1
|
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
|
2
|
Rodrigues D, Wezalis S. Clinical Assessment of Drug Transporter Inhibition Using Biomarkers: Review of the Literature (2015-2024). J Clin Pharmacol 2025. [PMID: 39828904 DOI: 10.1002/jcph.6183] [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: 10/28/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
As part of a narrative review of various publications describing the clinical use of urine- and plasma-based drug transporter biomarkers, it was determined that the utilization of coproporphyrin I, a hepatic organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 biomarker, has been reported for 28 different drug-drug interaction (DDI) perpetrator drugs. Similarly, biomarkers for liver organic cation transporter 1 (isobutyryl-l-carnitine, N = 7 inhibitors), renal organic cation transporter 2 and multidrug and toxin extrusion proteins (N1-methylnicotinamide, N = 13 inhibitors), renal organic anion transporter (OAT) 1 and 3 (pyridoxic acid, N = 7 inhibitors), and breast cancer resistance protein (riboflavin, N = 3 inhibitors) have also been described. Increased use of biomarkers has also been accompanied by modeling efforts to enable DDI predictions and development of multiplexed methods to facilitate their bioanalysis. Overall, there is consensus that exploratory biomarkers such as coproporphyrin I can be integrated into decision trees encompassing in vitro transporter inhibition data, DDI risk assessments, and follow-up Phase 1 studies. Therefore, sponsors can leverage biomarkers to evaluate dose-dependent inhibition of selected transporters, use them jointly with drug probes to deconvolute DDI mechanisms, and integrate in vitro data packages to establish calibrated (biomarker informed) DDI risk assessment cutoffs. Although transporter biomarker science has progressed, reflected by its inclusion in the recently issued International Council for Harmonisation DDI guidance document (M12), some biomarkers still require further validation. There is also a need for biomarkers that can differentiate specific transporters (e.g., OATP1B3 vs OATP1B1 and OAT1 vs OAT3).
Collapse
Affiliation(s)
- David Rodrigues
- Drug Metabolism and Nonclinical Pharmacokinetics, Translational Medicine, Incyte, Wilmington, DE, USA
| | - Stephanie Wezalis
- Drug Metabolism and Nonclinical Pharmacokinetics, Translational Medicine, Incyte, Wilmington, DE, USA
| |
Collapse
|
3
|
Gessner A, König J, Wenisch P, Heinrich MR, Stopfer P, Fromm MF, Müller F. New Biomarkers for Renal Transporter-Mediated Drug-Drug Interactions: Metabolomic Effects of Cimetidine, Probenecid, Verapamil, and Rifampin in Humans. Clin Pharmacol Ther 2025; 117:130-142. [PMID: 39148267 PMCID: PMC11652812 DOI: 10.1002/cpt.3414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
The inhibition of renal transport proteins organic cation transporter 2 (OCT2), multidrug and toxin extrusion proteins (MATE1, MATE2-K), and organic anion transporters (OAT1, OAT3) causes clinically relevant drug-drug interactions (DDI). Endogenous biomarkers could be used to improve risk prediction of such renal DDIs. While a number of biomarkers for renal DDIs have been described so far, multiple criteria for valid biomarkers have frequently not been investigated, for example, specificity, metabolism, or food effects. Therefore, there is a need for novel biomarkers of renal DDIs. Here, we investigated the global metabolomic effects following the administration of two classical inhibitors of renal transport proteins [cimetidine (OCT2/MATEs), probenecid (OATs)] in human plasma and urine of healthy volunteers. Additionally, we investigated metabolomic effects of two inhibitors of other transporters [verapamil (P-glycoprotein), rifampin (organic anion transporting polypeptides)] as controls. This analysis shows that both cimetidine and probenecid affect compounds involved in caffeine metabolism, carnitines, and sulfates. Hierarchical cluster analysis of the effects of all four inhibitors on endogenous compounds identified multiple promising new sensitive and specific biomarker candidates for OCT2/MATE- or OAT-mediated DDIs. For OCT2/MATEs, 5-amino valeric acid betaine (median log2-fold change of estimated renal elimination: -3.62) presented itself as a promising candidate. For OATs, estimated renal elimination of 7-methyluric acid and cinnamoylglycine (median log2-fold changes -3.10 and -1.92, respectively) was both sensitive and specific. This study provides comprehensive information on metabolomic effects of transport protein inhibition in humans and identifies putative new sensitive and specific biomarkers for renal transporter-mediated DDIs.
