD approach can be extended to discover biomarkers of other transporters and enzymes. SUMMARY Using clinical and mechanistic in vitro approaches, 23 endogenous substrates of OAT1/3 were identified as potential clinical biomarkers of renal secretary elimination of organic anions.
Collapse
|
4
|
Zbib F, Deschamps A, Velly L, Blin O, Guilhaumou R, Gattacceca F. Physiologically Based Pharmacokinetic Model of Cefotaxime in Patients with Impaired Renal Function. Clin Pharmacokinet 2025; 64:257-273. [PMID: 39762592 DOI: 10.1007/s40262-024-01469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 02/01/2025]
Abstract
BACKGROUND Cefotaxime is a widely prescribed cephalosporin antibiotic used to treat various infections. It is mainly eliminated unchanged by the kidney through tubular secretion and glomerular filtration. Therefore, a reduction of kidney function may increase exposure to the drug and induce toxic side effects. OBJECTIVES The objectives of this study were to develop a physiologically based pharmacokinetic (PBPK) model of cefotaxime in healthy European adults, to mechanistically describe the impact of chronic kidney disease (CKD) on cefotaxime pharmacokinetics, and to assess the applicability of the model to patients requiring intensive care. METHODS Using PK-Sim® software, we developed a PBPK model for cefotaxime, including basolateral and apical renal transporters and renal esterases, in healthy subjects and then extrapolated to patients with CKD by incorporating pathophysiological changes and reductions in activity of drug-metabolizing enzymes and transporters into the model. We then evaluated the predictive performance of the model in patients requiring intensive care using clinical routine data. RESULTS Model predictions were considered adequate in healthy subjects and patients with CKD, with predicted-to-observed area under the curve ratios within the two-fold acceptance criterion. Mean prediction error and mean absolute prediction error did not exceed ± 30 and 30%, respectively, except in patients with stage 4 CKD, where they were 70.5 and 75.6%, respectively. The model showed good predictive performance when applied to patients requiring intensive care, but its clinical applicability in this population needs to be further evaluated. CONCLUSION We successfully developed whole-body PBPK models to predict cefotaxime pharmacokinetics in different populations. These models represent an additional step toward improving personalized cefotaxime dosing regimens in vulnerable populations.
Collapse
Affiliation(s)
- Fatima Zbib
- Aix Marseille University, APHM, INSERM, Service de Pharmacologie Clinique et Pharmacosurveillance, INS Institute Neuroscience Syst, Marseille, France
| | - Anthéa Deschamps
- Aix Marseille University, APHM, INSERM, Service de Pharmacologie Clinique et Pharmacosurveillance, INS Institute Neuroscience Syst, Marseille, France
- Inria-Inserm COMPO Team, Centre Inria Sophia Antipolis-Méditerranée, CRCM, Inserm U1068-CNRS UMR7258-Aix-Marseille University UM105, Marseille, France
| | - Lionel Velly
- Aix Marseille University, APHM, Department of Anaesthesiology and Critical Care Medicine, University Hospital Timone, Marseille, France
- Aix Marseille University, CNRS, INT, Institute Neuroscience Timone, UMR7289, Marseille, France
| | - Olivier Blin
- Aix Marseille University, APHM, INSERM, Service de Pharmacologie Clinique et Pharmacosurveillance, INS Institute Neuroscience Syst, Marseille, France
| | - Romain Guilhaumou
- Aix Marseille University, APHM, INSERM, Service de Pharmacologie Clinique et Pharmacosurveillance, INS Institute Neuroscience Syst, Marseille, France.
| | - Florence Gattacceca
- Inria-Inserm COMPO Team, Centre Inria Sophia Antipolis-Méditerranée, CRCM, Inserm U1068-CNRS UMR7258-Aix-Marseille University UM105, Marseille, France
| |
Collapse
|
5
|
Wang X, Mu J, Ma K, Ma Y. Challenges of Serum Creatinine Level in GFR Assessment and Drug Dosing Decisions in Kidney Injury. Adv Pharm Bull 2024; 14:745-758. [PMID: 40190670 PMCID: PMC11970497 DOI: 10.34172/apb.42345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 04/09/2025] Open
Abstract
Serum creatinine (SCr) is widely regarded as a standard biomarker for assessing glomerular filtration rate (GFR) and is commonly used to guide dose adjustments for renally eliminated drugs. However, the application of SCr as a marker for evaluating GFR and drug dosing in kidney injury has significant limitations that are often overlooked in clinical practice. This oversight can result in subtherapeutic drug concentrations or adverse drug reactions due to inappropriate dosing adjustments based on SCr levels alone. This review aimed to highlight the factors affecting serum creatinine (SCr) and the challenges associated with using SCr as a biomarker for assessing GFR and adjusting drug doses with regard to its limitations and variability. The findings of this review underscore the complexity of SCr regulation, which is affected by its synthesis, metabolism, and excretion processes (glomerular filtration, tubular secretion, tubular reabsorption and extra-renal elimination), and disease states (such as trauma-induced hyperfiltration and HIV) and the use of medications (drug-creatinine interactions) lead to altered renal excretion of creatinine, either increasing or decreasing its levels. Additionally, the renal excretion pathways for drugs and creatinine are not entirely the same, making it difficult to use creatinine to evaluate drug renal excretion. In conclusion, SCr is an imperfect index of GFR and adjusting drug dosing, and the development of multi-biomarker panels, incorporating biomarkers from different excretory pathways-particularly those involving tubular transport-holds promise for improving the evaluation of renal excretory function and ensuring safer and more effective drug dosing.
Collapse
Affiliation(s)
- Xinyi Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Jing Mu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Kexin Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yanrong Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
- Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
6
|
Shahidehpour A, Rashid M, Askari MR, Ahmadasas M, Abdel-Latif M, Fritschi C, Quinn L, Reutrakul S, Bronas UG, Cinar A. Modeling Metformin and Dapagliflozin Pharmacokinetics in Chronic Kidney Disease. AAPS J 2024; 26:94. [PMID: 39160349 DOI: 10.1208/s12248-024-00962-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/27/2024] [Indexed: 08/21/2024] Open
Abstract
Chronic kidney disease (CKD) is a complication of diabetes that affects circulating drug concentrations and elimination of drugs from the body. Multiple drugs may be prescribed for treatment of diabetes and co-morbidities, and CKD complicates the pharmacotherapy selection and dosing regimen. Characterizing variations in renal drug clearance using models requires large clinical datasets that are costly and time-consuming to collect. We propose a flexible approach to incorporate impaired renal clearance in pharmacokinetic (PK) models using descriptive statistics and secondary data with mechanistic models and PK first principles. Probability density functions were generated for various drug clearance mechanisms based on the degree of renal impairment and used to estimate the total clearance starting from glomerular filtration for metformin (MET) and dapagliflozin (DAPA). These estimates were integrated with PK models of MET and DAPA for simulations. MET renal clearance decreased proportionally with a reduction in estimated glomerular filtration rate (eGFR) and estimated net tubular transport rates. DAPA total clearance varied little with renal impairment and decreased proportionally to reported non-renal clearance rates. Net tubular transport rates were negative to partially account for low renal clearance compared with eGFR. The estimated clearance values and trends were consistent with MET and DAPA PK characteristics in the literature. Dose adjustment based on reduced clearance levels estimated correspondingly lower doses for MET and DAPA while maintaining desired dose exposure. Estimation of drug clearance rates using descriptive statistics and secondary data with mechanistic models and PK first principles improves modeling of CKD in diabetes and can guide treatment selection.
