1
|
Gittus M, Haley H, Harris T, Borrows S, Padmanabhan N, Gale D, Simms R, Williams T, Acquaye A, Wong A, Chan M, Lee E, Ong AC. Commentary: Tolvaptan for Autosomal Dominant Polycystic Kidney Disease (ADPKD) - an update. BMC Nephrol 2025; 26:79. [PMID: 39953521 PMCID: PMC11827152 DOI: 10.1186/s12882-025-03960-4] [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: 11/05/2024] [Accepted: 01/10/2025] [Indexed: 02/17/2025] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) affects up to 70 000 people in the UK and the most common inherited cause of end-stage kidney disease (ESKD). It is generally a late-onset multisystem disorder characterised by bilateral kidney cysts, liver cysts and an increased risk of intracranial aneurysms. Approximately 50% of people with ADPKD reach ESKD by age 60. Disease-associated pain, discomfort, fatigue, emotional distress and, impaired mobility can impact health-related quality of life. The approval of tolvaptan, a vasopressin V2 receptor antagonist, has greatly advanced the care for people with ADPKD, shifting the focus from general chronic kidney disease management to targeted therapeutic approaches. While guidance from NICE and SMC provides a foundational framework, this is not clear or comprehensive enough to offer practical guidance for healthcare professionals in real-world settings. This commentary expands on the previous United Kingdom Kidney Association (UKKA) commentary in 2016 with an updated evidence base, the incorporation of real-world data and expert opinion to provide practical guidance to healthcare professionals. Through co-development with people affected by ADPKD, it now incorporates valuable patient perspectives and offers practical recommendations for the UK kidney community seeking to harmonise the quality of care of all people with ADPKD.
Collapse
Affiliation(s)
- Matt Gittus
- University of Sheffield, Sheffield, United Kingdom.
- Sheffield Teaching Hospitals NHS Trust, Sheffield, United Kingdom.
| | - Helen Haley
- University Hospitals of North Midlands, Birmingham, United Kingdom
| | | | - Sarah Borrows
- Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | | | - Danny Gale
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Roslyn Simms
- Sheffield Teaching Hospitals NHS Trust, Sheffield, United Kingdom
| | | | - Aaron Acquaye
- Hull and East Yorkshire Hospitals NHS Trust, Hull, United Kingdom
| | - Alisa Wong
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Melanie Chan
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Eduardo Lee
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Albert Cm Ong
- University of Sheffield, Sheffield, United Kingdom.
- Sheffield Teaching Hospitals NHS Trust, Sheffield, United Kingdom.
| |
Collapse
|
2
|
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]
|
3
|
Chebib FT, Dahl N, Zhou X, Garbinsky D, Wang J, Nunna S, Oberdhan D, Fernandes AW. Tolvaptan and Autosomal Dominant Polycystic Kidney Disease Progression in Individuals Aged 18-35 Years: A Pooled Database Analysis. Kidney Med 2025; 7:100935. [PMID: 39810815 PMCID: PMC11731472 DOI: 10.1016/j.xkme.2024.100935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
Rational & Objective Data are limited regarding the long-term efficacy of tolvaptan in adults aged 18-35 years with autosomal dominant polycystic kidney disease (ADPKD) at increased risk of rapid progression. We assessed the effects of tolvaptan within a larger population of younger adults and over longer follow-up than individual clinical trials could provide. Study Design Pooled database study. Setting & Study Populations A consolidated clinical study database with ADPKD patients aged 18-35 years. Selection Criteria for Studies Studies that enrolled patients who received either tolvaptan or standard-of-care treatment not including tolvaptan. Data Extraction Annual rate of change in estimated glomerular filtration rate (eGFR) and time to kidney failure. Analytical Approach For individuals participating in multiple studies, their data were longitudinally linked to extend the follow-up period. We matched tolvaptan-treated patients with controls based on age, sex, chronic kidney disease stage, eGFR, and, where possible, Mayo Imaging Classification. We compared eGFR decline between groups using mixed-effects modeling. Results The matched analysis set encompassed 204 tolvaptan-treated individuals and 204 controls. Median follow-up was 4.6 years for the tolvaptan group and 1.7 years for controls. In the mixed-effects model, the eGFR decline rate (in mL/min/1.73 m2/year) was -2.58 for the tolvaptan cohort and -4.28 for controls. This indicates reduction in the eGFR decline rate by 1.69 mL/min/1.73 m2/year (95% confidence interval: 0.87-2.52; P < 0.001) with tolvaptan, a 40% improvement. Extrapolating eGFR over 35 years, tolvaptan could delay kidney failure onset by approximately 11 years. Limitations Median follow-up was shorter in the control cohort than the tolvaptan cohort. The projection of time to kidney failure assumed a linear model of eGFR decline. Conclusions This analysis offers insights into the anticipated treatment benefits of tolvaptan for young adults with ADPKD. These findings are crucial for weighing treatment benefits against any associated risks.
Collapse
Affiliation(s)
- Fouad T. Chebib
- Mayo Clinic Division of Nephrology and Hypertension, Jacksonville, FL
| | - Neera Dahl
- Mayo Clinic Division of Nephrology and Hypertension, Rochester, MN
| | - Xiaolei Zhou
- RTI Health Solutions, Research Triangle Park, NC
| | | | - Jinyi Wang
- RTI Health Solutions, Research Triangle Park, NC
| | - Sasikiran Nunna
- Otsuka Pharmaceutical Development & Commercialization, Inc, Rockville, MD
| | - Dorothee Oberdhan
- Otsuka Pharmaceutical Development & Commercialization, Inc, Rockville, MD
| | | |
Collapse
|
4
|
Wolff CA, Aiello V, Elhassan EAE, Cristalli C, Lerario S, Paccapelo A, Ciurli F, Montanari F, Conti A, Benson K, Seri M, Brigl CB, Münster JS, Sciascia N, Kursch S, de Fallois J, La Manna G, Eckardt KU, Rank N, Popp B, Schönauer R, Conlon PJ, Capelli I, Halbritter J. Integrated Use of Autosomal Dominant Polycystic Kidney Disease Prediction Tools for Risk Prognostication. Clin J Am Soc Nephrol 2024:01277230-990000000-00521. [PMID: 39705090 DOI: 10.2215/cjn.0000000625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/17/2024] [Indexed: 12/22/2024]
Abstract
Key Points
The Mayo clinic imaging classification and the predicting renal outcome in polycystic kidney disease score are used to assess the risk of progression to kidney failure in autosomal dominant polycystic kidney disease.Mayo imaging classification and predicting renal outcome in polycystic kidney disease show little concordance; combined use increased the ability to identify rapid progression especially among intermediate risk patients.Accurate risk prediction is key for determining indication for specific treatment.
Background
Autosomal dominant polycystic kidney disease is the most common genetic cause of kidney failure. Specific treatment is indicated on observed or predicted rapid progression. For the latter, risk stratification tools have been developed independently based on either total kidney volume or genotyping as well as clinical variables. This study aimed to improve risk prediction by combining both imaging and clinical-genetic scores.
Methods
We conducted a retrospective multicenter cohort study of 468 patients diagnosed with autosomal dominant polycystic kidney disease. Clinical, imaging, and genetic data were analyzed for risk prediction. We defined rapid disease progression as an eGFR slope ≥3 ml/min per 1.73 m2 per year over 2 years, Mayo imaging classification (MIC) 1D–1E, or a predicting renal outcome in polycystic kidney disease (PROPKD) score of ≥7 points. Using MIC, PROPKD, and rare exome variant ensemble learner scores, several combined models were designed to develop a new classification with improved risk stratification. Primary endpoints were the development of advanced CKD stages G4–G5, longitudinal changes in eGFR, and clinical variables such as hypertension or urological events. Statistically, logistic regression, survival, receiver operating characteristic analyses, linear mixed models, and Cox proportional hazards models were used.
Results
PKD1-genotype (P < 0.001), MIC class 1E (P < 0.001), early-onset hypertension (P < 0.001), and early-onset urological events (P = 0.003) correlated best with rapid progression in multivariable analysis. While the MIC showed satisfactory specificity (77%), the PROPKD was more sensitive (59%). Among individuals with an intermediate risk in one of the scores, integration of the other score (combined scoring) allowed for more accurate stratification.
Conclusions
The combined use of both risk scores was associated with higher ability to identify rapid progressors and resulted in a better stratification, notably among intermediate risk patients.
Collapse
Affiliation(s)
- Constantin A Wolff
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Valeria Aiello
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elhussein A E Elhassan
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Carlotta Cristalli
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Sarah Lerario
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alexandro Paccapelo
- Research and Innovation Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Ciurli
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Montanari
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Amalia Conti
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Katherine Benson
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland
| | - Marco Seri
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Carolin B Brigl
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Julia S Münster
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Sciascia
- Pediatric and Adult Cardio-Thoracic and Vascular, Oncohematologic and Emergency Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Sebastian Kursch
- Division of Nephrology, University Hospital Leipzig, Leipzig, Germany
| | | | - Gaetano La Manna
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nina Rank
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Bernt Popp
- Center of Functional Genomics, Berlin Institute of Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ria Schönauer
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Peter J Conlon
- Department of Nephrology and Transplantation, Beaumont Hospital, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Irene Capelli
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Jan Halbritter
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
5
|
Jdiaa SS, Mustafa RA, Yu ASL. Treatment of Autosomal-Dominant Polycystic Kidney Disease. Am J Kidney Dis 2024:S0272-6386(24)01032-1. [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] [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
|
6
|
Pérez-Segovia A, Cojuc-Konigsberg G, Reul-Linares E, Hernández-Paredes EN, Chapa-Ibargüengoitia M, Ramírez-Sandoval JC. Kidney growth progression patterns in autosomal dominant polycystic kidney disease. Arch Med Res 2024; 56:103099. [PMID: 39393160 DOI: 10.1016/j.arcmed.2024.103099] [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: 04/24/2024] [Revised: 08/13/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Prognosis for autosomal dominant polycystic kidney disease (ADPKD), the main inherited cause of kidney failure, relies on estimating cystic growth using linear formulas derived from height-adjusted total kidney volume (Ht-TKV). However, nonlinear renal growth patterns may occur in typical ADPKD. AIMS To determine kidney outcomes of subjects diagnosed with typical ADPKD exhibiting nonlinear, and unpredictable cystic growth during follow-up. METHODS Retrospective cohort study. We categorized TKV changes in individuals with typical ADPKD according to observed kidney growth trajectories. Ht-TKV was calculated from consecutive CT or MRI using the ellipsoid method. We compared estimated glomerular filtration rate (eGFR) trajectories with linear mixed models. RESULTS We included 83 individuals with ADPKD (67% women; age 47 ± 12 years; follow-up 5.2 years [IQR 2.8-9.0]). Three kidney growth patterns were observed: slow progression (24%, <3%/year linear increase), fast progression (39%, ≥3%/year linear increase), and atypical progression (37%, nonlinear growth). Adjusted ht-TKV change in mL/m/year was +1.4 (IQR -4.5 to +10.0), +40.3 (+16.9 to +89.3), and +32.8 (+15.9 to +85.9) for slow, fast, and atypical progressors, respectively (p < 0.001). Atypical progressors exhibited a significantly greater decline in eGFR in mL/min/m²/year (-7.9, 95% CI -6.5, -3.9) compared to slow (-0.5, 95% CI -3.1 to +0.5) and fast progressors (-3.4, 95% CI -7.9, -2.0; between-group p < 0.001). Atypical progressors had a higher proportion of acute complications, including hemorrhages, infections, and urolithiasis (84%), compared to slow (20%) and fast progressors (31%) (p < 0.001). CONCLUSION In typical ADPKD, nonlinear, abrupt, and unpredictable cyst growth occurs frequently, leading to a higher risk of acute complications and kidney function decline.
Collapse
Affiliation(s)
- Aaron Pérez-Segovia
- Department of Radiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Gabriel Cojuc-Konigsberg
- Departament of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Estefania Reul-Linares
- Departament of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Elisa Naomi Hernández-Paredes
- Departament of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Mónica Chapa-Ibargüengoitia
- Department of Radiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Juan C Ramírez-Sandoval
- Department of Radiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
7
|
Minatoguchi S, Hayashi H, Umeda R, Koide S, Hasegawa M, Tsuboi N. Additional renoprotective effect of the SGLT2 inhibitor dapagliflozin in a patient with ADPKD receiving tolvaptan treatment. CEN Case Rep 2024; 13:419-424. [PMID: 38494546 PMCID: PMC11444039 DOI: 10.1007/s13730-024-00859-1] [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: 01/09/2024] [Accepted: 02/09/2024] [Indexed: 03/19/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a major cause of end-stage kidney disease (ESKD). Vasopressin plays a pivotal role in ADPKD progression; therefore, the selective vasopressin V2 receptor antagonist tolvaptan is used as a key drug in the management of ADPKD. On the other hand, sodium-glucose cotransporter-2 inhibitors (SGLT2i), which may possibly stimulate vasopressin secretion due to the diuretic effect of the drug, have been shown to have both renal and cardioprotective effects in various populations, including those with non-diabetic chronic kidney disease. However, the effect of SGLT2i in patients with ADPKD have not been fully elucidated. Herein, we report the case of a patient with ADPKD on tolvaptan who was administered the SGLT2i dapagliflozin. The patient was a Japanese woman diagnosed with ADPKD at age 30. Despite the treatment with tolvaptan, eGFR was gradually declined from 79.8 to 50 ml/min/1.73 m2 in almost 5 years and 10 mg of dapagliflozin was initiated in the hope of renoprotective effects. Although a small increase in vasopressin levels was observed, eGFR decline rate was moderated after dapagliflozin initiation. This case suggested an additional renoprotective effect of dapagliflozin in patient with ADPKD receiving tolvaptan. Although there is no evidence about the renal protective effect of SGLT2i in patients with ADPKD, we hereby report a case successfully treated with dapagliflozin for approximately 2 years. Further research, including clinical trials, is needed to evaluate whether SGLT2i are effective in patients with ADPKD.