Collapse
Affiliation(s)
- Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Pia Wenisch
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Markus R. Heinrich
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Peter Stopfer
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der RissGermany
| | - Martin F. Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der RissGermany
| |
Collapse
|
4
|
Granados JC, Nigam SK. Organic anion transporters in remote sensing and organ crosstalk. Pharmacol Ther 2024; 263:108723. [PMID: 39284369 DOI: 10.1016/j.pharmthera.2024.108723] [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: 05/01/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 11/05/2024]
Abstract
The organic anion transporters, OAT1 and OAT3, regulate the movement of drugs, toxins, and endogenous metabolites. In 2007, we proposed that OATs and other SLC22 transporters are involved in "remote sensing" and organ crosstalk. This is now known as the Remote Sensing and Signaling Theory (RSST). In the proximal tubule of the kidney, OATs regulate signaling molecules such as fatty acids, bile acids, indoxyl sulfate, kynurenine, alpha-ketoglutarate, urate, flavonoids, and antioxidants. OAT1 and OAT3 function as key hubs in a large homeostatic network involving multi-, oligo- and monospecific transporters, enzymes, and nuclear receptors. The Remote Sensing and Signaling Theory emphasizes the functioning of OATs and other "drug" transporters in the network at multiple biological scales (inter-organismal, organism, organ, cell, organelle). This network plays an essential role in the homeostasis of urate, bile acids, prostaglandins, sex steroids, odorants, thyroxine, gut microbiome metabolites, and uremic toxins. The transported metabolites have targets in the kidney and other organs, including nuclear receptors (e.g., HNF4a, AHR), G protein-coupled receptors (GPCRs), and protein kinases. Feed-forward and feedback loops allow OAT1 and OAT3 to mediate organ crosstalk as well as modulate energy metabolism, redox state, and remote sensing. Furthermore, there is intimate inter-organismal communication between renal OATs and the gut microbiome. Extracellular vesicles containing microRNAs and proteins (exosomes) play a key role in the Remote Sensing and Signaling System as does the interplay with the neuroendocrine, hormonal, and immune systems. Perturbation of function with OAT-interacting drugs (e.g., probenecid, diuretics, antivirals, antibiotics, NSAIDs) can lead to drug-metabolite interactions. The RSST has general applicability to other multi-specific SLC and ABC "drug" transporters (e.g., OCT1, OCT2, SLCO1B1, SLCO1B3, ABCG2, P-gp, ABCC2, ABCC3, ABCC4). Recent high-resolution structures of SLC22 and other transporters, together with chemoinformatic and artificial intelligence methods, will aid drug development and also lead to a deeper mechanistic understanding of polymorphisms.
Collapse
Affiliation(s)
- Jeffry C Granados
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Sanjay K Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine (Nephrology), University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
5
|
Hîncu S, Apetroaei MM, Ștefan G, Fâcă AI, Arsene AL, Mahler B, Drăgănescu D, Tăerel AE, Stancu E, Hîncu L, Zamfirescu A, Udeanu DI. Drug-Drug Interactions in Nosocomial Infections: An Updated Review for Clinicians. Pharmaceutics 2024; 16:1137. [PMID: 39339174 PMCID: PMC11434876 DOI: 10.3390/pharmaceutics16091137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Prevention, assessment, and identification of drug-drug interactions (DDIs) represent a challenge for healthcare professionals, especially in nosocomial settings. This narrative review aims to provide a thorough assessment of the most clinically significant DDIs for antibiotics used in healthcare-associated infections. Complex poly-pharmaceutical regimens, targeting multiple pathogens or targeting one pathogen in the presence of another comorbidity, have an increased predisposition to result in life-threatening DDIs. Recognising, assessing, and limiting DDIs in nosocomial infections offers promising opportunities for improving health outcomes. The objective of this review is to provide clinicians with practical advice to prevent or mitigate DDIs, with the aim of increasing the safety and effectiveness of therapy. DDI management is of significant importance for individualising therapy according to the patient, disease status, and associated comorbidities.
Collapse
Affiliation(s)
- Sorina Hîncu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
- Fundeni Clinical Institute, 258, Fundeni Street, 022328 Bucharest, Romania
| | - Miruna-Maria Apetroaei
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
| | - Gabriela Ștefan
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
| | - Anca Ionela Fâcă
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
- Marius Nasta Institute of Pneumophthisiology, 90, Viilor Street, 050159 Bucharest, Romania;
| | - Andreea Letiția Arsene
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
- Marius Nasta Institute of Pneumophthisiology, 90, Viilor Street, 050159 Bucharest, Romania;
| | - Beatrice Mahler
- Marius Nasta Institute of Pneumophthisiology, 90, Viilor Street, 050159 Bucharest, Romania;
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8, Eroii Sanitari Street, 050474 Bucharest, Romania
| | - Doina Drăgănescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
| | - Adriana-Elena Tăerel
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
| | - Emilia Stancu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
| | - Lucian Hîncu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
| | - Andreea Zamfirescu
- Faculty of Midwifery and Nursing, Carol Davila University of Medicine and Pharmacy, 8, Street, 050474 Bucharest, Romania;
| | - Denisa Ioana Udeanu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania; (S.H.); (G.Ș.); (A.I.F.); (A.L.A.); (D.D.); (A.-E.T.); (E.S.); (L.H.); (D.I.U.)
- Marius Nasta Institute of Pneumophthisiology, 90, Viilor Street, 050159 Bucharest, Romania;
| |
Collapse
|
6
|
Falah K, Zhang P, Nigam AK, Maity K, Chang G, Granados JC, Momper JD, Nigam SK. In Vivo Regulation of Small Molecule Natural Products, Antioxidants, and Nutrients by OAT1 and OAT3. Nutrients 2024; 16:2242. [PMID: 39064685 PMCID: PMC11280313 DOI: 10.3390/nu16142242] [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: 05/02/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The organic anion transporters OAT1 (SLC22A6) and OAT3 (SLC22A8) are drug transporters that are expressed in the kidney, with well-established roles in the in vivo transport of drugs and endogenous metabolites. A comparatively unexplored potential function of these drug transporters is their contribution to the in vivo regulation of natural products (NPs) and their effects on endogenous metabolism. This is important for the evaluation of potential NP interactions with other compounds at the transporter site. Here, we have analyzed the NPs present in several well-established databases from Asian (Chinese, Indian Ayurvedic) and other traditions. Loss of OAT1 and OAT3 in murine knockouts caused serum alterations of many NPs, including flavonoids, vitamins, and indoles. OAT1- and OAT3-dependent NPs were largely separable based on a multivariate analysis of chemical properties. Direct binding to the transporter was confirmed using in vitro transport assays and protein binding assays. Our in vivo and in vitro results, considered in the context of previous data, demonstrate that OAT1 and OAT3 play a pivotal role in the handling of non-synthetic small molecule natural products, NP-derived antioxidants, phytochemicals, and nutrients (e.g., pantothenic acid, thiamine). As described by remote sensing and signaling theory, drug transporters help regulate redox states by meditating the movement of endogenous antioxidants and nutrients between organs and organisms. Our results demonstrate how dietary antioxidants and other NPs might feed into these inter-organ and inter-organismal pathways.