Collapse
Affiliation(s)
- Andrew Shahidehpour
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Mudassir Rashid
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Mohammad Reza Askari
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Mohammad Ahmadasas
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Mahmoud Abdel-Latif
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois, USA
| | - Cynthia Fritschi
- Department of Biobehavioral Nursing Science, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lauretta Quinn
- Department of Biobehavioral Nursing Science, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Sirimon Reutrakul
- College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ulf G Bronas
- School of Nursing and Rehabilitation Medicine, Columbia University in New York City, New York, New York, USA
| | - Ali Cinar
- Department of Chemical and Biological Engineering, Illinois Institute of Technology, Chicago, Illinois, USA.
| |
Collapse
|
7
|
Ma Y, Wang X, Gou X, Wu X. Identification and characterization of an endogenous biomarker of the renal vectorial transport (OCT2-MATE1). Biopharm Drug Dispos 2024; 45:43-57. [PMID: 38305087 DOI: 10.1002/bdd.2382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/29/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024]
Abstract
The renal tubular organic cation transporter 2 (OCT2) and multidrug and toxin extrusion protein 1 (MATE1) mediate the vectorial elimination of many drugs and toxins from the kidney, and endogenous biomarkers for vectorial transport (OCT2-MATE1) would allow more accurate drug dosing and help to characterize drug-drug interactions and toxicity. Human serum uptake in OCT2-overexpressing cells and metabolomics analysis were carried out. Potential biomarkers were verified in vitro and in vivo. The specificity of biomarkers was validated in renal transporter overexpressing cells and the sensitivity was investigated by Km . The results showed that the uptake of thiamine, histamine, and 5-hydroxytryptamine was significantly increased in OCT2-overexpressing cells. In vitro assays confirmed that thiamine, histamine, and 5-hydroxytryptamine were substrates of both OCT2 and MATE1. In vivo measurements indicated that the serum thiamine level was increased significantly in the presence of the rOCT2 inhibitor cimetidine, and the level in renal tissue was increased significantly by the rMATE1 inhibitor pyrimethamine. There were no significant changes in the uptake or efflux of thiamine in cell lines overexpressed OAT1, OAT2, OAT3, MRP4, organic anion transporting polypeptide 4C1, P-gp, peptide transporter 2, urate transporter 1, and OAT4. The Km for thiamine with OCT2 and MATE1 were 71.2 and 10.8 μM, respectively. In addition, the cumulative excretion of thiamine at 2 and 4 h was strongly correlated with metformin excretion (R2 > 0.6). Thus, thiamine is preferentially secreted by the OCT2 and MATE1 in renal tubules and can provide a reference value for evaluating the function of the renal tubular OCT2-MATE1.
Collapse
Affiliation(s)
- Yanrong Ma
- The First Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xinyi Wang
- The First Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xueyan Gou
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Xinan Wu
- The First Clinical Medical School, Lanzhou University, Lanzhou, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
8
|
Teles F, Peçanha de Miranda Coelho JA, Albino RM, Verçosa Pacheco FC, Rodrigues de Oliveira E, Silveira MAD, Diógenes M. Feitosa A, Bezerra R. Effectiveness of thiazide and thiazide-like diuretics in advanced chronic kidney disease: a systematic review and meta-analysis. Ren Fail 2023; 45:2163903. [PMID: 36637019 PMCID: PMC9848247 DOI: 10.1080/0886022x.2022.2163903] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Thiazide diuretics are first-line drugs for the treatment of hypertension, but hypertension treatment guidelines have systematically discouraged their use in patients with advanced chronic kidney disease (CKD). For the first time, a systematic review and random-effects meta-analysis were performed to assess the effectiveness of thiazides and thiazide-like diuretics to treat hypertension in patients with stages 3b, 4, and 5 CKD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS A systematic review and random-effects meta-analysis that included a literature search using the following databases were performed: MEDLINE through PubMed, Cochrane Database of Systematic Reviews (CDSR) and Cochrane Central Register of Controlled Trials (CENTRAL) through the Cochrane Library, Embase, and ISI - Web of Science (all databases). Prospective studies that evaluated the effectiveness of thiazide and thiazide-like diuretics in individuals with a GFR < 45 mL/min/1.73 m2 were included. RESULTS Five clinical trials, totaling 214 participants, were included, and the mean GFR ranged from 13.0 ± 5.9 mL/min/1.73 m2 to 26.8 ± 8.8 mL/min/1.73 m2. There was evidence of a reduction in mean blood pressure and in GFR, as well as in fractional sodium excretion and fractional chloride excretion. CONCLUSION Thiazide and thiazide-like diuretics seem to maintain their effectiveness in lowering blood pressure in patients with advanced chronic kidney disease. These findings should spur new prospective randomized trials and spark discussions, particularly about upcoming hypertension guidelines.
Collapse
Affiliation(s)
- Flávio Teles
- School of Medicine, Federal University of Alagoas (UFAL), Maceió, Brazil,School of Medicine, State University of Health Sciences of Alagoas (UNCISAL), Maceió, Brazil
| | | | - Rosivânia Maria Albino
- School of Medicine, State University of Health Sciences of Alagoas (UNCISAL), Maceió, Brazil
| | | | | | | | | | - Rodrigo Bezerra
- Pernambuco Hypertension Service (SHIP), PROCAPE - University of Pernambuco (UPE), Recife, Brazil,Keizo Asami Laboratory of Immunopathology, Federal University of Pernambuco, Recife, Brazil,CONTACT Rodrigo Bezerra PROCAPE-University of Pernambuco, Brazil. Rua dos Palmares, S/N – Santo Amaro, Recife – PE - CEP: 74970-240, Brazil
| |
Collapse
|
9
|
Tan SPF, Willemin ME, Snoeys J, Shen H, Rostami-Hodjegan A, Scotcher D, Galetin A. Development of 4-Pyridoxic Acid PBPK Model to Support Biomarker-Informed Evaluation of OAT1/3 Inhibition and Effect of Chronic Kidney Disease. Clin Pharmacol Ther 2023; 114:1243-1253. [PMID: 37620246 DOI: 10.1002/cpt.3029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
Monitoring endogenous biomarkers is increasingly used to evaluate transporter-mediated drug-drug interactions (DDIs) in early drug development and may be applied to elucidate changes in transporter activity in disease. 4-pyridoxic acid (PDA) has been identified as the most sensitive plasma endogenous biomarker of renal organic anion transporters (OAT1/3). Increase in PDA baseline concentrations was observed after administration of probenecid, a strong clinical inhibitor of OAT1/3 and also in patients with chronic kidney disease (CKD). The aim of this study was to develop and verify a physiologically-based pharmacokinetic (PBPK) model of PDA, to predict the magnitude of probenecid DDI and predict the CKD-related changes in PDA baseline. The PBPK model for PDA was first developed in healthy population, building on from previous population pharmacokinetic modeling, and incorporating a mechanistic kidney model to consider OAT1/3-mediated renal secretion. Probenecid PBPK model was adapted from the Simcyp database and re-verified to capture its dose-dependent pharmacokinetics (n = 9 studies). The PBPK model successfully predicted the PDA plasma concentrations, area under the curve, and renal clearance in healthy subjects at baseline and after single/multiple probenecid doses. Prospective simulations in severe CKD predicted successfully the increase in PDA plasma concentration relative to healthy (within 2-fold of observed data) after accounting for 60% increase in fraction unbound in plasma and additional 50% decline in OAT1/3 activity beyond the decrease in glomerular filtration rate. The verified PDA PBPK model supports future robust evaluation of OAT1/3 DDI in drug development and increases our confidence in predicting exposure and renal secretion in patients with CKD.
Collapse
Affiliation(s)
- Shawn Pei Feng Tan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
| | - Marie-Emilie Willemin
- Janssen Pharmaceutical Companies of Johnson & Johnson, Janssen Research & Development, Beerse, Belgium
| | - Jan Snoeys
- Janssen Pharmaceutical Companies of Johnson & Johnson, Janssen Research & Development, Beerse, Belgium
| | - Hong Shen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
- Certara UK Limited (Simcyp Division), Sheffield, UK
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
10
|
Chang SY, Huang W, Chapron A, Quiñones AJL, Wang J, Isoherranen N, Shen DD, Kelly EJ, Himmelfarb J, Yeung CK. Incorporating Uremic Solute-mediated Inhibition of OAT1/3 Improves PBPK Prediction of Tenofovir Renal and Systemic Disposition in Patients with Severe Kidney Disease. Pharm Res 2023; 40:2597-2606. [PMID: 37704895 PMCID: PMC11901350 DOI: 10.1007/s11095-023-03594-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Dose modification of renally secreted drugs in patients with chronic kidney disease (CKD) has relied on serum creatinine concentration as a biomarker to estimate glomerular filtration (GFR) under the assumption that filtration and secretion decline in parallel. A discrepancy between actual renal clearance and predicted renal clearance based on GFR alone is observed in severe CKD patients with tenofovir, a compound secreted by renal OAT1/3. Uremic solutes that inhibit OAT1/3 may play a role in this divergence. METHODS To examine the impact of transporter inhibition by uremic solutes on tenofovir renal clearance, we determined the inhibitory potential of uremic solutes hippuric acid, indoxyl sulfate, and p-cresol sulfate. The inhibition parameters (IC50) were incorporated into a previously validated mechanistic kidney model; simulated renal clearance and plasma PK profile were compared to data from clinical studies. RESULTS Without the incorporation of uremic solute inhibition, the PBPK model failed to capture the observed data with an absolute average fold error (AAFE) > 2. However, when the inhibition of renal uptake transporters and uptake transporters in the slow distribution tissues were included, the AAFE value was within the pre-defined twofold model acceptance criterion, demonstrating successful model extrapolation to CKD patients. CONCLUSION A PBPK model that incorporates inhibition by uremic solutes has potential to better predict renal clearance and systemic disposition of secreted drugs in patients with CKD. Ongoing research is warranted to determine if the model can be expanded to include other OAT1/3 substrate drugs and to evaluate how these findings can be translated to clinical guidance for drug selection and dose optimization in patients with CKD.