Collapse
Affiliation(s)
- Shun Minatoguchi
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hiroki Hayashi
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Ryosuke Umeda
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Shigehisa Koide
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Midori Hasegawa
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Naotake Tsuboi
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
8
|
Ghanem A, Borghol AH, Munairdjy Debeh FG, Paul S, AlKhatib B, Harris PC, Garimella PS, Hanna C, Kline TL, Dahl NK, Chebib FT. Biomarkers of Kidney Disease Progression in ADPKD. Kidney Int Rep 2024; 9:2860-2882. [PMID: 39435347 PMCID: PMC11492289 DOI: 10.1016/j.ekir.2024.07.012] [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: 03/27/2024] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 10/23/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic kidney disorder and the fourth leading cause of kidney failure (KF) in adults. Characterized by a reduction in glomerular filtration rate (GFR) and increased kidney size, ADPKD exhibits significant variability in progression, highlighting the urgent need for reliable and predictive biomarkers to optimize management and treatment approaches. This review explores the roles of diverse biomarkers-including clinical, genetic, molecular, and imaging biomarkers-in evaluating disease progression and customizing treatments for ADPKD. Clinical biomarkers such as biological sex, the predicting renal outcome in polycystic kidney disease (PROPKD) score, and body mass index are shown to correlate with disease severity and progression. Genetic profiling, particularly distinguishing between truncating and non-truncating pathogenic variants in the PKD1 gene, refines risk assessment and prognostic precision. Advancements in imaging significantly enhance our ability to assess disease severity. Height-adjusted total kidney volume (htTKV) and the Mayo imaging classification (MIC) are foundational, whereas newer imaging biomarkers, including texture analysis, total cyst number (TCN), cyst-parenchyma surface area (CPSA), total cyst volume (TCV), and cystic index, focus on detailed cyst characteristics to offer deeper insights. Molecular biomarkers (including serum and urinary markers) shed light on potential therapeutic targets that could predict disease trajectory. Despite these advancements, there is a pressing need for the development of response biomarkers in both the adult and pediatric populations, which can evaluate the biological efficacy of treatments. The holistic evaluation of these biomarkers not only deepens our understanding of kidney disease progression in ADPKD, but it also paves the way for personalized treatment strategies aiming to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Ahmad Ghanem
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida, USA
| | - Abdul Hamid Borghol
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Stefan Paul
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida, USA
| | - Bassel AlKhatib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida, USA
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Pranav S. Garimella
- Division of Nephrology and Hypertension, University of California San Diego, San Diego, California, USA
| | - Christian Hanna
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Pediatric Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy L. Kline
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Neera K. Dahl
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Fouad T. Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
9
|
Watanabe EH, Onuchic LF. Visceral Abdominal Adiposity and Autosomal Dominant Polycystic Kidney Disease Progression: One More Step Toward Identifying Useful Biomarkers and Characterizing the Disease Metabolic Links. Am J Kidney Dis 2024; 84:263-266. [PMID: 39033453 DOI: 10.1053/j.ajkd.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/14/2024] [Indexed: 07/23/2024]
Affiliation(s)
- Elieser Hitoshi Watanabe
- Division of Nephrology, University of São Paulo School of Medicine, São Paulo, Brazil; Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Luiz Fernando Onuchic
- Division of Nephrology, University of São Paulo School of Medicine, São Paulo, Brazil; Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil.
| |
Collapse
|
10
|
Ivaturi V, Gobburu J, Leslie B, Wang X, Jadhav P. Urine Osmolality Is a Potential Marker of Longer-Term Efficacy of Tolvaptan in Autosomal Dominant Polycystic Kidney Disease: A Post Hoc Analysis. KIDNEY360 2024; 5:996-1001. [PMID: 38857379 PMCID: PMC11296543 DOI: 10.34067/kid.0000000000000485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
Key Points Post hoc analyses of the TEMPO 3:4 trial suggest that short-term reductions in urine osmolality with tolvaptan predict effects on total kidney volume and eGFR. Change in urine osmolality has potential as a biomarker of treatment response and may facilitate trial design and clinical decision making. Background Total kidney volume (TKV) and eGFR are measures of progression and treatment response in autosomal dominant polycystic kidney disease, but utility is limited by the long follow-up required for change assessment. In an analysis of data from the 3-year TEMPO 3:4 trial, we evaluated relationships among a short-term indicator of drug activity (change in urine osmolality [Uosm]) and longer-term outcomes to evaluate Uosm as a potential marker of efficacy. Methods Linear regression modeling and single-point analyses assessed relationships among change in Uosm to week 3, change in TKV to month 12, and change in eGFR to month 36 in participants treated with tolvaptan (n =961) or placebo (n =483). Multivariate models evaluated the proportion of the tolvaptan treatment effect on eGFR attributable to change in Uosm. Results Change in TKV to month 12 and Uosm to week 3 each correlated with change in eGFR to month 36, regardless of treatment assignment. A greater decrease in Uosm from baseline to week 3 was indicative of a slower decrease in eGFR to month 36 (slope estimate of −0.01, P < 0.00001). The effect of tolvaptan on Uosm accounted for 68.8% of the treatment effect on change in eGFR to month 36. Simulations of TEMPO 3:4 under the null hypothesis (i.e ., replacement of all values for change in Uosm from baseline to week 3 with values from the placebo arm only) yielded a type 1 error rate indicating an acceptable risk of falsely concluding treatment efficacy on the basis of change in Uosm as a trial end point. Conclusions Change in Uosm is a potential biomarker for long-term treatment outcome with tolvaptan and might expedite clinical trials and treatment decision making for drugs with similar mechanisms of action.
Collapse
Affiliation(s)
| | | | - Bruce Leslie
- Otsuka Pharmaceutical Development & Commercialization (OPDC), Inc., Princeton, New Jersey
| | - Xiaofeng Wang
- Otsuka Pharmaceutical Development & Commercialization (OPDC), Inc., Princeton, New Jersey
| | | |
Collapse
|
11
|
Sim JJ, Shu YH, Bhandari SK, Chen Q, Harrison TN, Lee MY, Munis MA, Morrissette K, Sundar S, Pareja K, Nourbakhsh A, Willey CJ. Data driven approach to characterize rapid decline in autosomal dominant polycystic kidney disease. PLoS One 2024; 19:e0298484. [PMID: 38837988 DOI: 10.1371/journal.pone.0298484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a genetic kidney disease with high phenotypic variability. Furthering insights into patients' ADPKD progression could lead to earlier detection, management, and alter the course to end stage kidney disease (ESKD). We sought to identify patients with rapid decline (RD) in kidney function and to determine clinical factors associated with RD using a data-driven approach. A retrospective cohort study was performed among patients with incident ADPKD (1/1/2002-12/31/2018). Latent class mixed models were used to identify RD patients using differences in eGFR trajectories over time. Predictors of RD were selected based on agreements among feature selection methods, including logistic, regularized, and random forest modeling. The final model was built on the selected predictors and clinically relevant covariates. Among 1,744 patients with incident ADPKD, 125 (7%) were identified as RD. Feature selection included 42 clinical measurements for adaptation with multiple imputations; mean (SD) eGFR was 85.2 (47.3) and 72.9 (34.4) in the RD and non-RD groups, respectively. Multiple imputed datasets identified variables as important features to distinguish RD and non-RD groups with the final prediction model determined as a balance between area under the curve (AUC) and clinical relevance which included 6 predictors: age, sex, hypertension, cerebrovascular disease, hemoglobin, and proteinuria. Results showed 72%-sensitivity, 70%-specificity, 70%-accuracy, and 0.77-AUC in identifying RD. 5-year ESKD rates were 38% and 7% among RD and non-RD groups, respectively. Using real-world routine clinical data among patients with incident ADPKD, we observed that six variables highly predicted RD in kidney function.
Collapse
Affiliation(s)
- John J Sim
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States of America
- Division of Nephrology and Hypertension, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA, United States of America
- Departments of Health Systems and Clinical Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, United States of America
| | - Yu-Hsiang Shu
- Biostatistics and Programming Clinical Affairs, Inari Medical, Irvine, CA, United States of America
| | - Simran K Bhandari
- Department of Internal Medicine, Bellflower Medical Center, Bellflower, CA, United States of America
| | - Qiaoling Chen
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States of America
| | - Teresa N Harrison
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States of America
| | - Min Young Lee
- Division of Nephrology and Hypertension, Kaiser Permanente Los Angeles Medical Center, Los Angeles, CA, United States of America
| | - Mercedes A Munis
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States of America
| | - Kerresa Morrissette
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA, United States of America
| | - Shirin Sundar
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, United States of America
| | - Kristin Pareja
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, United States of America
| | - Ali Nourbakhsh
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, United States of America
| | - Cynthia J Willey
- College of Pharmacy, University of Rhode Island, Kingston, RI, United States of America
| |
Collapse
|
12
|
Martin ALAR, Pereira RLS, Rocha JE, Farias PAM, Freitas TS, Caldas FRDL, Figueredo FG, Sampaio NFL, Oliveira-Tintino CDDM, Tintino SR, da Hora GCA, Lima MCP, de Menezes IRA, Carvalho DT, Coutinho HDM, Fonteles MMF. Unlocking bacterial defense: Exploring the potent inhibition of NorA efflux pump by coumarin derivatives in Staphylococcus aureus. Microb Pathog 2024; 190:106608. [PMID: 38503396 DOI: 10.1016/j.micpath.2024.106608] [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: 11/24/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 03/21/2024]
Abstract
The occurrence of bacterial resistance has been increasing, compromising the treatment of various infections. The high virulence of Staphylococcus aureus allows for the maintenance of the infectious process, causing many deaths and hospitalizations. The MepA and NorA efflux pumps are transporter proteins responsible for expelling antimicrobial agents such as fluoroquinolones from the bacterial cell. Coumarins are phenolic compounds that have been studied for their diverse biological actions, including against bacteria. A pharmacokinetic in silico characterization of compounds C10, C11, C13, and C14 was carried out according to the principles of Lipinski's Rule of Five, in addition to searching for similarity in ChemBL and subsequent search for publications in CAS SciFinder. All compounds were evaluated for their in vitro antibacterial and modulatory activity against standard and multidrug-resistant Gram-positive and Gram-negative strains. The effect of coumarins C9, C10, C11, C13, and C14 as efflux pump inhibitors in Staphylococcus aureus strains was evaluated using the microdilution method (MepA or NorA) and fluorimetry (NorA). The behavior of coumarins regarding the efflux pump was determined from their interaction properties with the membrane and coumarin-protein using molecular docking and molecular dynamics simulations. Only the isolated coumarin compound C13 showed antibacterial activity against standard strains of Staphylococcus aureus and Escherichia coli. However, the other tested coumarins showed modulatory capacity for fluoroquinolone and aminoglycoside antibacterials. Compounds C10, C13, and C14 were effective in reducing the MIC of both antibiotics for both multidrug-resistant strains, while C11 potentiated the effect of norfloxacin and gentamicin for Gram-positive and Gram-negative bacteria and only norfloxacin for Gram-negative. Only coumarin C14 produced synergistic effects when associated with ciprofloxacin in MepA-carrying strains. All tested coumarins have the ability to inhibit the NorA efflux pump present in Staphylococcus aureus, both in reducing the MIC and inducing increased ethidium bromide fluorescence emission in fluorimetry. The findings of this study offer an atomistic perspective on the potential of coumarins as active inhibitors of the NorA pump, highlighting their specific mode of action mainly targeting protein inhibition. In molecular docking, it was observed that coumarins are capable of interacting with various amino acid residues of the NorA pump. The simulation showed that coumarin C10 can cross the bilayer; however, the other coumarins interacted with the membrane but were unable to cross it. Coumarins demonstrated their potentiating role in the effect of norfloxacin through a dual mechanism: efflux pump inhibition through direct interaction with the protein (C9, C10, C11, and C13) and increased interaction with the membrane (C10 and C13). In the context of pharmacokinetic prediction studies, the studied structures have a suitable chemical profile for possible oral use. We suggest that coumarin derivatives may be an interesting alternative in the future for the treatment of resistant bacterial infections, with the possibility of a synergistic effect with other antibacterials, although further studies are needed to characterize their therapeutic effects and toxicity.
