1
|
Moka MK, Rathakrishnan D, Sriram DK, George M. Advancing therapeutic approaches and shaping future perspectives in the management of autosomal dominant polycystic kidney disease (ADPKD): Insights from clinical trials. Eur J Pharmacol 2025; 997:177634. [PMID: 40246136 DOI: 10.1016/j.ejphar.2025.177634] [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: 12/17/2024] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is a genetically inherited disorder characterized by progressive renal cyst formation, leading to end-stage renal disease (ESRD). The disease causes a continuous decline in renal function, resulting in significantly enlarged kidneys that adversely affect the patient's quality of life (QOL). Despite the identification of causative gene mutations, no approved therapeutic intervention halts cystogenesis or prevents renal failure. OBJECTIVES This review aims to evaluate the potential of emerging pharmacological interventions and investigational treatments to decelerate cyst growth and disease progression in ADPKD, with a focus on clinical trial outcomes and regulatory advancements. METHODS Data from clinical trials (phases I-IV) were systematically reviewed to assess the effectiveness and safety of various therapeutic strategies for ADPKD. Mechanism-agnostic approaches, biomarker integration, and regulatory developments were also analyzed to understand their role in improving clinical management. RESULTS Clinical studies indicate that pharmacological interventions and investigational therapies show promise in slowing cyst growth and disease progression. The U.S. FDA's adoption of mechanism-agnostic strategies and biomarker-based approaches has enhanced the framework for managing ADPKD. However, genetic heterogeneity and disease complexity remain significant barriers. CONCLUSION While a definitive cure for ADPKD remains elusive, recent therapeutic advancements provide hope for improving disease management and patient outcomes. Continued innovation and research are essential to overcome challenges in cystogenesis prevention and develop curative strategies.
Collapse
Affiliation(s)
- Murali Krishna Moka
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India.
| | - Deepalaxmi Rathakrishnan
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India.
| | - D K Sriram
- Department of Diabetology and Endocrinology, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India.
| | - Melvin George
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, Chennai, 600045, Tamil Nadu, India.
| |
Collapse
|
2
|
Sethi J. Reassessing Metformin's Potential in Autosomal Dominant Polycystic Kidney Disease (ADPKD): A Call for Further Research. Indian J Nephrol 2025; 35:444-445. [PMID: 40352866 PMCID: PMC12065590 DOI: 10.25259/ijn_542_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 05/14/2025] Open
Affiliation(s)
- Jasmine Sethi
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
3
|
Venkatasubramanian V, Sethi J, Kumar V, Yadav AK, Lal A, Kohli HS. Metformin Versus Standard of Care in Patients with Autosomal Dominant Polycystic Kidney Disease - A Randomized Control Trial. Indian J Nephrol 2025; 35:410-416. [PMID: 40352872 PMCID: PMC12065615 DOI: 10.25259/ijn_100_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/04/2024] [Indexed: 05/14/2025] Open
Abstract
Background Autosomal dominant kidney disease (ADPKD) is the most common monogenic disorder leading to renal failure with limited therapeutic options. We aimed to assess the efficacy and safety of metformin in nondiabetic ADPKD patients and its role in slowing disease progression. Materials and Methods We conducted a prospective, randomized controlled, open labelled clinical trial and enrolled 52 nondiabetic adults aged 18-60 years with typical ADPKD, estimated glomerular filtration rate (eGFR) > 45 mL/min/m2, and no risk factors of rapid disease progression. Participants were randomized in a 1:1 ratio by a computer-generated random number table into metformin + standard of care group (metformin arm) and standard of care group (Control arm). Primary outcome of the study was to evaluate the effects of metformin versus control arm on the percentage and absolute change in eGFR over a 6-month period. Results Mean (SD) age of the cohort was 37.15 (10.16) years with half of them being females. The mean (SD) baseline htTKV and eGFR were 335.67 (153.3) mL/m and 100.23 (25.95) mL/min/m2, respectively. Clinical exome sequencing was available in nine (17.3%) patients of which two-thirds had PKD1 mutation. Baseline characteristics were distributed equally across randomized groups. Baseline proteinuria was significantly higher in the metformin arm (p = 0.014). The eGFR difference and percentage change in eGFR was not different between the groups at 6 months (p = 0.53 and 0.48, respectively). There was no statistically significant difference in htTKV and percentage change in htTKV at 6 months between the groups, although an increase in htTKV was numerically smaller in the metformin group (p = 0.769, 0.805). Blood pressure, body weight, body mass index (BMI), and proteinuria also did not differ between the two groups. Only half of the cohort tolerated the maximum dose of metformin. Around two-thirds of patients reported adverse effects, most commonly asthenia. Conclusion Metformin appears to be safe and well tolerated in nondiabetic patients with ADPKD.
Collapse
Affiliation(s)
| | - Jasmine Sethi
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Vivek Kumar
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashok Kumar Yadav
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anupam Lal
- Department of Radiodiagnosis, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Harbir Singh Kohli
- Department of Nephrology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
4
|
Patil AN, Singh MP, Rawat P, Sah S, Satapathy P. Challenges and Considerations in Metformin Use for ADPKD: A Commentary on Recent Findings. Indian J Nephrol 2025; 35:443. [PMID: 40352899 PMCID: PMC12065576 DOI: 10.25259/ijn_513_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 09/15/2024] [Indexed: 05/14/2025] Open
Affiliation(s)
- Amol N Patil
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Mahendra Pratap Singh
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Pramod Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Sanjit Sah
- Department of Pediatrics, Sanjeevani Hospital, Kalyanpur, Siraha, Nepal
| | - Prakasini Satapathy
- University Center for Research and Development, Chandigarh University, Punjab, India
| |
Collapse
|
5
|
Giblin J, Simon R, Zarate-Diaz J, Lee B, Chung EJ. Targeting Dysregulated Epigenetic Modifiers With Kidney-Targeted Nanotherapeutics for Polycystic Kidney Disease. J Biomed Mater Res A 2025; 113:e37909. [PMID: 40200735 DOI: 10.1002/jbm.a.37909] [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: 10/15/2024] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic kidney disease worldwide. The one small molecule drug available to patients, tolvaptan, is associated with off-target side effects and high discontinuation rates, necessitating the development of new therapeutic strategies. Previous work has shown that the epigenome is altered in ADPKD; however, the identification and targeting of dysregulated epigenetic modulators has yet to be explored for human ADPKD therapy. Using cells derived from cysts of ADPKD patients, we tested the gene expression of several epigenetic modulators. We found Brd4 and BMi1 are upregulated and observed that their inhibition using small molecule drugs, AZD-5153 and PTC-209, significantly slowed the proliferation of ADPKD patient cells. To enhance the delivery of AZD-5153 and PTC-209 to renal cells, we loaded the drugs into kidney-targeting micelles (KM) and assessed their therapeutic effects in vitro. Combining AZD-5153 and PTC-209 in KMs had a synergistic effect on reducing the proliferation in ADPKD patient cells and in a 3D PKD cyst model. These findings were also consistent in murine in vitro models using Pkd1 null renal proximal tubule cells. In summary, we demonstrate Brd4 and BMi1 as novel targets in ADPKD and targeting the epigenome using kidney nanomedicine as a novel therapeutic strategy in ADPKD.
Collapse
Affiliation(s)
- Joshua Giblin
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Rowan Simon
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Jose Zarate-Diaz
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Brenton Lee
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Eun Ji Chung
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
- Bridge Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Jdiaa SS, Mustafa RA, Yu ASL. Treatment of Autosomal-Dominant Polycystic Kidney Disease. Am J Kidney Dis 2025; 85:491-500. [PMID: 39424253 DOI: 10.1053/j.ajkd.2024.08.008] [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: 03/16/2024] [Revised: 07/17/2024] [Accepted: 08/07/2024] [Indexed: 10/21/2024]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is a chronic systemic disease that affects all races and ethnicities. It is the fourth leading cause of end-stage kidney disease, and it has a heterogenous phenotype ranging from mild to severe disease. Identifying patients with ADPKD who are at risk of rapid progression can guide therapeutic decisions. Several tools to predict disease severity are available, based on features such as total kidney volume assessed with magnetic resonance imaging, PKD genotype, estimated glomerular filtration rate (eGFR) trajectory, and the occurrence of hypertension and urologic complications early in life. During the past decade, more evidence has emerged regarding optimal ADPKD management. The HALT PKD (Halt Progression of Polycystic Kidney Disease) trial supported intensive blood pressure control in patients younger than 50 years of age with preserved kidney function. A healthy lifestyle, including maintaining a healthy weight, salt restriction, and smoking cessation, is likely to be beneficial. Tolvaptan, the only disease-modifying agent for patients with ADPKD at risk of rapid progression, is gaining wider use, but is still limited by its side effects. This is an exciting time for the ADPKD community because multiple promising interventions are in the pipeline and being investigated.