Collapse
Affiliation(s)
- Shih-Yu Chang
- Department of Pharmacy, School of Pharmacy, University of Washington, 1959 NE Pacific St. H375, Box 357630, Seattle, WA, 98195, USA
- Janssen Research and Development, Raritan, NJ, USA
| | - Weize Huang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
- Genentech Inc, South San Francisco, CA, USA
| | - Alenka Chapron
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Antonio J López Quiñones
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
- Revolution Medicines, San Francisco, CA, USA
| | - Joanne Wang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, 98195, USA
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, WA, 98195, USA
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, WA, 98195, USA
| | - Catherine K Yeung
- Department of Pharmacy, School of Pharmacy, University of Washington, 1959 NE Pacific St. H375, Box 357630, Seattle, WA, 98195, USA.
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
11
|
Arakawa H, Kato Y. Emerging Roles of Uremic Toxins and Inflammatory Cytokines in the Alteration of Hepatic Drug Disposition in Patients with Kidney Dysfunction. Drug Metab Dispos 2023; 51:1127-1135. [PMID: 36854605 DOI: 10.1124/dmd.122.000967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/12/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Patients with kidney dysfunction exhibit distinct pharmacokinetic profiles compared to those with normal kidney function. Hence, it is desirable to monitor the drug efficacy and toxicity caused by fluctuations in plasma drug concentrations associated with kidney dysfunction. Recently, pharmacokinetic information of drugs excreted mainly through the urine of patients with kidney dysfunction has been reported via drug-labeling information. Pharmacokinetic changes in drugs mainly eliminated by the liver cannot be overlooked as drug metabolism and/or transport activity in the liver may also be altered in patients with kidney dysfunction; however, the underlying mechanisms remain unclear. To plan an appropriate dosage regimen, it is necessary to clarify the underlying processes of functional changes in pharmacokinetic proteins. In recent years, uremic toxins have been shown to reduce the activity and/or expression of renal and hepatic transporters. This inhibitory effect has been reported to be time-dependent. In addition, inflammatory cytokines, such as interleukin-6, released from immune cells activated by uremic toxins and/or kidney injury can reduce the expression levels of drug-metabolizing enzymes and transporters in human hepatocytes. In this mini-review, we have summarized the renal and hepatic pharmacokinetic changes as well as the potential underlying mechanisms in kidney dysfunction, such as the chronic kidney disease and acute kidney injury. SIGNIFICANCE STATEMENT: Patients with kidney dysfunction exhibit distinct pharmacokinetic profiles compared to those with normal kidney function. Increased plasma concentrations of uremic toxins and inflammatory cytokines during kidney disease may potentially affect the activities and/or expression levels of drug-metabolizing enzymes and transporters in the liver and kidneys.
Collapse
Affiliation(s)
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
12
|
Behal ML, Flannery AH, Barreto EF. Medication Management in the Critically Ill Patient with Acute Kidney Injury. Clin J Am Soc Nephrol 2023; 18:1080-1088. [PMID: 36723347 PMCID: PMC10564345 DOI: 10.2215/cjn.0000000000000101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 02/02/2023]
Abstract
ABSTRACT AKI occurs frequently in critically ill patients. Patients with AKI, including those who require KRT, experience multiple pharmacokinetic and pharmacodynamic perturbations that dynamically influence medication effectiveness and safety. Patients with AKI may experience both subtherapeutic drug concentrations, which lead to ineffective therapy, and supratherapeutic drug concentrations, which increase the risk for toxicity. In critically ill patients with AKI not requiring KRT, conventional GFR estimation equations, especially those based on serum creatinine, have several limitations that can limit the accuracy when used for medication dosing. Alternative methods to estimate kidney function may be informative, including use of measured urinary creatinine clearance, kinetic eGFR, and equations that integrate novel kidney biomarkers. For critically ill patients with AKI requiring KRT, physicochemical properties of the drug, the KRT prescription and circuit configuration, and patient-specific factors each contribute to medication clearance. Evidence-based guidance for medication dosing during AKI requiring KRT is often limited. A working knowledge of the basic tenets of drug elimination during KRT can provide a framework for how to approach decision making when the literature is lacking. Iterative re-evaluation of a patient's progress toward therapeutic goals with a medication must occur over the arc of critical illness, including and especially in the setting of dynamic kidney function.
Collapse
Affiliation(s)
- Michael L. Behal
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky
| | - Alexander H. Flannery
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky
| | | |
Collapse
|
13
|
Dong J, Liu J, Liu Y, Yao J, Lu Y, Jiao Z, Li W. Physiologically based pharmacokinetic modeling to predict OAT3-mediated drug-drug interactions of meropenem in varying stages of chronic kidney disease. Eur J Pharm Sci 2023; 183:106395. [PMID: 36716979 DOI: 10.1016/j.ejps.2023.106395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/31/2022] [Accepted: 01/26/2023] [Indexed: 01/30/2023]
Affiliation(s)
- Jing Dong
- Department of Pharmacy, Shanghai Pudong New Area Gongli Hospital, The Second Military Medical University, 219 Miaopu Road, Shanghai 200135, PR China
| | - Jinyao Liu
- Ningxia Medical University, 1160 Shengli Street, Ningxia, Yinchuan 750004, PR China
| | - Yanhui Liu
- Department of Pharmacy, Shanghai Pudong New Area Gongli Hospital, The Second Military Medical University, 219 Miaopu Road, Shanghai 200135, PR China
| | - Jiachen Yao
- Department of Pharmacy, Shanghai Pudong New Area Gongli Hospital, The Second Military Medical University, 219 Miaopu Road, Shanghai 200135, PR China
| | - Yan Lu
- Department of Pharmacy, Shanghai Pudong New Area Gongli Hospital, The Second Military Medical University, 219 Miaopu Road, Shanghai 200135, PR China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, 241 West Huaihai Road, Shanghai 200030, PR China.
| | - Wenyan Li
- Department of Pharmacy, Shanghai Pudong New Area Gongli Hospital, The Second Military Medical University, 219 Miaopu Road, Shanghai 200135, PR China.
| |
Collapse
|
14
|
Sakai M, Hirai T, Shitara S, Iwamoto T, Shiga T. Comparison of creatinine-based equations for estimating renal function for digoxin dose adjustment in patients with atrial fibrillation and heart failure. Pharmacol Res Perspect 2023; 11:e01050. [PMID: 36628508 PMCID: PMC9832284 DOI: 10.1002/prp2.1050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/08/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023] Open
Abstract
The aim of this study was to determine an appropriate equation for estimating renal function to dose regulate the serum digoxin trough concentration to a target of <0.9 ng/ml in patients with atrial fibrillation (AF) and heart failure (HF). All patients received 0.125 mg oral digoxin daily. The estimated glomerular filtration rate by the Modification of Diet in Renal Disease (eGFRMDRD ) equation deindexed based on body surface area had the highest correlation with digoxin trough concentrations (r = -0.450) compared to the Cockcroft-Gault equation (r = -0.415) or deindexed eGFR based on the Chronic Kidney Disease Epidemiology Collaboration (eGFRCKD-EPI ) equation (r = -0.416). The median digoxin trough concentrations were 0.60, 0.77, 0.97 and 1.30 ng/ml in patients with a deindexed eGFRMDRD ≥ 60, 45-59, 30-44 and < 30 ml/min, respectively. The deindexed eGFRMDRD is an appropriate equation for digoxin dose adjustment in patients with AF and HF.