Collapse
Affiliation(s)
- Ana Luíza A R Martin
- Department of Physiology and Pharmacology, Federal University of Ceará - UFC, 60430-160, Fortaleza, Brazil; Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | | | - Janaína Esmeraldo Rocha
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | - Pablo A M Farias
- School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil; CECAPE College, 63024-015, Juazeiro do Norte, Brazil
| | - Thiago S Freitas
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | | | - Fernando G Figueredo
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | - Nadghia Figueiredo Leite Sampaio
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil; School of Medicine, Medical Education Institute - IDOMED, 63048-080, Juazeiro do Norte, Brazil
| | | | - Saulo Relison Tintino
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | | | | | - Irwin Rose A de Menezes
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil
| | - Diogo T Carvalho
- School of Pharmacy, Federal University of Alfenas - UNIFAL, 37130-001, Alfenas, Brazil
| | - Henrique D M Coutinho
- Department of Biological Chemistry, Regional University of Cariri - URCA. 63105-000, Crato, Brazil.
| | - Marta M F Fonteles
- Department of Physiology and Pharmacology, Federal University of Ceará - UFC, 60430-160, Fortaleza, Brazil
| |
Collapse
|
13
|
Bais T, Geertsema P, Knol MGE, van Gastel MDA, de Haas RJ, Meijer E, Gansevoort RT. Validation of the Mayo Imaging Classification System for Predicting Kidney Outcomes in ADPKD. Clin J Am Soc Nephrol 2024; 19:591-601. [PMID: 38407866 PMCID: PMC11108249 DOI: 10.2215/cjn.0000000000000427] [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: 09/15/2023] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND The Mayo Imaging Classification was developed to predict the rate of disease progression in patients with autosomal dominant polycystic kidney disease. This study aimed to validate its ability to predict kidney outcomes in a large multicenter autosomal dominant polycystic kidney disease cohort. METHODS Included were patients with ≥1 height-adjusted total kidney volume (HtTKV) measurement and ≥3 eGFR values during ≥1-year follow-up. Mayo HtTKV class stability, kidney growth rates, and eGFR decline rates were calculated. The observed eGFR decline was compared with predictions from the Mayo Clinic future eGFR equation. The future eGFR prediction equation was also tested for nonlinear eGFR decline. Kaplan-Meier survival analysis and Cox regression models were used to assess time to kidney failure using Mayo HtTKV class as a predictor variable. RESULTS We analyzed 618 patients with a mean age of 47±11 years and mean eGFR of 64±25 ml/min per 1.73 m 2 at baseline. Most patients (82%) remained in their baseline Mayo HtTKV class. During a mean follow-up of 5.1±2.2 years, the mean total kidney volume growth rates and eGFR decline were 5.33%±3.90%/yr and -3.31±2.53 ml/min per 1.73 m 2 per year, respectively. Kidney growth and eGFR decline showed considerable overlap between the classes. The observed annual eGFR decline was not significantly different from the predicted values for classes 1A, 1B, 1C, and 1D but significantly slower for class 1E. This was also observed in patients aged younger than 40 years and older than 60 years and those with PKD2 mutations. A polynomial model allowing nonlinear eGFR decline provided more accurate slope predictions. Ninety-seven patients (16%) developed kidney failure during follow-up. The classification predicted the development of kidney failure, although the sensitivity and positive predictive values were limited. CONCLUSIONS The Mayo Imaging Classification demonstrated acceptable stability and generally predicted kidney failure and eGFR decline rate. However, there was marked interindividual variability in the rate of disease progression within each class.
Collapse
Affiliation(s)
- Thomas Bais
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Paul Geertsema
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martine G E Knol
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maatje D A van Gastel
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robbert J de Haas
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Esther Meijer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ron T Gansevoort
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
14
|
Ali H, Alahmad B, Senum SR, Warsame S, Bahbahani Y, Abu-Farha M, Abubaker J, Alqaddoumi M, Al-Mulla F, Harris PC. PKD1 Truncating Mutations Accelerate eGFR Decline in Autosomal Dominant Polycystic Kidney Disease Patients. Am J Nephrol 2024; 55:380-388. [PMID: 38194940 PMCID: PMC11151966 DOI: 10.1159/000536165] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic disease characterized by the accumulation of fluid-filled cysts in the kidneys, leading to renal volume enlargement and progressive kidney function impairment. Disease severity, though, may vary due to allelic and genetic heterogeneity. This study aimed to determine genotype-phenotype correlations between PKD1 truncating and non-truncating mutations and kidney function decline in ADPKD patients. METHODS We established a single-center retrospective cohort study in Kuwait where we followed every patient with a confirmed PKD1-ADPKD diagnosis clinically and genetically. Renal function tests were performed annually. We fitted generalized additive mixed effects models with random intercepts for each individual to analyze repeated measures of kidney function across mutation type. We then calculated survival time to kidney failure in a cox proportional hazards model. Models were adjusted for sex, age at visit, and birth year. RESULTS The study included 22 truncating and 20 non-truncating (42 total) patients followed for an average of 6.6 years (range: 1-12 years). Those with PKD1 truncating mutations had a more rapid rate of eGFR decline (-4.7 mL/min/1.73 m2 per year; 95% CI: -5.0, -4.4) compared to patients with PKD1 non-truncating mutations (-3.5 mL/min/1.73 m2 per year; 95% CI: -4.0, -3.1) (p for interaction <0.001). Kaplan-Meier survival analysis of time to kidney failure showed that patients with PKD1 truncating mutations had a shorter renal survival time (median 51 years) compared to those with non-truncating mutations (median 56 years) (P for log-rank = 0.008). CONCLUSION In longitudinal and survival analyses, patients with PKD1 truncating mutations showed a faster decline in kidney function compared to patients PKD1 non-truncating mutations. Early identification of patients with PKD1 truncating mutations can, at best, inform early clinical interventions or, at least, help suggest aggressive monitoring.
Collapse
Affiliation(s)
- Hamad Ali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
| | - Barrak Alahmad
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sarah R. Senum
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Samia Warsame
- Medical Division, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Yousif Bahbahani
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
- Medical Division, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Malak Alqaddoumi
- Department of Pathology, Faculty of Medicine, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
15
|
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: 8] [Impact Index Per Article: 4.0] [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
|
16
|
Wigerinck S, Gregory AV, Smith BH, Iliuta IA, Hanna C, Chedid M, Kaidbay HDN, Senum SR, Shukoor S, Harris PC, Torres VE, Kline TL, Chebib FT. Evaluation of advanced imaging biomarkers at kidney failure in patients with ADPKD: a pilot study. Clin Kidney J 2023; 16:1691-1700. [PMID: 37779848 PMCID: PMC10539251 DOI: 10.1093/ckj/sfad114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Indexed: 10/03/2023] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) presents with variable disease severity and progression. Advanced imaging biomarkers may provide insights into cystic and non-cystic processes leading to kidney failure in different age groups. Methods This pilot study included 39 ADPKD patients with kidney failure, stratified into three age groups (<46, 46-56, >56 years old). Advanced imaging biomarkers were assessed using an automated instance cyst segmentation tool. The biomarkers were compared with an age- and sex-matched ADPKD cohort in early chronic kidney disease (CKD). Results Ht-total parenchymal volume correlated negatively with age at kidney failure. The median Ht-total parenchymal volume was significantly lower in patients older than 56 years. Cystic burden was significantly higher at time of kidney failure, especially in patients who reached it before age 46 years. The cyst index at kidney failure was comparable across age groups and Mayo Imaging Classes. Advanced imaging biomarkers showed higher correlation with Ht-total kidney volume in early CKD than at kidney failure. Cyst index and parenchymal index were relatively stable over 5 years prior to kidney failure, whereas Ht-total cyst volume and cyst parenchymal surface area increased significantly. Conclusion Age-related differences in advanced imaging biomarkers suggest variable pathophysiological mechanisms in ADPKD patients with kidney failure. Further studies are needed to validate the utility of these biomarkers in predicting disease progression and guiding treatment strategies.
Collapse
Affiliation(s)
- Stijn Wigerinck
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Faculty of Medicine, Catholic University of Leuven, Leuven, Belgium
| | | | - Byron H Smith
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ioan-Andrei Iliuta
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL, USA
| | - Christian Hanna
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Division of Pediatric Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Maroun Chedid
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | | | - Sarah R Senum
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shebaz Shukoor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | | | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
17
|
Caroli A, Villa G, Brambilla P, Trillini M, Sharma K, Sironi S, Remuzzi G, Perico N, Remuzzi A. Diffusion magnetic resonance imaging for kidney cyst volume quantification and non-cystic tissue characterisation in ADPKD. Eur Radiol 2023; 33:6009-6019. [PMID: 37017703 DOI: 10.1007/s00330-023-09601-4] [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: 10/03/2022] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 04/06/2023]
Abstract
OBJECTIVES Beyond total kidney and cyst volume (TCV), non-cystic tissue plays an important role in autosomal dominant polycystic kidney disease (ADPKD) progression. This study aims at presenting and preliminarily validating a diffusion MRI (DWI)-based TCV quantification method and providing evidence of DWI potential in characterising non-cystic tissue microstructure. METHODS T2-weighted MRI and DWI scans (b = 0, 15, 50, 100, 200, 350, 500, 700, 1000; 3 directions) were acquired from 35 ADPKD patients with CKD stage 1 to 3a and 15 healthy volunteers on a 1.5 T scanner. ADPKD classification was performed using the Mayo model. DWI scans were processed by mono- and segmented bi-exponential models. TCV was quantified on T2-weighted MRI by the reference semi-automatic method and automatically computed by thresholding the pure diffusivity (D) histogram. The agreement between reference and DWI-based TCV values and the differences in DWI-based parameters between healthy and ADPKD tissue components were assessed. RESULTS There was strong correlation between DWI-based and reference TCV (rho = 0.994, p < 0.001). Non-cystic ADPKD tissue had significantly higher D, and lower pseudo-diffusion and flowing fraction than healthy tissue (p < 0.001). Moreover, apparent diffusion coefficient and D values significantly differed by Mayo imaging class, both in the whole kidney (Wilcoxon p = 0.007 and p = 0.004) and non-cystic tissue (p = 0.024 and p = 0.007). CONCLUSIONS DWI shows potential in ADPKD to quantify TCV and characterise non-cystic kidney tissue microstructure, indicating the presence of microcysts and peritubular interstitial fibrosis. DWI could complement existing biomarkers for non-invasively staging, monitoring, and predicting ADPKD progression and evaluating the impact of novel therapies, possibly targeting damaged non-cystic tissue besides cyst expansion. CLINICAL RELEVANCE STATEMENT This study shows diffusion-weighted MRI (DWI) potential to quantify total cyst volume and characterise non-cystic kidney tissue microstructure in ADPKD. DWI could complement existing biomarkers for non-invasively staging, monitoring, and predicting ADPKD progression and evaluating the impact of novel therapies, possibly targeting damaged non-cystic tissue besides cyst expansion. KEY POINTS • Diffusion magnetic resonance imaging shows potential to quantify total cyst volume in ADPKD. • Diffusion magnetic resonance imaging might allow to non-invasively characterise non-cystic kidney tissue microstructure. • Diffusion magnetic resonance imaging-based biomarkers significantly differ by Mayo imaging class, suggesting their possible prognostic value.
Collapse
Affiliation(s)
- Anna Caroli
- Clinical Research Center for Rare Diseases "Aldo & Cele Daccò", Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Camozzi 3, 24020, Bergamo, Ranica, Italy.
| | - Giulia Villa
- Clinical Research Center for Rare Diseases "Aldo & Cele Daccò", Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Camozzi 3, 24020, Bergamo, Ranica, Italy
| | - Paolo Brambilla
- Department of Diagnostic Radiology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Matias Trillini
- Clinical Research Center for Rare Diseases "Aldo & Cele Daccò", Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Camozzi 3, 24020, Bergamo, Ranica, Italy
| | - Kanishka Sharma
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Sandro Sironi
- Department of Diagnostic Radiology, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- School of Medicine, University of Milano-Bicocca, Milan, Italy
| | - Giuseppe Remuzzi
- Clinical Research Center for Rare Diseases "Aldo & Cele Daccò", Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Camozzi 3, 24020, Bergamo, Ranica, Italy
| | - Norberto Perico
- Clinical Research Center for Rare Diseases "Aldo & Cele Daccò", Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Camozzi 3, 24020, Bergamo, Ranica, Italy
| | - Andrea Remuzzi
- Department of Management, Information and Production Engineering, University of Bergamo, Dalmine, BG, Italy
| |
Collapse
|
18
|
Oh YK, Ryu H, Ahn C, Park HC, Ma Y, Xu D, Ecder T, Kao TW, Huang JW, Rangan GK. Clinical Characteristics of Rapid Progression in Asia-Pacific Patients With ADPKD. Kidney Int Rep 2023; 8:1801-1810. [PMID: 37705904 PMCID: PMC10496076 DOI: 10.1016/j.ekir.2023.06.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 09/15/2023] Open
Abstract
Introduction This study aimed to determine the utility of different methods to predict rapid progressors (RPs) and their clinical characteristics in Asia-Pacific patients with autosomal dominant polycystic kidney disease (ADPKD). Methods This was a multinational retrospective observational cohort study of patients with ADPKD in the Asia-Pacific region. Five hospitals from Australia, China, South Korea, Taiwan, and Turkey participated in this study. RP was defined by European Renal Association-European Dialysis and Transplantation Association (ERA-EDTA) guidelines and compared to slow progressors (SPs). Results Among 768 patients, 426 patients were RPs. Three hundred six patients met only 1 criterion and 120 patients satisfied multiple criteria for RP. Historical estimated glomerular filtration rate (eGFR) decline fulfilled the criteria for RP in 210 patients. Five patients met the criteria for a historical increase in height-adjusted total kidney volume (TKV). The 210 patients satisfied the criteria for based on kidney volume. During the follow-up period, cyst infections, cyst hemorrhage, and proteinuria occurred more frequently in RP; and 13.9% and 2.1% of RPs and SPs, respectively, progressed to end-stage kidney disease (ESKD). RP criteria based on historical eGFR decline had the strongest correlation with eGFR change over a 2-year follow-up. Conclusion Various assessment strategies should be used for identifying RPs among Asian-Pacific patients with ADPKD in real-world clinical practice during the follow-up period, cyst infections, cyst hemorrhage, and proteinuria occurred more frequently; and more patients progressed to ESKD in RPs compared with SPs.