Collapse
Affiliation(s)
- Sara S Jdiaa
- Division of Nephrology, University of Toronto, Toronto, ON, Canada
| | - Reem A Mustafa
- Division of Nephrology and Hypertension and Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Alan S L Yu
- Division of Nephrology and Hypertension and Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
7
|
Kuo IC, Lin MY, Tsao YH, Chiu YW, Lee JJ. Metformin Use and Clinical Outcomes in Autosomal Dominant Polycystic Kidney Disease: A Nationwide Cohort Study. Biomedicines 2025; 13:635. [PMID: 40149611 PMCID: PMC11940305 DOI: 10.3390/biomedicines13030635] [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: 01/09/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Autosomal dominant polycystic kidney disease (ADPKD) is a progressive genetic disorder marked by bilateral renal cysts and extrarenal manifestations, ultimately resulting in renal failure. Emerging research indicates that metformin might influence the intracellular mechanisms of ADPKD, though its clinical significance remains uncertain. Methods: We applied the Taiwan National Health Insurance Database (NHIRD) to investigate the clinical impact of metformin utilization in ADPKD patients in real-world practice. The metformin user group was defined by more than 90 days of usage. To mitigate selection bias, we established a non-user group with a 1:2 ratio, matching for age, sex, and comorbidities by a propensity score matching method. Results: A total of 10,222 ADPKD cases were identified in the NHIRD between 2009 and 2018. After matching, the metformin user group was composed of 778 cases with a mean age of 59.5 ± 13.9 years, and the non-user group of 1546 cases with a mean age of 59.3 ± 14.4 years. The time from the index date to the outcome of ESKD in ADPKD was 5.3 ± 2.2 years in the metformin user group and 5.3 ± 2.3 years in the metformin non-user group, respectively. The metformin user group exhibited a significant reduction in the risk of end-stage kidney disease (ESKD), as indicated in the fully adjusted model (0.75, 95% CI 0.58-0.97, p = 0.03). A decreased risk of major adverse cardiovascular events (MACEs) was noted in metformin users, with an adjusted hazard ratio (HR) of 0.78 (95% CI 0.65-0.95, p = 0.01). Sensitivity analysis showed similar results by excluding late-stage CKD (CKD stage 5 or erythropoietin-stimulating agents use). Conclusions: Metformin usage in real-world practice showed lower hazards of ESKD and MACEs in patients with ADPKD, except for those with advanced CKD.
Collapse
Affiliation(s)
- I-Ching Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807017, Taiwan;
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807017, Taiwan; (M.-Y.L.); (Y.-W.C.)
| | - Ming-Yen Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807017, Taiwan; (M.-Y.L.); (Y.-W.C.)
| | - Yu-Hsiang Tsao
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807017, Taiwan;
- Division of Medical Statistics and Bioinformatics, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807017, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807017, Taiwan; (M.-Y.L.); (Y.-W.C.)
| | - Jia-Jung Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807017, Taiwan; (M.-Y.L.); (Y.-W.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807017, Taiwan
| |
Collapse
|
8
|
Devuyst O, Ahn C, Barten TR, Brosnahan G, Cadnapaphornchai MA, Chapman AB, Cornec-Le Gall E, Drenth JP, Gansevoort RT, Harris PC, Harris T, Horie S, Liebau MC, Liew M, Mallett AJ, Mei C, Mekahli D, Odland D, Ong AC, Onuchic LF, P-C Pei Y, Perrone RD, Rangan GK, Rayner B, Torra R, Mustafa R, Torres VE. KDIGO 2025 Clinical Practice Guideline for the Evaluation, Management, and Treatment of Autosomal Dominant Polycystic Kidney Disease (ADPKD). Kidney Int 2025; 107:S1-S239. [PMID: 39848759 DOI: 10.1016/j.kint.2024.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 01/25/2025]
|
9
|
Xu W, Zhu Y, Wang S, Liu J, Li H. From Adipose to Ailing Kidneys: The Role of Lipid Metabolism in Obesity-Related Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:1540. [PMID: 39765868 PMCID: PMC11727289 DOI: 10.3390/antiox13121540] [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: 10/31/2024] [Revised: 12/01/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
Obesity has emerged as a significant public health crisis, closely linked to the pathogenesis and progression of chronic kidney disease (CKD). This review explores the intricate relationship between obesity-induced lipid metabolism disorders and renal health. We discuss how excessive free fatty acids (FFAs) lead to lipid accumulation in renal tissues, resulting in cellular lipotoxicity, oxidative stress, and inflammation, ultimately contributing to renal injury. Key molecular mechanisms, including the roles of transcriptional regulators like PPARs and SREBP-1, are examined for their implications in lipid metabolism dysregulation. The review also highlights the impact of glomerular and tubular lipid overload on kidney pathology, emphasizing the roles of podocytes and tubular cells in maintaining kidney function. Various therapeutic strategies targeting lipid metabolism, including pharmacological agents such as statins and SGLT2 inhibitors, as well as lifestyle modifications, are discussed for their potential to mitigate CKD progression in obese individuals. Future research directions are suggested to better understand the mechanisms linking lipid metabolism to kidney disease and to develop personalized therapeutic approaches. Ultimately, addressing obesity-related lipid metabolism disorders may enhance kidney health and improve outcomes for individuals suffering from CKD.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuting Zhu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siyuan Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
- Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
10
|
St Pierre K, Cashmore BA, Bolignano D, Zoccali C, Ruospo M, Craig JC, Strippoli GF, Mallett AJ, Green SC, Tunnicliffe DJ. Interventions for preventing the progression of autosomal dominant polycystic kidney disease. Cochrane Database Syst Rev 2024; 10:CD010294. [PMID: 39356039 PMCID: PMC11445802 DOI: 10.1002/14651858.cd010294.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is the leading inherited cause of kidney disease. Clinical management has historically focused on symptom control and reducing associated complications. Improved understanding of the molecular and cellular mechanisms involved in kidney cyst growth and disease progression has resulted in new pharmaceutical agents targeting disease pathogenesis and preventing disease progression. However, the role of disease-modifying agents for all people with ADPKD is unclear. This is an update of a review first published in 2015. OBJECTIVES We aimed to evaluate the benefits and harms of interventions to prevent the progression of ADPKD and the safety based on patient-important endpoints, defined by the Standardised Outcomes in NephroloGy-Polycystic Kidney Disease (SONG-PKD) core outcome set, and general and specific adverse effects. SEARCH METHODS We searched the Cochrane Kidney and Transplants Register of Studies up to 13 August 2024 through contact with the Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal, and ClinicalTrials.gov. SELECTION CRITERIA Randomised controlled trials (RCTs) comparing any interventions for preventing the progression of ADPKD with other interventions, placebo, or standard care were considered for inclusion. DATA COLLECTION AND ANALYSIS Two authors independently assessed study risks of bias and extracted data. Summary estimates of effects were obtained using a random-effects model, and results were expressed as risk ratios (RR) and their 95% confidence intervals (CI) for dichotomous outcomes and mean difference (MD) or standardised mean difference (SMD) and 95% CI for continuous outcomes. Confidence in the evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS We included 57 studies (8016 participants) that investigated 18 pharmacological interventions (vasopressin 2 receptor (V2R) antagonists, antihypertensive therapy, mammalian target of rapamycin (mTOR) inhibitors, somatostatin analogues, antiplatelet agents, eicosapentaenoic acids, statins, kinase inhibitors, diuretics, anti-diabetic agents, water intake, dietary intervention, and supplements) in this review. Compared to placebo, the V2R antagonist tolvaptan probably preserves eGFR (3 studies, 2758 participants: MD 1.26 mL/min/1.73 m2, 95% CI 0.73 to 1.78; I2 = 0%) and probably slows total kidney volume (TKV) growth in adults (1 study, 1307 participants: MD -2.70 mL/cm, 95% CI -3.24 to -2.16) (moderate certainty evidence). However, there was insufficient evidence to determine tolvaptan's impact on kidney failure and death. There may be no difference in serious adverse events; however, treatment probably increases nocturia, fatigue and liver enzymes, may increase dry mouth and thirst, and may decrease hypertension and urinary and upper respiratory tract infections. Data on the impact of other therapeutic interventions were largely inconclusive. Compared to placebo, somatostatin analogues probably decrease TKV (6 studies, 500 participants: SMD -0.33, 95% CI -0.51 to -0.16; I2 = 11%), probably have little or no effect on eGFR (4 studies, 180 participants: MD 4.11 mL/min/1.73 m3, 95% CI -3.19 to 11.41; I2 = 0%) (moderate certainty evidence), and may have little or no effect on kidney failure (2 studies, 405 participants: RR 0.64, 95% CI 0.16 to 2.49; I2 = 39%; low certainty evidence). Serious adverse events may increase (2 studies, 405 participants: RR 1.81, 95% CI 1.01 to 3.25; low certainty evidence). Somatostatin analogues probably increase alopecia, diarrhoea or abnormal faeces, dizziness and fatigue but may have little or no effect on anaemia or infection. The effect on death is unclear. Targeted low blood pressure probably results in a smaller per cent annual increase in TKV (1 study, 558 participants: MD -1.00, 95% CI -1.67 to -0.33; moderate certainty evidence) compared to standard blood pressure targets, had uncertain effects on death, but probably do not impact other outcomes such as change in eGFR or adverse events. Kidney failure was not reported. Data comparing antihypertensive agents, mTOR inhibitors, eicosapentaenoic acids, statins, vitamin D compounds, metformin, trichlormethiazide, spironolactone, bosutinib, curcumin, niacinamide, prescribed water intake and antiplatelet agents were sparse and inconclusive. An additional 23 ongoing studies were also identified, including larger phase III RCTs, which will be assessed in a future update of this review. AUTHORS' CONCLUSIONS Although many interventions have been investigated in patients with ADPKD, at present, there is little evidence that they improve patient outcomes. Tolvaptan is the only therapeutic intervention that has demonstrated the ability to slow disease progression, as assessed by eGFR and TKV change. However, it has not demonstrated benefits for death or kidney failure. In order to confirm the role of other therapeutic interventions in ADPKD management, large RCTs focused on patient-centred outcomes are needed. The search identified 23 ongoing studies, which may provide more insight into the role of specific interventions.