Collapse
Affiliation(s)
| | - Toshinori Hirai
- Department of Clinical Pharmacology and TherapeuticsThe Jikei University School of MedicineTokyoJapan
- Department of PharmacyMie University Hospital, Faculty of Medicine, Mie UniversityTsuMieJapan
| | - Satoshi Shitara
- Department of Clinical Pharmacology and TherapeuticsThe Jikei University School of MedicineTokyoJapan
| | - Takuya Iwamoto
- Department of PharmacyMie University Hospital, Faculty of Medicine, Mie UniversityTsuMieJapan
| | - Tsuyoshi Shiga
- Department of Clinical Pharmacology and TherapeuticsThe Jikei University School of MedicineTokyoJapan
- Department of CardiologyTokyo Women's Medical UniversityTokyoJapan
| |
Collapse
|
15
|
Dubinsky S, Malik P, Hajducek DM, Edginton A. Determining the Effects of Chronic Kidney Disease on Organic Anion Transporter1/3 Activity Through Physiologically Based Pharmacokinetic Modeling. Clin Pharmacokinet 2022; 61:997-1012. [PMID: 35508593 DOI: 10.1007/s40262-022-01121-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND OBJECTIVE The renal excretion of drugs via organic anion transporters 1 and 3 (OAT1/3) is significantly decreased in patients with renal impairment. This study uses physiologically based pharmacokinetic models to quantify the reduction in OAT1/3-mediated secretion of drugs throughout varying stages of chronic kidney disease. METHODS Physiologically based pharmacokinetic models were constructed for four OAT1/3 substrates in healthy individuals: acyclovir, meropenem, furosemide, and ciprofloxacin. Observed data from drug-drug interaction studies with probenecid, a potent OAT1/3 inhibitor, were used to parameterize the contribution of OAT1/3 to the renal elimination of each drug. The models were then translated to patients with chronic kidney disease by accounting for changes in glomerular filtration rate, kidney volume, renal blood flow, plasma protein binding, and hematocrit. Additionally, a relationship was derived between the estimated glomerular filtration rate and the reduction in OAT1/3-mediated secretion of drugs based on the renal extraction ratios of ƿ-aminohippuric acid in patients with varying degrees of renal impairment. The relationship was evaluated in silico by evaluating the predictive performance of each final model in describing the pharmacokinetics (PK) of drugs across stages of chronic kidney disease. RESULTS OAT1/3-mediated renal excretion of drugs was found to be decreased by 27-49%, 50-68%, and 70-96% in stage 3, stage 4, and stage 5 of chronic kidney disease, respectively. In support of the parameterization, physiologically based pharmacokinetic models of four OAT1/3 substrates were able to adequately characterize the PK in patients with different degrees of renal impairment. Total exposure after intravenous administration was predicted within a 1.5-fold error and 85% of the observed data points fell within a 1.5-fold prediction error. The models modestly under-predicted plasma concentrations in patients with end-stage renal disease undergoing intermittent hemodialysis. However, results should be interpreted with caution because of the limited number of molecules analyzed and the sparse sampling in observed chronic kidney disease pharmacokinetic studies. CONCLUSIONS A quantitative understanding of the reduction in OAT1/3-mediated excretion of drugs in differing stages of renal impairment will contribute to better predictive accuracy for physiologically based pharmacokinetic models in drug development, assisting with clinical trial planning and potentially sparing this population from unnecessary toxic exposures.
Collapse
Affiliation(s)
- Samuel Dubinsky
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Paul Malik
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | | | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
16
|
Tan SPF, Scotcher D, Rostami-Hodjegan A, Galetin A. Effect of Chronic Kidney Disease on the Renal Secretion via Organic Anion Transporters 1/3: Implications for Physiologically-Based Pharmacokinetic Modeling and Dose Adjustment. Clin Pharmacol Ther 2022; 112:643-652. [PMID: 35569107 PMCID: PMC9540491 DOI: 10.1002/cpt.2642] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/07/2022] [Indexed: 12/14/2022]
Abstract
There is growing evidence that active tubular secretory clearance (CLs) may not decline proportionally with the glomerular filtration rate (GFR) in chronic kidney disease (CKD), leading to the overestimation of renal clearance (CLr) when using solely GFR to approximate disease effect on renal elimination. The clinical pharmacokinetic data of 33 renally secreted OAT1/3 substrates were collated to investigate the impact of mild, moderate, and severe CKD on CLr, tubular secretion and protein binding (fu,p). The fu,p of the collated substrates ranged from 0.0026 to 1.0 in healthy populations; observed CKD‐related increase in the fu,p (up to 2.7‐fold) of 8 highly bound substrates (fu,p ≤ 0.2) was accounted for in the analysis. Use of prediction equation based on disease‐related changes in albumin resulted in underprediction of the CKD‐related increase in fu,p of highly bound substrates, highlighting the necessity to measure protein binding in severe CKD. The critical analysis of clinical data for 33 OAT1/3 probes established that decrease in OAT1/3 activity proportional to the changes in GFR was insufficient to recapitulate effects of severe CKD on unbound tubular secretion clearance. OAT1/3‐mediated CLs was estimated to decline by an additional 50% relative to the GFR decline in severe CKD, whereas change in active secretion in mild and moderate CKD was proportional to GFR. Consideration of this additional 50% decline in OAT1/3‐mediated CLs is recommended for physiologically‐based pharmacokinetic models and dose adjustment of OAT1/3 substrates in severe CKD, especially for substrates with high contribution of the active secretion to CLr.
Collapse
Affiliation(s)
- Shawn Pei Feng Tan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK.,Certara UK (Simcyp Division), Sheffield, UK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
17
|
Hall AM, Trepiccione F, Unwin RJ. Drug toxicity in the proximal tubule: new models, methods and mechanisms. Pediatr Nephrol 2022; 37:973-982. [PMID: 34050397 PMCID: PMC9023418 DOI: 10.1007/s00467-021-05121-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/23/2021] [Accepted: 05/05/2021] [Indexed: 10/28/2022]
Abstract
The proximal tubule (PT) reabsorbs most of the glomerular filtrate and plays an important role in the uptake, metabolism and excretion of xenobiotics. Some therapeutic drugs are harmful to the PT, and resulting nephrotoxicity is thought to be responsible for approximately 1 in 6 of cases of children hospitalized with acute kidney injury (AKI). Clinically, PT dysfunction leads to urinary wasting of important solutes normally reabsorbed by this nephron segment, leading to systemic complications such as bone demineralization and a clinical scenario known as the renal Fanconi syndrome (RFS). While PT defects can be diagnosed using a combination of blood and urine markers, including urinary excretion of low molecular weight proteins (LMWP), standardized definitions of what constitutes clinically significant toxicity are lacking, and identifying which patients will go on to develop progressive loss of kidney function remains a major challenge. In addition, much of our understanding of cellular mechanisms of drug toxicity is still limited, partly due to the constraints of available cell and animal models. However, advances in new and more sophisticated in vitro models of the PT, along with the application of high-content analytical methods that can provide readouts more relevant to the clinical manifestations of nephrotoxicity, are beginning to extend our knowledge. Such technical progress should help in discovering new biomarkers that can better detect nephrotoxicity earlier and predict its long-term consequences, and herald a new era of more personalized medicine.
Collapse
Affiliation(s)
- Andrew M. Hall
- grid.7400.30000 0004 1937 0650Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland ,grid.412004.30000 0004 0478 9977Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Francesco Trepiccione
- grid.9841.40000 0001 2200 8888Department of Translational Medical Science, University of Campania ‘Luigi Vanvitelli’, Naples, Italy ,grid.428067.f0000 0004 4674 1402Biogem Research Institute, Ariano Irpino, Italy
| | - Robert J. Unwin
- grid.83440.3b0000000121901201Department of Renal Medicine, University College London, London, UK
| |
Collapse
|
18
|
Scotcher D, Galetin A. PBPK Simulation-Based Evaluation of Ganciclovir Crystalluria Risk Factors: Effect of Renal Impairment, Old Age, and Low Fluid Intake. AAPS J 2021; 24:13. [PMID: 34907479 PMCID: PMC8816528 DOI: 10.1208/s12248-021-00654-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/02/2021] [Indexed: 11/30/2022] Open
Abstract
Dosing guidance is often lacking for chronic kidney disease (CKD) due to exclusion of such patients from pivotal clinical trials. Physiologically based pharmacokinetic (PBPK) modelling supports model-informed dosing when clinical data are lacking, but application of these approaches to patients with impaired renal function is not yet at full maturity. In the current study, a ganciclovir PBPK model was developed for patients with normal renal function and extended to CKD population. CKD-related changes in tubular secretion were explored in the mechanistic kidney model and implemented either as proportional or non-proportional decline relative to GFR. Crystalluria risk was evaluated in different clinical settings (old age, severe CKD and low fluid intake) by simulating ganciclovir medullary collecting duct (MCD) concentrations. The ganciclovir PBPK model captured observed changes in systemic pharmacokinetic endpoints in mild-to-severe CKD; these trends were evident irrespective of assumed pathophysiological mechanism of altered active tubular secretion in the model. Minimal difference in simulated ganciclovir MCD concentrations was noted between young adult and geriatric populations with normal renal function and urine flow (1 mL/min), with lower concentrations predicted for severe CKD patients. High crystalluria risk was identified at reduced urine flow (0.1 mL/min) as simulated ganciclovir MCD concentrations exceeded its solubility (2.6–6 mg/mL), irrespective of underlying renal function. The analysis highlighted the importance of appropriate distribution of virtual subjects’ systems data in CKD populations. The ganciclovir PBPK model illustrates the ability of this translational tool to explore individual and combined effects of age, urine flow, and renal impairment on local drug renal exposure.