Collapse
Affiliation(s)
- Yun Kyu Oh
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hayne C. Park
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Seoul, Korea
| | - Yiyi Ma
- Department of Nephrology, Kidney Institute, Second Affiliated Hospital, Navy Medical University, Shanghai, China
| | - Dechao Xu
- Department of Nephrology, Kidney Institute, Second Affiliated Hospital, Navy Medical University, Shanghai, China
| | - Tevfik Ecder
- Department of Internal Medicine, Division of Nephrology, Faculty of Medicine, Istanbul Bilim University, Istanbul, Turkey
| | - Tze-Wah Kao
- Division of Nephrology, Department of Internal Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Jeng-Wen Huang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Gopala K. Rangan
- Michael Stern Laboratory for Polycystic Kidney Disease, Westmead Institute for Medical Research, The University of Sydney and the Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Westmead, New South Wales, Australia
| |
Collapse
|
19
|
Schellekens P, Verjans M, Janssens P, Dachy A, De Rechter S, Breysem L, Allegaert K, Bammens B, Vennekens R, Vermeersch P, Pottel H, Mekahli D. Low agreement between various eGFR formulae in pediatric and young adult ADPKD patients. Pediatr Nephrol 2023; 38:3043-3053. [PMID: 36939917 DOI: 10.1007/s00467-023-05926-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/21/2023]
Abstract
BACKGROUND Young autosomal dominant polycystic kidney disease (ADPKD) patients are becoming the new target population for the development of new treatment options. Determination of a reliable equation for estimated glomerular filtration rate (eGFR) from early stages is needed with the promising potential interventional therapies. METHODS Prospective and longitudinal study on a cohort of 68 genotyped ADPKD patients (age range 0-23 years) with long-term follow-up. Commonly used equations for eGFR were compared for their relative performance. RESULTS The revised Schwartz formula (CKiD) showed a highly significant decline in eGFR with aging (- 3.31 mL/min/1.73 m2/year, P < 0.0001). The recently updated equation by the Schwartz group (CKiDU25) showed a smaller (- 0.90 mL/min/1.73 m2/year) but significant (P = 0.001) decline in eGFR with aging and also showed a significant sex difference (P < 0.0001), not observed by the other equations. In contrast, the full age spectrum (FAS) equations (FAS-SCr, FAS-CysC, and the combined) showed no age and sex dependency. The prevalence of hyperfiltration is highly dependent on the formula used, and the highest prevalence was observed with the CKiD Equation (35%). CONCLUSIONS The most widely used methods to calculate eGFR in ADPKD children (CKiD and CKiDU25 equations) were associated with unexpected age or sex differences. The FAS equations were age- and sex-independent in our cohort. Hence, the switch from the CKiD to CKD-EPI equation at the transition from pediatric to adult care causes implausible jumps in eGFR, which could be misinterpreted. Having reliable methods to calculate eGFR is indispensable for clinical follow-up and clinical trials. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Pieter Schellekens
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Louvain, Belgium
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Louvain, Belgium
| | - Marcelien Verjans
- Department of Pediatric Nephrology, University Hospitals Leuven, Louvain, Belgium
| | - Peter Janssens
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
- Department of Nephrology and Arterial Hypertension, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel, Brussels, Belgium
| | - Angélique Dachy
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
- Department of Pediatrics, U Liège Academic Hospital, Liège, Belgium
| | - Stéphanie De Rechter
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
- Department of Pediatric Nephrology, University Hospitals Leuven, Louvain, Belgium
| | - Luc Breysem
- Department of Radiology, University Hospitals Leuven, Louvain, Belgium
| | - Karel Allegaert
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000, Louvain, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000, Louvain, Belgium
- Department of Clinical Pharmacy, Erasmus MC, 3000, CA, Rotterdam, the Netherlands
| | - Bert Bammens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Louvain, Belgium
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Louvain, Belgium
| | - Rudi Vennekens
- Department of Cellular and Molecular Medicine, VIB Centre for Brain and Disease Research, Laboratory of Ion Channel Research, KU Leuven, Louvain, Belgium
| | - Pieter Vermeersch
- Department of Laboratory of Laboratory Medicine, University Hospitals Leuven, Louvain, Belgium
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Djalila Mekahli
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium.
- Department of Pediatric Nephrology, University Hospitals Leuven, Louvain, Belgium.
| |
Collapse
|
20
|
Caroli A, Kline TL. Abdominal Imaging in ADPKD: Beyond Total Kidney Volume. J Clin Med 2023; 12:5133. [PMID: 37568535 PMCID: PMC10420262 DOI: 10.3390/jcm12155133] [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: 05/26/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
In the context of autosomal dominant polycystic kidney disease (ADPKD), measurement of the total kidney volume (TKV) is crucial. It acts as a marker for tracking disease progression, and evaluating the effectiveness of treatment strategies. The TKV has also been recognized as an enrichment biomarker and a possible surrogate endpoint in clinical trials. Several imaging modalities and methods are available to calculate the TKV, and the choice depends on the purpose of use. Technological advancements have made it possible to accurately assess the cyst burden, which can be crucial to assessing the disease state and helping to identify rapid progressors. Moreover, the development of automated algorithms has increased the efficiency of total kidney and cyst volume measurements. Beyond these measurements, the quantification and characterization of non-cystic kidney tissue shows potential for stratifying ADPKD patients early on, monitoring disease progression, and possibly predicting renal function loss. A broad spectrum of radiological imaging techniques are available to characterize the kidney tissue, showing promise when it comes to non-invasively picking up the early signs of ADPKD progression. Radiomics have been used to extract textural features from ADPKD images, providing valuable information about the heterogeneity of the cystic and non-cystic components. This review provides an overview of ADPKD imaging biomarkers, focusing on the quantification methods, potential, and necessary steps toward a successful translation to clinical practice.
Collapse
Affiliation(s)
- Anna Caroli
- Bioengineering Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24020 Ranica, BG, Italy
| | | |
Collapse
|
21
|
Gregory AV, Chebib FT, Poudyal B, Holmes HL, Yu ASL, Landsittel DP, Bae KT, Chapman AB, Frederic RO, Mrug M, Bennett WM, Harris PC, Erickson BJ, Torres VE, Kline TL. Utility of new image-derived biomarkers for autosomal dominant polycystic kidney disease prognosis using automated instance cyst segmentation. Kidney Int 2023; 104:334-342. [PMID: 36736536 PMCID: PMC10363210 DOI: 10.1016/j.kint.2023.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 02/03/2023]
Abstract
New image-derived biomarkers for patients affected by autosomal dominant polycystic kidney disease are needed to improve current clinical management. The measurement of total kidney volume (TKV) provides critical information for clinicians to drive care decisions. However, patients with similar TKV may present with very different phenotypes, often requiring subjective decisions based on other factors (e.g., appearance of healthy kidney parenchyma, a few cysts contributing significantly to overall TKV, etc.). In this study, we describe a new technique to individually segment cysts and quantify biometric parameters including cyst volume, cyst number, parenchyma volume, and cyst parenchyma surface area. Using data from the Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease (CRISP) study the utility of these new parameters was explored, both quantitatively as well as visually. Total cyst number and cyst parenchyma surface area showed superior prediction of the slope of estimated glomerular filtration rate decline, kidney failure and chronic kidney disease stages 3A, 3B, and 4, compared to TKV. In addition, presentations such as a few large cysts contributing significantly to overall kidney volume were shown to be much better stratified in terms of outcome predictions. Thus, these new image biomarkers, which can be obtained automatically, will have great utility in future studies and clinical care for patients affected by autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Adriana V Gregory
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Bhavya Poudyal
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Heather L Holmes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Alan S L Yu
- Jared Grantham Kidney Institute, Kansas University Medical Center, Kansas City, Kansas, USA; Division of Nephrology and Hypertension, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Douglas P Landsittel
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kyongtae T Bae
- Department of Diagnostic Radiology, University of Hong Kong, Hong Kong
| | - Arlene B Chapman
- Division of Nephrology, University of Chicago School of Medicine, Chicago, Illinois, USA
| | | | - Michal Mrug
- Division of Nephrology, University of Alabama and the Department of Veterans Affairs Medical Center, Birmingham, Alabama, USA
| | - William M Bennett
- Legacy Transplant Services, Legacy Good Samaritan Hospital, Portland, Oregon, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Bradley J Erickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy L Kline
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
22
|
Kistler AD. Function follows form: the quest for the best prognostic imaging biomarker in ADPKD. Kidney Int 2023; 104:239-241. [PMID: 37479385 DOI: 10.1016/j.kint.2023.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 07/23/2023]
Abstract
Total kidney volume represents the most solid prognostic biomarker for autosomal dominant polycystic kidney disease, because it mirrors cyst growth that precedes kidney function decline. Considerable variability of glomerular filtration rate trajectories, however, remains unexplained by total kidney volume, and its calculation is time-consuming. Using deep learning algorithms, Gregory et al. determined total kidney volume and other, novel, imaging-based biomarkers. They achieved automation and improved prognostic accuracy for long-term kidney function loss, yet the study leaves some open questions and room for further improvement.
Collapse
Affiliation(s)
- Andreas D Kistler
- Department of Medicine, Cantonal Hospital Frauenfeld, Spital Thurgau AG, Frauenfeld, Switzerland; University of Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Chebib FT, Zhou X, Garbinsky D, Davenport E, Nunna S, Oberdhan D, Fernandes A. Tolvaptan and Kidney Function Decline in Older Individuals With Autosomal Dominant Polycystic Kidney Disease: A Pooled Analysis of Randomized Clinical Trials and Observational Studies. Kidney Med 2023; 5:100639. [PMID: 37250503 PMCID: PMC10220412 DOI: 10.1016/j.xkme.2023.100639] [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] [Indexed: 05/31/2023] Open
Abstract
Rationale & Objective Tolvaptan is indicated for treatment of patients with autosomal dominant polycystic kidney disease (ADPKD) at risk of rapid progression. Participants aged 56-65 years constituted a small proportion of the Replicating Evidence of Preserved Renal Function: an Investigation of Tolvaptan Safety and Efficacy in ADPKD (REPRISE) trial population. We assessed effects of tolvaptan on estimated glomerular filtration rate (eGFR) decline in participants aged >55 years. Study Design This was a pooled data analysis from 8 studies of tolvaptan or non-tolvaptan standard of care (SOC). Setting & Participants Participants aged >55 years with ADPKD were included. Data on participants in >1 study were linked longitudinally for maximum follow-up duration, with matching for age, sex, eGFR, and chronic kidney disease (CKD) stage to minimize confounding. Interventions Tolvaptan or non-tolvaptan SOC. Outcomes Treatment effects on annualized eGFR decline were compared using mixed models with fixed effects for treatment, time, treatment-by-time interaction, and baseline eGFR. Results In the pooled studies, 230 tolvaptan-treated and 907 SOC participants were aged >55 years at baseline. Ninety-five participant pairs from each treatment group were matched, all in CKD G3 or G4, ranging from 56.0 to 65.0 years (tolvaptan) or from 55.1 to 67.0 years (SOC). The eGFR annual decline rate was significantly reduced by 1.66 mL/min/1.73 m2 (95% CI, 0.43-2.90; P = 0.009) in the tolvaptan group compared with SOC (-2.33 versus -3.99 mL/min/1.73 m2) over 3 years. Limitations Limitations include potential bias because of study population differences (bias risk was reduced through matching and multiple regression adjustment); vascular disease history data was not uniformly collected, and therefore not adjusted; and natural history of ADPKD precludes evaluating certain clinical endpoints within the study time frame. Conclusions In individuals aged 56-65 years with CKD G3 or G4, compared to a SOC group with mean GFR rate of decline ≥3 mL/min/1.73 m2/year, tolvaptan was associated with efficacy similar to that observed in the overall indication. Funding Otsuka Pharmaceutical Development & Commercialization, Inc (Rockville, MD). Trial Registration TEMPO 2:4 (NCT00413777); phase 1 tolvaptan trial (no NCT number; trial number 156-06-260); phase 2 tolvaptan trial (NCT01336972); TEMPO 4:4 (NCT01214421); REPRISE (NCT02160145); long-term tolvaptan safety extension trial (NCT02251275); OVERTURE (NCT01430494); HALT Progression of Polycystic Kidney Disease (HALT-PKD) study B (NCT01885559).