Collapse
Affiliation(s)
- Kian Falah
- Department of Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Patrick Zhang
- Department of Biology, University of California San Diego, La Jolla, CA 92093, USA
| | - Anisha K. Nigam
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Koustav Maity
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Geoffrey Chang
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jeffry C. Granados
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Jeremiah D. Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Sanjay K. Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Department of Medicine (Nephrology), University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Wu J, Aga L, Tang L, Li H, Wang N, Yang L, Zhang N, Wang X, Wang X. Lacticaseibacillus paracasei JS-3 Isolated from "Jiangshui" Ameliorates Hyperuricemia by Regulating Gut Microbiota and iTS Metabolism. Foods 2024; 13:1371. [PMID: 38731742 PMCID: PMC11083236 DOI: 10.3390/foods13091371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Background: A diet high in purines can impair the function of the gut microbiota and disrupt purine metabolism, which is closely associated with the onset of hyperuricemia. Dietary regulation and intestinal health maintenance are key approaches for controlling uric acid (UA) levels. Investigating the impacts of fermented foods offers potential dietary interventions for managing hyperuricemia. Methods: In this study, we isolated a strain with potent UA-degrading capabilities from "Jiangshui", a fermented food product from Gansu, China. We performed strain identification and assessed its probiotic potential. Hyperuricemic quails, induced by a high-purine diet, were used to assess the UA degradation capability of strain JS-3 by measuring UA levels in serum and feces. Additionally, the UA degradation pathways were elucidated through analyses of the gut microbiome and fecal metabolomics. Results: JS-3, identified as Lacticaseibacillus paracasei, was capable of eliminating 16.11% of uric acid (UA) within 72 h, rapidly proliferating and producing acid within 12 h, and surviving in the gastrointestinal tract. Using hyperuricemic quail models, we assessed JS-3's UA degradation capacity. Two weeks after the administration of JS-3 (2 × 108 cfu/d per quail), serum uric acid (SUA) levels significantly decreased to normal levels, and renal damage in quails was markedly improved. Concurrently, feces from the JS-3 group demonstrated a significant degradation of UA, achieving up to 49% within 24 h. 16S rRNA sequencing revealed JS-3's role in gut microbiota restoration by augmenting the probiotic community (Bifidobacterium, Bacteroides unclassified_f-Lachnospiraceae, and norank_fynorank_o-Clostridia_UCG-014) and diminishing the pathogenic bacteria (Macrococus and Lactococcus). Corresponding with the rise in short-chain fatty acid (SCFA)-producing bacteria, JS-3 significantly increased SCFA levels (p < 0.05, 0.01). Additionally, JS-3 ameliorated metabolic disturbances in hyperuricemic quails, influencing 26 abnormal metabolites predominantly linked to purine, tryptophan, and bile acid metabolism, thereby enhancing UA degradation and renal protection. Conclusions: For the first time, we isolated and identified an active probiotic strain, JS-3, from the "Jiangshui" in Gansu, used for the treatment of hyperuricemia. It modulates host-microbiome interactions, impacts the metabolome, enhances intestinal UA degradation, reduces levels of SUA and fecal UA, alleviates renal damage, and effectively treats hyperuricemia without causing gastrointestinal damage. In summary, JS-3 can serve as a probiotic with potential therapeutic value for the treatment of hyperuricemia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xueyong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China; (J.W.); (L.T.); (H.L.); (N.W.)
| |
Collapse
|
8
|
Hou Z, Ma A, Mao J, Song D, Zhao X. Overview of the pharmacokinetics and pharmacodynamics of URAT1 inhibitors for the treatment of hyperuricemia and gout. Expert Opin Drug Metab Toxicol 2023; 19:895-909. [PMID: 37994776 DOI: 10.1080/17425255.2023.2287477] [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: 05/17/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
INTRODUCTION Hyperuricemia is a common metabolic disease, which is a risk factor for gouty arthritis and ureteral stones and may also lead to cardiovascular and chronic kidney disease (CDK). Therefore, hyperuricemia should be treated early. Xanthine oxidase inhibitors (XOIs) and uricosuric agents (UAs), which target uric acid, are two types of medications that are used to treat gout and hyperuricemia. XOIs stop the body from producing excessive uric acid, while UAs eliminate it rapidly via the kidneys. Urate transporter 1 (URAT1) belongs to the organic anion transporter family (OAT) and is specifically localized to the apical membrane of the epithelial cells of proximal tubules. Unlike other organic anion transporter family members, URAT1 identifies and transports organic anions that are primarily responsible for urate transport. AREAS COVERED This article reviews the pharmacokinetics and pharmacodynamics of the existing URAT1 inhibitors to serve as a reference for subsequent drug studies. EXPERT OPINION The URAT1 inhibitors that are currently used as clinical drugs mainly include dotinurad, benzbromarone, and probenecid. Results indicate that RDEA3170 may be the most promising inhibitor, in addition to SHR4640, URC-102, and MBX-102, which are in the early stages of development.