Collapse
Affiliation(s)
- Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
19
|
Mair RD, Lee S, Plummer NS, Sirich TL, Meyer TW. Impaired Tubular Secretion of Organic Solutes in Advanced Chronic Kidney Disease. J Am Soc Nephrol 2021; 32:2877-2884. [PMID: 34408065 PMCID: PMC8806100 DOI: 10.1681/asn.2021030336] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/29/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The clearance of solutes removed by tubular secretion may be altered out of proportion to the GFR in CKD. Recent studies have described considerable variability in the secretory clearance of waste solutes relative to the GFR in patients with CKD. METHODS To test the hypothesis that secretory clearance relative to GFR is reduced in patients approaching dialysis, we used metabolomic analysis to identify solutes in simultaneous urine and plasma samples from 16 patients with CKD and an eGFR of 7±2 ml/min per 1.73 m2 and 16 control participants. Fractional clearances were calculated as the ratios of urine to plasma levels of each solute relative to those of creatinine and urea in patients with CKD and to those of creatinine in controls. RESULTS Metabolomic analysis identified 39 secreted solutes with fractional clearance >3.0 in control participants. Fractional clearance values in patients with CKD were reduced on average to 65%±27% of those in controls. These values were significantly lower for 18 of 39 individual solutes and significantly higher for only one. Assays of the secreted anions phenylacetyl glutamine, p-cresol sulfate, indoxyl sulfate, and hippurate confirmed variable impairment of secretory clearances in advanced CKD. Fractional clearances were markedly reduced for phenylacetylglutamine (4.2±0.6 for controls versus 2.3±0.6 for patients with CKD; P<0.001), p-cresol sulfate (8.6±2.6 for controls versus 4.1±1.5 for patients with CKD; P<0.001), and indoxyl sulfate (23.0±7.3 versus 7.5±2.8; P<0.001) but not for hippurate (10.2±3.8 versus 8.4±2.6; P=0.13). CONCLUSIONS Secretory clearances for many solutes are reduced more than the GFR in advanced CKD. Impaired secretion of these solutes might contribute to uremic symptoms as patients approach dialysis.
Collapse
Affiliation(s)
- Robert D. Mair
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, Veterans Affairs Palo Alto Healthcare System, Palo Alto, California
| | - Seolhyun Lee
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, Veterans Affairs Palo Alto Healthcare System, Palo Alto, California
| | - Natalie S. Plummer
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, Veterans Affairs Palo Alto Healthcare System, Palo Alto, California
| | - Tammy L. Sirich
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, Veterans Affairs Palo Alto Healthcare System, Palo Alto, California
| | - Timothy W. Meyer
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, Veterans Affairs Palo Alto Healthcare System, Palo Alto, California
| |
Collapse
|
20
|
Ganesan LL, O’Brien FJ, Sirich TL, Plummer NS, Sheth R, Fajardo C, Brakeman P, Sutherland SM, Meyer TW. Association of Plasma Uremic Solute Levels with Residual Kidney Function in Children on Peritoneal Dialysis. Clin J Am Soc Nephrol 2021; 16:1531-1538. [PMID: 34233922 PMCID: PMC8499013 DOI: 10.2215/cjn.01430121] [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: 01/28/2021] [Accepted: 07/01/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Residual native kidney function confers health benefits in patients on dialysis. It can facilitate control of extracellular volume and inorganic ion concentrations. Residual kidney function can also limit the accumulation of uremic solutes. This study assessed whether lower plasma concentrations of uremic solutes were associated with residual kidney function in pediatric patients on peritoneal dialysis. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Samples were analyzed from 29 pediatric patients on peritoneal dialysis, including 13 without residual kidney function and ten with residual kidney function. Metabolomic analysis by untargeted mass spectrometry compared plasma solute levels in patients with and without residual kidney function. Dialytic and residual clearances of selected solutes were also measured by assays using chemical standards. RESULTS Metabolomic analysis showed that plasma levels of 256 uremic solutes in patients with residual kidney function averaged 64% (interquartile range, 51%-81%) of the values in patients without residual kidney function who had similar total Kt/Vurea. The plasma levels were significantly lower for 59 of the 256 solutes in the patients with residual kidney function and significantly higher for none. Assays using chemical standards showed that residual kidney function provides a higher portion of the total clearance for nonurea solutes than it does for urea. CONCLUSIONS Concentrations of many uremic solutes are lower in patients on peritoneal dialysis with residual kidney function than in those without residual kidney function receiving similar treatment as assessed by Kt/Vurea.
Collapse
Affiliation(s)
- Lakshmi L. Ganesan
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California,Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| | - Frank J. O’Brien
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Tammy L. Sirich
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California,Department of Medicine, Stanford University, Palo Alto, California
| | - Natalie S. Plummer
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California,Department of Medicine, Stanford University, Palo Alto, California
| | - Rita Sheth
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California
| | - Cecile Fajardo
- Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California,Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Paul Brakeman
- Department of Pediatrics, University of California, San Francisco, California
| | - Scott M. Sutherland
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| | - Timothy W. Meyer
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California,Department of Medicine, Stanford University, Palo Alto, California
| |
Collapse
|
21
|
Pradhan S, Duffull SB, Wilson LC, Kuan IHS, Walker RJ, Putt TL, Schollum JBW, Wright DFB. Does the intact nephron hypothesis provide a reasonable model for metformin dosing in chronic kidney disease? Br J Clin Pharmacol 2021; 87:4868-4876. [PMID: 34004027 DOI: 10.1111/bcp.14919] [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: 02/07/2021] [Revised: 04/20/2021] [Accepted: 05/09/2021] [Indexed: 11/27/2022] Open
Abstract
This research explored the intact nephron hypothesis (INH) as a model for metformin dosing in patients with chronic kidney disease (CKD). The INH assumes that glomerular filtration rate (GFR) will account for all kidney drug handling even for drugs eliminated by tubular secretion like metformin. We conducted two studies: (1) a regression analysis to explore the relationship between metformin clearance and eGFR metrics, and (2) a joint population pharmacokinetic analysis to test the relationship between metformin renal clearance and gentamicin clearance. The relationship between metformin renal clearance and eGFR metrics and gentamicin clearance was found to be linear, suggesting that a proportional dose reduction based on GFR in patients with CKD is reasonable.
Collapse
Affiliation(s)
- Sudeep Pradhan
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | | - Luke C Wilson
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | | | - Robert J Walker
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Tracey L Putt
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
22
|
Challenges of reducing protein-bound uremic toxin levels in chronic kidney disease and end stage renal disease. Transl Res 2021; 229:115-134. [PMID: 32891787 DOI: 10.1016/j.trsl.2020.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022]
Abstract
The prevalence of chronic kidney disease (CKD) in the worldwide population is currently estimated between 11% and 13%. Adequate renal clearance is compromised in these patients and the accumulation of a large number of uremic retention solutes results in an irreversible worsening of renal function which can lead to end stage renal disease (ESRD). Approximately three million ESRD patients currently receive renal replacement therapies (RRTs), such as hemodialysis, which only partially restore kidney function, as they are only efficient in removing mainly small, unbound solutes from the circulation while leaving larger and protein-bound uremic toxins (PBUTs) untouched. The accumulation of PBUTs in patients highly increases the risk of cardiovascular events and is associated with higher mortality and morbidity in CKD and ESRD. In this review, we address several strategies currently being explored toward reducing PBUT concentrations, including clinical and medical approaches, therapeutic techniques, and recent developments in RRT technology. These include preservation of renal function, limitation of colon derived PBUTs, oral sorbents, adsorbent RRT technology, and use of albumin displacers. Despite the promising results of the different approaches to promote enhanced removal of a small percentage of the more than 30 identified PBUTs, on their own, none of them provide a treatment with the required efficiency, safety and cost-effectiveness to prevent CKD-related complications and decrease mortality and morbidity in ESRD.
Collapse
|
23
|
Chen Y, Zelnick LR, Hoofnagle AN, Yeung CK, Shireman LM, Phillips B, Brauchla CC, de Boer I, Manahan L, Heckbert SR, Himmelfarb J, Kestenbaum BR. Prediction of Kidney Drug Clearance: A Comparison of Tubular Secretory Clearance and Glomerular Filtration Rate. J Am Soc Nephrol 2021; 32:459-468. [PMID: 33239392 PMCID: PMC8054886 DOI: 10.1681/asn.2020060833] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/23/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Although proximal tubular secretion is the primary mechanism of kidney drug elimination, current kidney drug dosing strategies are on the basis of eGFR. METHODS In a dedicated pharmacokinetic study to compare GFR with tubular secretory clearance for predicting kidney drug elimination, we evaluated stable outpatients with eGFRs ranging from 21 to 140 ml/min per 1.73 m2. After administering single doses of furosemide and famciclovir (metabolized to penciclovir), we calculated their kidney clearances on the basis of sequential plasma and timed urine measurements. Concomitantly, we quantified eight endogenous secretory solutes in plasma and urine using liquid chromatography-tandem mass spectrometry and measured GFR by iohexol clearance (iGFR). We computed a summary secretion score as the scaled average of the secretory solute clearances. RESULTS Median iGFR of the 54 participants was 73 ml/min per 1.73 m2. The kidney furosemide clearance correlated with iGFR (r=0.84) and the summary secretion score (r=0.86). The mean proportionate error (MPE) between iGFR-predicted and measured furosemide clearance was 30.0%. The lowest MPE was observed for the summary secretion score (24.1%); MPEs for individual secretory solutes ranged from 27.3% to 48.0%. These predictive errors were statistically indistinguishable. Penciclovir kidney clearance was correlated with iGFR (r=0.83) and with the summary secretion score (r=0.91), with similar predictive accuracy of iGFR and secretory clearances. Combining iGFR with the summary secretion score yielded only modest improvements in the prediction of the kidney clearance of furosemide and penciclovir. CONCLUSIONS Secretory solute clearance measurements can predict kidney drug clearances. However, tight linkage between GFR and proximal tubular secretory clearance in stable outpatients provides some reassurance that GFR, even when estimated, is a useful surrogate for predicting secretory drug clearances in such patients.