Collapse
Affiliation(s)
- Fouad T. Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL
| | - Xiaolei Zhou
- RTI Health Solutions, Research Triangle Park, NC
| | | | | | - Sasikiran Nunna
- Otsuka Pharmaceutical Development & Commercialization, Inc, Rockville, MD
| | - Dorothee Oberdhan
- Otsuka Pharmaceutical Development & Commercialization, Inc, Rockville, MD
| | - Ancilla Fernandes
- Otsuka Pharmaceutical Development & Commercialization, Inc, Rockville, MD
| |
Collapse
|
24
|
Singh S, Sreenidhi HC, Das P, Devi C. Predicting the Risk of Progression in Indian ADPKD Cohort using PROPKD Score - A Single-Center Retrospective Study. Indian J Nephrol 2023; 33:195-201. [PMID: 37448904 PMCID: PMC10337231 DOI: 10.4103/ijn.ijn_69_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/01/2022] [Accepted: 04/30/2022] [Indexed: 07/18/2023] Open
Abstract
Background With the variable genotype-phenotype expression of autosomal dominant polycystic kidney disease (ADPKD) and availability of novel targeted therapies, it is important to find predictors for rapid progression. The PROPKD score, consisting of genetic and clinical parameters like sex, hypertension, and urological events, is a useful tool in predicting the risk of progression. This study was aimed to determine the risk of ADPKD progression in Indian patients using the PROPKD score. Materials and Methods A retrospective study was done from 2006 to 2021. ADPKD patients with ESRD were included in the study. Scoring was done as per the PROPKD score as follows: male sex: 1, onset of hypertension before 35 years: 2, first urological event before 35 years: 2, PKD1 truncating mutation: 4, PKD1 non-truncating mutation: 2, and PKD2 mutation: 0. Two types of risk classifications were done as follows: (a) considering the clinical variables in all 73 patients (male sex, onset of hypertension before 35 years, and first urological event before 35 years), they were classified into three risk groups: low-risk group (0-1), intermediate-risk group (2-3), and high-risk group (4-5) and (b) considering the clinical variables and type of mutation in 39 patients, they were classified into three risk groups: low-risk group (0-3), intermediate-risk group (4-6), and high-risk group (7-9). Results Total number of patients included was 73, with the median age at ESRD being 54 years. High-risk group of clinical variables with hazard ratio (HR) of 4.570 (2.302-9.075, P < 0.001) and high-risk group of the PROPKD score with HR of 6.594 (1.868-23.284, P = 0.003) were associated with early ESRD. High-risk groups of both classifications were associated with early ESRD. Conclusion High-risk groups based on the PROPKD scoring and clinical variables were associated with early progression to ESRD.
Collapse
Affiliation(s)
- Shivendra Singh
- Department of Nephrology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - HC Sreenidhi
- Department of Nephrology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Parimal Das
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Chandra Devi
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
25
|
Yu ASL, Landsittel DP. Biomarkers in Polycystic Kidney Disease: Are We There? ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:285-293. [PMID: 37088529 DOI: 10.1053/j.akdh.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 04/25/2023]
Abstract
This article describes the use of prognostic, predictive, and response biomarkers that have been developed for autosomal dominant polycystic kidney disease and their use in clinical care or drug development. We focus on biochemical markers that can be assayed in patients' blood and urine and their association with the outcome of decreased glomerular filtration rate. There have been several studies on prognostic biomarkers. The most promising ones have been markers of tubular injury, inflammation, metabolism, or the vasopressin-urinary concentration axis. So far, none have been shown to be superior to kidney volume-based biomarkers. Several biomarkers are additive to kidney volume and genotype in prognostic models, but there have been few direct comparisons between the biochemical markers to identify the best ones. Moreover, there is a lack of uniformity in the statistical tools used to assess and compare biomarkers. There have been few reports of predictive and response biomarkers, and none are suitable surrogate endpoints. The U.S. Food and Drug Administration's Biomarker Qualification Program provides a regulatory pathway to approve biomarkers for use across multiple drug-development programs.
Collapse
Affiliation(s)
- Alan S L Yu
- Division of Nephrology and Hypertension and the Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS.
| | - Douglas P Landsittel
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, IN
| |
Collapse
|
26
|
Perrone RD, Oberdhan D, Ouyang J, Bichet DG, Budde K, Chapman AB, Gitomer BY, Horie S, Ong AC, Torres VE, Turner AN, Krasa H. OVERTURE: A Worldwide, Prospective, Observational Study of Disease Characteristics in Patients With ADPKD. Kidney Int Rep 2023; 8:989-1001. [PMID: 37180499 PMCID: PMC10166786 DOI: 10.1016/j.ekir.2023.02.1073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction The course of autosomal dominant polycystic kidney disease (ADPKD) varies greatly among affected individuals, necessitating natural history studies to characterize the determinants and effects of disease progression. Therefore, we conducted an observational, longitudinal study (OVERTURE; NCT01430494) of patients with ADPKD. Methods This prospective study enrolled a large international population (N = 3409) encompassing a broad spectrum of ages (12-78 years), chronic kidney disease (CKD) stages (G1-G5), and Mayo imaging classifications (1A-1E). Outcomes included kidney function, complications, quality of life, health care resource utilization, and work productivity. Results Most subjects (84.4%) completed ≥12 months of follow-up. Consistent with earlier findings, each additional l/m of height-adjusted total kidney volume (htTKV) on magnetic resonance imaging (MRI) was associated with worse outcomes, including lower estimated glomerular filtration rate (eGFR) (regression coefficient 17.02, 95% confidence interval [CI] 15.94-18.11) and greater likelihood of hypertension (odds ratio [OR] 1.25, 95% CI 1.17-1.34), kidney pain (OR 1.22, 95% CI 1.11-1.33), and hematuria (OR 1.35, 95% CI 1.21-1.51). Greater baseline htTKV was also associated with worse patient-reported health-related quality of life (e.g., ADPKD Impact Scale physical score, regression coefficient 1.02, 95% CI 0.65-1.39), decreased work productivity (e.g., work days missed, regression coefficient 0.55, 95% CI 0.18-0.92), and increased health care resource utilization (e.g., hospitalizations, OR 1.48, 95% CI 1.33-1.64) during follow-up. Conclusion Although limited by a maximum 3-year duration of follow-up, this observational study characterized the burden of ADPKD in a broad population and indicated the predictive value of kidney volume for outcomes other than kidney function.
Collapse
Affiliation(s)
- Ronald D. Perrone
- Division of Nephrology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Dorothee Oberdhan
- Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland, USA
| | - John Ouyang
- Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, Maryland, USA
| | - Daniel G. Bichet
- Division of Nephrology, Département de Médecine, Pharmacologie et Physiologie, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Quebec, Canada
| | - Klemens Budde
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Arlene B. Chapman
- Section of Nephrology, University of Chicago School of Medicine, Chicago, Illinois, USA
| | - Berenice Y. Gitomer
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Albert C.M. Ong
- Kidney Genetics Group, Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, UK
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - A. Neil Turner
- Renal and Autoimmunity Group, MRC Center for Inflammation, University of Edinburgh, Edinburgh, UK
| | - Holly Krasa
- Blue Persimmon Group LLC, Washington, District of Columbia, USA
| |
Collapse
|
27
|
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
|
28
|
Luo L, Roy S, Li L, Ma M. Polycystic kidney disease: novel insights into polycystin function. Trends Mol Med 2023; 29:268-281. [PMID: 36805211 DOI: 10.1016/j.molmed.2023.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/17/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a life-threatening monogenic disease caused by mutations in PKD1 and PKD2 that encode polycystin 1 (PC1) and polycystin 2 (PC2). PC1/2 localize to cilia of renal epithelial cells, and their function is believed to embody an inhibitory activity that suppresses the cilia-dependent cyst activation (CDCA) signal. Consequently, PC deficiency results in activation of CDCA and stimulates cyst growth. Recently, re-expression of PCs in established cysts has been shown to reverse PKD. Thus, the mode of action of PCs resembles a 'counterbalance in cruise control' to maintain lumen diameter within a designated range. Herein we review recent studies that point to novel arenas for future PC research with therapeutic potential for ADPKD.
Collapse
Affiliation(s)
- Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, 400715, China
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, 61 Biopolis Drive, Singapore, 138673, Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore, 119288, Singapore; Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Li Li
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, 400715, China; Research Center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ming Ma
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, 400715, China.
| |
Collapse
|
29
|
Ryu H, Hong Y, Kang E, Kang M, Kim J, Park HC, Oh YK, Chin HJ, Park SK, Jung JY, Hyun YY, Sung SA, Ahn C, Oh KH. Comparison of outcomes of chronic kidney disease based on etiology: a prospective cohort study from KNOW-CKD. Sci Rep 2023; 13:3570. [PMID: 36864195 PMCID: PMC9981888 DOI: 10.1038/s41598-023-29844-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/10/2023] [Indexed: 03/04/2023] Open
Abstract
The causes of chronic kidney disease (CKD) affects its outcomes. However, the relative risks for adverse outcomes according to specific causes of CKD is not well established. In a prospective cohort study from KNOW-CKD, a cohort was analyzed using overlap propensity score weighting methods. Patients were grouped into four categories according to the cause of CKD: glomerulonephritis (GN), diabetic nephropathy (DN), hypertensive nephropathy (HTN), or polycystic kidney disease (PKD). From a total of 2070 patients, the hazard ratio of kidney failure, the composite of cardiovascular disease (CVD) and mortality, and the slope of the estimated glomerular filtration rate (eGFR) decline according to the cause of CKD were compared between causative groups in a pairwise manner. There were 565 cases of kidney failure and 259 cases of composite CVD and death over 6.0 years of follow-up. Patients with PKD had a significantly increased risk for kidney failure compared to those with GN [Hazard ratio (HR) 1.82], HTN (HR 2.23), and DN (HR 1.73). For the composite outcome of CVD and death, the DN group had increased risks compared to the GN (HR 2.07), and HTN (HR 1.73) groups but not to the PKD group. The adjusted annual eGFR change for the DN and PKD groups were - 3.07 and - 3.37 mL/min/1.73 m2 per year, respectively, and all of these values were significantly different than those of the GN and HTN groups (- 2.16 and - 1.42 mL/min/1.73 m2 per year, respectively). In summary, the risk of kidney disease progression was relatively higher in patients with PKD compared to other causes of CKD. However, the composite of CVD and death was relatively higher in patients with DN-related CKD than in those with GN- and HTN-related CKD.
Collapse
Grants
- 2011E3300300, 2012E3301100, 2013E3301600, 2013E3301601, 2013E3301602, 2016E3300200, 2016E3300201, 2016E3300202, and 2019E320100 Korea Centers for Disease Control and Prevention
- 2011E3300300, 2012E3301100, 2013E3301600, 2013E3301601, 2013E3301602, 2016E3300200, 2016E3300201, 2016E3300202, and 2019E320100 Korea Centers for Disease Control and Prevention
- 2011E3300300, 2012E3301100, 2013E3301600, 2013E3301601, 2013E3301602, 2016E3300200, 2016E3300201, 2016E3300202, and 2019E320100 Korea Centers for Disease Control and Prevention
- 2017M3A9E4044649 National Research Foundation (NRF)& funded by the Korean government (MSIT)
- National Research Foundation (NRF)& funded by the Korean government (MSIT)
Collapse
Affiliation(s)
- Hyunjin Ryu
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yeji Hong
- Rehabilitation Medical Research Center, Korea Workers' Compensation and Welfare Service Incheon Hospital, Incheon, Republic of Korea
| | - Eunjeong Kang
- Department of Internal Medicine, Ewha Womans University Medical Center, Seoul, Republic of Korea
| | - Minjung Kang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jayoun Kim
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hayne Cho Park
- Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Ho Jun Chin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sue K Park
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Yong Jung
- Division of Nephrology, Department of Internal Medicine, Gachon University of Gil Medical Center, Gachon University School of Medicine, Incheon, Korea
| | - Young Youl Hyun
- Department of Internal Medicine, Kangbuk Samsung Hospital, College of Medicine, Sungkyunkwan University, Seoul, Korea
| | - Su Ah Sung
- Department of Internal Medicine, Eulji Medical Center, Eulji University, Seoul, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | | |
Collapse
|
30
|
Ortiz A, Ars E, Bernis C, Fraga G, Furlano M, Martínez V, Martins J, Pérez-Gómez MV, Rodríguez-Pérez JC, Sans L, Torra R. Reply to Comments on the SENefro Consensus Document on Autosomal Dominant Polycystic Kidney Disease. Nefrologia 2023; 43:152-153. [PMID: 37217372 DOI: 10.1016/j.nefroe.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 05/24/2023] Open
Affiliation(s)
- Alberto Ortiz
- Servicio de Nefrología, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, RICORS2040, Madrid, Spain
| | - Elisabet Ars
- Laboratorio de Biología Molecular, Fundació Puigvert, Institut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Carmen Bernis
- Servicio de Nefrología, Hospital de la Princesa, Instituto de Investigación Carlos III, Madrid, Spain
| | - Gloria Fraga
- Sección de Nefrología Pediátrica, Hospital de la Santa Creu i Sant Pau, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Mónica Furlano
- Enfermedades Renales Hereditarias, Servicio de Nefrología, Fundació Puigvert, Institut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau), Universidad Autónoma de Barcelona (Departamento de Medicina), Barcelona, Spain
| | - Víctor Martínez
- Servicio de Nefrología, Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Judith Martins
- Servicio de Nefrología, Hospital Universitario de Getafe, Universidad Europea de Madrid, Getafe, Madrid, Spain
| | - Maria Vanessa Pérez-Gómez
- Servicio de Nefrología, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, RICORS2040, Madrid, Spain
| | - José Carlos Rodríguez-Pérez
- Servicio de Nefrología, Hospital Universitario de Gran Canaria Dr. Negrín, Universidad de Las Palmas de Gran Canaria, Spain
| | - Laia Sans
- Servicio de Nefrología, Hospital del Mar, Barcelona, Spain
| | - Roser Torra
- Enfermedades Renales Hereditarias, Servicio de Nefrología, Fundació Puigvert, Institut d'Investigacions Biomèdiques Sant Pau (IIB-Sant Pau), Universidad Autónoma de Barcelona (Departamento de Medicina), Barcelona, Spain.
| |
Collapse
|
31
|
Espinosa Cabello M, Ansio Vázquez I, Espejo Portero I, Rodriguez Fuentes D, Rabasco Ruiz C, Espinosa Hernández M. The natural history of autosomal dominant polycystic kidney disease. A strategy for grouping families and mutations. Nefrologia 2023; 43:120-125. [PMID: 37268502 DOI: 10.1016/j.nefroe.2023.03.010] [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: 12/20/2021] [Accepted: 05/17/2022] [Indexed: 06/04/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a main cause of end-stage renal disease. Today, knowledge of its genetic basis has made it possible to develop strategies that prevent the transmission of the disease. OBJECTIVES The objective of the study was to analyze the natural history of ADPKD in the province of Córdoba and to design a database that allows grouping families with different mutations. PATIENTS AND METHODS All patients (n = 678) diagnosed with ADPKD followed by the Córdoba nephrology service are included. Various clinical variables (age and sex), genetic variables (mutation in PKD1, PKD2) and the need for renal replacement therapy (RRT) were retrospectively analyzed. RESULTS The prevalence was 61 cases per 100,000 inhabitants. Median renal survival was significantly worse in PKD1 (57.5 years) than in PKD2 (70 years) (log-rank p = 0.000). We have genetically identified 43.8% of the population, detecting PKD1 mutations in 61.2% and PKD2 mutations in 37.4% of cases, respectively. The most frequent mutation, in PKD2 (c.2159del), appeared in 68 patients belonging to 10 different families. The one with the worst renal prognosis was a truncating mutation in PKD1 (c.9893 G > A). These patients required RRT at a median age of 38.7 years. CONCLUSIONS Renal survival of ADPKD in the province of Córdoba is similar to that described in the literature. We detected PKD2 mutations in 37.4% of cases. This strategy allows us to know the genetic basis of a large proportion of our population while saving resources. This is essential to be able to offer primary prevention of ADPKD through preimplantation genetic diagnosis.
