1
|
Bezerra ROF, Strufaldi FL, Dantas PP, Andrade F, Caires RA, Costalonga EC, Filho GBC, Gil LA, Sapienza MT, Cerri GG, Burdmann EA, Costa E Silva VT. Total kidney volume as a predictor of measured glomerular filtration rate in patients with solid tumors: a prospective cross-sectional analysis. J Nephrol 2025:10.1007/s40620-025-02231-7. [PMID: 40314881 DOI: 10.1007/s40620-025-02231-7] [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: 04/13/2024] [Accepted: 01/24/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Although previous data demonstrate that total kidney volume (TKV) correlates with measured glomerular filtration rate (mGFR), evidence is however scarce in the oncology setting. The aim of this is study is to evaluate whether adding TKV to the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) estimating equations improves the prediction of measured glomerular filtration rate (mGFR) in patients with cancer. METHODS We evaluated patients with solid tumors between April 2015 and September 2017 who had undergone contrast computed tomography and GFR measurement through the plasma clearance of 51Cr-EDTA. Estimated GFR (eGFR) was determined through CKD-EPI equations based on serum creatinine (eGFRcr) and combined with serum cystatin C (eGFRcr-cys). We used the 2009 eGFRcr and 2012 eGFRcr-cys equations and the race-free 2021 eGFRcr, and eGFRcr-cys. TKV was measured using a semi-automatic segmentation program, excluding non-functional tissues. Linear regression models were built, with TKV and eGFR equations as predictors and mGFR as the outcome. RESULTS We included 189 patients (median age 58.0 [48.0-65.0] years, 49.2% male). Median mGFR and TKV were 82.7 (66.3-94.5) mL/min and 303.1 (257.7-351.8) cm3, respectively. TKV improved the coefficient of determination (R2) when added to 2009 eGFRcr and 2012 eGFRcr-cys equations from 0.62 to 0.73 and 0.73 to 0.80, respectively. For the 2021 eGFRcr and eGFRcr-cys equations, R2 improved from 0.65 to 0.75 and 0.75 to 0.82, respectively. CONCLUSION These results suggest that TKV measurement improves the prediction of mGFR in association with the CKD-EPI equations in patients with solid tumors.
Collapse
Affiliation(s)
- Regis Otaviano Franca Bezerra
- Serviço de Radiologia do Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo, 25, São Paulo, São Paulo, 01246-000, Brazil.
| | - Fernando Louzada Strufaldi
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Patricia Perola Dantas
- Serviço de Radiologia do Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo, 25, São Paulo, São Paulo, 01246-000, Brazil
| | - Filipe Andrade
- Serviço de Radiologia do Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo, 25, São Paulo, São Paulo, 01246-000, Brazil
| | - Renato A Caires
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Elerson Carlos Costalonga
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - George Barbério Coura Filho
- Serviço de Medicina Nuclear, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz A Gil
- Laboratório de Investigação Médica (LIM) 66, Serviço de Geriatria, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo Tatit Sapienza
- Serviço de Radiologia do Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo, 25, São Paulo, São Paulo, 01246-000, Brazil
| | - Giovanni Guido Cerri
- Serviço de Radiologia do Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Av. Dr. Arnaldo, 25, São Paulo, São Paulo, 01246-000, Brazil
| | - Emmanuel A Burdmann
- Laboratório de Investigação Médica (LIM) 12, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Veronica Torres Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Investigação Médica (LIM) 16, Serviço de Nefrologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Madero M, Levin A, Ahmed SB, Carrero JJ, Foster B, Francis A, Hall RK, Herrington WG, Hill G, Inker LA, Kazancıoğlu R, Lamb E, Lin P, McIntyre N, Morrow K, Roberts G, Sabanayagam D, Shlipak M, Shroff R, Tangri N, Thanachayanont T, Ulasi I, Wong G, Yang CW, Zhang L, Robinson KA, Wilson LM, Wilson RF, Kasiske BL, Cheung M, Earley A, Stevens PE, Schaeffner E. Evaluation and Management of Chronic Kidney Disease: Synopsis of the Kidney Disease: Improving Global Outcomes 2024 Clinical Practice Guideline. Ann Intern Med 2025; 178:705-713. [PMID: 40063957 DOI: 10.7326/annals-24-01926] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/21/2025] Open
Abstract
DESCRIPTION The Kidney Disease: Improving Global Outcomes (KDIGO) organization updated its existing clinical practice guideline in 2024 to provide guidance on the evaluation, management, and treatment of chronic kidney disease (CKD) in adults and children who are not receiving kidney replacement therapy. METHODS The KDIGO CKD Guideline Work Group defined the scope of the guideline and determined topics for systematic review. An independent Evidence Review Team systematically reviewed the evidence and graded the certainty of evidence for each of the review topics. Latest searches of the English-language literature were done in July 2023. Final modification of the guideline was informed by a public review process during summer of 2023 involving registered stakeholders. RECOMMENDATIONS The full guideline included 28 recommendations and 141 practice points. This synopsis focuses on the recommendations that have the greatest evidence. Practice points reflect the expert opinion of the group where evidence is not that strong. Recommendations include greater emphasis on cystatin C for assessment of glomerular filtration rate, point-of-care testing in remote areas, a shift to an individualized risk-based approach to predict kidney failure, sodium-glucose cotransporter-2 inhibitors for some patients with CKD with and without diabetes, and statin use for adults older than 50 years and CKD. Together the recommendations and practice points provide guidance for how to evaluate and manage persons with CKD.
Collapse
Affiliation(s)
- Magdalena Madero
- Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico (M.M.)
| | - Adeera Levin
- University of British Columbia, Vancouver, British Columbia, Canada (A.L.)
| | - Sofia B Ahmed
- University of Alberta, Edmonton, Alberta, Canada (S.B.A.)
| | | | | | - Anna Francis
- Queensland Children's Hospital, Brisbane, Australia (A.F.)
| | | | - William G Herrington
- Renal Studies Group, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom (W.G.H.)
| | - Guy Hill
- Patient partner, Manchester, United Kingdom (G.H.)
| | | | | | - Edmund Lamb
- East Kent Hospitals University NHS Foundation Trust, Canterbury, United Kingdom (E.L.)
| | - Peter Lin
- Canadian Heart Research Center, Toronto, Ontario, Canada (P.L.)
| | - Natasha McIntyre
- Western University, London Health Sciences Centre-Victoria Hospital, London, Ontario, Canada (N.M.)
| | - Kelly Morrow
- Bastyr University, Osher Center for Integrative Medicine, University of Washington, Kenmore, Washington (K.M.)
| | - Glenda Roberts
- UW Center for Dialysis Innovation & Kidney Research Institute, Seattle, Washington (G.R.)
| | | | - Michael Shlipak
- University of California, San Francisco, San Francisco, California (M.S.)
| | - Rukshana Shroff
- UCL Great Ormond Street Hospital Institute of Child Health, London, United Kingdom (R.S.)
| | - Navdeep Tangri
- University of Manitoba, Winnipeg, Manitoba, Canada (N.T.)
| | | | - Ifeoma Ulasi
- University of Nigeria, Ituku-Ozalla Campus, Enugu, Nigeria (I.U.)
| | - Germaine Wong
- University of Sydney, Sydney, Australia (D.S., G.W.)
| | - Chih-Wei Yang
- Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan (C.-W.Y.)
| | - Luxia Zhang
- Peking University First Hospital, Beijing, China (L.Z.)
| | - Karen A Robinson
- The Johns Hopkins University Evidence-based Practice Center, Johns Hopkins University, Baltimore, Maryland (K.A.R., L.M.W., R.F.W.)
| | - Lisa M Wilson
- The Johns Hopkins University Evidence-based Practice Center, Johns Hopkins University, Baltimore, Maryland (K.A.R., L.M.W., R.F.W.)
| | - Renee F Wilson
- The Johns Hopkins University Evidence-based Practice Center, Johns Hopkins University, Baltimore, Maryland (K.A.R., L.M.W., R.F.W.)
| | - Bertram L Kasiske
- Hennepin County Medical Center, University of Minnesota, Minneapolis, Minnesota (B.L.K.)
| | | | | | - Paul E Stevens
- Kent Kidney Care Centre, East Kent Hospitals University NHS Foundation Trust, Canterbury, United Kingdom (P.E.S.)
| | | |
Collapse
|
3
|
Yamamoto PA, Rodriguez-Vera L, Telles JP, Moreira FL, Roy S, Korn O, Coelho D, Migotto KC, de Carvalho FRT, Caruso P, França E Silva IL, Vozmediano V, de Moraes NV. A Bayesian Framework for Optimizing Amikacin Therapy in Critically Ill Patients With Cancer. Ther Drug Monit 2025:00007691-990000000-00338. [PMID: 40209112 DOI: 10.1097/ftd.0000000000001324] [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: 09/27/2024] [Accepted: 01/08/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Amikacin (AMK) is used to treat gram-negative bacterial infections in intensive care unit (ICU) patients. However, its narrow therapeutic range and high interindividual variability can lead to toxicity and ineffectiveness. This study aimed to establish a roadmap for AMK therapeutic drug monitoring in critically ill patients with cancer to provide a Bayesian estimator of bedside applicability. METHODS An observational retrospective study was conducted on oncological patients admitted to the ICU, treated with AMK as a 30-min intravenous infusion at 5.8-39.2 mg/kg. The plasma concentrations were analyzed using a nonlinear mixed-effects modeling approach. Covariate analyses were performed using anthropometric and laboratory data, concomitant drugs, and comorbidities. The model predictive performance was compared with previous AMK dosing approaches using the Bland-Altman method. RESULTS The concentration-time profiles were best described using a one-compartment model with linear elimination. The estimated glomerular filtration rate was a significant covariate of clearance (CL), explaining 16% of the interpatient variability. Body weight was positively correlated with the volume of distribution, accounting for 4% of the variability. Our model reduced the bias in the estimates of individual CL values compared with that of other available methods and was further implemented in DoseMeRx for real-time application at the bedside. CONCLUSIONS This study provides an effective example of a Bayesian estimation method for individualizing AMK doses in critically ill patients with cancer. Collecting more comprehensive patient information, including additional biomarkers for renal function, could further refine the model and improve its predictive performance in this special population.