Collapse
Affiliation(s)
- Zihan Hou
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Aijinxiu Ma
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Jiale Mao
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Danni Song
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Xu Zhao
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
9
|
Thakur A, Saradhi Mettu V, Singh DK, Prasad B. Effect of probenecid on blood levels and renal elimination of furosemide and endogenous compounds in rats: Discovery of putative organic anion transporter biomarkers. Biochem Pharmacol 2023; 218:115867. [PMID: 37866801 PMCID: PMC10900896 DOI: 10.1016/j.bcp.2023.115867] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Transporter-mediated drug-drug interactions (DDIs) are assessed using probe drugs and in vitro and in vivo models during drug development. The utility of endogenous metabolites as transporter biomarkers is emerging for prediction of DDIs during early phases of clinical trials. Endogenous metabolites such as pyridoxic acid and kynurenic acid have shown potential to predict DDIs mediated by organic anion transporters (OAT1 and OAT3). However, these metabolites have not been assessed in rats as potential transporter biomarkers. We carried out a rat pharmacokinetic DDI study using probenecid and furosemide as OAT inhibitor and substrate, respectively. Probenecid administration led to a 3.8-fold increase in the blood concentrations and a 3-fold decrease in renal clearance of furosemide. High inter-individual and intra-day variability in pyridoxic acid and kynurenic acid, and no or moderate effect of probenecid administration on these metabolites suggest their limited utility for prediction of Oat-mediated DDI in rats. Therefore, rat blood and urine samples were further analysed using untargeted metabolomics. Twenty-one m/z features (out of >8000 detected features) were identified as putative biomarkers of rat Oat1 and Oat3 using a robust biomarker qualification approach. These m/z features belong to metabolic pathways such as fatty acid analogues, peptides, prostaglandin analogues, bile acid derivatives, flavonoids, phytoconstituents, and steroids, and can be used as a panel to decrease variability caused by processes other than Oats. When validated, these putative biomarkers will be useful in predicting DDIs caused by Oats in rats.
Collapse
Affiliation(s)
- Aarzoo Thakur
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Vijaya Saradhi Mettu
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Dilip K Singh
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
10
|
Terkeltaub R. Emerging Urate-Lowering Drugs and Pharmacologic Treatment Strategies for Gout: A Narrative Review. Drugs 2023; 83:1501-1521. [PMID: 37819612 DOI: 10.1007/s40265-023-01944-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/13/2023]
Abstract
Hyperuricemia with consequent monosodium urate crystal deposition leads to gout, characterized by painful, incapacitating inflammatory arthritis flares that are also associated with increased cardiovascular event and related mortality risk. This narrative review focuses on emerging pharmacologic urate-lowering treatment (ULT) and management strategies in gout. Undertreated, gout can progress to palpable tophi and joint damage. In oral ULT clinical trials, target serum urate of < 6.0 mg/dL can be achieved in ~ 80-90% of subjects, with flare burden reduction by 1-2 years. However, real-world ULT results are far less successful, due to both singular patient nonadherence and prescriber undertreatment, particularly in primary care, where most patients are managed. Multiple dose titrations commonly needed to optimize first-line allopurinol ULT monotherapy, and substantial potential toxicities and other limitations of approved, marketed oral monotherapy ULT drugs, promote hyperuricemia undertreatment. Common gout comorbidities with associated increased mortality (e.g., moderate-severe chronic kidney disease [CKD], type 2 diabetes, hypertension, atherosclerosis, heart failure) heighten ULT treatment complexity and emphasize unmet needs for better and more rapid clinically significant outcomes, including attenuated gout flare burden. The gout drug armamentarium will be expanded by integrating sodium-glucose cotransporter-2 (SGLT2) inhibitors with uricosuric and anti-inflammatory properties as well as clinically indicated antidiabetic, nephroprotective, and/or cardioprotective effects. The broad ULT developmental pipeline is loaded with multiple uricosurics that selectively target uric acid transporter 1 (URAT1). Evolving ULT approaches include administering selected gut anaerobic purine degrading bacteria (PDB), modulating intestinal urate transport, and employing liver-targeted xanthine oxidoreductase mRNA knockdown. Last, emerging measures to decrease the immunogenicity of systemically administered recombinant uricases should simplify treatment regimens and further improve outcomes in managing the most severe gout phenotypes.
Collapse
Affiliation(s)
- Robert Terkeltaub
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
11
|
Gessner A, Müller F, Wenisch P, Heinrich MR, König J, Stopfer P, Fromm MF. A Metabolomic Analysis of Sensitivity and Specificity of 23 Previously Proposed Biomarkers for Renal Transporter-Mediated Drug-Drug Interactions. Clin Pharmacol Ther 2023; 114:1058-1072. [PMID: 37540045 DOI: 10.1002/cpt.3017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Endogenous biomarkers are discussed as tools for detection of drug-drug interactions (DDIs) mediated by renal transport proteins, such as organic cation transporter 2 (OCT2), multidrug and toxin extrusion proteins (MATE1 and MATE2-K) and organic anion transporters (OAT1 and OAT3). Whereas sensitivity of some endogenous biomarkers against at least one clinical transporter inhibitor has frequently been shown, intra-study comparisons of the extent of effects of inhibitors on different biomarkers are frequently lacking. Moreover, in vivo specificity of such discussed biomarkers has frequently not been studied. We therefore investigated changes of 10 previously described putative biomarkers for inhibition of OCT2/MATEs, as well as 15 previously described putative biomarkers for OATs in human plasma and urine samples of healthy volunteers in response to treatment with 4 inhibitors of transport proteins [verapamil (P-glycoprotein), rifampin (organic anion transporting polypeptides), cimetidine (OCT2/MATEs), and probenecid (OATs)]. Two of the putative biomarkers had been suggested for both OCT2/MATEs and OATs. All substances were unequivocally identified in an untargeted metabolomics assay. The OCT2/MATE biomarkers choline and trimethylamine N-oxide were both sensitive and specific (median log2-fold changes -1.18 in estimated renal elimination and -0.85 in urinary excretion, respectively). For renal OATs, indoleacetyl glutamine and indoleacetic acid (median log2-fold changes -3.77 and -2.85 in estimated renal elimination, respectively) were the candidates for sensitive and specific biomarkers with the most extensive change, followed by taurine, indolelactic acid, and hypoxanthine. This comprehensive study adds further knowledge on sensitivity and specificity of 23 previously described biomarkers of renal OCT2/MATE- and OAT-mediated DDIs.