Collapse
Affiliation(s)
- Kitty St Pierre
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Pharmacy Department, Gold Coast University Hospital, Gold Coast, Australia
| | - Brydee A Cashmore
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Davide Bolignano
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Carmine Zoccali
- Institute of Clinical Physiology, CNR - Italian National Council of Research, Reggio Calabria, Italy
| | - Marinella Ruospo
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Jonathan C Craig
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Giovanni Fm Strippoli
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Andrew J Mallett
- Department of Renal Medicine, Townsville Hospital and Health Service, Townsville, Australia
- Australasian Kidney Trials Network, The University of Queensland, Herston, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Suetonia C Green
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - David J Tunnicliffe
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| |
Collapse
|
11
|
Rosati E, Condello G, Tacente C, Mariani I, Tommolini V, Calvaruso L, Fulignati P, Grandaliano G, Pesce F. Potential Add-On Benefits of Dietary Intervention in the Treatment of Autosomal Dominant Polycystic Kidney Disease. Nutrients 2024; 16:2582. [PMID: 39203719 PMCID: PMC11357151 DOI: 10.3390/nu16162582] [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: 06/17/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of renal failure. The pathogenesis of the disease encompasses several pathways and metabolic alterations, including the hyperactivation of mTOR and suppression of AMPK signaling pathways, as well as mitochondrial dysfunction. This metabolic reprogramming makes epithelial cyst-lining cells highly dependent on glucose for energy and unable to oxidize fatty acids. Evidence suggests that high-carbohydrate diets may worsen the progression of ADPKD, providing the rationale for treating ADPKD patients with calorie restriction and, in particular, with ketogenic dietary interventions, already used for other purposes such as in overweight/obese patients or in the treatment of refractory epilepsy in children. Preclinical studies have demonstrated that calorie restriction may prevent and/or slow disease progression by inducing ketosis, particularly through increased beta-hydroxybutyrate (BHB) levels, which may modulate the metabolic signaling pathways altered in ADKPK. In these patients, although limited, ketogenic intervention studies have shown promising beneficial effects. However, larger and longer randomized controlled trials are needed to confirm their tolerability and safety in long-term maintenance and their additive role in the therapy of polycystic kidney disease.
Collapse
Affiliation(s)
- Erica Rosati
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giulia Condello
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Chiara Tacente
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Ilaria Mariani
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Tommolini
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Luca Calvaruso
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Pierluigi Fulignati
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giuseppe Grandaliano
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (E.R.); (G.C.); (C.T.); (I.M.); (V.T.); (L.C.); (P.F.); (G.G.)
- Unità Operativa Complessa di Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Pesce
- Division of Renal Medicine, Ospedale Isola Tiberina—Gemelli Isola, 00186 Rome, Italy
| |
Collapse
|
12
|
Wang W, You Z, Steele CN, Gitomer B, Chonchol M, Nowak KL. Changes in tubular biomarkers with dietary intervention and metformin in patients with autosomal dominant polycystic kidney disease: a post-hoc analysis of two clinical trials. BMC Nephrol 2024; 25:206. [PMID: 38918734 PMCID: PMC11200847 DOI: 10.1186/s12882-024-03643-6] [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: 01/15/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Tubular biomarkers, which reflect tubular dysfunction or injury, are associated with incident chronic kidney disease and kidney function decline. Several tubular biomarkers have also been implicated in the progression of autosomal dominant polycystic kidney disease (ADPKD). We evaluated changes in multiple tubular biomarkers in four groups of patients with ADPKD who participated in one of two clinical trials (metformin therapy and diet-induced weight loss), based on evidence suggesting that such interventions could reduce tubule injury. METHODS 66 participants (26 M/40 F) with ADPKD and an estimated glomerular filtration rate (eGFR) ≥ 30 ml/min/1.73m2 who participated in either a metformin clinical trial (n = 22 metformin; n = 23 placebo) or dietary weight loss study (n = 10 daily caloric restriction [DCR]; n = 11 intermittent fasting [IMF]) were included in assessments of urinary tubular biomarkers (kidney injury molecule-1 [KIM-1], fatty-acid binding protein [FABP], interleukin-18 [IL-18], monocyte chemoattractant protein-1 [MCP-1], neutrophil gelatinase-associated lipocalin [NGAL], clusterin, and human cartilage glycoprotein-40 [YKL-40]; normalized to urine creatinine), at baseline and 12 months. The association of baseline tubular biomarkers with both baseline and change in height-adjusted total kidney volume (HtTKV; percent change from baseline to 12 months) and estimated glomerular filtration rate (eGFR; absolute change at 12 months vs. baseline), with covariate adjustment, was also assessed using multiple linear regression. RESULTS Mean ± s.d. age was 48 ± 8 years, eGFR was 71 ± 16 ml/min/1.73m2, and baseline BMI was 30.5 ± 5.9 kg/m2. None of the tubular biomarkers changed with any intervention as compared to placebo. Additionally, baseline tubular biomarkers were not associated with either baseline or change in eGFR or HtTKV over 12 months, after adjustments for demographics, group assignment, and clinical characteristics. CONCLUSIONS Tubular biomarkers did not change with dietary-induced weight loss or metformin, nor did they associate with kidney disease progression, in this cohort of patients with ADPKD.
Collapse
Affiliation(s)
- Wei Wang
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zhiying You
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Michel Chonchol
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen L Nowak
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
13
|
El-Damanawi R, Stanley IK, Staatz C, Pascoe EM, Craig JC, Johnson DW, Mallett AJ, Hawley CM, Milanzi E, Hiemstra TF, Viecelli AK. Metformin for preventing the progression of chronic kidney disease. Cochrane Database Syst Rev 2024; 6:CD013414. [PMID: 38837240 PMCID: PMC11152183 DOI: 10.1002/14651858.cd013414.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
BACKGROUND Metformin has been used in the management of diabetes for decades. It is an effective, low-cost intervention with a well-established safety profile. Emerging evidence suggests that metformin targets a number of pathways that lead to chronic kidney damage, and long-term use may, therefore, slow the rate of kidney function decline and chronic kidney disease (CKD) progression. OBJECTIVES To evaluate the effect of metformin therapy on kidney function decline in patients with CKD with or without diabetes mellitus and assess the safety and dose tolerability in this population. SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies up to 19 July 2023 with assistance from an Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA We included randomised controlled trials (RCTs) that reported kidney-related outcomes with a minimum duration of 12 months delivery of the metformin intervention and whose eligibility criteria included adult participants with either i) a diagnosis of CKD of any aetiology and/or ii) those with a diagnosis of diabetes mellitus. Comparisons included placebo, no intervention, non-pharmacological interventions, other antidiabetic medications or any other active control. Studies that included patients on any modality of kidney replacement therapy were excluded. DATA COLLECTION AND ANALYSIS Two authors independently carried out data extraction using a standard data extraction form. The methodological quality of the included studies was assessed using the Cochrane risk of bias tool. Summary estimates of effect were obtained using a random-effects model, and results were expressed as risk ratios (RR) and their 95% confidence intervals (CI) for dichotomous outcomes and mean difference (MD) and 95% CI for continuous outcomes. Confidence in the evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS This review included 11 studies reporting on 8449 randomised participants. Studies were conducted in patient populations with Autosomal Dominant Polycystic Kidney Disease (ADPKD) (four studies) or diabetes mellitus (seven studies). Six studies compared metformin with no active control, four studies compared metformin with active controls (rosiglitazone, glyburide, pioglitazone, or glipizide), and one study included treatment arms that randomised to either metformin, diet and lifestyle modifications, or other antidiabetic therapies. The risk of bias in included studies varied; two studies were abstract-only publications and were judged to have a high risk of bias in most domains. Other included publications were judged to have a low risk of bias in most domains. Across comparisons, GRADE evaluations for most outcomes were judged as low or very low certainty, except for those relating to side effects, tolerance, and withdrawals, which were judged as moderate certainty. The evidence suggests that compared to placebo, metformin may result in i) a slightly smaller decline in kidney function (3 studies, 505 participants: MD 1.92 mL/min, 95% CI 0.33 to 3.51; I2 = 0%; low certainty), ii) very uncertain effects on the incidence of kidney failure (1 study, 753 participants: RR 1.20, 95% CI 0.17 to 8.49), iii) little or no effect on death (3 studies, 865 participants: RR 1.00, 95% CI 0.76 to 1.32; I2 = 0%; moderate certainty), iv) little or no effect on the incidence of serious adverse events (3 studies, 576 participants: RR 1.15, 95% CI 0.76 to 1.72; I2 = 0%; moderate certainty), and v) likely higher incidence of intolerance leading to study withdrawal than placebo (4 studies, 646 participants: RR 2.19, 95% CI 1.46 to 3.27; I2 = 0%; moderate certainty). The certainty of the evidence for proteinuria was very uncertain. Compared to other active controls (rosiglitazone, glyburide, pioglitazone, or glipizide), metformin i) demonstrated very uncertain effects on kidney function decline, ii) may result in little or no difference in death (3 studies, 5608 participants: RR 0.95 95% CI 0.63 to 1.43; I2 = 0%; low certainty), iii) probably results in little or no difference in intolerance leading to study withdrawal (3 studies, 5593 participants: RR 0.92, 95% CI, 0.79 to 1.08; I2 = 0%; moderate certainty), iv) probably results in little or no difference in the incidence of serious adverse events (2 studies, 5545 participants: RR 1.16, 95% CI 0.79 to 1.71; I2 = 0%; moderate certainty), and v) may increase the urinary albumin-creatinine ratio (2 studies, 3836 participants: MD 14.61, 95% CI 8.17 to 21.05; I2 = 0%; low certainty). No studies reported the incidence of kidney failure. AUTHORS' CONCLUSIONS This review highlights the lack of RCTs reporting on the effects of metformin on kidney function, particularly in patients with CKD. Future research in this field requires adequately powered RCTs comparing metformin to placebo or standard care in those with CKD. Seven ongoing studies were identified in this review, and future updates, including their findings, may further inform the results of this review.