Collapse
Affiliation(s)
- Priscila Akemi Yamamoto
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| | - Leyanis Rodriguez-Vera
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
- Model Informed Development, Regulatory Development and Consulting, CTI Laboratories, Covington, Kentucky
| | | | - Fernanda Lima Moreira
- Laboratory of Pharmacometrics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | - Valvanera Vozmediano
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
- Model Informed Development, Regulatory Development and Consulting, CTI Laboratories, Covington, Kentucky
| | - Natalia Valadares de Moraes
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| |
Collapse
|
4
|
Titan SM, Lieske JC, Meeusen JW, Thorson S, Lin Y, Nowakowski GS, Barreto JN, Barreto EF, Ruddy KJ, Leung N, Rule AD, Herrmann SM. Performance of Creatinine and Cystatin-Based Equations on Estimating Measured GFR in People with Hematological and Solid Cancers. Clin J Am Soc Nephrol 2025; 20:358-366. [PMID: 39874103 PMCID: PMC11905996 DOI: 10.2215/cjn.0000000616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
Key Points Studies on GFR assessment in people with cancer are needed. In this study, the creatinine–cystatin C equations performed better than other equations, and this was observed in solid and hematological cancers. Our findings give support to the preferential use of creatinine and cystatin C–based equations in people with cancer. Background GFR assessment is important in clinical practice with implications for diagnosis, prognostication, and drug dosing. People with cancer are at risk of imprecision in GFR estimation. This cross-sectional study evaluated the performance of various creatinine and cystatin C–based equations in comparison with measured GFR (mGFR) in people with cancer. Methods We retrieved data for all adult patients who had mGFR by urinary iothalamate clearance between 2011 and 2023 at Mayo Clinic and use of an electronic health record diagnosis code for cancer within 2 years before mGFR. The CKD Epidemiology Collaboration (CKD-EPI), European Kidney Function Consortium, and Cockcroft–Gault equations were computed, along with performance metrics (bias, precision, and root mean square error [RMSE]. Confidence intervals were generated by bootstrapping, and analysis were stratified by solid and hematological cancers. Results From all adults with cancer and mGFR, 1145 had both creatinine and cystatin C available within 7 days of mGFR. Among all equations, the creatinine–cystatin C CKD-EPI equation provided the best performance, with small bias (median, 3.0; 95% confidence interval, 2.3 to 3.8) and higher precision (RMSE, 14.5) compared with creatinine-only or cystatin C–only equations (RMSE varying from 16.6 to 20), and this was also true in solid and hematological cancers. The creatinine–cystatin European Kidney Function Consortium equation had a similar performance to CKD-EPI, Cockcroft–Gault showed worst precision (30% of people with errors above 30%), and cystatin C CKD-EPI equation was the most biased, prone to underestimation of mGFR. Conclusions In our cohort of patients with mGFR and cancer, the CKD-EPI creatinine–cystatin C equation performed best for GFR assessment, and this was true for both solid and hematological cancers. Our findings give support for the preferential use of creatinine and cystatin C–based equations instead of creatinine-only or cystatin C–only equations in people with cancer.
Collapse
Affiliation(s)
- Silvia M. Titan
- Nephrology and Hypertension Division, Mayo Clinic, Rochester, Minnesota
| | - John C. Lieske
- Nephrology and Hypertension Division, Mayo Clinic, Rochester, Minnesota
| | | | - Stacy Thorson
- Biomedical Informatics, Center for Clinical and Translational Science, Mayo Clinic, Rochester, Minnesota
| | - Yi Lin
- Hematology Division, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | - Nelson Leung
- Nephrology and Hypertension Division, Mayo Clinic, Rochester, Minnesota
| | - Andrew D. Rule
- Nephrology and Hypertension Division, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
5
|
Navaneethan SD, Bansal N, Cavanaugh KL, Chang A, Crowley S, Delgado C, Estrella MM, Ghossein C, Ikizler TA, Koncicki H, St Peter W, Tuttle KR, William J. KDOQI US Commentary on the KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of CKD. Am J Kidney Dis 2025; 85:135-176. [PMID: 39556063 DOI: 10.1053/j.ajkd.2024.08.003] [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: 07/18/2024] [Accepted: 08/04/2024] [Indexed: 11/19/2024]
Abstract
The Kidney Disease Outcomes Quality Initiative (KDOQI) convened a work group to review the 2024 KDIGO (Kidney Disease: Improving Global Outcomes) guideline for the management of chronic kidney disease (CKD). The KDOQI Work Group reviewed the KDIGO guideline statements and practice points and provided perspective for implementation within the context of clinical practice in the United States. In general, the KDOQI Work Group concurs with several recommendations and practice points proposed by the KDIGO guidelines regarding CKD evaluation, risk assessment, and management options (both lifestyle and medications) for slowing CKD progression, addressing CKD-related complications, and improving cardiovascular outcomes. The KDOQI Work Group acknowledges the growing evidence base to support the use of several novel agents such as sodium/glucose cotransporter 2 inhibitors for several CKD etiologies, and glucagon-like peptide 1 receptor agonists and nonsteroidal mineralocorticoid receptor antagonists for type 2 CKD in setting of diabetes. Further, KDIGO guidelines emphasize the importance of team-based care which was also recognized by the work group as a key factor to address the growing CKD burden. In this commentary, the Work Group has also assessed and discussed various barriers and potential opportunities for implementing the recommendations put forth in the 2024 KDIGO guidelines while the scientific community continues to focus on enhancing early identification of CKD and discovering newer therapies for managing kidney disease.
Collapse
Affiliation(s)
- Sankar D Navaneethan
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health and Institute of Clinical and Translational Research, Baylor College of Medicine, Houston, Texas; Section of Nephrology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas.
| | - Nisha Bansal
- Cardiovascular Health Research Unit, Department of Medicine, Washington
| | - Kerri L Cavanaugh
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alexander Chang
- Department of Population Health Sciences, Geisinger, Danville, Pennsylvania
| | - Susan Crowley
- Section of Nephrology, Department of Medicine, School of Medicine, Yale University, New Haven, Connecticut; Kidney Medicine Section, Medical Services, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Cynthia Delgado
- Nephrology Section, San Francisco Veterans Affairs Health Care System, San Francisco, California; Division of Nephrology, University of California-San Francisco, San Francisco, California
| | - Michelle M Estrella
- Nephrology Section, San Francisco Veterans Affairs Health Care System, San Francisco, California; Division of Nephrology, University of California-San Francisco, San Francisco, California
| | - Cybele Ghossein
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - T Alp Ikizler
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Holly Koncicki
- Division of Nephrology, Mount Sinai Health System, New York, New York
| | - Wendy St Peter
- College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | - Katherine R Tuttle
- Institute of Translational Health Sciences, Kidney Research Institute, and Nephrology Division, Washington; School of Medicine, University of Washington, Seattle, and Providence Medical Research Center, Providence Inland Northwest Health, Spokane, Washington
| | - Jeffrey William
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
6
|
Mistry K, Sadarangani S, Moreno D, Mejia SM, Moledina DG, Sise ME. Novel Biomarkers and Imaging Tests for Acute Kidney Injury Diagnosis in Patients with Cancer. KIDNEY360 2025; 6:167-174. [PMID: 39575585 PMCID: PMC11793192 DOI: 10.34067/kid.0000000660] [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: 07/22/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025]
Abstract
The lack of noninvasive urine and blood-based biomarkers for the diagnosis of AKI in patients with cancer is an area of significant unmet clinical need. Traditional noninvasive diagnostic tools that are currently used in the clinic, such as creatinine and cystatin C-based eGFR measurements, urinalysis, urine sediment examination, urine protein quantification, and urine electrolyte measurement, lack the sensitivity and specificity to distinguish between the various underlying etiologies of AKI in patients with cancer. Imaging-based diagnostics can be helpful to rule out urinary obstruction, but also lack sensitivity and specificity to diagnose the etiology of AKI. Kidney biopsy is often required for definitive diagnosis. As our scientific understanding of the biological pathways that are dysregulated in AKI has advanced, there has been considerable interest in developing new biomarkers for AKI. For example, the diagnosis of acute interstitial nephritis, which can occur in patients treated with immune checkpoint inhibitors, promises to be revolutionized by the incorporation of urinary testing for inflammatory biomarkers, such as C-X-C motif ligand 9, TNF- α , and IL-9. In the case of cisplatin administration, biomarkers such as neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 may improve prognostication, differentiating between persistent AKI resulting from acute tubular injury versus prerenal azotemia. The development and validation of blood, urine, and imaging biomarkers into widely used diagnostic tests will require a concerted effort, but could improve diagnosis, management, and prognostication for a growing group of patients who are at high risk of developing AKI during the course of their illness.