Collapse
Affiliation(s)
- Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Pia Wenisch
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus R Heinrich
- Department of Chemistry and Pharmacy, Pharmaceutical Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Stopfer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Dou T, Lian T, Shu S, He Y, Jiang J. The substrate and inhibitor binding mechanism of polyspecific transporter OAT1 revealed by high-resolution cryo-EM. Nat Struct Mol Biol 2023; 30:1794-1805. [PMID: 37845412 PMCID: PMC11406556 DOI: 10.1038/s41594-023-01123-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023]
Abstract
Organic anion transporters (OATs) of the SLC22 family have crucial roles in the transport of organic anions, including metabolites and therapeutic drugs, and in transporter-mediated drug-drug interactions. In the kidneys, OATs facilitate the elimination of metabolic waste products and xenobiotics. However, their transport activities can lead to the accumulation of certain toxic compounds within cells, causing kidney damage. Moreover, OATs are important drug targets, because their inhibition modulates the elimination or retention of substrates linked to diseases. Despite extensive research on OATs, the molecular basis of their substrate and inhibitor binding remains poorly understood. Here we report the cryo-EM structures of rat OAT1 (also known as SLC22A6) and its complexes with para-aminohippuric acid and probenecid at 2.1, 2.8 and 2.9 Å resolution, respectively. Our findings reveal a highly conserved substrate binding mechanism for SLC22 transporters, wherein four aromatic residues form a cage to accommodate the polyspecific binding of diverse compounds.
Collapse
Affiliation(s)
- Tongyi Dou
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Tengfei Lian
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Shi Shu
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Yi He
- Fermentation Facility, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Jiansen Jiang
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
13
|
Peng X, Li X, Xie B, Lai Y, Sosnik A, Boucetta H, Chen Z, He W. Gout therapeutics and drug delivery. J Control Release 2023; 362:728-754. [PMID: 37690697 DOI: 10.1016/j.jconrel.2023.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Gout is a common inflammatory arthritis caused by persistently elevated uric acid levels. With the improvement of people's living standards, the consumption of processed food and the widespread use of drugs that induce elevated uric acid, gout rates are increasing, seriously affecting the human quality of life, and becoming a burden to health systems worldwide. Since the pathological mechanism of gout has been elucidated, there are relatively effective drug treatments in clinical practice. However, due to (bio)pharmaceutical shortcomings of these drugs, such as poor chemical stability and limited ability to target the pathophysiological pathways, traditional drug treatment strategies show low efficacy and safety. In this scenario, drug delivery systems (DDS) design that overcome these drawbacks is urgently called for. In this review, we initially describe the pathological features, the therapeutic targets, and the drugs currently in clinical use and under investigation to treat gout. We also comprehensively summarize recent research efforts utilizing lipid, polymeric and inorganic carriers to develop advanced DDS for improved gout management and therapy.
Collapse
Affiliation(s)
- Xiuju Peng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Alejandro Sosnik
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Hamza Boucetta
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
14
|
Lai Y. The Role of Coproporphyrins As Endogenous Biomarkers for Organic Anion Transporting Polypeptide 1B Inhibition-Progress from 2016 to 2023. Drug Metab Dispos 2023; 51:950-961. [PMID: 37407093 DOI: 10.1124/dmd.122.001012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 07/07/2023] Open
Abstract
Since the initial clinical study investigating coproporphyrins I and III (CP-I and CP-III) as endogenous biomarkers for organic anion transporting polypeptide (OATP) inhibition drug-drug interactions (DDIs) published in 2016, significant progress has been made in confirming the usefulness of the CPs, particularly CP-I, as biomarkers in assessing OATP functions. CP-I exhibits selectivity toward OATP1B activity in human subjects with genetic variants of OATP1B1. Its sensitivity to a broad spectrum of clinical OATP1B inhibitors has been established from weak to vigorous. Dose-dependent CP-I changes in healthy human subjects show agreement with DDI magnitudes of probe substrates by rifampin treatment. Physiologically based pharmacokinetic models have been established for concentration changes of plasma CP-I with OATP inhibitors, demonstrating the usefulness of supporting the quantitative translation of the effect of CP-I levels into the DDI risk assessment of potential OATP inhibitors. As plasma CP-I's sensitivity, specificity, and selectivity have been validated in humans, monitoring CP-I levels in single and multiple clinical phase I dose escalation studies is recommended for early assessment of DDI risks and understanding the full dose-response of an investigational drug to OATP inhibitions. A decision tree is proposed to preclude the need to conduct a dedicated DDI study by administering a probe substrate drug to human subjects. SIGNIFICANCE STATEMENT: The minireview summarized the validation paths of coproporphyrins I and III (CP-I and CP-III) as biomarkers of organic anion transporting polypeptide 1B (OATP1B) inhibition in humans for their selectivity, specificity, and sensitivity. The utility of monitoring CP-I to assess drug-drug interactions of OATP1B inhibition in early drug development is proposed. Changes in plasma CP-I in phase I dose range studies can be used to frame plans for late-stage development and facilitate the mechanistic understanding of complex drug-drug interactions.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| |
Collapse
|
15
|
Hou H, Horikawa M, Narita Y, Jono H, Kakizoe Y, Izumi Y, Kuwabara T, Mukoyama M, Saito H. Suppression of Indoxyl Sulfate Accumulation Reduces Renal Fibrosis in Sulfotransferase 1a1-Deficient Mice. Int J Mol Sci 2023; 24:11329. [PMID: 37511089 PMCID: PMC10380001 DOI: 10.3390/ijms241411329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Renal fibrosis is the final manifestation of chronic kidney disease (CKD); its prevention is vital for controlling CKD progression. Indoxyl sulfate (IS), a typical sulfate-conjugated uremic solute, is produced in the liver via the enzyme sulfotransferase (SULT) 1A1 and accumulates significantly during CKD. We investigated the toxicopathological role of IS in renal fibrosis using Sult1a1-KO mice and the underlying mechanisms. The unilateral ureteral obstruction (UUO) model was created; kidney IS concentrations, inflammation, and renal fibrosis were assessed on day 14. After UUO treatment, inflammation and renal fibrosis were exacerbated in WT mice, with an accumulation of IS in the kidney. However, they were significantly suppressed in Sult1a1-KO mice. CD206+ expression was upregulated, and β-catenin expression was downregulated in Sult1a1-KO mice. To confirm the impact of erythropoietin (EPO) on renal fibrosis, we evaluated the time-dependent expression of EPO. In Sult1a1-KO mice, EPO mRNA expression was improved considerably; UUO-induced renal fibrosis was further attenuated by recombinant human erythropoietin (rhEPO). Thus, UUO-induced renal fibrosis was alleviated in Sult1a1-KO mice with a decreased accumulation of IS. Our findings confirmed the pathological role of IS in renal fibrosis and identified SULT1A1 as a new therapeutic target enzyme for preventing and attenuating renal fibrosis.