Collapse
Affiliation(s)
| | | | - Christine Staatz
- School of Pharmacy, The University of Queensland, Brisbane, Australia
| | - Elaine M Pascoe
- Centre for Health Services Research, The University of Queensland, Brisbane, Australia
| | - Jonathan C Craig
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - David W Johnson
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, Australia
- Australasian Kidney Trials Network, The University of Queensland, Herston, Australia
- Translational Research Institute, Brisbane, Australia
| | - Andrew J Mallett
- Australasian Kidney Trials Network, The University of Queensland, Herston, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia
- Department of Renal Medicine, Townsville Hospital & Health Service, Townsville, Australia
| | - Carmel M Hawley
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, Australia
- Australasian Kidney Trials Network, The University of Queensland, Herston, Australia
- Translational Research Institute, Brisbane, Australia
| | - Elasma Milanzi
- School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Thomas F Hiemstra
- Cambridge Clinical Trials Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Andrea K Viecelli
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, Australia
- Australasian Kidney Trials Network, The University of Queensland, Herston, Australia
| |
Collapse
|
14
|
Stanley IK, Palma AM, Viecelli AK, Johnson DW, Hawley CM, Staatz CE, Mallett AJ. A secondary analysis of concurrent use of metformin and tolvaptan in ADPKD tolvaptan trials. J Nephrol 2024; 37:1417-1419. [PMID: 38512373 PMCID: PMC11405464 DOI: 10.1007/s40620-024-01906-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/15/2024] [Indexed: 03/23/2024]
Affiliation(s)
- I Kitty Stanley
- Centre for Health Services Research, The University of Queensland, Brisbane, QLD, Australia.
| | - Anton M Palma
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, USA
| | - Andrea K Viecelli
- Centre for Health Services Research, The University of Queensland, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, Australia
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Australia
| | - David W Johnson
- Centre for Health Services Research, The University of Queensland, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, Australia
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Australia
| | - Carmel M Hawley
- Centre for Health Services Research, The University of Queensland, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, Australia
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Australia
| | | | - Andrew J Mallett
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Australia.
- Department of Renal Medicine, Townsville University Hospital, Townsville, Australia.
- College of Medicine and Dentistry, James Cook University, Townsville, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
15
|
Brownjohn PW, Zoufir A, O’Donovan DJ, Sudhahar S, Syme A, Huckvale R, Porter JR, Bange H, Brennan J, Thompson NT. Computational drug discovery approaches identify mebendazole as a candidate treatment for autosomal dominant polycystic kidney disease. Front Pharmacol 2024; 15:1397864. [PMID: 38846086 PMCID: PMC11154008 DOI: 10.3389/fphar.2024.1397864] [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: 03/08/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a rare genetic disorder characterised by numerous renal cysts, the progressive expansion of which can impact kidney function and lead eventually to renal failure. Tolvaptan is the only disease-modifying drug approved for the treatment of ADPKD, however its poor side effect and safety profile necessitates the need for the development of new therapeutics in this area. Using a combination of transcriptomic and machine learning computational drug discovery tools, we predicted that a number of existing drugs could have utility in the treatment of ADPKD, and subsequently validated several of these drug predictions in established models of disease. We determined that the anthelmintic mebendazole was a potent anti-cystic agent in human cellular and in vivo models of ADPKD, and is likely acting through the inhibition of microtubule polymerisation and protein kinase activity. These findings demonstrate the utility of combining computational approaches to identify and understand potential new treatments for traditionally underserved rare diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hester Bange
- Crown Bioscience Netherlands B.V., Biopartner Center Leiden JH, Leiden, Netherlands
| | | | | |
Collapse
|
16
|
Geng Y, Wang Z, Xu X, Sun X, Dong X, Luo Y, Sun X. Extensive therapeutic effects, underlying molecular mechanisms and disease treatment prediction of Metformin: a systematic review. Transl Res 2024; 263:73-92. [PMID: 37567440 DOI: 10.1016/j.trsl.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Metformin (Met), a first-line management for type 2 diabetes mellitus, has been expansively employed and studied with results indicating its therapeutic potential extending beyond glycemic control. Beyond its established role, this therapeutic drug demonstrates a broad spectrum of action encompassing over 60 disorders, encompassing metabolic conditions, inflammatory disorders, carcinomas, cardiovascular diseases, and cerebrovascular pathologies. There is clear evidence of Met's action targeting specific nodes in the molecular pathways of these diseases and, intriguingly, interactions with the intestinal microbiota and epigenetic processes have been explored. Furthermore, novel Met derivatives with structural modifications tailored to diverse diseases have been synthesized and assessed. This manuscript proffers a comprehensive thematic review of the diseases amenable to Met treatment, elucidates their molecular mechanisms, and employs informatics technology to prospect future therapeutic applications of Met. These data and insights gleaned considerably contribute to enriching our understanding and appreciation of Met's far-reaching clinical potential and therapeutic applicability.
Collapse
Affiliation(s)
- Yifei Geng
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Zhen Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Xiaoyu Xu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Xiao Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Xi Dong
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China.
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Diabetes Research Center, Chinese Academy of Medical Sciences, China; Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, China.
| |
Collapse
|
17
|
Cukoski S, Lindemann CH, Arjune S, Todorova P, Brecht T, Kühn A, Oehm S, Strubl S, Becker I, Kämmerer U, Torres JA, Meyer F, Schömig T, Hokamp NG, Siedek F, Gottschalk I, Benzing T, Schmidt J, Antczak P, Weimbs T, Grundmann F, Müller RU. Feasibility and impact of ketogenic dietary interventions in polycystic kidney disease: KETO-ADPKD-a randomized controlled trial. Cell Rep Med 2023; 4:101283. [PMID: 37935200 PMCID: PMC10694658 DOI: 10.1016/j.xcrm.2023.101283] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/21/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023]
Abstract
Ketogenic dietary interventions (KDIs) are beneficial in animal models of autosomal-dominant polycystic kidney disease (ADPKD). KETO-ADPKD, an exploratory, randomized, controlled trial, is intended to provide clinical translation of these findings (NCT04680780). Sixty-six patients were randomized to a KDI arm (ketogenic diet [KD] or water fasting [WF]) or the control group. Both interventions induce significant ketogenesis on the basis of blood and breath acetone measurements. Ninety-five percent (KD) and 85% (WF) report the diet as feasible. KD leads to significant reductions in body fat and liver volume. Additionally, KD is associated with reduced kidney volume (not reaching statistical significance). Interestingly, the KD group exhibits improved kidney function at the end of treatment, while the control and WF groups show a progressive decline, as is typical in ADPKD. Safety-relevant events are largely mild, expected (initial flu-like symptoms associated with KD), and transient. Safety assessment is complemented by nuclear magnetic resonance (NMR) lipid profile analyses.
Collapse
Affiliation(s)
- Sadrija Cukoski
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christoph Heinrich Lindemann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Sita Arjune
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Center for Rare Diseases Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases, Cologne, Germany
| | - Polina Todorova
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Theresa Brecht
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Adrian Kühn
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Simon Oehm
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Sebastian Strubl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Ingrid Becker
- Institute of Medical Statistics and Computational Biology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Ulrike Kämmerer
- Department of Obstetrics and Gynecology, University Hospital of Würzburg, Würzburg, Germany
| | - Jacob Alexander Torres
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Franziska Meyer
- University of Cologne, Faculty of Medicine and University Hospital, Institute of Diagnostic and Interventional Radiology, Cologne, Germany
| | - Thomas Schömig
- University of Cologne, Faculty of Medicine and University Hospital, Institute of Diagnostic and Interventional Radiology, Cologne, Germany
| | - Nils Große Hokamp
- University of Cologne, Faculty of Medicine and University Hospital, Institute of Diagnostic and Interventional Radiology, Cologne, Germany
| | - Florian Siedek
- University of Cologne, Faculty of Medicine and University Hospital, Institute of Diagnostic and Interventional Radiology, Cologne, Germany
| | - Ingo Gottschalk
- University of Cologne, Faculty of Medicine and University Hospital, Division of Prenatal Medicine, Department of Obstetrics and Gynecology, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Center for Rare Diseases Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases, Cologne, Germany
| | - Johannes Schmidt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Bonacci GmbH, Cologne, Germany
| | - Philipp Antczak
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases, Cologne, Germany
| | - Thomas Weimbs
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Franziska Grundmann
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Center for Rare Diseases Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases, Cologne, Germany.