Collapse
Affiliation(s)
- Kavita Mistry
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Sagar Sadarangani
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Daiana Moreno
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Sherley M. Mejia
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Dennis G. Moledina
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Internal Medicine, Clinical and Translational Research Accelerator, New Haven, Connecticut
| | - Meghan E. Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
7
|
Delecluse S, Harder F, Keller F, Zeier M, Zschäbitz S. Onconephrology: The Significance of Renal Function for the Development, Diagnosis, and Treatment of Cancer. DEUTSCHES ARZTEBLATT INTERNATIONAL 2024; 121:793-799. [PMID: 39417369 DOI: 10.3238/arztebl.m2024.0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Modern treatment strategies have markedly improved the chances of survival for patients with cancer. As the population ages, cancer is becoming more common, as is chronic kidney disease (CKD). CKD increases the risk of cancer; conversely, cancer treatments can cause CKD. METHODS This review is based on publications retrieved by a selective literature search concerning the epidemiology and comorbidities of cancer and kidney diseases, the renal side effects of new anticancer drugs, and the need to consider renal function in cancer treatment. RESULTS The prevalence of severe CKD in Germany is 2.3%. Persons who have CKD, are on dialysis, or have undergone kidney transplantation are 1.2 to 3.5 times more likely to develop cancer than the general population. For patients who have CKD or are dialysis-dependent, the doses of approximately 67% of anticancer drugs need to be adjusted on the basis of their glomerular filtration rate and the renally excreted fraction of the drug. The optimal efficacy of therapeutic drugs, as well as of those used for diagnostic purposes, and the minimization of side effects, depend critically on adapted dosing and on proper timing of administration before or after dialysis. Modern anticancer drugs can also cause acute kidney damage (incidence with checkpoint inhibitors: 2-16%). CONCLUSION Patients who have CKD, are on dialysis, or have undergone kidney transplantation make up a considerable fraction of persons being treated for cancer, and they need interdisciplinary treatment.
Collapse
Affiliation(s)
- Susanne Delecluse
- Heidelberg Kidney Center, Heidelberg, Germany; German Cancer Research Center (DKFZ), Unit D400, Heidelberg, Germany; Institute of Experimental and Clinical Pharmacology, Toxicology, and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany; Department of Internal Medicine VI, Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
8
|
Pichler R, Fritz J, Mari A, Cadenar A, von Deimling M, Marcq G, Del Giudice F, Leonardo C, Bologna E, Mori K, Tahbaz R, De Santis M, Klatte T, Erber B, Lackner F, Kronbichler A, Seeber A, Fisch M, Moschini M, Pradere B, Mertens LS. Cisplatin eligibility in the neoadjuvant setting of patients with muscle-invasive bladder cancer undergoing radical cystectomy. Oncologist 2024; 29:e1511-e1522. [PMID: 38956801 PMCID: PMC11546640 DOI: 10.1093/oncolo/oyae160] [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: 02/06/2024] [Accepted: 05/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND To examine the agreement of different calculated estimated glomerular filtration rate (eGFR) formulas and measured creatinine clearance (CrCI) at the primary diagnosis of muscle-invasive bladder cancer (MIBC). MATERIALS AND METHODS We performed a multicenter analysis of patients with MIBC, treated with cisplatin-based neoadjuvant chemotherapy (NAC) and radical cystectomy (RC), or with RC alone, between 2011 and 2021. Baseline eGFR was computed using 4 calculated serum equations including Cockcroft-Gault (CG), MDRD, CKD-EPI 2009, and race-free CKD-EPI 2021. To examine the association between calculated eGFR and measured CrCI, subgroup analyses were performed among patients in whom measured 24-hour urine CrCl was determined. Cisplatin-ineligibility was defined as CrCI and/or eGFR < 60 mL/minute per 1.73 m2. RESULTS Of 956 patients, 30.0%, 33.3%, 31.9%, and 27.7% were found to be cisplatin-ineligible by the CG, MDRD, CKD-EPI, and race-free CKD-EPI equations (P = .052). The concordance between calculated eGFR formulas was rated substantial (Cohen's kappa (k): 0.66-0.95). Among the subgroup (n = 245) with measured CrCl, 37 (15.1%) patients had a CrCI less than 60 mL/minute. Concordance between measured CrCl and calculated eGFR was poor (ĸ: 0.29-0.40). All calculated eGFR formulas markedly underestimated the measured CrCI. Specifically, 78%-87.5% of patients with a calculated eGFR between 40 and 59 mL/minute exhibited a measured CrCI ≥ 60 mL/minute. CONCLUSIONS Comparing calculated eGFR formulas, similar percentages of patients with MIBC were deemed cisplatin-ineligible. However, a significant number of patients could be upgraded by being cisplatin-fit based on measured CrCI, particularly when the calculated eGFR was falling within the gray range of 40-59 mL/minute.
Collapse
Affiliation(s)
- Renate Pichler
- Department of Urology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Josef Fritz
- Institute of Medical Statistics and Informatics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Andrea Mari
- Department of Experimental and Clinical Medicine, University of Florence, Unit of Oncologic Minimally-Invasive Urology and Andrology, Careggi Hospital, Florence, Italy
| | - Anna Cadenar
- Department of Experimental and Clinical Medicine, University of Florence, Unit of Oncologic Minimally-Invasive Urology and Andrology, Careggi Hospital, Florence, Italy
| | - Markus von Deimling
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg 20249, Germany
| | - Gautier Marcq
- Department of Urology, Claude Huriez Hospital, CHU Lille, Lille 59037, France
| | - Francesco Del Giudice
- Department of Maternal Infant and Urologic Sciences, "Sapienza" University of Rome, Policlinico Umberto I Hospital, Rome 00185, Italy
| | - Costantino Leonardo
- Department of Maternal Infant and Urologic Sciences, "Sapienza" University of Rome, Policlinico Umberto I Hospital, Rome 00185, Italy
| | - Eugenio Bologna
- Department of Maternal Infant and Urologic Sciences, "Sapienza" University of Rome, Policlinico Umberto I Hospital, Rome 00185, Italy
| | - Keiichiro Mori
- Department of Urology, The Jikei University School of Medicine, 105-8461 Tokyo, Japan
| | - Rana Tahbaz
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin 10117, Germany
- Department of Urology, Medical University Vienna, Vienna 1090, Austria
| | - Tobias Klatte
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Barbara Erber
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Felizian Lackner
- Department of Urology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Andreas Kronbichler
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- Department of Internal Medicine IV, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Margit Fisch
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg 20249, Germany
| | - Marco Moschini
- Department of Urology, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Benjamin Pradere
- Department of Urology, La Croix du Sud Hospital, 31130 Quint-Fonsegrives, France
| | - Laura S Mertens
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam 1006, The Netherlands
| |
Collapse
|
9
|
Vanhoutte T, Van Craenenbroeck AH, Sprangers B. Combined creatinine/cystatin C equations for estimation of GFR in patients with cancer: the future is now! Nephrol Dial Transplant 2024; 39:1744-1746. [PMID: 38782727 DOI: 10.1093/ndt/gfae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- Thomas Vanhoutte
- Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Amaryllis H Van Craenenbroeck
- Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Ben Sprangers
- Department of Nephrology, General Hospital Oost-Limburg, Genk, Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium
| |
Collapse
|
10
|
Gueutin V, Cardineau A, Mathian A, Lanot A, Comoz F, Brocheriou I, Izzedine H. Renal involvement in solid cancers: epidemiological, clinical and histological characteristics study of 154 onconephrology patients. BMC Nephrol 2024; 25:367. [PMID: 39427142 PMCID: PMC11490999 DOI: 10.1186/s12882-024-03812-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Onconephrology is a growing discipline that aims to improve the management of patients with cancer and kidney disease. If kidney histology is an essential key, the anatomopathological data remain weak although essential to this complex management. METHODS Patients with active cancer who had a kidney biopsy (KB) between 2014 and 2020 were included, and their clinicobiological and histological data were analyzed retrospectively. RESULTS Our cohort consisted of 154 patients (83 women) with a mean age of 58 years. One hundred twelve patients presented with proteinuria, 95 with acute kidney injury, and 59 with arterial hypertension. Histologically, interstitial fibrosis was found in 74% of KBs, tubular atrophy in 55.1%, arteriolar hyalinosis in 58.4%, and fibrous endarteritis in 54.4%. Regarding the main acute lesions, thrombotic microangiopathy (TMA) was found in 29.9% of biopsies, acute tubular necrosis (ATN) in 51.3%, and acute interstitial nephritis in 24.8%. The etiological diagnosis most often made was the nephrotoxicity of anticancer drugs (87 patients), followed by a pre-renal (15 patients) and kidney disease unrelated to cancer (13 patients). Sixty-seven patients presented with at least 2 associated diagnoses reflecting the complexity of kidney damage in cancer. Different clusters were found, highlighting that immunotherapy and anti-VEGF were the most commonly involved drugs. CONCLUSIONS During onconephrology practice, kidney toxicity of treatments is the most common etiology. Several mechanisms can be involved, underscoring the importance of kidney biopsy and the complexity of its management. Chronic histological lesions were very common.
Collapse
Affiliation(s)
- Victor Gueutin
- Service de néphrologie-dialyse-transplantation CHU de CAEN, Côte de Nacre, Caen, France.