Collapse
Affiliation(s)
- Huixian Hou
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Mai Horikawa
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Yuki Narita
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Hirofumi Jono
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Yutaka Kakizoe
- Department of Nephrology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Yuichiro Izumi
- Department of Nephrology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Hideyuki Saito
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| |
Collapse
|
16
|
Chen Y, Yang Y, Xu S, Wang C, Shu P, Zhang X, Huang Q, Kim JS, Jiao Z. Model Informed Development of SIM0295 in Patients with Gout and Hyperuricemia and Healthy Volunteers Using a Population Pharmacokinetics/ Pharmacodynamics Approach. Expert Opin Investig Drugs 2023:1-10. [PMID: 37183631 DOI: 10.1080/13543784.2023.2212153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND SIM0295, a novel inhibitor of human uric acid transporter 1 (hURAT1), is used to treat patients with gout and hyperuricemia. This study aimed to develop population pharmacokinetics and pharmacodynamics (popPK/PD) models of SIM0295 and explore potential covariates to inform clinical drug development. RESEARCH DESIGN AND METHODS Data were obtained from four phase I studies conducted in healthy Korean and Chinese subjects and two phase II studies conducted in Korean patients with gout and hyperuricemia. The popPK/PD model of SIM0295 was developed using nonlinear mixed effects modeling. RESULTS SIM0295 pharmacokinetics was described using a two-compartment model with the absorption of four transit compartments and first-order elimination. PK parameters were normalized to weight via allometric scaling. Food was identified as a factor significantly affecting the absorption rate, with no clinical relevance. The sigmoid Emax model with a semi-mechanism of inhibition of serum uric acid (sUA) reabsorption was used to describe the exposure-response relationship. Additionally, Monte Carlo simulations demonstrated that approimately 9 mg/day of SIM0295 for 7 days could achieve the maximum decrease in sUA. CONCLUSION The established popPK/PD model characterized the dose-exposure-response relationship for SIM0295 in healthy subjects and patients with gout and hyperuricemia and could be used to inform the drug development.
Collapse
Affiliation(s)
- Yueting Chen
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Yang
- Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Shansen Xu
- Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Chenyu Wang
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pan Shu
- Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Xiaoyu Zhang
- Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Qin Huang
- Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Jin Sook Kim
- JW Pharmaceutical Corporation, Seoul, Korea, Republic of South
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Jamshidi N, Nigam KB, Nigam SK. Loss of the Kidney Urate Transporter, Urat1, Leads to Disrupted Redox Homeostasis in Mice. Antioxidants (Basel) 2023; 12:antiox12030780. [PMID: 36979028 PMCID: PMC10045411 DOI: 10.3390/antiox12030780] [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: 01/15/2023] [Revised: 02/28/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
High uric acid is associated with gout, hypertension, metabolic syndrome, cardiovascular disease, and kidney disease. URAT1 (SLC22A12), originally discovered in mice as Rst, is generally considered a very selective uric acid transporter compared to other closely-related kidney uric acid transporters such as OAT1 (SLC22A6, NKT) and OAT3 (SLC22A8). While the role of URAT1 in regulating human uric acid is well-established, in recent studies the gene has been linked to redox regulation in flies as well as progression of renal cell carcinoma. We have now identified over twenty metabolites in the Urat1 knockout that are generally distinct from metabolites accumulating in the Oat1 and Oat3 knockout mice, with distinct molecular properties as revealed by chemoinformatics and machine learning analysis. These metabolites are involved in seemingly disparate aspects of cellular metabolism, including pyrimidine, fatty acid, and amino acid metabolism. However, through integrative systems metabolic analysis of the transcriptomic and metabolomic data using a human metabolic reconstruction to build metabolic genome-scale models (GEMs), the cellular response to loss of Urat1/Rst revealed compensatory processes related to reactive oxygen species handling and maintaining redox state balances via Vitamin C metabolism and cofactor charging reactions. These observations are consistent with the increasingly appreciated role of the antioxidant properties of uric acid. Collectively, the results highlight the role of Urat1/Rst as a transporter strongly tied to maintaining redox homeostasis, with implications for metabolic side effects from drugs that block its function.