Collapse
Affiliation(s)
| | | | - Isabel Espejo Portero
- UGC de Análisis Clínicos, Genética-Molecular, Hospital Universitario Reina Sofía. Córdoba, Spain
| | | | | | | |
Collapse
|
32
|
Mader G, Mladsi D, Sanon M, Purser M, Barnett CL, Oberdhan D, Watnick T, Seliger S. A disease progression model estimating the benefit of tolvaptan on time to end-stage renal disease for patients with rapidly progressing autosomal dominant polycystic kidney disease. BMC Nephrol 2022; 23:334. [PMID: 36258169 PMCID: PMC9578187 DOI: 10.1186/s12882-022-02956-8] [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: 05/11/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background Tolvaptan was approved in the United States in 2018 for patients with autosomal dominant polycystic kidney disease (ADPKD) at risk of rapid progression as assessed in a 3-year phase 3 clinical trial (TEMPO 3:4). An extension study (TEMPO 4:4) showed continued delay in progression at 2 years, and a trial in patients with later-stage disease (REPRISE) provided confirmatory evidence of efficacy. Given the relatively shorter-term duration of the clinical trials, estimating the longer-term benefit associated with tolvaptan via extrapolation of the treatment effect is an important undertaking. Methods A model was developed to simulate a cohort of patients with ADPKD at risk of rapid progression and predict their long-term outcomes using an algorithm organized around the Mayo Risk Classification system, which has five subclasses (1A through 1E) based on estimated kidney growth rates. The model base-case population represents 1280 patients enrolled in TEMPO 3:4 beginning in chronic kidney disease (CKD) stages G1, G2, and G3 across Mayo subclasses 1C, 1D, and 1E. The algorithm was used to predict longer-term natural history health outcomes. The estimated treatment effect of tolvaptan from TEMPO 3:4 was applied to the natural history to predict the longer-term treatment benefit of tolvaptan. For the cohort, analyzed once reflecting natural history and once assuming treatment with tolvaptan, the model estimated lifetime progression through CKD stages, end-stage renal disease (ESRD), and death. Results When treated with tolvaptan, the model cohort was predicted to experience a 3.1-year delay of ESRD (95% confidence interval: 1.8 to 4.4), approximately a 23% improvement over the estimated 13.7 years for patients not receiving tolvaptan. Patients beginning tolvaptan treatment in CKD stages G1, G2, and G3 were predicted to experience estimated delays of ESRD, compared with patients not receiving tolvaptan, of 3.8 years (21% improvement), 3.0 years (24% improvement), and 2.1 years (28% improvement), respectively. Conclusions The model estimated that patients treated with tolvaptan versus no treatment spent more time in earlier CKD stages and had later onset of ESRD. Findings highlight the potential long-term value of early intervention with tolvaptan in patients at risk of rapid ADPKD progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02956-8.
Collapse
Affiliation(s)
- Gregory Mader
- RTI Health Solutions, Research Triangle Park, NC, USA
| | | | - Myrlene Sanon
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, USA
| | - Molly Purser
- RTI Health Solutions, Research Triangle Park, NC, USA
| | | | - Dorothee Oberdhan
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, USA
| | - Terry Watnick
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephen Seliger
- University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Echocardiographic Abnormalities in Autosomal Dominant Polycystic Kidney Disease (ADPKD) Patients. J Clin Med 2022; 11:jcm11205982. [PMID: 36294302 PMCID: PMC9604303 DOI: 10.3390/jcm11205982] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/21/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular abnormalities, such as left ventricular hypertrophy and valvular disorders, particularly mitral valve prolapse, have been described as highly prevalent among adult patients with autosomal dominant polycystic kidney disease (ADPKD). The present study aimed to assess echocardiographic parameters in a large sample of both normotensive and hypertensive ADPKD patients, regardless of kidney function level, and evaluate their association with clinical and laboratorial parameters. A retrospective study consisted of the analysis of clinical, laboratorial, and transthoracic echocardiograms data retrieved from the medical records of young adult ADPKD outpatients. A total of 294 patients (120 M/174 F, 41.0 ± 13.8 years old, 199 hypertensive and 95 normotensive) with a median estimated glomerular filtration rate (eGFR) of 75.5 mL/min/1.73 m2 were included. The hypertensive group (67.6%) was significantly older and exhibited significantly lower eGFR than the normotensive one. Increased left ventricular mass index (LVMI) was seen in 2.0%, mitral valve prolapse was observed in 3.4%, mitral valve regurgitation in 15.3%, tricuspid valve regurgitation in 16.0%, and aortic valve regurgitation in 4.8% of the whole sample. The present study suggested that the prevalence of mitral valve prolapse was much lower than previously reported, and increased LVMI was not seen in most adult ADPKD patients.
Collapse
|
34
|
Perrone RD, Hariri A, Minini P, Ahn C, Chapman AB, Horie S, Knebelmann B, Mrug M, Ong AC, Pei YP, Torres VE, Modur V, Gansevoort RT. The STAGED-PKD 2-Stage Adaptive Study With a Patient Enrichment Strategy and Treatment Effect Modeling for Improved Study Design Efficiency in Patients With ADPKD. Kidney Med 2022; 4:100538. [PMID: 36204243 PMCID: PMC9529969 DOI: 10.1016/j.xkme.2022.100538] [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] [Indexed: 11/15/2022] Open
Abstract
Rationale & Objective Venglustat, a glucosylceramide synthase inhibitor, inhibits cyst growth and reduces kidney failure in mouse models of autosomal dominant polycystic kidney disease (ADPKD). STAGED-PKD aims to determine the safety and efficacy of venglustat and was designed using patient enrichment for progression to end-stage kidney disease and modeling from prior ADPKD trials. Study Design STAGED-PKD is a 2-stage, international, double-blind, randomized, placebo-controlled trial in adults with ADPKD (Mayo Class 1C-1E) and estimated glomerular filtration rate (eGFR) 45-<90 mL/min/1.73 m2 at risk of rapidly progressive disease. Enrichment for rapidly progressing patients was identified based on retrospective analysis of total kidney volume (TKV) and eGFR slope from the combined Consortium for Radiologic Imaging Studies of Polycystic Kidney Disease and HALT Progression of Polycystic Kidney Disease A studies. Setting & Participants Target enrollment in stages 1 and 2 was 240 and 320 patients, respectively. Interventions Stage 1 randomizes patients 1:1:1 to venglustat 8 mg or 15 mg once daily or placebo. Stage 2 randomizes patients 1:1 to placebo or venglustat, with the preferred dose based on stage 1 safety data. Outcomes Primary endpoints are TKV growth rate over 18 months in stage 1 and eGFR slope over 24 months in stage 2. Secondary endpoints include: annualized rate of change in eGFR from baseline to 18 months (stage 1); annualized rate of change in TKV based on magnetic resonance imaging from baseline to 18 months (stage 2); and safety, tolerability, pain, and fatigue (stages 1 and 2). Limitations If stage 1 is unsuccessful, patients enrolled in the trial may develop drug-related adverse events that can have long-lasting effects. Conclusions Modeling allows the design and powering of a 2-stage combined study to assess venglustat’s impact on TKV growth and eGFR slope. Stage 1 TKV assessment via a nested approach allows early evaluation of efficacy and increased efficiency of the trial design by reducing patient numbers and trial duration. Funding This study was funded by Sanofi. Trial registration STAGED-PKD has been registered at ClinicalTrials.gov with study number NCT03523728.
Collapse
Affiliation(s)
- Ronald D. Perrone
- Division of Nephrology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts
| | - Ali Hariri
- Eloxx Pharmaceuticals, Watertown, Massachusetts
| | | | - Curie Ahn
- Department of Internal Medicine, Seoul National University, Seoul, Republic of Korea
| | | | - Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Bertrand Knebelmann
- Université de Paris, AP-HP, Service de Néphrologie, Hôpital Necker-Enfants Malades, Paris, France
| | - Michal Mrug
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama and Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - Albert C.M. Ong
- Academic Nephrology Unit, Department of Infection Immunity & Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - York P.C. Pei
- Division of Nephrology, University of Toronto, Toronto, Ontario, Canada
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Vijay Modur
- Eloxx Pharmaceuticals, Watertown, Massachusetts
| | - Ronald T. Gansevoort
- Department of Nephrology, University Medical Center Groningen, The Netherlands
- Address for Correspondence: Ronald T. Gansevoort, MD, PhD, Department of Nephrology, University Medical Center Groningen, The Netherlands.
| |
Collapse
|
35
|
Sharbatdaran A, Romano D, Teichman K, Dev H, Raza SI, Goel A, Moghadam MC, Blumenfeld JD, Chevalier JM, Shimonov D, Shih G, Wang Y, Prince MR. Deep Learning Automation of Kidney, Liver, and Spleen Segmentation for Organ Volume Measurements in Autosomal Dominant Polycystic Kidney Disease. Tomography 2022; 8:1804-1819. [PMID: 35894017 PMCID: PMC9326744 DOI: 10.3390/tomography8040152] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/01/2022] [Accepted: 07/08/2022] [Indexed: 12/02/2022] Open
Abstract
Organ volume measurements are a key metric for managing ADPKD (the most common inherited renal disease). However, measuring organ volumes is tedious and involves manually contouring organ outlines on multiple cross-sectional MRI or CT images. The automation of kidney contouring using deep learning has been proposed, as it has small errors compared to manual contouring. Here, a deployed open-source deep learning ADPKD kidney segmentation pipeline is extended to also measure liver and spleen volumes, which are also important. This 2D U-net deep learning approach was developed with radiologist labeled T2-weighted images from 215 ADPKD subjects (70% training = 151, 30% validation = 64). Additional ADPKD subjects were utilized for prospective (n = 30) and external (n = 30) validations for a total of 275 subjects. Image cropping previously optimized for kidneys was included in training but removed for the validation and inference to accommodate the liver which is closer to the image border. An effective algorithm was developed to adjudicate overlap voxels that are labeled as more than one organ. Left kidney, right kidney, liver and spleen labels had average errors of 3%, 7%, 3%, and 1%, respectively, on external validation and 5%, 6%, 5%, and 1% on prospective validation. Dice scores also showed that the deep learning model was close to the radiologist contouring, measuring 0.98, 0.96, 0.97 and 0.96 on external validation and 0.96, 0.96, 0.96 and 0.95 on prospective validation for left kidney, right kidney, liver and spleen, respectively. The time required for manual correction of deep learning segmentation errors was only 19:17 min compared to 33:04 min for manual segmentations, a 42% time saving (p = 0.004). Standard deviation of model assisted segmentations was reduced to 7, 5, 11, 5 mL for right kidney, left kidney, liver and spleen respectively from 14, 10, 55 and 14 mL for manual segmentations. Thus, deep learning reduces the radiologist time required to perform multiorgan segmentations in ADPKD and reduces measurement variability.
Collapse
Affiliation(s)
- Arman Sharbatdaran
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Dominick Romano
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Kurt Teichman
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Hreedi Dev
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Syed I. Raza
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Akshay Goel
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Mina C. Moghadam
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Jon D. Blumenfeld
- The Rogosin Institute and Department of Medicine Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (J.D.B.); (J.M.C.); (D.S.)
| | - James M. Chevalier
- The Rogosin Institute and Department of Medicine Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (J.D.B.); (J.M.C.); (D.S.)
| | - Daniil Shimonov
- The Rogosin Institute and Department of Medicine Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (J.D.B.); (J.M.C.); (D.S.)
| | - George Shih
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
| | - Yi Wang
- Departments of Radiology at Weill Cornell Medicine and Biomedical Engineering, Cornell University, New York, NY 10065, USA;
| | - Martin R. Prince
- Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; (A.S.); (D.R.); (K.T.); (H.D.); (S.I.R.); (A.G.); (M.C.M.); (G.S.)