- Service de néphrologie-dialyse, hôpital Jacques-Monod, Rue Eugène-Garnier, Flers, France.
| | - Aurore Cardineau
- Service de néphrologie-dialyse CH Mémorial France Etats-Unis, 715 rue Henri Dunant, Saint Lô, France
| | - Alexis Mathian
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié- Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des anti-phospholipides et autres maladies auto-immunes rares, Service de Médecine Interne 2, Institut E3M, Paris, France
| | - Antoine Lanot
- Service de néphrologie-dialyse-transplantation CHU de CAEN, Côte de Nacre, Caen, France
- Service de néphrologie-dialyse, hôpital Jacques-Monod, Rue Eugène-Garnier, Flers, France
| | - François Comoz
- Service d'Anatomie et Cytologie Pathologiques, CHU Côte de Nacre, Caen, France
| | - Isabelle Brocheriou
- Service d'Anatomie et Cytologie Pathologiques, La Pitié Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
- UMRS 1155, Institut National de la Santé et de la Recherche Médicale, Hôpital Tenon, Sorbonne Université, Paris, France
| | - Hassan Izzedine
- Department of Nephrology, Peupliers Private Hospital, Paris, France
| |
Collapse
|
11
|
Wang ZS, Wang SF, Zhao MY, He QN. [Current clinical application of glomerular filtration rate assessment methods in pediatric populations]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:1002-1008. [PMID: 39267519 PMCID: PMC11404467 DOI: 10.7499/j.issn.1008-8830.2401011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Glomerular filtration rate (GFR) is a critical indicator of renal function assessment, which exhibits age-dependency in children and may differ from adults under various disease conditions. In recent years, there has been a growing focus on GFR among scholars, with an increasing number of clinical studies dedicated to refining and optimizing GFR estimation to span all pediatric age groups. However, the methods and assessment equations for estimating GFR may vary under different disease conditions, affecting the accuracy and applicability of assessments. This article reviews the peculiarities of renal function in children, explores GFR measurement methods, and evaluates the application of various GFR assessment equations in pediatric clinical practice, providing a reference for clinical assessment of renal function in children.
Collapse
Affiliation(s)
- Zi-Sai Wang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| | - Sheng-Feng Wang
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| | - Ming-Yi Zhao
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| | - Qing-Nan He
- Department of Pediatrics, Third Xiangya Hospital, Central South University, Changsha 410013, China(He Q-N, . cn)
| |
Collapse
|
12
|
Costa E Silva VT, Gil LA, Inker LA, Caires RA, Costalonga E, Coura-Filho G, Sapienza MT, Castro G, Estevez-Diz MDP, Zanetta DMT, Antonângelo L, Marçal L, Tighiouart H, Miao S, Mathew P, Levey AS, Burdmann EA. Glomerular Filtration Rate Estimation Using β 2-Microglobulin and β-Trace Protein in Adults With Solid Tumors: A Prospective Cross-Sectional Study. Am J Kidney Dis 2024; 84:339-348.e1. [PMID: 38537905 DOI: 10.1053/j.ajkd.2024.01.532] [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: 08/10/2023] [Revised: 12/06/2023] [Accepted: 01/02/2024] [Indexed: 05/26/2024]
Abstract
RATIONALE & OBJECTIVE β2-Microglobulin (B2M) and β-trace protein (BTP) are novel endogenous filtration markers that may improve the accuracy of estimated glomerular filtration rate (eGFR) beyond creatinine and cystatin C (eGFRcr-cys), but they have not been assessed in patients with cancer. STUDY DESIGN Cross-sectional analysis. SETTING & PARTICIPANTS Prospective cohort of 1,200 patients with active solid tumors recruited between April 2015 and September 2017. EXPOSURE CKD-EPI equations without race combining B2M and/or BTP with creatinine with or without cystatin C (2-, 3-, or 4-marker panel eGFR). OUTCOME Performance of equations compared with eGFRcr-cys and non-GFR determinants of serum B2M and BTP (SB2M, and SBTP, respectively). Measured GFR (mGFR) was determined using the plasma clearance of chromium-51 labeled ethylenediamine tetraacetic acid (51Cr-EDTA). ANALYTICAL APPROACH Bias was defined as the median of the differences between mGFR and eGFR, and 1-P30 was defined as the percentage of estimates that differed by more than 30% from the mGFR (1-P30). Linear regression was used to assess association of clinical and laboratory variables with SB2M, and SBTP after adjustment for mGFR. RESULTS Mean age and mGFR were 58.8±13.2 SD years and 78.4±21.7 SD mL/min/1.73m2, respectively. Performance of the 3-marker and 4-marker panel equations was better than eGFRcr-cys (lesser bias and 1-P30). Performance of 2-marker panel equations was as good as eGFRcr-cys (lesser bias and similar 1-P30). SB2M and SBTP were not strongly influenced by cancer site. LIMITATIONS Participants may have had better clinical performance status than the general population of patients with solid tumors. CONCLUSIONS B2M and BTP can improve the accuracy of eGFR and may be useful as confirmatory tests in patients with solid tumors, either by inclusion in a multimarker panel equation with creatinine and cystatin C, or by substituting for cystatin C in combination with creatinine. PLAIN-LANGUAGE SUMMARY The most accurate method to assess estimate kidney function is estimated glomerular filtration rate (eGFR) using creatinine and cystatin C (eGFRcr-cys). We studied whether using β2-microglobulin (B2M) and/or β-trace protein (BTP) with creatinine with or without cystatin C (2-, 3-, or 4-marker panel eGFR) might be useful in patients with active solid tumors. The performance of the 3-marker and 4-marker panel equations was better than eGFRcr-cys. Performance of 2-marker panel equations was as good as eGFRcr-cys. We conclude that B2M and BTP can improve the accuracy of eGFR and may be useful as a confirmatory test in patients with solid tumors either by inclusion in multimarker panel equation with creatinine and cystatin C or by substituting for cystatin C in combination with creatinine.
Collapse
Affiliation(s)
- Verônica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Laboratório de Investigação Médica (LIM) 16, Serviço de Nefrologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| | - Luiz A Gil
- Laboratório de Investigação Médica (LIM) 66, Serviço de Geriatria, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Renato A Caires
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Elerson Costalonga
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - George Coura-Filho
- Serviço de Medicina Nuclear, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo T Sapienza
- Radiology and Oncology Department, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Gilberto Castro
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Maria D P Estevez-Diz
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Dirce Maria T Zanetta
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Leila Antonângelo
- Laboratório de Investigação Médica (LIM) 03, Division of Clinical Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Lia Marçal
- Laboratório de Investigação Médica (LIM) 03, Division of Clinical Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Hocine Tighiouart
- Institute for Clinical Research and Health Policy Studies, Biostatistics, Epidemiology, and Research Design Center, Tufts Medical Center, Boston, Massachusetts
| | - Shiyuan Miao
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Paul Mathew
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Emmanuel A Burdmann
- Laboratório de Investigação Médica (LIM) 12, Serviço de Nefrologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Kitchlu A, Silva VTCE, Anand S, Kala J, Abudayyeh A, Inker LA, Rosner MH, Karam S, Gudsoorkar P, Gupta S, Chen S, Klomjit N, Leung N, Milanez T, Motwani SS, Khalid SB, Srinivasan V, Wanchoo R, Beumer JH, Liu G, Tannir NM, Orchanian-Cheff A, Geng Y, Herrmann SM. Assessment of GFR in Patients with Cancer: A Statement from the American Society of Onco-Nephrology. Clin J Am Soc Nephrol 2024; 19:1061-1072. [PMID: 38848131 PMCID: PMC11321742 DOI: 10.2215/cjn.0000000000000508] [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: 11/09/2023] [Accepted: 06/03/2024] [Indexed: 07/03/2024]
Abstract
Accurate assessment of GFR is crucial to guiding drug eligibility, dosing of systemic therapy, and minimizing the risks of both undertreatment and toxicity in patients with cancer. Up to 32% of patients with cancer have baseline CKD, and both malignancy and treatment may cause kidney injury and subsequent CKD. To date, there has been lack of guidance to standardize approaches to GFR estimation in the cancer population. In this two-part statement from the American Society of Onco-Nephrology, we present key messages for estimation of GFR in patients with cancer, including the choice of GFR estimating equation, use of race and body surface area adjustment, and anticancer drug dose-adjustment in the setting of CKD. These key messages are based on a systematic review of studies assessing GFR estimating equations using serum creatinine and cystatin C in patients with cancer, against a measured GFR comparator. The preponderance of current data involving validated GFR estimating equations involves the CKD Epidemiology Collaboration (CKD-EPI) equations, with 2508 patients in whom CKD-EPI using serum creatinine and cystatin C was assessed (eight studies) and 15,349 in whom CKD-EPI with serum creatinine was assessed (22 studies). The former may have improved performance metrics and be less susceptible to shortfalls of eGFR using serum creatinine alone. Since included studies were moderate quality or lower, the American Society of Onco-Nephrology Position Committee rated the certainty of evidence as low. Additional studies are needed to assess the accuracy of other validated eGFR equations in patients with cancer. Given the importance of accurate and timely eGFR assessment, we advocate for the use of validated GFR estimating equations incorporating both serum creatinine and cystatin C in patients with cancer. Measurement of GFR via exogenous filtration markers should be considered in patients with cancer for whom eGFR results in borderline eligibility for therapies or clinical trials.