| |
Collapse
|
18
|
Chen LC, Chu YC, Lu T, Lin HYH, Chan TC. Cardiometabolic comorbidities in autosomal dominant polycystic kidney disease: a 16-year retrospective cohort study. BMC Nephrol 2023; 24:333. [PMID: 37946153 PMCID: PMC10637020 DOI: 10.1186/s12882-023-03382-0] [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: 03/20/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Autosomal-dominant polycystic kidney disease (ADPKD) is the most prevalent hereditary kidney disease and the fourth leading cause of end-stage renal disease (ESRD) requiring renal replacement therapy (RRT). Nevertheless, there is a paucity of epidemiological research examining the risk factors and survival on RRT for ADPKD. Thus, we aimed to investigate the cumulative effects of cardiometabolic comorbidities, including hypertension (HTN), type 2 diabetes mellitus (DM), and dyslipidemia (DLP) to clinical outcomes in ADPKD. METHODS We identified 6,142 patients with ADPKD aged ≥ 20 years from 2000 to 2015 using a nationwide population-based database. HTN, DM, and DLP diagnoses before or at the time of ADPKD diagnosis and different combinations of the three diagnoses were used as the predictors for the outcomes. Survival analyses were used to estimate the adjusted mortality risk from cardiometabolic comorbidities and the risk for renal survival. RESULTS Patients with ADPKD who developed ESRD had the higher all-cause mortality (HR, 5.14; [95% CI: 3.88-6.80]). Patients with all three of the diseases had a significantly higher risk of entering ESRD (HR:4.15, [95% CI:3.27-5.27]), followed by those with HTN and DM (HR:3.62, [95% CI:2.82-4.65]), HTN and DLP (HR:3.54, [95% CI:2.91-4.31]), and HTN alone (HR:3.10, [95% CI:2.62-3.66]) compared with those without any three cardiometabolic comorbidities. CONCLUSIONS Our study discovered the cumulative effect of HTN, DM, and DLP on the risk of developing ESRD, which reinforces the urgency of proactive prevention of cardiometabolic comorbidities to improve renal outcomes and overall survival in ADPKD patients.
Collapse
Affiliation(s)
- Li-Chi Chen
- Research Center for Humanities and Social Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Yi-Chi Chu
- Research Center for Humanities and Social Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Tzongshi Lu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hugo Y-H Lin
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, No.68, Jhonghua 3rd Road, Cianjin, Kaohsiung, 807, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Ta-Chien Chan
- Research Center for Humanities and Social Sciences, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan.
- Institute of Public Health, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
19
|
Oto OA, Atwood DJ, Chaudhary A, He Z, Li AS, Wempe MF, Edelstein CL. Metformin does not slow cyst growth in the PCK rat model of polycystic kidney disease. Physiol Rep 2023; 11:e15776. [PMID: 37653564 PMCID: PMC10471794 DOI: 10.14814/phy2.15776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 09/02/2023] Open
Abstract
Metformin (MET) has the potential to activate p-AMPK and block mTORC1-induced proliferation of tubular cells in PKD kidneys. The aim of this study was to determine the effects of MET on cyst growth, kidney function, AMPK and mTOR signaling, and lactate levels in male PCK rats, a Pkhd1 gene mutation model of human autosomal recessive polycystic kidney disease (ARPKD). MET 300 mg/kg/day IP from days 28 to 84 of age resulted in a mean serum metformin level that was 10 times the upper limit of therapeutic, no effect on cyst indices, nephrotoxicity, and increased serum lactate. MET 150 mg/kg resulted in a therapeutic serum metformin level but had no effect on kidney weight, cyst indices, kidney function, or mTOR and autophagy proteins. In summary, a standard dose of MET was ineffective in reducing PKD, did not activate p-AMPK or suppress mTOR and the higher dose resulted in increased lactate levels and nephrotoxicity. In conclusion, the study dampens enthusiasm for human studies of MET in PKD. Doubling the metformin dose resulted in a 10-fold increase in mean blood levels and toxicity suggesting that the dosage range between therapeutic and toxic is narrow.
Collapse
Affiliation(s)
- Ozgur A. Oto
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Daniel J. Atwood
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Anjana Chaudhary
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Zhibin He
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Amy S. Li
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Michael F. Wempe
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Charles L. Edelstein
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
20
|
Sieben CJ, Harris PC. Experimental Models of Polycystic Kidney Disease: Applications and Therapeutic Testing. KIDNEY360 2023; 4:1155-1173. [PMID: 37418622 PMCID: PMC10476690 DOI: 10.34067/kid.0000000000000209] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
Polycystic kidney diseases (PKDs) are genetic disorders characterized by the formation and expansion of numerous fluid-filled renal cysts, damaging normal parenchyma and often leading to kidney failure. Although PKDs comprise a broad range of different diseases, with substantial genetic and phenotypic heterogeneity, an association with primary cilia represents a common theme. Great strides have been made in the identification of causative genes, furthering our understanding of the genetic complexity and disease mechanisms, but only one therapy so far has shown success in clinical trials and advanced to US Food and Drug Administration approval. A key step in understanding disease pathogenesis and testing potential therapeutics is developing orthologous experimental models that accurately recapitulate the human phenotype. This has been particularly important for PKDs because cellular models have been of limited value; however, the advent of organoid usage has expanded capabilities in this area but does not negate the need for whole-organism models where renal function can be assessed. Animal model generation is further complicated in the most common disease type, autosomal dominant PKD, by homozygous lethality and a very limited cystic phenotype in heterozygotes while for autosomal recessive PKD, mouse models have a delayed and modest kidney disease, in contrast to humans. However, for autosomal dominant PKD, the use of conditional/inducible and dosage models have resulted in some of the best disease models in nephrology. These have been used to help understand pathogenesis, to facilitate genetic interaction studies, and to perform preclinical testing. Whereas for autosomal recessive PKD, using alternative species and digenic models has partially overcome these deficiencies. Here, we review the experimental models that are currently available and most valuable for therapeutic testing in PKD, their applications, success in preclinical trials, advantages and limitations, and where further improvements are needed.
Collapse
Affiliation(s)
- Cynthia J Sieben
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
21
|
Hanna C, Iliuta IA, Besse W, Mekahli D, Chebib FT. Cystic Kidney Diseases in Children and Adults: Differences and Gaps in Clinical Management. Semin Nephrol 2023; 43:151434. [PMID: 37996359 DOI: 10.1016/j.semnephrol.2023.151434] [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] [Indexed: 11/25/2023]
Abstract
Cystic kidney diseases, when broadly defined, have a wide differential diagnosis extending from recessive diseases with a prenatal or pediatric diagnosis, to the most common autosomal-dominant polycystic kidney disease primarily affecting adults, and several other genetic or acquired etiologies that can manifest with kidney cysts. The most likely diagnoses to consider when assessing a patient with cystic kidney disease differ depending on family history, age stratum, radiologic characteristics, and extrarenal features. Accurate identification of the underlying condition is crucial to estimate the prognosis and initiate the appropriate management, identification of extrarenal manifestations, and counseling on recurrence risk in future pregnancies. There are significant differences in the clinical approach to investigating and managing kidney cysts in children compared with adults. Next-generation sequencing has revolutionized the diagnosis of inherited disorders of the kidney, despite limitations in access and challenges in interpreting the data. Disease-modifying treatments are lacking in the majority of kidney cystic diseases. For adults with rapid progressive autosomal-dominant polycystic kidney disease, tolvaptan (V2-receptor antagonist) has been approved to slow the rate of decline in kidney function. In this article, we examine the differences in the differential diagnosis and clinical management of cystic kidney disease in children versus adults, and we highlight the progress in molecular diagnostics and therapeutics, as well as some of the gaps meriting further attention.
Collapse
Affiliation(s)
- Christian Hanna
- Division of Pediatric Nephrology and Hypertension, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN; Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN.
| | - Ioan-Andrei Iliuta
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Whitney Besse
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Djalila Mekahli
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL.
| |
Collapse
|
22
|
Dachy A, Van Loo L, Mekahli D. Autosomal Dominant Polycystic Kidney Disease in Children and Adolescents: Assessing and Managing Risk of Progression. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:236-244. [PMID: 37088526 DOI: 10.1053/j.akdh.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/07/2023] [Accepted: 01/19/2023] [Indexed: 04/25/2023]
Abstract
The clinical management of autosomal dominant polycystic kidney disease (ADPKD) in adults has shifted from managing complications to delaying disease progression through newly emerging therapies. Regarding pediatric management of the disease, there are still specific hurdles related to the management of children and adolescents with ADPKD and, unlike adults, there are no specific therapies for pediatric ADPKD or stratification models to identify children and young adults at risk of rapid decline in kidney function. Therefore, early identification and management of factors that may modify disease progression, such as hypertension and obesity, are of most importance for young children with ADPKD. Many of these risk factors could promote disease progression in both ADPKD and chronic kidney disease. Hence, nephroprotective measures applied early in life can represent a window of opportunity to prevent the decline of the glomerular filtration rate especially in young patients with ADPKD. In this review, we highlight current challenges in the management of patients with pediatric ADPKD, the importance of early modifying factors in disease progression as well as the gaps and future perspectives in the pediatric ADPKD research field.