Collapse
Affiliation(s)
- Neema Jamshidi
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA;
- Correspondence:
| | - Kabir B. Nigam
- Department of Psychiatry, Brigham and Women’s Hospital, Boston, MA 02130, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA 02130, USA
| | - Sanjay K. Nigam
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA;
- Departments of Pediatrics and Medicine (Nephrology), University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
18
|
Adomako EA, Maalouf NM. Type 4 renal tubular acidosis and uric acid nephrolithiasis: two faces of the same coin? Curr Opin Nephrol Hypertens 2023; 32:145-152. [PMID: 36683539 PMCID: PMC9881823 DOI: 10.1097/mnh.0000000000000859] [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] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW The present review summarizes findings of recent studies examining the epidemiology, pathophysiology, and treatment of type 4 renal tubular acidosis (RTA) and uric acid nephrolithiasis, two conditions characterized by an abnormally acidic urine. RECENT FINDINGS Both type 4 RTA and uric acid nephrolithiasis disproportionately occur in patients with type 2 diabetes and/or chronic kidney disease. Biochemically, both conditions are associated with reduced renal ammonium excretion resulting in impaired urinary buffering and low urine pH. Reduced ammoniagenesis is postulated to result from hyperkalemia in type 4 RTA and from insulin resistance and fat accumulation in the renal proximal tubule in uric acid nephrolithiasis. The typical biochemical findings of hyperkalemia and systemic acidosis of type 4 RTA are rarely reported in uric acid stone formers. Additional clinical differences between the two conditions include findings of higher urinary uric acid excretion and consequent urinary uric acid supersaturation in uric acid stone formers but not in type 4 RTA. SUMMARY Type 4 RTA and uric acid nephrolithiasis share several epidemiological, clinical, and biochemical features. Although both conditions may be manifestations of diabetes mellitus and thus have a large at-risk population, the means to the shared biochemical finding of overly acidic urine are different. This difference in pathophysiology may explain the dissimilarity in the prevalence of kidney stone formation.
Collapse
Affiliation(s)
- Emmanuel A. Adomako
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Naim M. Maalouf
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
19
|
Qi X, Ma Y, Guan K, Liu C, Wang R, Ma Y, Niu T. Whey protein peptide PEW attenuates hyperuricemia and associated renal inflammation in potassium oxonate and hypoxanthine-induced rat. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2022.102311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Granados JC, Watrous JD, Long T, Rosenthal SB, Cheng S, Jain M, Nigam SK. Regulation of Human Endogenous Metabolites by Drug Transporters and Drug Metabolizing Enzymes: An Analysis of Targeted SNP-Metabolite Associations. Metabolites 2023; 13:171. [PMID: 36837791 PMCID: PMC9958903 DOI: 10.3390/metabo13020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Drug transporters and drug-metabolizing enzymes are primarily known for their role in the absorption, distribution, metabolism, and excretion (ADME) of small molecule drugs, but they also play a key role in handling endogenous metabolites. Recent cross-tissue co-expression network analyses have revealed a "Remote Sensing and Signaling Network" of multispecific, oligo-specific, and monospecific transporters and enzymes involved in endogenous metabolism. This includes many proteins from families involved in ADME (e.g., SLC22, SLCO, ABCC, CYP, UGT). Focusing on the gut-liver-kidney axis, we identified the endogenous metabolites potentially regulated by this network of ~1000 proteins by associating SNPs in these genes with the circulating levels of thousands of small, polar, bioactive metabolites, including free fatty acids, eicosanoids, bile acids, and other signaling metabolites that act in part via G-protein coupled receptors (GPCRs), nuclear receptors, and kinases. We identified 77 genomic loci associated with 7236 unique metabolites. This included metabolites that were associated with multiple, distinct loci, indicating coordinated regulation between multiple genes (including drug transporters and drug-metabolizing enzymes) of specific metabolites. We analyzed existing pharmacogenomic data and noted SNPs implicated in endogenous metabolite handling (e.g., rs4149056 in SLCO1B1) also affecting drug ADME. The overall results support the existence of close relationships, via interactions with signaling metabolites, between drug transporters and drug-metabolizing enzymes that are part of the Remote Sensing and Signaling Network, and with GPCRs and nuclear receptors. These analyses highlight the potential for drug-metabolite interactions at the interfaces of the Remote Sensing and Signaling Network and the ADME protein network.
Collapse
Affiliation(s)
- Jeffry C. Granados
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Jeramie D. Watrous
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - Tao Long
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
| | - Susan Cheng
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mohit Jain
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - Sanjay K. Nigam
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
21
|
Granados JC, Ermakov V, Maity K, Vera DR, Chang G, Nigam SK. The kidney drug transporter OAT1 regulates gut microbiome-dependent host metabolism. JCI Insight 2023; 8:e160437. [PMID: 36692015 PMCID: PMC9977316 DOI: 10.1172/jci.insight.160437] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/30/2022] [Indexed: 01/24/2023] Open
Abstract
Organic anion transporter 1 (OAT1/SLC22A6, NKT) is a multispecific drug transporter in the kidney with numerous substrates, including pharmaceuticals, endogenous metabolites, natural products, and uremic toxins. Here, we show that OAT1 regulates levels of gut microbiome-derived metabolites. We depleted the gut microbiome of Oat1-KO and WT mice and performed metabolomics to analyze the effects of genotype (KO versus WT) and microbiome depletion. OAT1 is an in vivo intermediary between the host and the microbes, with 40 of the 162 metabolites dependent on the gut microbiome also impacted by loss of Oat1. Chemoinformatic analysis revealed that the altered metabolites (e.g., indoxyl sulfate, p-cresol sulfate, deoxycholate) had more ring structures and sulfate groups. This indicates a pathway from gut microbes to liver phase II metabolism, to renal OAT1-mediated transport. The idea that multiple gut-derived metabolites directly interact with OAT1 was confirmed by in vitro transport and magnetic bead binding assays. We show that gut microbiome-derived metabolites dependent on OAT1 are impacted in a chronic kidney disease (CKD) model and human drug-metabolite interactions. Consistent with the Remote Sensing and Signaling Theory, our results support the view that drug transporters (e.g., OAT1, OAT3, OATP1B1, OATP1B3, MRP2, MRP4, ABCG2) play a central role in regulating gut microbe-dependent metabolism, as well as interorganismal communication between the host and microbiome.