Collapse
Affiliation(s)
- Abhijat Kitchlu
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Verônica T. Costa E. Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Investigação Médica (LIM) 16, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Shuchi Anand
- Department of Medicine (Nephrology), Stanford University, Stanford, California
| | - Jaya Kala
- Division of Renal Diseases and Hypertension, University of Texas Health Science Center at Houston-McGovern Medical School, Houston, Texas
| | - Ala Abudayyeh
- Section of Nephrology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Lesley A. Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Mitchell H. Rosner
- Division of Nephrology, University of Virginia Health, Charlottesville, Virginia
| | - Sabine Karam
- Division of Nephrology and Hypertension, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Prakash Gudsoorkar
- Division of Nephrology, Kidney C.A.R.E. Program, University of Cincinnati, Cincinnati, Ohio
| | - Shruti Gupta
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sheldon Chen
- Section of Nephrology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Nattawat Klomjit
- Division of Nephrology and Hypertension, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Tomaz Milanez
- Institute of Oncology Ljubljana and Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenija
| | - Shveta S. Motwani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Sheikh B. Khalid
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Vinay Srinivasan
- Division of Nephrology, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, New Jersey
| | - Rimda Wanchoo
- Division of Kidney Diseases and Hypertension, Glomerular Center at Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York
| | - Jan H. Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Geoffrey Liu
- Princess Margaret Cancer Centre and University Health Network, University of Toronto, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Nizar M. Tannir
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ani Orchanian-Cheff
- Library and Information Services, University Health Network, Toronto, Ontario, Canada
| | - Yimin Geng
- Section of Nephrology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
- Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sandra M. Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
14
|
Gomes AC, Coura Filho GB, Gil Junior LA, Caires RA, Burdmann EA, Buchpiguel CA, Costa E Silva VT, Sapienza MT. Comparison of plasma clearance of [ 51Cr]CrEDTA based on three, two and single samples to measure the glomerular filtration rate in patients with solid tumors: a prospective cross-sectional analysis. Clinics (Sao Paulo) 2024; 79:100427. [PMID: 38945113 PMCID: PMC11261263 DOI: 10.1016/j.clinsp.2024.100427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/02/2024] Open
Abstract
OBJECTIVES [51Cr]CrEDTA is used to measure the Glomerular Filtration Rate (GFR) in different clinical conditions. However, there is no consensus on the ideal number of blood samples to be taken and at what time points to measure its clearance. This study aimed to compare Slope Intercept (SI) and Single-Sample (SS) methods for measuring GFR in patients with solid tumors, stratified by age, GFR, and Body Mass Index (BMI). METHODS 1,174 patients with cancer were enrolled in this prospective study. GFR was calculated by the SI method using blood samples drawn 2-, 4-, and 6-hours after [51Cr]CrEDTA injection (246-GFR). GFR was also measured using the SI method with samples at 2 and 4 hours (24-GFR) and at 4 and 6 hours (46-GFR), and SS methods according to Groth (4Gr-GFR) and Fleming (4Fl-GFR). Statistical analysis was performed to assess the accuracy, precision, and bias of the methods. RESULTS Mean 246-GFR was 79.2 ± 21.9 mL/min/1.73 m2. ANOVA indicated a significant difference between 4Gr-GFR and the reference 246-GFR. Bias was lower than 5 mL/min/1.73 m2 for all methods, except for SS methods in subgroups BMI > 40 kg/m2; GFR > 105 or < 45. Precision was adequate and accuracy of 30 % was above 98% for all methods, except for SS methods in subgroup GFR < 45. CONCLUSION 46-GFR and 246-GFR have high agreement and may be used to evaluate kidney function in patients with solid tumors. Single-sample methods can be adopted in specific situations, for non-obese patients with expected normal GFR.
Collapse
Affiliation(s)
- Anne C Gomes
- Divisão de Medicina Nuclear, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - George B Coura Filho
- Divisão de Medicina Nuclear, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Luiz A Gil Junior
- Laboratório de Investigação Médica (LIM 66), Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Renato A Caires
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Emmanuel A Burdmann
- Laboratório de Investigação Médica (LIM 12), Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Carlos A Buchpiguel
- Laboratório de Investigação Médica (LIM 43), Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Veronica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Investigação Médica (LIM 16), Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Marcelo T Sapienza
- Laboratório de Investigação Médica (LIM 43), Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil.
| |
Collapse
|
15
|
Hall RK, Kazancıoğlu R, Thanachayanont T, Wong G, Sabanayagam D, Battistella M, Ahmed SB, Inker LA, Barreto EF, Fu EL, Clase CM, Carrero JJ. Drug stewardship in chronic kidney disease to achieve effective and safe medication use. Nat Rev Nephrol 2024; 20:386-401. [PMID: 38491222 PMCID: PMC11929520 DOI: 10.1038/s41581-024-00823-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
People living with chronic kidney disease (CKD) often experience multimorbidity and require polypharmacy. Kidney dysfunction can also alter the pharmacokinetics and pharmacodynamics of medications, which can modify their risks and benefits; the extent of these changes is not well understood for all situations or medications. The principle of drug stewardship is aimed at maximizing medication safety and effectiveness in a population of patients through a variety of processes including medication reconciliation, medication selection, dose adjustment, monitoring for effectiveness and safety, and discontinuation (deprescribing) when no longer necessary. This Review is aimed at serving as a resource for achieving optimal drug stewardship for patients with CKD. We describe special considerations for medication use during pregnancy and lactation, during acute illness and in patients with cancer, as well as guidance for the responsible use of over-the-counter drugs, herbal remedies, supplements and sick-day rules. We also highlight inequities in medication access worldwide and suggest policies to improve access to quality and essential medications for all persons with CKD. Further strategies to promote drug stewardship include patient education and engagement, the use of digital health tools, shared decision-making and collaboration within interdisciplinary teams. Throughout, we position the person with CKD at the centre of all drug stewardship efforts.
Collapse
Affiliation(s)
- Rasheeda K Hall
- Division of Nephrology, Department of Medicine, and Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | | | | | - Germaine Wong
- Sydney School of Public Health, University of Sydney, Sydney, Australia
| | | | | | - Sofia B Ahmed
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Lesley A Inker
- Division of Nephrology, Department of Internal Medicine, Tufts Medical Center, Boston, MA, USA
| | | | - Edouard L Fu
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Catherine M Clase
- Department of Medicine and Health Research Methods, Evidence and Impact, McMaster University, Ontario, Canada
| | - Juan J Carrero
- Medical Epidemiology and Biostatistics, Karolinska Institutet, and Division of Nephrology, Department of Clinical Sciences, Danderyd Hospital, Stockholm, Sweden.
| |
Collapse
|
16
|
Hanna PE, Ouyang T, Tahir I, Katz‐Agranov N, Wang Q, Mantz L, Strohbehn I, Moreno D, Harden D, Dinulos JE, Cosar D, Seethapathy H, Gainor JF, Shah SJ, Gupta S, Leaf DE, Fintelmann FJ, Sise ME. Sarcopenia, adiposity and large discordance between cystatin C and creatinine-based estimated glomerular filtration rate in patients with cancer. J Cachexia Sarcopenia Muscle 2024; 15:1187-1198. [PMID: 38646842 PMCID: PMC11154767 DOI: 10.1002/jcsm.13469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 12/15/2023] [Accepted: 03/06/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Creatinine-based estimated glomerular filtration rate (eGFRCRE) may overestimate kidney function in patients with sarcopenia. While cystatin C-based eGFR (eGFRCYS) is less affected by muscle mass, it may underestimate kidney function in patients with obesity. We sought to evaluate the relationship between body composition defined by computed tomography (CT) scans and discordance between creatinine, eGFRCRE and eGFRCYS in adult patients with cancer. METHODS This study is a cross-sectional study of consecutive adults with cancer with an abdominal CT scan performed within 90 days of simultaneous eGFRCRE and eGFRCYS measurements between May 2010 and January 2022. Muscle and adipose tissue cross-sectional areas were measured at the level of the third lumbar vertebral body using a validated deep-learning pipeline. CT-defined sarcopenia was defined using independent sex-specific cut-offs for skeletal muscle index (<39 cm2/m2 for women and <55 cm2/m2 for men). High adiposity was defined as the highest sex-specific quartile of the total (visceral plus subcutaneous) adiposity index in the cohort. The primary outcome was eGFR discordance, defined by eGFRCYS > 30% lower than eGFRCRE; the secondary outcome was eGFRCYS > 50% lower than eGFRCRE. The odds of eGFR discordance were estimated using multivariable logistic regression modelling. Unadjusted spline regression was used to evaluate the relationship between skeletal muscle index and the difference between eGFRCYS and eGFRCRE. RESULTS Of the 545 included patients (mean age 63 ± 14 years, 300 [55%] females, 440 [80.7%] non-Hispanic white), 320 (58.7%) met the criteria for CT-defined sarcopenia, and 136 (25%) had high adiposity. A total of 259 patients (48%) had >30% eGFR discordance, and 122 (22.4%) had >50% eGFR discordance. After adjustment for potential confounders, CT-defined sarcopenia and high adiposity were both associated with >30% eGFR discordance (adjusted odds ratio [aOR] 1.90, 95% confidence interval [CI] 1.12-3.24; aOR 2.01, 95% CI 1.15-3.52, respectively) and >50% eGFR discordance (aOR 2.34, 95% CI 1.21-4.51; aOR 2.23, 95% CI 1.19-4.17, respectively). A spline model demonstrated that as skeletal muscle index decreases, the predicted difference between eGFRCRE and eGFRCYS widens considerably. CONCLUSIONS CT-defined sarcopenia and high adiposity are both independently associated with large eGFR discordance. Incorporating valuable information from body composition analysis derived from CT scans performed as a part of routine cancer care can impact the interpretation of GFR estimates.