Collapse
Affiliation(s)
- Angélique Dachy
- PKD Research Group, Department of Cellular and MoleculMedar icine, KU Leuven, Leuven, Belgium; Department of Pediatrics, ULiège Academic Hospital, Liège, Belgium; Laboratory of Translational Research in Nephrology (LTRN), GIGA Cardiovascular Sciences, ULiège, Liège, Belgium
| | - Liselotte Van Loo
- Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium.
| | - Djalila Mekahli
- PKD Research Group, Department of Cellular and MoleculMedar icine, KU Leuven, Leuven, Belgium; Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
23
|
Zhou JX, Torres VE. Autosomal Dominant Polycystic Kidney Disease Therapies on the Horizon. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:245-260. [PMID: 37088527 DOI: 10.1053/j.akdh.2023.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 04/25/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the formation of numerous kidney cysts which leads to kidney failure. ADPKD is responsible for approximately 10% of patients with kidney failure. Overwhelming evidence supports that vasopressin and its downstream cyclic adenosine monophosphate signaling promote cystogenesis, and targeting vasopressin 2 receptor with tolvaptan and other antagonists ameliorates cyst growth in preclinical studies. Tolvaptan is the only drug approved by Food and Drug Administration to treat ADPKD patients at the risk of rapid disease progression. A major limitation of the widespread use of tolvaptan is aquaretic events. This review discusses the potential strategies to improve the tolerability of tolvaptan, the progress on the use of an alternative vasopressin 2 receptor antagonist lixivaptan, and somatostatin analogs. Recent advances in understanding the pathophysiology of PKD have led to new approaches of treatment via targeting different signaling pathways. We review the new pharmacotherapies and dietary interventions of ADPKD that are promising in the preclinical studies and investigated in clinical trials.
Collapse
|
24
|
Chebib FT, Perrone RD. Drug Development in Autosomal Dominant Polycystic Kidney Disease: Opportunities and Challenges. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:261-284. [PMID: 37088528 DOI: 10.1053/j.akdh.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/20/2022] [Accepted: 01/06/2023] [Indexed: 04/25/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a hereditary disorder characterized by relentless growth of innumerable renal cysts bilaterally, associated with decline in glomerular filtration rate over the course of decades. The burden of ADPKD and its treatment is associated with a significant economic and societal cost. Despite several clinical studies conducted over the past decade, only one treatment has been approved by regulatory agencies to slow disease progression in ADPKD. Elucidating feasible endpoints and clear regulatory pathway may stimulate interest in developing and translating novel therapeutics. This review summarizes the recent progress, challenges, and opportunities in drug development for ADPKD. We discuss the traditional and accelerated regulatory approval pathways, the various clinical trials endpoints, and biomarkers in ADPKD. Furthermore, we propose strategies that could optimize the clinical trial design in ADPKD. Finally, we owe it to our ADPKD patient community to strive for international collaborative studies geared toward discovery and validation of surrogate endpoints and to rally for funded infrastructure that would allow phase 3 master protocols in ADPKD. These advances will serve to derisk and potentially accelerate the development of therapies and eventually bring hope to patients and families who endure through this devastating disease.
Collapse
Affiliation(s)
- Fouad T Chebib
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL.
| | - Ronald D Perrone
- Division of Nephrology, Department of Medicine, Tufts Medical Center and Tufts University School of Medicine, Boston, MA.
| |
Collapse
|
25
|
Zhou JX, Torres VE. Drug repurposing in autosomal dominant polycystic kidney disease. Kidney Int 2023; 103:859-871. [PMID: 36870435 DOI: 10.1016/j.kint.2023.02.010] [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: 11/18/2022] [Revised: 01/23/2023] [Accepted: 02/07/2023] [Indexed: 03/06/2023]
Abstract
Autosomal dominant polycystic kidney disease is characterized by progressive kidney cyst formation that leads to kidney failure. Tolvaptan, a vasopressin 2 receptor antagonist, is the only drug approved to treat patients with autosomal dominant polycystic kidney disease who have rapid disease progression. The use of tolvaptan is limited by reduced tolerability from aquaretic effects and potential hepatotoxicity. Thus, the search for more effective drugs to slow down the progression of autosomal dominant polycystic kidney disease is urgent and challenging. Drug repurposing is a strategy for identifying new clinical indications for approved or investigational medications. Drug repurposing is increasingly becoming an attractive proposition because of its cost-efficiency and time-efficiency and known pharmacokinetic and safety profiles. In this review, we focus on the repurposing approaches to identify suitable drug candidates to treat autosomal dominant polycystic kidney disease and prioritization and implementation of candidates with high probability of success. Identification of drug candidates through understanding of disease pathogenesis and signaling pathways is highlighted.
Collapse
Affiliation(s)
- Julie Xia Zhou
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Mayo Clinic Robert M. and Billie Kelley Pirnie Translational Polycystic Kidney Disease Center, Rochester, Minnesota, USA.
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Mayo Clinic Robert M. and Billie Kelley Pirnie Translational Polycystic Kidney Disease Center, Rochester, Minnesota, USA.
| |
Collapse
|
26
|
Dagorn PG, Buchholz B, Kraus A, Batchuluun B, Bange H, Blockken L, Steinberg GR, Moller DE, Hallakou-Bozec S. A novel direct adenosine monophosphate kinase activator ameliorates disease progression in preclinical models of Autosomal Dominant Polycystic Kidney Disease. Kidney Int 2023; 103:917-929. [PMID: 36804411 DOI: 10.1016/j.kint.2023.01.026] [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: 06/16/2022] [Revised: 01/13/2023] [Accepted: 01/27/2023] [Indexed: 02/21/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) mainly results from mutations in the PKD1 gene, which encodes polycystin 1. It is the most common inherited kidney disease and is characterized by a progressive bilateral increase in cyst number and size, often leading to kidney failure. The cellular energy sensor and regulator adenosine monophosphate stimulated protein kinase (AMPK) has been implicated as a promising new therapeutic target. To address this hypothesis, we determined the effects of a potent and selective clinical stage direct allosteric AMPK activator, PXL770, in canine and patient-derived 3D cyst models and an orthologous mouse model of ADPKD. PXL770 induced AMPK activation and dose-dependently reduced cyst growth in principal-like Madin-Darby Canine Kidney cells stimulated with forskolin and kidney epithelial cells derived from patients with ADPKD stimulated with desmopressin. In an inducible, kidney epithelium-specific Pkd1 knockout mouse model, PXL770 produced kidney AMPK pathway engagement, prevented the onset of kidney failure (reducing blood urea by 47%), decreased cystic index by 26% and lowered the kidney weight to body weight ratio by 35% compared to untreated control Pkd1 knockout mice. These effects were accompanied by a reduction of markers of cell proliferation (-48%), macrophage infiltration (-53%) and tissue fibrosis (-37%). Thus, our results show the potential of direct allosteric AMPK activation in the treatment of ADPKD and support the further development of PXL770 for this indication.
Collapse
Affiliation(s)
| | - Bjoern Buchholz
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andre Kraus
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Battsetseg Batchuluun
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Hester Bange
- Crown Bioscience Netherlands B.V., The Netherlands
| | | | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
27
|
Jiang K, Huang Y, Chung EJ. Combining Metformin and Drug-Loaded Kidney-Targeting Micelles for Polycystic Kidney Disease. Cell Mol Bioeng 2023; 16:55-67. [PMID: 36660586 PMCID: PMC9842834 DOI: 10.1007/s12195-022-00753-9] [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: 08/09/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease that leads to eventual renal failure. Metformin (MET), an AMP-activated protein kinase (AMPK) activator already approved for type 2 diabetes, is currently investigated for ADPKD treatment. However, despite high tolerability, MET showed varying therapeutic efficacy in preclinical ADPKD studies. Thus, newer strategies have combined MET with other ADPKD small molecule drug candidates, thereby targeting multiple ADPKD-associated signaling pathways to enhance therapeutic outcomes through potential drug synergy. Unfortunately, the off-target side effects caused by these additional drug candidates pose a major hurdle. To address this, our group has previously developed kidney-targeting peptide amphiphile micelles (KMs), which displayed significant kidney accumulation in vivo, for delivering drugs to the site of the disease. Methods To mitigate the adverse effects of ADPKD drugs and evaluate their therapeutic potential in combination with MET, herein, we loaded KMs with ADPKD drug candidates including salsalate, octreotide, bardoxolone methyl, rapamycin, tolvaptan, and pioglitazone, and tested their in vitro therapeutic efficacy when combined with free MET. Specifically, after determining the 40% inhibitory concentration for each drug (IC40), the size, morphology, and surface charge of drug-loaded KMs were characterized. Next, drug-loaded KMs were applied in combination with MET to treat renal proximal tubule cells derived from Pkd1flox/-:TSLargeT mice in 2D proliferation and 3D cyst model. Results MET combined with all drug-loaded KMs demonstrated significantly enhanced efficacy as compared to free drugs in inhibiting cell proliferation and cyst growth. Notably, synergistic effects were found for MET and KMs loaded with either salsalate or rapamycin as determined by Bliss synergy scores. Conclusion Together, we show drug synergy using drug-loaded nanoparticles and free MET for the first time and present a novel nanomedicine-based combinatorial therapeutic approach for ADPKD with enhanced efficacy. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00753-9.