- Columbia College of Physicians and Surgeons, Cornell University, New York, NY 10027, USA
| |
Collapse
|
36
|
Radhakrishnan Y, Duriseti P, Chebib FT. Management of autosomal dominant polycystic kidney disease in the era of disease-modifying treatment options. Kidney Res Clin Pract 2022; 41:422-431. [PMID: 35354242 PMCID: PMC9346401 DOI: 10.23876/j.krcp.21.309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 11/04/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the reported etiology in 10% of end-stage kidney disease (ESKD) patients and has an estimated prevalence of 12.5 million cases worldwide across all ethnicities. There have been major advancements over the last two decades in understanding the pathogenesis and development of disease-modifying treatment options for ADPKD, culminating in regulatory approval of tolvaptan for ADPKD patients at risk of rapid progression to kidney failure. This review highlights the genetic mutations associated with ADPKD, defines patients at risk of rapid progression to ESKD, and focuses on the management of ADPKD in the era of disease-modifying agents.
Collapse
Affiliation(s)
- Yeshwanter Radhakrishnan
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Parikshit Duriseti
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Fouad T. Chebib
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
37
|
Ortiz A, Ars E, Bernis C, Fraga G, Furlano M, Martínez V, Martins J, Pérez-Gómez MV, Rodríguez-Pérez JC, Sans L, Torra R. Respuesta a Comentarios sobre el Documento de Consenso de Poliquistosis Renal Autosómica Dominante de la SENefro. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
38
|
Historia natural de la poliquistosis renal autosómica dominante en Córdoba: utilidad de una base de datos para agrupar familias y mutaciones. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
39
|
Rasmussen R, Sanford T, Parwani AV, Pedrosa I. Artificial Intelligence in Kidney Cancer. Am Soc Clin Oncol Educ Book 2022; 42:1-11. [PMID: 35580292 DOI: 10.1200/edbk_350862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Artificial intelligence is rapidly expanding into nearly all facets of life, particularly within the field of medicine. The diagnosis, characterization, management, and treatment of kidney cancer is ripe with areas for improvement that may be met with the promises of artificial intelligence. Here, we explore the impact of current research work in artificial intelligence for clinicians caring for patients with renal cancer, with a focus on the perspectives of radiologists, pathologists, and urologists. Promising preliminary results indicate that artificial intelligence may assist in the diagnosis and risk stratification of newly discovered renal masses and help guide the clinical treatment of patients with kidney cancer. However, much of the work in this field is still in its early stages, limited in its broader applicability, and hampered by small datasets, the varied appearance and presentation of kidney cancers, and the intrinsic limitations of the rigidly structured tasks artificial intelligence algorithms are trained to complete. Nonetheless, the continued exploration of artificial intelligence holds promise toward improving the clinical care of patients with kidney cancer.
Collapse
Affiliation(s)
- Robert Rasmussen
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Thomas Sanford
- Department of Urology, Upstate Medical University, Syracuse, NY
| | - Anil V Parwani
- Department of Pathology, The Ohio State University, Columbus, OH
| | - Ivan Pedrosa
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX.,Department of Urology, The University of Texas Southwestern Medical Center, Dallas, TX.,Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
40
|
Akbari P, Nasri F, Deng S, Khowaja S, Lee S, Warnica W, Lu H, Rattansingh A, Atri M, Khalili K, Pei Y. Total Kidney Volume Measurements in ADPKD by 3D and Ellipsoid Ultrasound in Comparison to Magnetic Resonance Imaging. Clin J Am Soc Nephrol 2022; 17:827-834. [PMID: 35383043 PMCID: PMC9269662 DOI: 10.2215/cjn.14931121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/23/2022] [Indexed: 11/23/2022]
Abstract
Background and Objectives: Total kidney volume (TKV) is a validated prognostic biomarker for autosomal dominant polycystic kidney disease (ADPKD). TKV by magnetic resonance imaging (MRI) and manual segmentation is considered the "reference standard", but is time-consuming and not readily accessible. By contrast, 3-dimensional ultrasound (3D ultrasound) provides a promising technology for TKV measurements with unknown potential. Here, we report a comparative study of TKV measurements by 3D ultrasound vs. the conventional methods by ultrasound ellipsoid and MRI ellipsoid. Design, setting, participants, and measurements: Single-center prospective study of 142 patients who completed a standardized 3D ultrasound and MRI. TKV by 3D ultrasound and ultrasound ellipsoid were compared to those by MRI. We assessed the agreement of TKV measurements by Bland-Altman plots and misclassification of the Mayo Clinic Imaging Classes (MCIC) between the different imaging methods, and prediction of MCIC 1C-1E by average ultrasound kidney length >16.5 cm. Results: Compared to MRI manual segmentation, MRI ellipsoid, 3D ultrasound, and ultrasound ellipsoid underestimated TKV (mean difference: -3.2%, -9.1%, and -11.0%) with MCIC misclassified in 11%, 21% and 22% of patients, respectively; most misclassified cases by MRI ellipsoid (11/16), 3D ultrasound (23/30), and ultrasound ellipsoid (26/31) were placed into a lower MCIC. Prediction of the high-risk MCIC (1C-1E) by MRI ellipsoid, 3D ultrasound, and ultrasound ellipsoid all yielded high positive predictive value (96%, 95%, 98%), and specificity (96%, 96%, 99%). However, both negative predictive value (90%, 88%, 95%) and sensitivity (88%, 85%, 94%) were lower for 3D ultrasound and ultrasound ellipsoid compared to MRI ellipsoid. An average ultrasound kidney length >16.5 cm was highly predictive of MCIC 1C-1E only in patients aged <45 years. Conclusions: TKV measurements in ADPKD by 3D ultrasound and ultrasound ellipsoid displayed similar bias, variability, and are less accurate than MRI ellipsoid. Prediction of high-risk MCIC (1C-1E) by all three methods provides high positive predictive value, but ultrasound ellipsoid is simpler to use and more readily available.
Collapse
Affiliation(s)
- Pedram Akbari
- P Akbari, Division of Nephrology, University Health Network, Toronto, Canada
| | - Fatemeh Nasri
- F Nasri, Department of Medical Imaging, University Health Network, Toronto, Canada
| | - Shirley Deng
- S Deng, Division of Nephrology, University Health Network, Toronto, Canada
| | - Saima Khowaja
- S Khowaja, Division of Nephrology, University Health Network, Toronto, Canada
| | - Seung Lee
- S Lee, Division of Nephrology, University Health Network, Toronto, Canada
| | - William Warnica
- W Warnica, Department of Medical Imaging, University Health Network, Toronto, Canada
| | - Hua Lu
- H Lu, Cell Biology Program, Hospital for Sick Children Research Institute, Toronto, Canada
| | - Anand Rattansingh
- A Rattansingh, Department of Medical Imaging, University Health Network, Toronto, Canada
| | - Mostafa Atri
- M Atri, Department of Medical Imaging, University Health Network, Toronto, Canada
| | - Korosh Khalili
- K Khalili, Department of Medical Imaging, University Health Network, Toronto, Canada
| | - York Pei
- Y Pei, Division of Nephrology, University Health Network, Toronto, Canada
| |
Collapse
|
41
|
Park HC, Hong Y, Yeon JH, Ryu H, Kim YC, Lee J, Kim YH, Chae DW, Chung W, Ahn C, Oh KH, Oh YK. Mayo imaging classification is a good predictor of rapid progress among Korean patients with autosomal dominant polycystic kidney disease: results from the KNOW-CKD study. Kidney Res Clin Pract 2022; 41:432-441. [PMID: 35286789 PMCID: PMC9346394 DOI: 10.23876/j.krcp.21.261] [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: 11/03/2021] [Accepted: 12/29/2021] [Indexed: 11/25/2022] Open
Abstract
Background Mayo imaging classification (MIC) is a useful biomarker to predict disease progression in autosomal dominant polycystic kidney disease (ADPKD). This study was performed to validate MIC in the prediction of renal outcome in a prospective Korean ADPKD cohort and evaluate clinical parameters associated with rapid disease progression. Methods A total of 178 ADPKD patients were enrolled and prospectively observed for an average duration of 6.2 ± 1.9 years. Rapid progressor was defined as MIC 1C through 1E while slow progressor was defined as 1A through 1B. Renal composite outcome (doubling of serum creatinine, 50% decline of estimated glomerular filtration rate [eGFR], or initiation of renal replacement therapy) as well as the annual percent change of height-adjusted total kidney volume (mHTKV-α) and eGFR decline (mGFR-α) were compared between groups. Results A total of 110 patients (61.8%) were classified as rapid progressors. These patients were younger and showed a higher proportion of male patients. Rapid progressor was an independent predictor for renal outcome (hazard ratio, 4.09; 95% confidence interval, 1.23–13.54; p = 0.02). The mGFR-α was greater in rapid progressors (–3.58 mL/min per year in 1C, –3.7 in 1D, and –4.52 in 1E) compared with that in slow progressors (–1.54 in 1A and –2.06 in 1B). The mHTKV-α was faster in rapid progressors (5.3% per year in 1C, 9.4% in 1D, and 11.7% in 1E) compared with that in slow progressors (1.2% in 1A and 3.8% in 1B). Conclusion MIC is a good predictive tool to define rapid progressors in Korean ADPKD patients.
Collapse
Affiliation(s)
- Hayne Cho Park
- Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Republic of Korea
- Kidney Research Institute, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Yeji Hong
- Rehabilitation Medical Research Center, Korea Workers’ Compensation and Welfare Service Incheon Hospital, Incheon, Republic of Korea
| | - Jeong-Heum Yeon
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yong-Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Joongyub Lee
- Department of Prevention and Management, Inha University School of Medicine, Incheon, Republic of Korea
| | - Yeong Hoon Kim
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Dong-Wan Chae
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - WooKyung Chung
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Curie Ahn
- Department of Internal Medicine, National Medical Center, Seoul, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
- Correspondence: Yun Kyu Oh Department of Internal Medicine, SMG-SNU Boramae Medical Center, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul 07061, Republic of Korea. E-mail:
| |
Collapse
|
42
|
Pickel L, Iliuta IA, Scholey J, Pei Y, Sung HK. Dietary Interventions in Autosomal Dominant Polycystic Kidney Disease. Adv Nutr 2022; 13:652-666. [PMID: 34755831 PMCID: PMC8970828 DOI: 10.1093/advances/nmab131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the progressive growth of renal cysts, leading to the loss of functional nephrons. Recommendations for individuals with ADPKD to maintain a healthy diet and lifestyle are largely similar to those for the general population. However, recent evidence from preclinical models suggests that more tightly specified dietary regimens, including caloric restriction, intermittent fasting, and ketogenic diets, hold promise to slow disease progression, and the results of ongoing human clinical trials are eagerly awaited. These dietary interventions directly influence nutrient signaling and substrate availability in the cystic kidney, while also conferring systemic metabolic benefits. The present review focuses on the importance of local and systemic metabolism in ADPKD and summarizes current evidence for dietary interventions to slow disease progression and improve quality of life.
Collapse
Affiliation(s)
- Lauren Pickel
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ioan-Andrei Iliuta
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - James Scholey
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - York Pei
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
43
|
Zhou X, Davenport E, Ouyang J, Hoke ME, Garbinsky D, Agarwal I, Krasa HB, Oberdhan D. Pooled Data Analysis of the Long-Term Treatment Effects of Tolvaptan in ADPKD. Kidney Int Rep 2022; 7:1037-1048. [PMID: 35570988 PMCID: PMC9091612 DOI: 10.1016/j.ekir.2022.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/11/2022] [Accepted: 02/07/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Methods Results Conclusion
Collapse
|
44
|
Müller RU, Messchendorp AL, Birn H, Capasso G, Cornec-Le Gall E, Devuyst O, van Eerde A, Guirchoun P, Harris T, Hoorn EJ, Knoers NVAM, Korst U, Mekahli D, Le Meur Y, Nijenhuis T, Ong ACM, Sayer JA, Schaefer F, Servais A, Tesar V, Torra R, Walsh SB, Gansevoort RT. An update on the use of tolvaptan for ADPKD: Consensus statement on behalf of the ERA Working Group on Inherited Kidney Disorders (WGIKD), the European Rare Kidney Disease Reference Network (ERKNet) and Polycystic Kidney Disease International (PKD-International). Nephrol Dial Transplant 2021; 37:825-839. [PMID: 35134221 PMCID: PMC9035348 DOI: 10.1093/ndt/gfab312] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Indexed: 12/02/2022] Open
Abstract
Approval of the vasopressin V2 receptor antagonist tolvaptan—based on the landmark TEMPO 3:4 trial—marked a transformation in the management of autosomal dominant polycystic kidney disease (ADPKD). This development has advanced patient care in ADPKD from general measures to prevent progression of chronic kidney disease to targeting disease-specific mechanisms. However, considering the long-term nature of this treatment, as well as potential side effects, evidence-based approaches to initiate treatment only in patients with rapidly progressing disease are crucial. In 2016, the position statement issued by the European Renal Association (ERA) was the first society-based recommendation on the use of tolvaptan and has served as a widely used decision-making tool for nephrologists. Since then, considerable practical experience regarding the use of tolvaptan in ADPKD has accumulated. More importantly, additional data from REPRISE, a second randomized clinical trial (RCT) examining the use of tolvaptan in later-stage disease, have added important evidence to the field, as have post hoc studies of these RCTs. To incorporate this new knowledge, we provide an updated algorithm to guide patient selection for treatment with tolvaptan and add practical advice for its use.