Collapse
Affiliation(s)
- Paul E. Hanna
- Division of Nephrology, Department of MedicineMedical College of WisconsinMilwaukeeWIUSA
| | - Tianqi Ouyang
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Ismail Tahir
- Department of RadiologyMassachusetts General HospitalBostonMAUSA
| | - Nurit Katz‐Agranov
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Qiyu Wang
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Lea Mantz
- Department of RadiologyMassachusetts General HospitalBostonMAUSA
- Department of Diagnostic and Interventional RadiologyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Ian Strohbehn
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Daiana Moreno
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Destiny Harden
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - James E. Dinulos
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Duru Cosar
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Harish Seethapathy
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Justin F. Gainor
- Division of Hematology and Oncology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Sachin J. Shah
- Division of General Internal Medicine, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | - Shruti Gupta
- Division of Renal Medicine, Department of MedicineBrigham and Women's HospitalBostonMAUSA
- Adult Survivorship ProgramDana‐Farber Cancer InstituteBostonMAUSA
| | - David E. Leaf
- Division of General Internal Medicine, Department of MedicineMassachusetts General HospitalBostonMAUSA
| | | | - Meghan E. Sise
- Division of Nephrology, Department of MedicineMassachusetts General HospitalBostonMAUSA
| |
Collapse
|
17
|
Stevens PE, Ahmed SB, Carrero JJ, Foster B, Francis A, Hall RK, Herrington WG, Hill G, Inker LA, Kazancıoğlu R, Lamb E, Lin P, Madero M, McIntyre N, Morrow K, Roberts G, Sabanayagam D, Schaeffner E, Shlipak M, Shroff R, Tangri N, Thanachayanont T, Ulasi I, Wong G, Yang CW, Zhang L, Levin A. KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int 2024; 105:S117-S314. [PMID: 38490803 DOI: 10.1016/j.kint.2023.10.018] [Citation(s) in RCA: 515] [Impact Index Per Article: 515.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 03/17/2024]
|
18
|
Butrovich MA, Qin J, Xue X, Ivy SP, Nolin TD, Beumer JH. Impact of the 2021 CKD-EPI equation on anticancer pharmacotherapy in black and non-black cancer patients. Cancer Lett 2024; 586:216679. [PMID: 38307411 PMCID: PMC10939791 DOI: 10.1016/j.canlet.2024.216679] [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: 12/09/2023] [Revised: 01/14/2024] [Accepted: 01/24/2024] [Indexed: 02/04/2024]
Abstract
Cancer and kidney disease disproportionately impact Black patients. The CKD-EPI2021 equation was developed to estimate glomerular filtration rate (eGFR) without using race. We assessed the impact of using CKD-EPI2021 instead of CKD-EPI2009 or Cockcroft-Gault (CG) on dosing and eligibility of anticancer drugs in Black and non-Black patients. Utilizing the National Cancer Institute Theradex database, deindexed eGFR (mL/min) was calculated for 3931 patients (8.6 % Black) using CKD-EPI2021, CKD-EPI2009, and CG. Dosing simulations based on each eGFR were performed for ten anticancer drugs with kidney function-based eligibility or dosing cutoffs. eGFR differences using CKD-EPI2021 versus CKD-EPI2009 varied between Black and non-Black patients (p < 0.001); on average, Black patients had 10.3 mL/min lower eGFR and non-Black patients had 4.2 mL/min higher eGFR using CKD-EPI2021. This corresponded to a difference in relative odds of cisplatin ineligibility using CKD-EPI2021 versus CKD-EPI2009; Black patients had 48 % higher odds of ineligibility and non-Black patients had 27 % lower odds of ineligibility using CKD-EPI2021 (p < 0.001). When using CKD-EPI2021 versus CG, eGFR differences were similar between Black and non-Black patients (p = 0.679) and relative difference in odds of cisplatin ineligibility did not vary. Using CKD-EPI2021 versus CKD-EPI2009 differentially impacts Black versus non-Black cancer patients; Black patients have lower calculated eGFR and are less likely to receive full doses of drug using CKD-EPI2021. From the historical default of CG, adopting CKD-EPI2021 would not disparately impact patients based on race, but would result in Black patients being less likely to receive full doses of drug than if CKD-EPI2009 were used.
Collapse
Affiliation(s)
- Morgan A Butrovich
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jiyue Qin
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY, 10461, USA; Division of Biostatistics and Bioinformatics, Herbert Wertheim School of Public Health, University of California San Diego, La Jolla, CA, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY, 10461, USA
| | - S Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Thomas D Nolin
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Jan H Beumer
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Hematology/Oncology Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
19
|
Levin A, Ahmed SB, Carrero JJ, Foster B, Francis A, Hall RK, Herrington WG, Hill G, Inker LA, Kazancıoğlu R, Lamb E, Lin P, Madero M, McIntyre N, Morrow K, Roberts G, Sabanayagam D, Schaeffner E, Shlipak M, Shroff R, Tangri N, Thanachayanont T, Ulasi I, Wong G, Yang CW, Zhang L, Robinson KA, Wilson L, Wilson RF, Kasiske BL, Cheung M, Earley A, Stevens PE. Executive summary of the KDIGO 2024 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease: known knowns and known unknowns. Kidney Int 2024; 105:684-701. [PMID: 38519239 DOI: 10.1016/j.kint.2023.10.016] [Citation(s) in RCA: 69] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 03/24/2024]
Abstract
The Kidney Disease: Improving Global Outcomes (KDIGO) Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease (CKD) updates the KDIGO 2012 guideline and has been developed with patient partners, clinicians, and researchers around the world, using robust methodology. This update, based on a substantially broader base of evidence than has previously been available, reflects an exciting time in nephrology. New therapies and strategies have been tested in large and diverse populations that help to inform care; however, this guideline is not intended for people receiving dialysis nor those who have a kidney transplant. The document is sensitive to international considerations, CKD across the lifespan, and discusses special considerations in implementation. The scope includes chapters dedicated to the evaluation and risk assessment of people with CKD, management to delay CKD progression and its complications, medication management and drug stewardship in CKD, and optimal models of CKD care. Treatment approaches and actionable guideline recommendations are based on systematic reviews of relevant studies and appraisal of the quality of the evidence and the strength of recommendations which followed the "Grading of Recommendations Assessment, Development, and Evaluation" (GRADE) approach. The limitations of the evidence are discussed. The guideline also provides practice points, which serve to direct clinical care or activities for which a systematic review was not conducted, and it includes useful infographics and describes an important research agenda for the future. It targets a broad audience of people with CKD and their healthcare, while being mindful of implications for policy and payment.
Collapse
Affiliation(s)
- Adeera Levin
- Division of Nephrology, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Sofia B Ahmed
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Juan Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Bethany Foster
- Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Anna Francis
- Department of Medicine, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Rasheeda K Hall
- Division of Nephrology, Duke School of Medicine, Durham, North Carolina, USA
| | - Will G Herrington
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | | | - Edmund Lamb
- Department of Clinical Biochemistry, East Kent Hospitals University NHS Foundation Trust, Canterbury, UK
| | - Peter Lin
- Director of Primary Care Initiatives, Canadian Heart Research Center, Toronto, Ontario, Canada
| | - Magdalena Madero
- Division of Nephrology, Instituto Nacional de Cardiología Ignacio Chavéz, Mexico City, Mexico
| | - Natasha McIntyre
- London Health Sciences Centre-Victoria Hospital, Western University, London, Ontario, Canada
| | - Kelly Morrow
- Department of Nutrition and Exercise Science, Bastyr University, Kenmore, Washington, USA; Osher Center for Integrative Medicine, University of Washington, Kenmore, Washington, USA
| | - Glenda Roberts
- UW Center for Dialysis Innovation & Kidney Research Institute, Seattle, Washington, USA
| | | | - Elke Schaeffner
- Division of Nephrology and Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Shlipak
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Rukshana Shroff
- Department of Paediatric Nephrology, UCL Great Ormond Street Hospital Institute of Child Health, London, UK
| | - Navdeep Tangri
- Division of Nephrology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Ifeoma Ulasi
- Department of Medicine, Ituku-Ozalla Campus, University of Nigeria, Enugu, Nigeria
| | - Germaine Wong
- Western Renal Service, University of Sydney, Sydney, New South Wales, Australia
| | - Chih-Wei Yang
- Division of Nephrology, Chang Gung University, Taoyuan, Taiwan
| | - Luxia Zhang
- Renal Division, Peking University First Hospital, Beijing, China
| | - Karen A Robinson
- The Johns Hopkins University Evidence-based Practice Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lisa Wilson
- The Johns Hopkins University Evidence-based Practice Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Renee F Wilson
- The Johns Hopkins University Evidence-based Practice Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bertram L Kasiske
- Hennepin County Medical Center, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | - Paul E Stevens
- Department of Nephrology, Kent Kidney Care Centre, East Kent Hospitals University NHS Foundation Trust, Canterbury, UK.
| |
Collapse
|
20
|
Fino NF, Adingwupu OM, Coresh J, Greene T, Haaland B, Shlipak MG, Costa E Silva VT, Kalil R, Mindikoglu AL, Furth SL, Seegmiller JC, Levey AS, Inker LA. Evaluation of novel candidate filtration markers from a global metabolomic discovery for glomerular filtration rate estimation. Kidney Int 2024; 105:582-592. [PMID: 38006943 PMCID: PMC10932836 DOI: 10.1016/j.kint.2023.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 11/27/2023]
Abstract
Creatinine and cystatin-C are recommended for estimating glomerular filtration rate (eGFR) but accuracy is suboptimal. Here, using untargeted metabolomics data, we sought to identify candidate filtration markers for a new targeted assay using a novel approach based on their maximal joint association with measured GFR (mGFR) and with flexibility to consider their biological properties. We analyzed metabolites measured in seven diverse studies encompasing 2,851 participants on the Metabolon H4 platform that had Pearson correlations with log mGFR and used a stepwise approach to develop models to < -0.5 estimate mGFR with and without inclusion of creatinine that enabled selection of candidate markers. In total, 456 identified metabolites were present in all studies, and 36 had correlations with mGFR < -0.5. A total of 2,225 models were developed that included these metabolites; all with lower root mean square errors and smaller coefficients for demographic variables compared to estimates using untargeted creatinine. Seventeen metabolites were chosen, including 12 new candidate filtration markers. The selected metabolites had strong associations with mGFR and little dependence on demographic factors. Candidate metabolites were identified with maximal joint association with mGFR and minimal dependence on demographic variables across many varied clinical settings. These metabolites are excreted in urine and represent diverse metabolic pathways and tubular handling. Thus, our data can be used to select metabolites for a multi-analyte eGFR determination assay using mass spectrometry that potentially offers better accuracy and is less prone to non-GFR determinants than the current eGFR biomarkers.