Collapse
Affiliation(s)
- Kairui Jiang
- Department of Biomedical Engineering, University of Southern California, 1002 Childs Way, MCB 357, Los Angeles, CA 90089 USA
| | - Yi Huang
- Department of Biomedical Engineering, University of Southern California, 1002 Childs Way, MCB 357, Los Angeles, CA 90089 USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, 1002 Childs Way, MCB 357, Los Angeles, CA 90089 USA
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA USA
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA USA
- Bridge Institute, University of Southern California, Los Angeles, CA USA
| |
Collapse
|
28
|
Hallows KR, Li H, Saitta B, Sepehr S, Huang P, Pham J, Wang J, Mancino V, Chung EJ, Pinkosky SL, Pastor-Soler NM. Beneficial effects of bempedoic acid treatment in polycystic kidney disease cells and mice. Front Mol Biosci 2022; 9:1001941. [PMID: 36504724 PMCID: PMC9730828 DOI: 10.3389/fmolb.2022.1001941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022] Open
Abstract
ADPKD has few therapeutic options. Tolvaptan slows disease but has side effects limiting its tolerability. Bempedoic acid (BA), an ATP citrate-lyase (ACLY) inhibitor FDA-approved for hypercholesterolemia, catalyzes a key step in fatty acid/sterol synthesis important for cell proliferation. BA is activated by very long-chain acyl-CoA synthetase (FATP2) expressed primarily in kidney and liver. BA also activates AMPK. We hypothesized that BA could be a novel ADPKD therapy by inhibiting cyst growth, proliferation, injury, and metabolic dysregulation via ACLY inhibition and AMPK activation. Pkd1-null kidney cell lines derived from mouse proximal tubule (PT) and collecting duct (IMCD) were grown in 2D or 3D Matrigel cultures and treated ± BA, ± SB-204990 (another ACLY inhibitor) or with Acly shRNA before cyst analysis, immunoblotting or mitochondrial assays using MitoSox and MitoTracker staining. Pkd1 fl/fl ; Pax8-rtTA; Tet-O-Cre C57BL/6J mice were induced with doxycycline injection on postnatal days 10 and 11 (P10-P11) and then treated ± BA (30 mg/kg/d) ± tolvaptan (30-100 mg/kg/d) by gavage from P12-21. Disease severity was determined by % total-kidney-weight-to-bodyweight (%TKW/BW) and BUN levels at euthanasia (P22). Kidney and liver homogenates were immunoblotted for expression of key biomarkers. ACLY expression and activity were upregulated in Pkd1-null PT and IMCD-derived cells vs. controls. Relative to controls, both BA and SB-204990 inhibited cystic growth in Pkd1-null kidney cells, as did Acly knockdown. BA inhibited mitochondrial superoxide production and promoted mitochondrial elongation, suggesting improved mitochondrial function. In ADPKD mice, BA reduced %TKW/BW and BUN to a similar extent as tolvaptan vs. untreated controls. Addition of BA to tolvaptan caused a further reduction in %TKW/BW and BUN vs. tolvaptan alone. BA generally reduced ACLY and stimulated AMPK activity in kidneys and livers vs. controls. BA also inhibited mTOR and ERK signaling and reduced kidney injury markers. In liver, BA treatment, both alone and together with tolvaptan, increased mitochondrial biogenesis while inhibiting apoptosis. We conclude that BA and ACLY inhibition inhibited cyst growth in vitro, and BA decreased ADPKD severity in vivo. Combining BA with tolvaptan further improved various ADPKD disease parameters. Repurposing BA may be a promising new ADPKD therapy, having beneficial effects alone and along with tolvaptan.
Collapse
Affiliation(s)
- Kenneth R. Hallows
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Hui Li
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Biagio Saitta
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Saman Sepehr
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Polly Huang
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Jessica Pham
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Jonathan Wang
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Valeria Mancino
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Eun Ji Chung
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | | | - Núria M. Pastor-Soler
- Division of Nephrology and Hypertension and USC/UKRO Kidney Research Center, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States,*Correspondence: Núria M. Pastor-Soler,
| |
Collapse
|
29
|
Zhou X, Torres VE. Emerging therapies for autosomal dominant polycystic kidney disease with a focus on cAMP signaling. Front Mol Biosci 2022; 9:981963. [PMID: 36120538 PMCID: PMC9478168 DOI: 10.3389/fmolb.2022.981963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), with an estimated genetic prevalence between 1:400 and 1:1,000 individuals, is the third most common cause of end stage kidney disease after diabetes mellitus and hypertension. Over the last 3 decades there has been great progress in understanding its pathogenesis. This allows the stratification of therapeutic targets into four levels, gene mutation and polycystin disruption, proximal mechanisms directly caused by disruption of polycystin function, downstream regulatory and signaling pathways, and non-specific pathophysiologic processes shared by many other diseases. Dysfunction of the polycystins, encoded by the PKD genes, is closely associated with disruption of calcium and upregulation of cyclic AMP and protein kinase A (PKA) signaling, affecting most downstream regulatory, signaling, and pathophysiologic pathways altered in this disease. Interventions acting on G protein coupled receptors to inhibit of 3',5'-cyclic adenosine monophosphate (cAMP) production have been effective in preclinical trials and have led to the first approved treatment for ADPKD. However, completely blocking cAMP mediated PKA activation is not feasible and PKA activation independently from cAMP can also occur in ADPKD. Therefore, targeting the cAMP/PKA/CREB pathway beyond cAMP production makes sense. Redundancy of mechanisms, numerous positive and negative feedback loops, and possibly counteracting effects may limit the effectiveness of targeting downstream pathways. Nevertheless, interventions targeting important regulatory, signaling and pathophysiologic pathways downstream from cAMP/PKA activation may provide additive or synergistic value and build on a strategy that has already had success. The purpose of this manuscript is to review the role of cAMP and PKA signaling and their multiple downstream pathways as potential targets for emergent therapies for ADPKD.
Collapse
Affiliation(s)
- Xia Zhou
- Mayo Clinic, Department of Nephrology, Rochester, MN, United States
| | | |
Collapse
|
30
|
Dachy A, Decuypere JP, Vennekens R, Jouret F, Mekahli D. Is autosomal dominant polycystic kidney disease an early sweet disease? Pediatr Nephrol 2022; 37:1945-1955. [PMID: 34988697 DOI: 10.1007/s00467-021-05406-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 10/19/2022]
Abstract
The clinical course of autosomal dominant polycystic kidney disease (ADPKD) starts in childhood. Evidence of the beneficial impact of early nephron-protective strategies and lifestyle modifications on ADPKD prognosis is accumulating. Recent studies have described the association of overweight and obesity with rapid disease progression in adults with ADPKD. Moreover, defective glucose metabolism and metabolic reprogramming have been reported in distinct ADPKD models highlighting these pathways as potential therapeutic targets in ADPKD. Several "metabolic" approaches are currently under evaluation in adults, including ketogenic diet, food restriction, and metformin therapy. No data are available on the impact of these approaches in childhood thus far. Yet, according to World Health Organization (WHO), we are currently facing a childhood obesity crisis with an increased prevalence of overweight/obesity in the pediatric population associated with a cardio-metabolic risk profile. The present review summarizes the knowledge about the role of glucose metabolism in the pathophysiology of ADPKD and underscores the possible harm of overweight and obesity in ADPKD especially in terms of long-term cardiovascular outcomes and renal prognosis.
Collapse
Affiliation(s)
- Angélique Dachy
- PKD Research Group, GPURE, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pediatrics, ULiège Academic Hospital, Liège, Belgium.,Laboratory of Translational Research in Nephrology (LTRN), GIGA Cardiovascular Sciences, ULiège, Liège, Belgium
| | - Jean-Paul Decuypere
- PKD Research Group, GPURE, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
| | - François Jouret
- Laboratory of Translational Research in Nephrology (LTRN), GIGA Cardiovascular Sciences, ULiège, Liège, Belgium.,Division of Nephrology, Department of Internal Medicine, ULiège Academic Hospital, Liège, Belgium
| | - Djalila Mekahli
- PKD Research Group, GPURE, Department of Development and Regeneration, KU Leuven, Leuven, Belgium. .,Department of Pediatric Nephrology, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| |
Collapse
|
31
|
Zhang Y, Daniel EA, Metcalf J, Dai Y, Reif GA, Wallace DP. CaMK4 overexpression in polycystic kidney disease promotes mTOR-mediated cell proliferation. J Mol Cell Biol 2022; 14:6674767. [PMID: 36002021 PMCID: PMC9802383 DOI: 10.1093/jmcb/mjac050] [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: 11/04/2021] [Revised: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 01/14/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by progressive enlargement of fluid-filled cysts, causing nephron loss and a decline in renal function. Mammalian target of rapamycin (mTOR) is overactive in cyst-lining cells and contributes to abnormal cell proliferation and cyst enlargement; however, the mechanism for mTOR stimulation remains unclear. We discovered that calcium/calmodulin (CaM) dependent kinase IV (CaMK4), a multifunctional kinase, is overexpressed in the kidneys of ADPKD patients and PKD mouse models. In human ADPKD cells, CaMK4 knockdown reduced mTOR abundance and the phosphorylation of ribosomal protein S6 kinase (S6K), a downstream target of mTOR. Pharmacologic inhibition of CaMK4 with KN-93 reduced phosphorylated S6K and S6 levels and inhibited cell proliferation and in vitro cyst formation of ADPKD cells. Moreover, inhibition of calcium/CaM-dependent protein kinase kinase-β and CaM, two key upstream regulators of CaMK4, also decreased mTOR signaling. The effects of KN-93 were independent of the liver kinase B1-adenosine monophosphate-activated protein kinase (AMPK) pathway, and the combination of KN-93 and metformin, an AMPK activator, had additive inhibitory effects on mTOR signaling and in vitro cyst growth. Our data suggest that increased CaMK4 expression and activity contribute to mTOR signaling and the proliferation of cystic cells of ADPKD kidneys.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA,Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - Emily A Daniel
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA,Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - July Metcalf
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA,Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - Yuqiao Dai
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA,Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | - Gail A Reif
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA,Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160-3018, USA
| | | |
Collapse
|
32
|
Iliuta IA, Song X, Pickel L, Haghighi A, Retnakaran R, Scholey J, Sung HK, Steinberg GR, Pei Y. Shared pathobiology identifies AMPK as a therapeutic target for obesity and autosomal dominant polycystic kidney disease. Front Mol Biosci 2022; 9:962933. [PMID: 36106024 PMCID: PMC9467623 DOI: 10.3389/fmolb.2022.962933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/21/2022] [Indexed: 12/02/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common Mendelian kidney disease, affecting approximately one in 1,000 births and accounting for 5% of end-stage kidney disease in developed countries. The pathophysiology of ADPKD is strongly linked to metabolic dysregulation, which may be secondary to defective polycystin function. Overweight and obesity are highly prevalent in patients with ADPKD and constitute an independent risk factor for progression. Recent studies have highlighted reduced AMP-activated protein kinase (AMPK) activity, increased mammalian target of rapamycin (mTOR) signaling, and mitochondrial dysfunction as shared pathobiology between ADPKD and overweight/obesity. Notably, mTOR and AMPK are two diametrically opposed sensors of energy metabolism that regulate cell growth and proliferation. However, treatment with the current generation of mTOR inhibitors is poorly tolerated due to their toxicity, making clinical translation difficult. By contrast, multiple preclinical and clinical studies have shown that pharmacological activation of AMPK provides a promising approach to treat ADPKD. In this narrative review, we summarize the pleiotropic functions of AMPK as a regulator of cellular proliferation, macromolecule metabolism, and mitochondrial biogenesis, and discuss the potential for pharmacological activation of AMPK to treat ADPKD and obesity-related kidney disease.