Collapse
Affiliation(s)
- Yan Chen
- Department of Epidemiology, University of Washington, Seattle, Washington,Kidney Research Institute, Seattle, Washington
| | - Leila R. Zelnick
- Kidney Research Institute, Seattle, Washington,Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Andrew N. Hoofnagle
- Kidney Research Institute, Seattle, Washington,Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Catherine K. Yeung
- Kidney Research Institute, Seattle, Washington,Department of Pharmacy, University of Washington, Seattle, Washington
| | - Laura M. Shireman
- Department of Pharmacy, University of Washington, Seattle, Washington
| | - Brian Phillips
- Department of Pharmacy, University of Washington, Seattle, Washington
| | | | - Ian de Boer
- Kidney Research Institute, Seattle, Washington,Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Linda Manahan
- Kidney Research Institute, Seattle, Washington,Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, Washington,Department of Pharmacy, University of Washington, Seattle, Washington
| | - Jonathan Himmelfarb
- Kidney Research Institute, Seattle, Washington,Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Bryan R. Kestenbaum
- Kidney Research Institute, Seattle, Washington,Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
24
|
Takita H, Scotcher D, Chinnadurai R, Kalra PA, Galetin A. Physiologically-Based Pharmacokinetic Modelling of Creatinine-Drug Interactions in the Chronic Kidney Disease Population. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:695-706. [PMID: 33049120 PMCID: PMC7762809 DOI: 10.1002/psp4.12566] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Elevated serum creatinine (SCr ) caused by the inhibition of renal transporter(s) may be misinterpreted as kidney injury. The interpretation is more complicated in patients with chronic kidney disease (CKD) due to altered disposition of creatinine and renal transporter inhibitors. A clinical study was conducted in 17 patients with CKD (estimated glomerular filtration rate 15-59 mL/min/1.73 m2 ); changes in SCr were monitored during trimethoprim treatment (100-200 mg/day), administered to prevent recurrent urinary infection, relative to the baseline level. Additional SCr -interaction data with trimethoprim, cimetidine, and famotidine in patients with CKD were collated from the literature. Our published physiologically-based creatinine model was extended to predict the effect of the CKD on SCr and creatinine-drug interaction. The creatinine-CKD model incorporated age/sex-related differences in creatinine synthesis, CKD-related glomerular filtration deterioration; change in transporter activity either proportional or disproportional to glomerular filtration rate (GFR) decline were explored. Optimized models successfully recovered baseline SCr from 64 patients with CKD (geometric mean fold-error of 1.1). Combined with pharmacokinetic models of inhibitors, the creatinine model was used to simulate transporter-mediated creatinine-drug interactions. Use of inhibitor unbound plasma concentrations resulted in 66% of simulated SCr interaction data within the prediction limits, with cimetidine interaction significantly underestimated. Assuming that transporter activity deteriorates disproportional to GFR decline resulted in higher predicted sensitivity to transporter inhibition in patients with CKD relative to healthy patients, consistent with sparse clinical data. For the first time, this novel modelling approach enables quantitative prediction of SCr in CKD and delineation of the effect of disease and renal transporter inhibition in this patient population.
Collapse
Affiliation(s)
- Hiroyuki Takita
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Laboratory for Safety Assessment and ADME, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rajkumar Chinnadurai
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Philip A Kalra
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
25
|
Huang W, Isoherranen N. Novel Mechanistic PBPK Model to Predict Renal Clearance in Varying Stages of CKD by Incorporating Tubular Adaptation and Dynamic Passive Reabsorption. CPT Pharmacometrics Syst Pharmacol 2020; 9:571-583. [PMID: 32977369 PMCID: PMC7577018 DOI: 10.1002/psp4.12553] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/22/2020] [Indexed: 11/13/2022] Open
Abstract
Chronic kidney disease (CKD) has significant effects on renal clearance (CLr ) of drugs. Physiologically-based pharmacokinetic (PBPK) models have been used to predict CKD effects on transporter-mediated renal active secretion and CLr for hydrophilic nonpermeable compounds. However, no studies have shown systematic PBPK modeling of renal passive reabsorption or CLr for hydrophobic permeable drugs in CKD. The goal of this study was to expand our previously developed and verified mechanistic kidney model to develop a universal model to predict changes in CLr in CKD for permeable and nonpermeable drugs that accounts for the dramatic nonlinear effect of CKD on renal passive reabsorption of permeable drugs. The developed model incorporates physiologically-based tubular changes of reduced water reabsorption/increased tubular flow rate per remaining functional nephron in CKD. The final adaptive kidney model successfully (absolute fold error (AFE) all < 2) predicted renal passive reabsorption and CLr for 20 permeable and nonpermeable test compounds across the stages of CKD. In contrast, use of proportional glomerular filtration rate reduction approach without addressing tubular adaptation processes in CKD to predict CLr generated unacceptable CLr predictions (AFE = 2.61-7.35) for permeable compounds in severe CKD. Finally, the adaptive kidney model accurately predicted CLr of para-amino-hippuric acid and memantine, two secreted compounds, in CKD, suggesting successful integration of active secretion into the model, along with passive reabsorption. In conclusion, the developed adaptive kidney model enables mechanistic predictions of in vivo CLr through CKD progression without any empirical scaling factors and can be used for CLr predictions prior to assessment of drug disposition in renal impairment.
Collapse
Affiliation(s)
- Weize Huang
- Department of PharmaceuticsSchool of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | - Nina Isoherranen
- Department of PharmaceuticsSchool of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
26
|
Pradhan S, Wright DF, Duffull SB. Evaluation of designs for renal drug studies based on the European Medicines Agency and Food and Drug Administration guidelines for drugs that are predominantly secreted. Br J Clin Pharmacol 2020; 87:1401-1410. [DOI: 10.1111/bcp.14536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 08/09/2020] [Accepted: 08/18/2020] [Indexed: 12/01/2022] Open
Affiliation(s)
- Sudeep Pradhan
- School of Pharmacy University of Otago Dunedin New Zealand
| | | | | |
Collapse
|
27
|
Zhang Y, Sherwin CM, Gonzalez D, Zhang Q, Khurana M, Fisher J, Burckart GJ, Wang Y, Yao LP, Ganley CJ, Wang J. Creatinine-Based Renal Function Assessment in Pediatric Drug Development: An Analysis Using Clinical Data for Renally Eliminated Drugs. Clin Pharmacol Ther 2020; 109:263-269. [PMID: 32696977 DOI: 10.1002/cpt.1991] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/30/2020] [Indexed: 11/09/2022]
Abstract
The estimated glomerular filtration rate (eGFR) equations based on serum creatinine (SCR) have been used for pediatric dose adjustment in drug labeling. This study evaluated the performance of those equations in estimating individual clearance of drugs that are predominantly eliminated by glomerular filtration, using clinical data from the renally eliminated drugs gadobutrol, gadoterate, amikacin, and vancomycin. The eGFR was compared with the observed drug clearance (CL) in 352 pediatric patients from birth to 12 years of age. Multiple eGFR equations overestimated the drug CL on average, including the original and bedside Schwartz equations, which showed an average eGFR/CL ratio between 1 and 3. Further analysis with bedside Schwartz equation showed a higher eGFR/CL ratio in the subjects with a lower SCR or CL. Supraphysiological eGFR as high as 380 mL/min/1.73 m2 was obtained using the bedside Schwartz equation for some of the subjects, most of whom are children < 2 years of age with SCR < 0.2 mg/dL. Excluding the subjects with supraphysiological eGFR from the analysis did not change the overall trend of overestimation. In conclusion, Schwartz equations led to an overestimation of drug clearance for the drugs evaluated. When greater precision is required in predicting eGFR for pediatric patients, such as in drug dosing, revised k constants for the Schwartz equation or new methods of glomerular filtration rate estimation may be necessary.