Collapse
Affiliation(s)
- Nora F Fino
- Division of Biostatistics, Department of Population Health Sciences, University of Utah Health, Salt Lake City, Utah, USA
| | - Ogechi M Adingwupu
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Josef Coresh
- Department of Population Health, NYU Langone, New York, New York, USA
| | - Tom Greene
- Division of Biostatistics, Department of Population Health Sciences, University of Utah Health, Salt Lake City, Utah, USA
| | - Ben Haaland
- Division of Biostatistics, Department of Population Health Sciences, University of Utah Health, Salt Lake City, Utah, USA
| | - Michael G Shlipak
- Kidney Health Research Collaborative, San Francisco Veterans Affair Medical Center and University of California, San Francisco, San Francisco, California, USA
| | - Veronica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Laboratório de Investigação Médica 16, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Roberto Kalil
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ayse L Mindikoglu
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA; Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, Texas, USA
| | - Susan L Furth
- Department of Pediatrics, Children's Hospital of Philadelphia, and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jesse C Seegmiller
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA.
| |
Collapse
|
21
|
Tanizaki J, Hayashi H. Unraveling Pseudo Kidney Injury: The Significance of Understanding Our "MATE" in Molecular-Targeted Therapies. J Thorac Oncol 2024; 19:15-17. [PMID: 38185510 DOI: 10.1016/j.jtho.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 01/09/2024]
Affiliation(s)
- Junko Tanizaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan.
| |
Collapse
|
22
|
Chen MF, Harada G, Liu D, DeMatteo R, Falcon C, Wilhelm C, Kris MG, Drilon A, Gutgarts V. Brief Report: Tyrosine Kinase Inhibitors for Lung Cancers That Inhibit MATE-1 Can Lead to "False" Decreases in Renal Function. J Thorac Oncol 2024; 19:153-159. [PMID: 37748692 PMCID: PMC10841070 DOI: 10.1016/j.jtho.2023.09.1444] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/11/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023]
Abstract
INTRODUCTION Select tyrosine kinase inhibitors (TKIs) used to treat oncogene-driven lung cancers also inhibit MATE-1. When MATE-1 is blocked, creatinine is retained in the serum. Elevated creatinine levels raise the specter of drug-induced intrarenal insufficiency despite the lack of true renal injury. We conducted a systematic analysis of MATE-1 inhibitor (MATEi)-treated patients to comprehensively characterize this phenomenon. METHODS Patients with oncogene-driven lung cancer treated with a wide variety of MATEi TKIs (brigatinib, cabozantinib, capmatinib, crizotinib, entrectinib, lorlatinib, pralsetinib, selpercatinib, and tepotinib) were eligible for an analysis of renal dysfunction. Acute kidney injury was classified on the basis of creatinine levels (Kidney Disease: Improving Global Outcomes criteria) as stage 1 (≥1.5× but <2× baseline), stage 2 (≥2× but <3× baseline), or stage 3 (>3× baseline). When available, cystatin C, a marker of kidney function unaffected by MATE-1, was used to evaluate the glomerular filtration rate (GFR). RESULTS We identified 863 patients receiving MATEi TKIs including crizotinib (39%, n = 333), lorlatinib (17%, n = 144), cabozantinib (10%, n = 87), selpercatinib (10%, n = 82), capmatinib (9%, n = 77), brigatinib (6%, n = 53), entrectinib (5%, n = 45), tepotinib (5%, n = 41), and pralsetinib (0.1%, n = 1). Of the 90 patients (10%) with acute kidney injury, Kidney Disease: Improving Global Outcomes stages 1, 2, and 3 were observed in 72% (n = 65), 21% (n = 19), and 7% (n = 6) of patients, respectively. Concurrently drawn creatinine and cystatin C levels on TKI therapy were available for 17 patients. In most cases (n = 15 of 17), the calculated GFR was higher using cystatin C versus creatinine. The percentage of patients whose GFR was higher using cystatin C versus creatinine by less than 10 mL/min, 10 to 19 mL/min, 20 to 29 mL/min, and more than or equal to 30 mL/min was 27% (n = four of 15), 20% (n = three of 15), 20% (n = three of 15), and 33% (n = five of 15), respectively. Long-term data in three patients that spanned 3 years revealed that GFR was higher using cystatin C versus creatinine in 96% (n = 49 of 51) of all time points. Using a virtual clinical trial GFR cutoff of 40 mL/min, the percentage of eligible patients rose from 41% (n = seven of 17) using creatinine calculations to 71% (n = 12 of 17) using cystatin C calculations. CONCLUSIONS The calculated GFR in patients with cancer receiving MATEi TKIs was higher in almost all cases when using cystatin C. When serum creatinine level seems elevated in patients receiving MATE-1 inhibitors, we recommend recalculating GFR using cystatin C before searching for other etiologies of kidney injury and reducing or stopping TKI therapy.
Collapse
Affiliation(s)
- Monica F Chen
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York; Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Guilherme Harada
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Dazhi Liu
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Ray DeMatteo
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Christina Falcon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York; Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Clare Wilhelm
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Mark G Kris
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York; Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York.
| | - Victoria Gutgarts
- Division of Nephrology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| |
Collapse
|
23
|
Lemire F, Fergusson DA, Knoll G, Morash C, Lavallée LT, Mallick R, Finelli A, Kapoor A, Pouliot F, Izawa J, Rendon R, Cagiannos I, Breau RH. Estimated glomerular filtration rate from the renal hypothermia trial: clinical implications. BJU Int 2023; 132:452-460. [PMID: 37409827 DOI: 10.1111/bju.16114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
OBJECTIVE To assess if estimated glomerular filtration rate (eGFR) can replace measured GFR (mGFR) in partial nephrectomy (PN) trials, using data from a randomised clinical trial. PATIENTS AND METHODS We conducted a post hoc analysis of the renal hypothermia trial. Patients underwent mGFR with diethylenetriaminepentaacetic acid (DTPA) plasma clearance preoperatively and 1 year after PN. The eGFR was calculated using the 2009 Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) creatinine equations incorporating age and sex, with and without race: 2009 eGFRcr(ASR) and 2009 eGFRcr(AS), and the 2021 equation that only incorporates age and sex: 2021 eGFRcr(AS). Performance was evaluated by determining the median bias, precision (interquartile range [IQR] of median bias), and accuracy (percentage of eGFR within 30% of mGFR). RESULTS Overall, 183 patients were included. Pre- and postoperative median bias and precision were similar between the 2009 eGFRcr(ASR) (-0.2 mL/min/1.73 m2 , 95% confidence interval [CI] -2.2 to 1.7, IQR 18.8; and -2.9, 95% CI -5.1 to -1.5, IQR 15, respectively) and 2009 eGFRcr(AS) (-0.3 mL/min/1.73 m2 , 95% CI -2.4 to 1.5, IQR 18.8; and -3.0, 95% CI -5.7 to -1.7, IQR 15.0, respectively). Bias and precision were worse for the 2021 eGFRcr(AS) (-8.8 mL/min/1.73 m2 , 95% CI -10.9 to -6.3, IQR 24.7; and -12.0, 95% CI -15.8 to -8.9, IQR 23.5, respectively). Similarly, pre- and postoperative accuracy was >90% for the 2009 eGFRcr(ASR) and 2009 eGFRcr(AS) equations. Accuracy was 78.6% preoperatively and 66.5% postoperatively for 2021 eGFRcr(AS). CONCLUSION The 2009 eGFRcr(AS) can accurately estimate GFR in PN trials and could be used instead of mGFR to reduce cost and patient burden.
Collapse
Affiliation(s)
- Francis Lemire
- Division of Urology, Department of Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Greg Knoll
- Division of Nephrology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Christopher Morash
- Division of Urology, Department of Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Luke T Lavallée
- Division of Urology, Department of Surgery, The Ottawa Hospital, Ottawa, ON, Canada
- Division of Nephrology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Ranjeeta Mallick
- Division of Nephrology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Antonio Finelli
- Division of Urology, Departments of Surgery and Surgical Oncology, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Anil Kapoor
- Division of Urology, McMaster University, Hamilton, ON, Canada
| | | | - Jonathan Izawa
- Division of Urology, Department of Surgery, Western University, London, ON, Canada
| | - Ricardo Rendon
- Department of Urology, Dalhousie University, Halifax, NS, Canada
| | - Ilias Cagiannos
- Division of Urology, Department of Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Rodney H Breau
- Division of Urology, Department of Surgery, The Ottawa Hospital, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
24
|
Hanna PE, Wang Q, Strohbehn IA, Moreno D, Harden D, Ouyang T, Katz-Agranov N, Seethapathy H, Reynolds KL, Gupta S, Leaf DE, Sise ME. Medication-Related Adverse Events and Discordancies in Cystatin C-Based vs Serum Creatinine-Based Estimated Glomerular Filtration Rate in Patients With Cancer. JAMA Netw Open 2023; 6:e2321715. [PMID: 37405775 PMCID: PMC10323710 DOI: 10.1001/jamanetworkopen.2023.21715] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/19/2023] [Indexed: 07/06/2023] Open
Abstract
Importance Serum creatinine-based estimated glomerular filtration rate (eGFRcr) may overestimate the glomerular filtration rate (GFR) in patients with cancer. Cystatin C-based eGFR (eGFRcys) is an alternative marker of GFR. Objective To determine whether the therapeutic drug levels and adverse events (AEs) associated with renally cleared medications were higher in patients with cancer whose eGFRcys was more than 30% lower than their eGFRcr. Design, Setting, and Participants This cohort study analyzed adult patients with cancer at 2 major academic cancer centers in Boston, Massachusetts. These patients had their creatinine and cystatin C measured on the same day between May 2010 and January 2022. The date of the first simultaneous eGFRcr and eGFRcys measurement was considered to be the baseline date. Exposure The primary exposure was eGFR discordance, defined as an eGFRcys that was more than 30% lower than the eGFRcr. Main Outcomes and Measures The primary outcome was risk of the following medication-related AEs within 90 days of the baseline date: (1) supratherapeutic vancomycin trough level greater than 30 μg/mL, (2) trimethoprim-sulfamethoxazole-related hyperkalemia (>5.5 mEq/L), (3) baclofen toxic effect, and (4) supratherapeutic digoxin level (>2.0 ng/mL). For the secondary outcome, a multivariable Cox proportional hazards regression model was used to compare 30-day survival of those with vs without eGFR discordance. Results A total of 1869 adult patients with cancer (mean [SD] age, 66 [14] years; 948 males [51%]) had simultaneous eGFRcys and eGFRcr measurement. There were 543 patients (29%) with an eGFRcys that was more than 30% lower than their eGFRcr. Patients with an eGFRcys that was more than 30% lower than their eGFRcr were more likely to experience medication-related AEs compared with patients with concordant eGFRs (defined as eGFRcys within 30% of eGFRcr), including vancomycin levels greater than 30 μg/mL (43 of 179 [24%] vs 7 of 77 [9%]; P = .01), trimethoprim-sulfamethoxazole-related hyperkalemia (29 of 129 [22%] vs 11 of 92 [12%]; P = .07), baclofen toxic effects (5 of 19 [26%] vs 0 of 11; P = .19), and supratherapeutic digoxin levels (7 of 24 [29%] vs 0 of 10; P = .08). The adjusted odds ratio for vancomycin levels more than 30 μg/mL was 2.59 (95% CI, 1.08-7.03; P = .04). Patients with an eGFRcys more than 30% lower than their eGFRcr had an increased 30-day mortality (adjusted hazard ratio, 1.98; 95% CI, 1.26-3.11; P = .003). Conclusions and relevance Results of this study suggest that among patients with cancer with simultaneous assessment of eGFRcys and eGFRcr, supratherapeutic drug levels and medication-related AEs occurred more commonly in those with an eGFRcys more than 30% lower than their eGFRcr. Future prospective studies are needed to improve and personalize GFR estimation and medication dosing in patients with cancer.