Collapse
Affiliation(s)
| | - A Lianne Messchendorp
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Henrik Birn
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Departments of Clinical Medicine and Biomedicine, Aarhus University, Aarhus, Denmark
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, Vanvitelli University, Naples, Italy
- Biogem Institute for Molecular Biology and Genetics, Ariano Irpino, Italy
| | | | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Division of Nephrology, UCL Medical School, Brussels, Belgium
| | - Albertien van Eerde
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nine V A M Knoers
- Department Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Uwe Korst
- PKD Familiäre Zystennieren e.V., Bensheim, Germany
| | - Djalila Mekahli
- PKD Research Group, Laboratory of Pediatrics, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pediatric Nephrology and Organ Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Yannick Le Meur
- Department of Nephrology, Hemodialysis and Renal Transplantation, CHU and University of Brest, Brest, France
| | - Tom Nijenhuis
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboudumc Center of Expertise for Rare Kidney Disorders, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Albert C M Ong
- Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, UK
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Aude Servais
- Nephrology and Transplantation Department, Necker University Hospital, APHP, Paris, France
| | - Vladimir Tesar
- Department of Nephrology, 1st Faculty of Medicine, General University Hospital, Prague, Czech Republic
| | - Roser Torra
- Inherited Kidney Diseases Nephrology Department, Fundació Puigvert Instituto de Investigaciones Biomédicas Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- REDINREN, Barcelona, Spain
| | - Stephen B Walsh
- Department of Renal Medicine, University College London, London, UK
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Strubl S, Oehm S, Torres JA, Grundmann F, Haratani J, Decker M, Vuong S, Kaur Bhandal A, Methot N, Haynie-Cion R, Meyer F, Siedek F, Korst U, Müller RU, Weimbs T. Ketogenic dietary interventions in autosomal dominant polycystic kidney disease-a retrospective case series study: first insights into feasibility, safety and effects. Clin Kidney J 2021; 15:1079-1092. [PMID: 35664270 PMCID: PMC9155228 DOI: 10.1093/ckj/sfab162] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Indexed: 01/13/2023] Open
Abstract
Background Our laboratory published the first evidence that nutritional ketosis, induced by a ketogenic diet (KD) or time-restricted diet (TRD), ameliorates disease progression in polycystic kidney disease (PKD) animal models. We reasoned that, due to their frequent use for numerous health benefits, some autosomal dominant PKD (ADPKD) patients may already have had experience with ketogenic dietary interventions (KDIs). This retrospective case series study is designed to collect the first real-life observations of ADPKD patients about safety, feasibility and possible benefits of KDIs in ADPKD as part of a translational project pipeline. Methods Patients with ADPKD who had already used KDIs were recruited to retrospectively collect observational and medical data about beneficial or adverse effects and the feasibility and safety of KDIs in questionnaire-based interviews. Results A total of 131 ADPKD patients took part in this study. About 74 executed a KD and 52 a TRD for 6 months on average. A total of 86% of participants reported that KDIs had improved their overall health, 67% described improvements in ADPKD-associated health issues, 90% observed significant weight loss, 64% of participants with hypertension reported improvements in blood pressure, 66% noticed adverse effects that are frequently observed with KDIs, 22 participants reported safety concerns like hyperlipidemia, 45 participants reported slight improvements in estimated glomerular filtration rate and 92% experienced KDIs as feasible while 53% reported breaks during their diet. Conclusions Our preliminary data indicate that KDIs may be safe, feasible and potentially beneficial for ADPKD patients, highlighting that prospective clinical trials are warranted to confirm these results in a controlled setting and elucidate the impact of KDIs specifically on kidney function and cyst progression.
Collapse
Affiliation(s)
- Sebastian Strubl
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA,Department II of Internal Medicine and Center for Molecular Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Simon Oehm
- Department II of Internal Medicine and Center for Molecular Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jacob A Torres
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Franziska Grundmann
- Department II of Internal Medicine and Center for Molecular Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jazmine Haratani
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Morgan Decker
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Sabrina Vuong
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Amrit Kaur Bhandal
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Nils Methot
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Rhianna Haynie-Cion
- Department of Molecular, Cellular, and Developmental Biology and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Franziska Meyer
- Institute of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Siedek
- Institute of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Uwe Korst
- PKD Familiäre Zystennieren e.V., Bensheim, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | | |
Collapse
|
46
|
Schirutschke H, Gross P, Paliege A, Hugo C. 10-Year Evaluation of Adherence and Satisfaction with Information about Tolvaptan in ADPKD: A Single-Center Pilot Study. Patient Prefer Adherence 2021; 15:1941-1952. [PMID: 34511889 PMCID: PMC8427080 DOI: 10.2147/ppa.s325738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Tolvaptan is the only approved drug for the treatment of autosomal dominant polycystic kidney disease (ADPKD) and causes significant polyuria with secondary polydipsia. Up to now, there is no study that examines tolvaptan adherence and satisfaction with information received about tolvaptan in ADPKD patients 10 years after starting tolvaptan therapy. PATIENTS AND METHODS This pilot study includes 12 ADPKD patients that were formerly enrolled in the tolvaptan registration trials and have continued to use tolvaptan thereafter. Data were collected once via questionnaires on patients' self-reported adherence (MARS-D: Medication Adherence Report Scale - German version) and satisfaction with the information received about tolvaptan (SIMS-D: Satisfaction with Information about Medicines Scale - German version) at the time of the present study. In addition, serum creatinine levels and clinical data were evaluated. RESULTS The MARS-D demonstrated strong adherence to tolvaptan (range of possible score: 5-25; median: 23.5; range of individual results: 5). The SIMS-D showed a high level of satisfaction with the information received about the action and usage of tolvaptan (SIMS-D AU subscale; range of possible score: 0-9; median: 9, range of individual results: 1), but also revealed dissatisfaction regarding the information received about potential problems of tolvaptan in 42% of the participants (SIMS-D PP subscale; range of possible score: 0-8; median: 8, range of individual results: 6). During treatment with tolvaptan, the eGFR decreased from 78.8 ± 15.9 mL/min/1.73 m2 to 48.3 ± 19.4 mL/min/1.73 m2 (P < 0.0001). CONCLUSION Although patients reported strong adherence to tolvaptan, there was still dissatisfaction with the information received about potential problems with tolvaptan. Therefore, our data suggest conduction of at least one patient survey on adherence and satisfaction with the information received about tolvaptan during any tolvaptan treatment to improve patient education regarding the use of tolvaptan in slowing down of ADPKD.
Collapse
Affiliation(s)
- Holger Schirutschke
- University Hospital Carl Gustav Carus at the Technische, Universität Dresden, Department of Internal Medicine III, Division of Nephrology, Dresden, Germany
| | - Peter Gross
- University Hospital Carl Gustav Carus at the Technische, Universität Dresden, Department of Internal Medicine III, Division of Nephrology, Dresden, Germany
| | - Alexander Paliege
- University Hospital Carl Gustav Carus at the Technische, Universität Dresden, Department of Internal Medicine III, Division of Nephrology, Dresden, Germany
| | - Christian Hugo
- University Hospital Carl Gustav Carus at the Technische, Universität Dresden, Department of Internal Medicine III, Division of Nephrology, Dresden, Germany
| |
Collapse
|
47
|
Patel DM, Dahl NK. Examining the Role of Novel CKD Therapies for the ADPKD Patient. KIDNEY360 2021; 2:1036-1041. [PMID: 35373079 PMCID: PMC8791369 DOI: 10.34067/kid.0007422020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/23/2021] [Indexed: 12/19/2022]
|
48
|
Issa N, Chedid M, Irazabal MV, Dean PG, Chebib FT. Twenty-Year Survival of Kidney Transplant From a Deceased Donor With Autosomal Dominant Polycystic Kidney Disease. Kidney Int Rep 2021; 6:2240-2242. [PMID: 34386677 PMCID: PMC8344107 DOI: 10.1016/j.ekir.2021.05.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 11/07/2022] Open
Affiliation(s)
- Naim Issa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Maroun Chedid
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Maria V Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Patrick G Dean
- Department of Transplantation Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
49
|
Phakdeekitcharoen B, Treesinchai W, Wibulpolprasert P, Boongird S, Klytrayong P. The correlation between kidney volume and measured glomerular filtration rate in an Asian ADPKD population: a prospective cohort study. BMC Nephrol 2021; 22:178. [PMID: 33992075 PMCID: PMC8126117 DOI: 10.1186/s12882-021-02392-0] [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: 01/12/2021] [Accepted: 05/06/2021] [Indexed: 11/16/2022] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disorder that leads to end stage renal disease (ESRD). Cyst expansion in ADPKD is strongly associated with the decline in renal function. However, the correlation between total kidney volume (TKV) and glomerular filtration rate (GFR) at an early stage has not been well demonstrated. There is growing evidence that utilization of estimated GFR (eGFR) may induce misleading information in a population with near normal renal function. Therefore, a more accurate method is essential. Methods A prospective cohort of ADPKD patients was conducted with clinical data and laboratory collection. Measured GFR (mGFR) was assessed by iohexol plasma clearance method using ultra performance liquid chromatography. eGFR was calculated using the CKD-EPI equation. Kidney volumes were evaluated using MRI imaging protocol. Results Thirty two patients completed the study. The mean age was 56 years old. The mean initial mGFR was 83.8 mL/min/1.73m2. The mean change in mGFR per year was –2.99 mL/min/1.73m2/year. The mean initial height-adjusted TKV (htTKV) was 681.0 mL/m. The mean percentage change in htTKV per year (%ΔhtTKV/y) was 4.77 %/year. mGFR had a better association with clinical parameters than eGFR. Initial mGFR was significantly and inversely correlated with initial htTKV and age. The percentage change in mGFR per year was significantly and inversely correlated with the %ΔhtTKV/y and 24-hr urine albumin. The %ΔhtTKV/y was significantly correlated with initial htTKV. Conclusions Our studies demonstrated that mGFR using iohexol is a more reliable and accurate method than eGFR for evaluating GFR changes in the early stages of ADPKD patients. There is a strong inverse correlation between kidney volume and mGFR in an Asian ADPKD population. The initial htTKV is a good predictor of kidney volume progression. The %ΔhtTKV/y is a good early surrogate marker for the decline in renal function. 24-hr urine albumin is also a good indicator for renal progression.
Collapse
Affiliation(s)
- Bunyong Phakdeekitcharoen
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 10400, Bangkok, Thailand.
| | - Watcharapong Treesinchai
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 10400, Bangkok, Thailand
| | - Pornphan Wibulpolprasert
- Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sarinya Boongird
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 10400, Bangkok, Thailand
| | - Pinkael Klytrayong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 10400, Bangkok, Thailand
| |
Collapse
|
50
|
El Ters M, Lu P, Mahnken JD, Stubbs JR, Zhang S, Wallace DP, Grantham JJ, Chapman AB, Torres VE, Harris PC, Bae KT, Landsittel DP, Rahbari-Oskoui FF, Mrug M, Bennett WM, Yu AS. Prognostic Value of Fibroblast Growth Factor 23 in Autosomal Dominant Polycystic Kidney Disease. Kidney Int Rep 2021; 6:953-961. [PMID: 33912745 PMCID: PMC8071629 DOI: 10.1016/j.ekir.2021.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Autosomal dominant polycystic kidney disease (ADPKD) is characterized by progressive cyst growth and a loss of functioning renal mass, but a decline in glomerular filtration rate (GFR) and onset of end-stage renal disease (ESRD) occur late in the disease course. There is therefore a great need for early prognostic biomarkers in this disorder. METHODS We measured baseline serum fibroblast growth factor 23 (FGF23) levels in 192 patients with ADPKD from the Consortium for Radiologic Imaging Studies of PKD (CRISP) cohort that were followed for a median of 13 years and tested the association between FGF23 levels and change over time in height-adjusted total kidney volume (htTKV), GFR, and time to the composite endpoints of ESRD, death, and doubling of serum creatinine. RESULTS Patients in the highest quartile for baseline FGF23 level had a higher rate of increase in htTKV (0.95% per year, P = 0.0016), and faster rate of decline in GFR (difference of -1.03 ml/min/1.73 m2 per year, P = 0.005) compared with the lowest quartile, after adjusting for other covariates, including htTKV and genotype. The highest quartile of FGF23 was also associated with a substantial increase in risk for the composite endpoint of ESRD, death, or doubling of serum creatinine (hazard ratio [HR] of 2.45 in the fully adjusted model, P = 0.03). CONCLUSION FGF23 is a prognostic biomarker for disease progression and clinically important outcomes in ADPKD, and has additive value to established imaging and genetic biomarkers.
Collapse
Affiliation(s)
- Mireille El Ters
- Division of Nephrology and Hypertension and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Pengcheng Lu
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jonathan D. Mahnken
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jason R. Stubbs
- Division of Nephrology and Hypertension and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Shiqin Zhang
- Division of Nephrology and Hypertension and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Darren P. Wallace
- Division of Nephrology and Hypertension and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jared J. Grantham
- Division of Nephrology and Hypertension and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Arlene B. Chapman
- Division of Nephrology, University of Chicago School of Medicine, Chicago, Illinois, USA
- Department of Internal Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kyongtae Ty Bae
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Douglas P. Landsittel
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Michal Mrug
- Division of Nephrology, University of Alabama and the Department of Veterans Affairs Medical Center, Birmingham, Alabama, USA
| | | | - Alan S.L. Yu
- Division of Nephrology and Hypertension and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|