Collapse
Affiliation(s)
| | | | - Koustav Maity
- Skaggs School of Pharmacy and Pharmaceutical Sciences
| | - David R. Vera
- Department of Radiology
- In Vivo Cancer and Molecular Imaging Program
| | - Geoffrey Chang
- Skaggs School of Pharmacy and Pharmaceutical Sciences
- Department of Pharmacology, School of Medicine
| | - Sanjay K. Nigam
- Department of Pediatrics, and
- Department of Medicine (Nephrology), UCSD, La Jolla, California, USA
| |
Collapse
|
22
|
Nigam SK, Granados JC. OAT, OATP, and MRP Drug Transporters and the Remote Sensing and Signaling Theory. Annu Rev Pharmacol Toxicol 2023; 63:637-660. [PMID: 36206988 DOI: 10.1146/annurev-pharmtox-030322-084058] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The coordinated movement of organic anions (e.g., drugs, metabolites, signaling molecules, nutrients, antioxidants, gut microbiome products) between tissues and body fluids depends, in large part, on organic anion transporters (OATs) [solute carrier 22 (SLC22)], organic anion transporting polypeptides (OATPs) [solute carrier organic (SLCO)], and multidrug resistance proteins (MRPs) [ATP-binding cassette, subfamily C (ABCC)]. Depending on the range of substrates, transporters in these families can be considered multispecific, oligospecific, or (relatively) monospecific. Systems biology analyses of these transporters in the context of expression patterns reveal they are hubs in networks involved in interorgan and interorganismal communication. The remote sensing and signaling theory explains how the coordinated functions of drug transporters, drug-metabolizing enzymes, and regulatory proteins play a role in optimizing systemic and local levels of important endogenous small molecules. We focus on the role of OATs, OATPs, and MRPs in endogenous metabolism and how their substrates (e.g., bile acids, short chain fatty acids, urate, uremic toxins) mediate interorgan and interorganismal communication and help maintain and restore homeostasis in healthy and disease states.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Department of Pediatrics and Medicine (Nephrology), University of California San Diego, La Jolla, California, USA;
| | - Jeffry C Granados
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
23
|
Jamshidi N, Nigam SK. Drug transporters OAT1 and OAT3 have specific effects on multiple organs and gut microbiome as revealed by contextualized metabolic network reconstructions. Sci Rep 2022; 12:18308. [PMID: 36316339 PMCID: PMC9622871 DOI: 10.1038/s41598-022-21091-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022] Open
Abstract
In vitro and in vivo studies have established the organic anion transporters OAT1 (SLC22A6, NKT) and OAT3 (SLC22A8) among the main multi-specific "drug" transporters. They also transport numerous endogenous metabolites, raising the possibility of drug-metabolite interactions (DMI). To help understand the role of these drug transporters on metabolism across scales ranging from organ systems to organelles, a formal multi-scale analysis was performed. Metabolic network reconstructions of the omics-alterations resulting from Oat1 and Oat3 gene knockouts revealed links between the microbiome and human metabolism including reactions involving small organic molecules such as dihydroxyacetone, alanine, xanthine, and p-cresol-key metabolites in independent pathways. Interestingly, pairwise organ-organ interactions were also disrupted in the two Oat knockouts, with altered liver, intestine, microbiome, and skin-related metabolism. Compared to older models focused on the "one transporter-one organ" concept, these more sophisticated reconstructions, combined with integration of a multi-microbial model and more comprehensive metabolomics data for the two transporters, provide a considerably more complex picture of how renal "drug" transporters regulate metabolism across the organelle (e.g. endoplasmic reticulum, Golgi, peroxisome), cellular, organ, inter-organ, and inter-organismal scales. The results suggest that drugs interacting with OAT1 and OAT3 can have far reaching consequences on metabolism in organs (e.g. skin) beyond the kidney. Consistent with the Remote Sensing and Signaling Theory (RSST), the analysis demonstrates how transporter-dependent metabolic signals mediate organ crosstalk (e.g., gut-liver-kidney) and inter-organismal communication (e.g., gut microbiome-host).
Collapse
Affiliation(s)
- Neema Jamshidi
- grid.19006.3e0000 0000 9632 6718Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, CA USA ,grid.266100.30000 0001 2107 4242Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA USA
| | - Sanjay K. Nigam
- grid.266100.30000 0001 2107 4242Departments of Pediatrics and Medicine (Nephrology), University of California, San Diego, La Jolla, CA USA
| |
Collapse
|
24
|
Nigam SK, Granados JC. A Biological Basis for Pharmacokinetics: The Remote Sensing and Signaling Theory. Clin Pharmacol Ther 2022; 112:456-460. [PMID: 35598078 PMCID: PMC9398931 DOI: 10.1002/cpt.2634] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/30/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Sanjay K. Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093
| | - Jeffry C. Granados
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093
| |
Collapse
|
25
|
Giacomini KM, Huang SM. More Than Pharmacokinetics: Transporters in Clinical Pharmacology. Clin Pharmacol Ther 2022; 112:423-426. [PMID: 35989454 DOI: 10.1002/cpt.2710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Shiew Mei Huang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|