Collapse
Affiliation(s)
- Ioan-Andrei Iliuta
- Division of Nephrology, Department of Medicine, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Xuewen Song
- Division of Nephrology, Department of Medicine, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Lauren Pickel
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Amirreza Haghighi
- Division of Nephrology, Department of Medicine, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ravi Retnakaran
- Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - James Scholey
- Division of Nephrology, Department of Medicine, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gregory R. Steinberg
- Department of Medicine, Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - York Pei
- Division of Nephrology, Department of Medicine, University Health Network and University of Toronto, Toronto, ON, Canada
- *Correspondence: York Pei,
| |
Collapse
|
33
|
Bais T, Gansevoort RT, Meijer E. Drugs in Clinical Development to Treat Autosomal Dominant Polycystic Kidney Disease. Drugs 2022; 82:1095-1115. [PMID: 35852784 PMCID: PMC9329410 DOI: 10.1007/s40265-022-01745-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 12/16/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by progressive cyst formation that ultimately leads to kidney failure in most patients. Approximately 10% of patients who receive kidney replacement therapy suffer from ADPKD. To date, a vasopressin V2 receptor antagonist (V2RA) is the only drug that has been proven to attenuate disease progression. However, aquaresis-related adverse events limit its widespread use. Data on the renoprotective effects of somatostatin analogues differ largely between studies and medications. This review discusses new drugs that are investigated in clinical trials to treat ADPKD, such as cystic fibrosis transmembrane conductance regulator (CFTR) modulators and micro RNA inhibitors, and drugs already marketed for other indications that are being investigated for off-label use in ADPKD, such as metformin. In addition, potential methods to improve the tolerability of V2RAs are discussed, as well as methods to select patients with (likely) rapid disease progression and issues regarding the translation of preclinical data into clinical practice. Since ADPKD is a complex disease with a high degree of interindividual heterogeneity, and the mechanisms involved in cyst growth also have important functions in various physiological processes, it may prove difficult to develop drugs that target cyst growth without causing major adverse events. This is especially important since long-standing treatment is necessary in this chronic disease. This review therefore also discusses approaches to targeted therapy to minimize systemic side effects. Hopefully, these developments will advance the treatment of ADPKD.
Collapse
|
34
|
Tsukamoto S, Urate S, Yamada T, Azushima K, Yamaji T, Kinguchi S, Uneda K, Kanaoka T, Wakui H, Tamura K. Comparative Efficacy of Pharmacological Treatments for Adults With Autosomal Dominant Polycystic Kidney Disease: A Systematic Review and Network Meta-Analysis of Randomized Controlled Trials. Front Pharmacol 2022; 13:885457. [PMID: 35662736 PMCID: PMC9158498 DOI: 10.3389/fphar.2022.885457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Tolvaptan is the gold standard treatment for autosomal dominant polycystic kidney disease (ADPKD), while several other drugs have the potential to inhibit the progression of ADPKD. However, individual clinical trials may not show sufficient differences in clinical efficacy due to small sample sizes. Furthermore, the differences in therapeutic efficacy among drugs are unclear. Herein, we investigated the effect of the ADPKD treatments. Methods: We systematically searched PubMed, Medline, EMBASE, and the Cochrane Library through January 2022 to identify randomized controlled trials in ADPKD patients that compared the effects of treatments with placebo or conventional therapy. A network meta-analysis was performed to compare the treatments indirectly. The primary outcomes were changes in kidney function and the rate of total kidney volume (TKV) growth. Results: Sixteen studies were selected with a total of 4,391 patients. Tolvaptan significantly preserved kidney function and inhibited TKV growth compared to the placebo {standardized mean difference (SMD) [95% confidence interval (CI)]: 0.24 (0.16; 0.31) and MD: -2.70 (-3.10; -2.30), respectively}. Tyrosine kinase inhibitors and mammalian target of rapamycin (mTOR) inhibitors inhibited TKV growth compared to the placebo; somatostatin analogs significantly inhibited TKV growth compared to the placebo and tolvaptan [MD: -5.69 (-7.34; -4.03) and MD: -2.99 (-4.69; -1.29), respectively]. Metformin tended to preserve renal function, although it was not significant [SMD: 0.28 (-0.05; 0.61), p = 0.09]. Conclusion: The therapeutic effect of tolvaptan was reasonable as the gold standard for ADPKD treatment, while somatostatin analogs also showed notable efficacy in inhibiting TKV growth. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/, identifier CRD42022300814.
Collapse
Affiliation(s)
- Shunichiro Tsukamoto
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shingo Urate
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takayuki Yamada
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takahiro Yamaji
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazushi Uneda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Department of Kampo Medicine, Aizu Medical Center, Fukushima Medical University School of Medicine, Aizuwakamatsu, Japan
| | - Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
35
|
Kramers BJ, Koorevaar IW, van Gastel MD, van Goor H, Hallows KR, Heerspink HL, Li H, Leonhard WN, Peters DJ, Qiu J, Touw DJ, Gansevoort RT, Meijer E. Effects of Hydrochlorothiazide and Metformin on Aquaresis and Nephroprotection by a Vasopressin V2 Receptor Antagonist in ADPKD: A Randomized Crossover Trial. Clin J Am Soc Nephrol 2022; 17:507-517. [PMID: 35314480 PMCID: PMC8993480 DOI: 10.2215/cjn.11260821] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/17/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVES The vasopressin V2 receptor antagonist tolvaptan is the only drug that has been proven to be nephroprotective in autosomal dominant polycystic kidney disease (ADPKD). Tolvaptan also causes polyuria, limiting tolerability. We hypothesized that cotreatment with hydrochlorothiazide or metformin may ameliorate this side effect. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We performed a clinical study and an animal study. In a randomized, controlled, double-blind, crossover trial, we included 13 tolvaptan-treated patients with ADPKD. Patients were treated for three 2-week periods with hydrochlorothiazide, metformin, or placebo in random order. Primary outcome was change in 24-hour urine volume. We also measured GFR and a range of metabolic and kidney injury markers. RESULTS Patients (age 45±8 years, 54% women, measured GFR of 55±11 ml/min per 1.73 m2) had a baseline urine volume on tolvaptan of 6.9±1.4 L/24 h. Urine volume decreased to 5.1 L/24 h (P<0.001) with hydrochlorothiazide and to 5.4 L/24 h (P<0.001) on metformin. During hydrochlorothiazide treatment, plasma copeptin (surrogate for vasopressin) decreased, quality of life improved, and several markers of kidney damage and glucose metabolism improved. Metformin did not induce changes in these markers or in quality of life. Given these results, the effect of adding hydrochlorothiazide to tolvaptan was investigated on long-term kidney outcome in an animal experiment. Water intake in tolvaptan-hydrochlorothiazide cotreated mice was 35% lower than in mice treated with tolvaptan only. Combination treatment was superior to "no treatment" on markers of disease progression (kidney weight, P=0.003 and cystic index, P=0.04) and superior or equal to tolvaptan alone. CONCLUSIONS Both metformin and hydrochlorothiazide reduced tolvaptan-caused polyuria in a short-term study. Hydrochlorothiazide also reduced polyuria in a long-term animal model without negatively affecting nephroprotection. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2022_03_21_CJN11260821.mp3.
Collapse
Affiliation(s)
- Bart J. Kramers
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Iris W. Koorevaar
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maatje D.A. van Gastel
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Kenneth R. Hallows
- Division of Nephrology and Hypertension, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California
- University of Southern California/University Kidney Research Organization Kidney Research Center, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Hiddo L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University, Hospital Groningen, Groningen, The Netherlands
| | - Hui Li
- Division of Nephrology and Hypertension, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California
- University of Southern California/University Kidney Research Organization Kidney Research Center, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Wouter N. Leonhard
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Dorien J.M. Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jiedong Qiu
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daan J. Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University, Hospital Groningen, Groningen, The Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Ron T. Gansevoort
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Esther Meijer
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|