Collapse
Affiliation(s)
- Yifei Zhang
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Catherine M Sherwin
- Department of Pediatrics, Wright State University School of Medicine, Dayton Children's Hospital, Dayton, Ohio, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Qunshu Zhang
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mona Khurana
- Division of Pediatric and Maternal Health, Office of Drug Evaluation IV, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jeffrey Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yaning Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lynne P Yao
- Division of Pediatric and Maternal Health, Office of Drug Evaluation IV, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Charles J Ganley
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jian Wang
- Office of Drug Evaluation IV, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
28
|
Schijvens AM, de Wildt SN, Schreuder MF. Pharmacokinetics in children with chronic kidney disease. Pediatr Nephrol 2020; 35:1153-1172. [PMID: 31375913 PMCID: PMC7248054 DOI: 10.1007/s00467-019-04304-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/20/2022]
Abstract
In children, the main causes of chronic kidney disease (CKD) are congenital diseases and glomerular disorders. CKD is associated with multiple physiological changes and may therefore influence various pharmacokinetic (PK) parameters. A well-known consequence of CKD on pharmacokinetics is a reduction in renal clearance due to a decrease in the glomerular filtration rate. The impact of renal impairment on pharmacokinetics is, however, not limited to a decreased elimination of drugs excreted by the kidney. In fact, renal dysfunction may lead to modifications in absorption, distribution, transport, and metabolism as well. Currently, insufficient evidence is available to guide dosing decisions on many commonly used drugs. Moreover, the impact of maturation on drug disposition and action should be taken into account when selecting and dosing drugs in the pediatric population. Clinicians should take PK changes into consideration when selecting and dosing drugs in pediatric CKD patients in order to avoid toxicity and increase efficiency of drugs in this population. The aim of this review is to summarize known PK changes in relation to CKD and to extrapolate available knowledge to the pediatric CKD population to provide guidance for clinical practice.
Collapse
Affiliation(s)
- Anne M Schijvens
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Radboud University Medical Center, Amalia Children's Hospital, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Michiel F Schreuder
- Radboud Institute for Molecular Life Sciences, Department of Pediatric Nephrology, Radboud University Medical Center, Amalia Children's Hospital, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
29
|
Zhang X, Rule AD, McCulloch CE, Lieske JC, Ku E, Hsu CY. Tubular secretion of creatinine and kidney function: an observational study. BMC Nephrol 2020; 21:108. [PMID: 32228497 PMCID: PMC7104490 DOI: 10.1186/s12882-020-01736-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 02/21/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Prior papers have been inconsistent regarding how much creatinine clearance (CrCl) overestimates glomerular filtration rate (GFR). A recent cross-sectional study suggested that measurement error alone could entirely account for the longstanding observation that CrCl/GFR ratio is larger when GFR is lower among patients with chronic kidney disease (CKD); but there have been no validation of this in other cohorts. METHODS To fill these gaps in knowledge regarding the relation between CrCl and GFR, we conducted cross-sectional and longitudinal analysis of the Modification of Diet in Renal Disease study (MDRD) and African American Study of Kidney Disease and Hypertension (AASK); and cross-sectional analysis of a clinical dataset from the Mayo Clinic of four different patient populations (CKD patients, kidney transplant recipients, post kidney donation subgroup and potential kidney donors). In the cross-sectional analyses (MDRD, AASK and Mayo Clinic cohort), we examined the relation between the CrCl/iothalamate GFR (iGFR) ratio at different categories of iGFR or different levels of CrCl. In the MDRD and AASK longitudinal analyses, we studied how the CrCl/iGFR ratio changed with those who had improvement in iGFR (CrCl) over time versus those who had worsening of iGFR (CrCl) over time. RESULTS Observed CrCl/iGFR ratios were generally on the lower end of the range reported in the literature for CKD (median 1.24 in MDRD, 1.13 in AASK and 1.25 in Mayo Clinic cohort). Among CKD patients in whom CrCl and iGFR were measured using different timed urine collections, CrCl/iGFR ratio were higher with lower iGFR categories but lower with lower CrCl categories. However, among CKD patients in whom CrCl and iGFR were measured using the same timed urine collections (which reduces dis-concordant measurement error), CrCl/iGFR ratio were higher with both lower iGFR categories and lower CrCl categories. CONCLUSIONS These data refute the recent suggestion that measurement error alone could entirely account for the longstanding observation that CrCl/GFR ratio increases as GFR decreases in CKD patients. They also highlight the lack of certainty in our knowledge with regard to how much CrCl actually overestimates GFR.
Collapse
Affiliation(s)
- Xuehan Zhang
- Department of Health Care, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, No. 1, Shuaifuyuan, Wangfujing St., Beijing, 100730, China.
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Charles E McCulloch
- Division of Biostatistics, Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - John C Lieske
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Elaine Ku
- Division of Nephrology, University of California, San Francisco, San Francisco, CA, USA
| | - Chi-Yuan Hsu
- Division of Nephrology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
30
|
Lew AK, Crass RL, Eschenauer G. Evolution of Equations for Estimating Renal Function and Their Application to the Dosing of New Antimicrobials. Ann Pharmacother 2019; 54:496-503. [PMID: 31762287 DOI: 10.1177/1060028019890346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objective: To address the background and rationale for the recent introduction of the Modification of Diet in Renal Disease (MDRD) equation for renal dose adjustment of antimicrobials and to provide recommendations for pharmacists dosing new antimicrobial agents. Data Sources: Comprehensive MEDLINE and EMBASE literature searches (from August 2018 to October 2019) were performed. Search terms included creatinine clearance, Cockcroft-Gault, MDRD, and glomerular filtration rate and a subsequent search included the preceding terms AND antimicrobials OR antibiotics. Study Selection and Data Extraction: Available English-language studies on the derivation and/or use of the Cockcroft-Gault (CG) and MDRD study equation were evaluated as well as those that specifically discussed their use for dosing antimicrobial agents. Data Synthesis: The US Food and Drug Administration (FDA) approval of delafloxacin and meropenem-vaborbactam in 2017 ushered in a new era in renal dosing of antibiotics, in that both agents are recommended to be dosed by the MDRD equation. Studies demonstrate that the CG and MDRD equations can result in discrepant dosing recommendations. Relevance to Patient Care and Clinical Practice: The renal estimation equation recommended in a new antibiotic label should dictate the dosing of that medication. It is noteworthy that these equations are not interchangeable. Conclusion: Recently approved antimicrobials utilizing the MDRD equation for renal dose adjustment will be interspersed with old and new antimicrobials utilizing the CG equation because of lack of singular guidance by the FDA. This requires pharmacists to be vigilant in evaluating drug labels to determine which equation is recommended and to understand the differences, strengths, and limitations of each equation.
Collapse
Affiliation(s)
- Alison K Lew
- University of Michigan Health System, Ann Arbor, MI, USA.,University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Ryan L Crass
- University of Michigan Health System, Ann Arbor, MI, USA.,University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Gregory Eschenauer
- University of Michigan Health System, Ann Arbor, MI, USA.,University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| |
Collapse
|
31
|
Evers R, Piquette-Miller M, Polli JW, Russel FGM, Sprowl JA, Tohyama K, Ware JA, de Wildt SN, Xie W, Brouwer KLR. Disease-Associated Changes in Drug Transporters May Impact the Pharmacokinetics and/or Toxicity of Drugs: A White Paper From the International Transporter Consortium. Clin Pharmacol Ther 2018; 104:900-915. [PMID: 29756222 PMCID: PMC6424581 DOI: 10.1002/cpt.1115] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022]
Abstract
Drug transporters are critically important for the absorption, distribution, metabolism, and excretion (ADME) of many drugs and endogenous compounds. Therefore, disruption of these pathways by inhibition, induction, genetic polymorphisms, or disease can have profound effects on overall physiology, drug pharmacokinetics, drug efficacy, and toxicity. This white paper provides a review of changes in transporter function associated with acute and chronic disease states, describes regulatory pathways affecting transporter expression, and identifies opportunities to advance the field.
Collapse
Affiliation(s)
- Raymond Evers
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey, USA
| | | | - Joseph W Polli
- Mechanistic Safety and Drug Disposition, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jason A Sprowl
- Department of Pharmaceutical, Social and Administrative Sciences, School of Pharmacy, D'Youville College School, Buffalo, New York, USA
| | - Kimio Tohyama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company, Fujisawa, Kanagawa, Japan
| | - Joseph A Ware
- Department of Small Molecule Pharmaceutical Sciences, Genentech, South San Francisco, California, USA
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology and Department of Intensive Care, Radboud University Medical Center, Nijmegen, The Netherlands, and Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
32
|
The intact nephron hypothesis as a model for renal drug handling. Eur J Clin Pharmacol 2018; 75:147-156. [DOI: 10.1007/s00228-018-2572-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/30/2018] [Indexed: 10/28/2022]
|
33
|
Wang K, Kestenbaum B. Proximal Tubular Secretory Clearance: A Neglected Partner of Kidney Function. Clin J Am Soc Nephrol 2018; 13:1291-1296. [PMID: 29490976 PMCID: PMC6086711 DOI: 10.2215/cjn.12001017] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The secretion of small molecules by the proximal tubules of the kidneys represents a vital homeostatic function for rapidly clearing endogenous solutes and medications from the circulation. After filtration at the glomerulus, renal blood flow is directed through a network of peritubular capillaries, where transporters of the proximal tubules actively secrete putative uremic toxins and hundreds of commonly prescribed drugs into the urine, including protein-bound substances that cannot readily cross the glomerular basement membrane. Despite its central physiologic importance, tubular secretory clearance is rarely measured or even estimated in clinical or research settings. Major barriers to estimating tubular solute clearance include uncertainty regarding optimal endogenous secretory markers and a lack of standardized laboratory assays. The creation of new methods to measure tubular secretion could catalyze advances in kidney disease research and clinical care. Differences in secretory clearance relative to the GFR could help distinguish among the causes of CKD, particularly for disorders that primarily affect the tubulointerstitium. As the primary mechanism by which the kidneys excrete medications, tubular secretory clearance offers promise for improving kidney medication dosing, which is currently exclusively on the basis of filtration. The differing metabolic profiles of retained solutes eliminated by secretion versus glomerular filtration suggest that secretory clearance could uniquely inform uremic toxicity, refine existing measures of residual kidney function, and improve prediction of cardiovascular and kidney disease outcomes. Interdisciplinary research across clinical, translational, and laboratory medicine is needed to bring this often neglected kidney function into the limelight.
Collapse
Affiliation(s)
- Ke Wang
- Division of Nephrology, Department of Medicine and
- Kidney Research Institute, University of Washington, Seattle, Washington
| | - Bryan Kestenbaum
- Division of Nephrology, Department of Medicine and
- Kidney Research Institute, University of Washington, Seattle, Washington
| |
Collapse
|
34
|
Lea-Henry TN, Carland JE, Stocker SL, Sevastos J, Roberts DM. Clinical Pharmacokinetics in Kidney Disease: Fundamental Principles. Clin J Am Soc Nephrol 2018; 13:1085-1095. [PMID: 29934432 PMCID: PMC6032582 DOI: 10.2215/cjn.00340118] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Kidney disease is an increasingly common comorbidity that alters the pharmacokinetics of many drugs. Prescribing to patients with kidney disease requires knowledge about the drug, the extent of the patient's altered physiology, and pharmacokinetic principles that influence the design of dosing regimens. There are multiple physiologic effects of impaired kidney function, and the extent to which they occur in an individual at any given time can be difficult to define. Although some guidelines are available for dosing in kidney disease, they may be on the basis of limited data or not widely applicable, and therefore, an understanding of pharmacokinetic principles and how to apply them is important to the practicing clinician. Whether kidney disease is acute or chronic, drug clearance decreases, and the volume of distribution may remain the same or increase. Although in CKD, these changes progress relatively slowly, they are dynamic in AKI, and recovery is possible depending on the etiology and treatments. This, and the use of kidney replacement therapies further complicate attempts to quantify drug clearance at the time of prescribing and dosing in AKI. The required change in the dosing regimen can be estimated or even quantitated in certain instances through the application of pharmacokinetic principles to guide rational drug dosing. This offers an opportunity to provide personalized medical care and minimizes adverse drug events from either under- or overdosing. We discuss the principles of pharmacokinetics that are fundamental for the design of an appropriate dosing regimen in this review.
Collapse
Affiliation(s)
- Tom N. Lea-Henry
- Nephrology and Transplantation Unit, John Hunter Hospital, Newcastle, New South Wales, Australia
- Department of Renal Medicine, The Canberra Hospital, Woden, Australian Capital Territory, Australia; and
| | - Jane E. Carland
- Departments of Clinical Pharmacology and Toxicology and
- Department of Medicine, St. Vincent’s Clinical School, St. Vincent’s Hospital, University of New South Wales, Sydney, New South Wales, Australia
| | - Sophie L. Stocker
- Departments of Clinical Pharmacology and Toxicology and
- Department of Medicine, St. Vincent’s Clinical School, St. Vincent’s Hospital, University of New South Wales, Sydney, New South Wales, Australia
| | - Jacob Sevastos
- Nephrology and Renal Transplantation, St. Vincent’s Hospital, Darlinghurst, New South Wales, Australia
- Department of Medicine, St. Vincent’s Clinical School, St. Vincent’s Hospital, University of New South Wales, Sydney, New South Wales, Australia
| | - Darren M. Roberts
- Departments of Clinical Pharmacology and Toxicology and
- Department of Renal Medicine, The Canberra Hospital, Woden, Australian Capital Territory, Australia; and
- Medical School, Australian National University, Acton, Australian Capital Territory, Australia
| |
Collapse
|
35
|
Uremic Toxin Clearance and Cardiovascular Toxicities. Toxins (Basel) 2018; 10:toxins10060226. [PMID: 29865226 PMCID: PMC6024759 DOI: 10.3390/toxins10060226] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/25/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
Uremic solutes contribute to cardiovascular disease in renal insufficiency. In this review we describe the clearance of selected uremic solutes, which have been associated with cardiovascular disease. These solutes—indoxyl sulfate (IS), p-cresol sulfate (PCS), phenylacetylglutamine (PAG), trimethylamine-n-oxide (TMAO), and kynurenine—exemplify different mechanisms of clearance. IS and PCS are protein-bound solutes efficiently cleared by the native kidney through tubular secretion. PAG and TMAO are not protein-bound but are also cleared by the native kidney through tubular secretion, while kynurenine is not normally cleared by the kidney. Increases in the plasma levels of the normally secreted solutes IS, PCS, TMAO, and PAG in chronic kidney disease (CKD) are attributable to a reduction in their renal clearances. Levels of each of these potential toxins are even higher in patients on dialysis than in those with advanced chronic kidney disease, which can be accounted for in part by a low ratio of dialytic to native kidney clearance. The rise in plasma kynurenine in CKD and dialysis patients, by contrast, remains to be explained. Our ability to detect lower levels of the potential uremic cardiovascular toxins with renal replacement therapy may be limited by the intermittency of treatment, by increases in solute production, and by the presence of non-renal clearance. Reduction in the levels of uremic cardiovascular toxins may in the future be achieved more effectively by inhibiting their production.
Collapse
|
36
|
Abstract
Uremic solutes contribute to cardiovascular disease in renal insufficiency. In this review we describe the clearance of selected uremic solutes, which have been associated with cardiovascular disease. These solutes-indoxyl sulfate (IS), p-cresol sulfate (PCS), phenylacetylglutamine (PAG), trimethylamine-n-oxide (TMAO), and kynurenine-exemplify different mechanisms of clearance. IS and PCS are protein-bound solutes efficiently cleared by the native kidney through tubular secretion. PAG and TMAO are not protein-bound but are also cleared by the native kidney through tubular secretion, while kynurenine is not normally cleared by the kidney. Increases in the plasma levels of the normally secreted solutes IS, PCS, TMAO, and PAG in chronic kidney disease (CKD) are attributable to a reduction in their renal clearances. Levels of each of these potential toxins are even higher in patients on dialysis than in those with advanced chronic kidney disease, which can be accounted for in part by a low ratio of dialytic to native kidney clearance. The rise in plasma kynurenine in CKD and dialysis patients, by contrast, remains to be explained. Our ability to detect lower levels of the potential uremic cardiovascular toxins with renal replacement therapy may be limited by the intermittency of treatment, by increases in solute production, and by the presence of non-renal clearance. Reduction in the levels of uremic cardiovascular toxins may in the future be achieved more effectively by inhibiting their production.
Collapse
Affiliation(s)
- Robert D Mair
- The Departments of Medicine, VA Palo Alto Healthcare System, 111R, 3801 Miranda Ave., Palo Alto, CA 94304, USA.
- Division of Nephrology, Stanford University, 777 Welch Road, Suite DE, Palo Alto, CA 94304, USA.
| | - Tammy L Sirich
- The Departments of Medicine, VA Palo Alto Healthcare System, 111R, 3801 Miranda Ave., Palo Alto, CA 94304, USA.
- Division of Nephrology, Stanford University, 777 Welch Road, Suite DE, Palo Alto, CA 94304, USA.
| | - Timothy W Meyer
- The Departments of Medicine, VA Palo Alto Healthcare System, 111R, 3801 Miranda Ave., Palo Alto, CA 94304, USA.
- Division of Nephrology, Stanford University, 777 Welch Road, Suite DE, Palo Alto, CA 94304, USA.
| |
Collapse
|