Collapse
Affiliation(s)
- Paul E. Hanna
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Qiyu Wang
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Ian A. Strohbehn
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Daiana Moreno
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Destiny Harden
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Tianqi Ouyang
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Nurit Katz-Agranov
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Harish Seethapathy
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Kerry L. Reynolds
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Shruti Gupta
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Adult Survivorship Program, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David E. Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Meghan E. Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| |
Collapse
|
25
|
Claudel SE, Gandhi M, Patel AB, Verma A. Estimating kidney function in patients with cancer: A narrative review. Acta Physiol (Oxf) 2023; 238:e13977. [PMID: 37057998 PMCID: PMC11839183 DOI: 10.1111/apha.13977] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/15/2023]
Abstract
AIM Accurate evaluation of glomerular filtration rate (GFR) is crucial in Oncology as drug eligibility and dosing depend on estimates of GFR. However, there are no clear guidelines on the optimal method of determining kidney function in patients with cancer. We aimed to summarize the evidence on estimation of kidney function in patients with cancer. METHODS We searched PubMed for literature discussing the performance of GFR estimating equations in patients with malignancy to create a table of the evidence for creatinine- and cystatin c-based equations. We further reviewed novel estimation techniques such as panel eGFR, real-time measured GFR, and functional magnetic resonance imaging. RESULTS The commonly used GFR estimating equations were derived from populations of patients without cancer. These equations may be less applicable in Oncology due to severe sarcopenia, inflammation, and other physiologic changes in patients with cancer. The Cockcroft-Gault equation currently dominates in clinical Oncology despite significant limitations and accumulating evidence for use of the CKD-EPICr formula. Additional considerations in the practice of Oncology include a recently developed equation (CamGFRv2, also called the Janowitz formula) and the use of cystatin c-based equations to overcome some of the barriers to accurate GFR estimation based on creatinine alone. CONCLUSION Overall, we suggest using the CKD-EPI equations (either cystatin c or creatinine-based) among patients with cancer in routine clinical practice and measured GFR for patients at a critical threshold for treatment decisions.
Collapse
Affiliation(s)
- Sophie E. Claudel
- Department of Internal Medicine, Boston Medical Center, Boston, Massachusetts, USA
| | - Malini Gandhi
- Harvard Medical School, Boston, Massachusetts, USA
- Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ankit B. Patel
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Ashish Verma
- Department of Medicine, Section on Nephrology, Boston Medical Center, Boston, Massachusetts, USA
- Amyloidosis Center, Boston Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Butrovich MA, Reaves AC, Heyward J, Moore TJ, Alexander GC, Inker LA, Nolin TD. Inclusion of Participants with CKD and Other Kidney-Related Considerations during Clinical Drug Development: Landscape Analysis of Anticancer Agents Approved from 2015 to 2019. Clin J Am Soc Nephrol 2023; 18:455-464. [PMID: 36723359 PMCID: PMC10103296 DOI: 10.2215/cjn.0000000000000105] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/18/2023] [Indexed: 02/02/2023]
Abstract
BACKGROUND The US Food and Drug Administration has prioritized efforts to expand availability of therapies, including anticancer agents, for patients with CKD. US Food and Drug Administration Guidance recommends inclusion of study participants with CKD in clinical trials, improving pharmacokinetic characterization in people with decreased GFR, and using contemporary GFR assessment methods during drug development. We performed a landscape analysis of anticancer agents approved from 2015 to 2019 to evaluate inclusion of study participants with CKD and GFR assessment methods used during drug development and subsequent translation to kidney-related safety and dosing data in product labeling. METHODS Oncology drugs approved from 2015 to 2019 and associated pivotal trials were identified. We evaluated inclusion of study participants with CKD in pivotal trials and pharmacokinetic analyses, investigated GFR assessment methods used for pivotal trial eligibility and renal pharmacokinetic analyses, and identified kidney-related adverse drug event and dosing information. RESULTS A total of 55 drugs and 74 pivotal trials were included. Of the pivotal trials, 95% contained kidney-related eligibility criteria, including 68% with GFR-based eligibility. The median lower limit of GFR required for inclusion was 45 ml/min or ml/min per 1.73 m 2 . Pharmacokinetic analyses were performed in CKD stages 4-5 and hemodialysis for only 29% and 6% of drugs, respectively. Estimated creatinine clearance was used in over 60% and 80% of pivotal trials and pharmacokinetic analyses, respectively. Reporting of kidney-related adverse drug events was highly variable. Product labeling for 49% of drugs contained no kidney dosing information. CONCLUSIONS Study participants with CKD continue to be excluded from anticancer drug development, and GFR estimation in pivotal trials and renal pharmacokinetic analyses remains imprecise and heterogeneous. Furthermore, kidney-related safety and dosing information is scarcely and inconsistently presented.
Collapse
Affiliation(s)
- Morgan A. Butrovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Allison C. Reaves
- William B. Schwartz, MD, Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Jamie Heyward
- Center for Drug Safety and Effectiveness, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Thomas J. Moore
- Center for Drug Safety and Effectiveness, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - G. Caleb Alexander
- Center for Drug Safety and Effectiveness, Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Lesley A. Inker
- William B. Schwartz, MD, Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Thomas D. Nolin
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
27
|
Vanhoutte T, Sprangers B. Pseudo-AKI associated with targeted anti-cancer agents-the truth is in the eye of the filtration marker. Clin Kidney J 2023; 16:603-610. [PMID: 37007700 PMCID: PMC10061433 DOI: 10.1093/ckj/sfad011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Indexed: 01/18/2023] Open
Abstract
Besides true acute kidney injury (AKI), the occurrence of pseudo-AKI has been associated with several targeted agents. To improve the management of cancer patients treated with targeted agents, we need to be aware of this and use diagnostic approaches to differentiate between pseudo-AKI and AKI. In an article by Wijtvliet et al. in this issue of CKJ, tepotinib is added to the list of targeted agents associated with pseudo-AKI. In this editorial we discuss the current literature regarding pseudo-AKI and true AKI associated with targeted agents, and subsequently propose a management strategy to monitor kidney function in patients treated with targeted agents.
Collapse
Affiliation(s)
- Thomas Vanhoutte
- Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Ben Sprangers
- Ziekenhuis Oost-Limburg, Genk, Belgium
- Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium
| |
Collapse
|
28
|
Costa E Silva VT, Gil LA, Inker LA, Caires RA, Costalonga E, Coura-Filho G, Sapienza MT, Castro G, Estevez-Diz MDP, Zanetta DMT, Antonângelo L, Marçal L, Tighiouart H, Miao S, Mathew P, Levey AS, Burdmann EA. A Prospective Cross-Sectional Study on the Performance of the 2021 CKD-EPI Equations Without Race in a Multiracial Population of Adults With Solid Tumors in Brazil. Am J Kidney Dis 2023:S0272-6386(23)00577-2. [PMID: 36965828 DOI: 10.1053/j.ajkd.2023.01.445] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/03/2023] [Indexed: 03/27/2023]
Affiliation(s)
- Verônica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, BR; LIM 12, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, BR.
| | - Luiz A Gil
- LIM 66, Serviço de Geriatria, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, BR
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Renato A Caires
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, BR
| | - Elerson Costalonga
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, BR
| | - George Coura-Filho
- Serviço de Medicina Nuclear, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, BR
| | - Marcelo T Sapienza
- Radiology and Oncology Department, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, BR
| | - Gilberto Castro
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, BR
| | - Maria D P Estevez-Diz
- Serviço de Oncologia Clínica, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, BR
| | - Dirce Maria T Zanetta
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, SP, BR
| | - Leila Antonângelo
- LIM 03, Division of Clinical Pathology, University of São Paulo Medical School, São Paulo, SP, BR
| | - Lia Marçal
- LIM 03, Division of Clinical Pathology, University of São Paulo Medical School, São Paulo, SP, BR
| | - Hocine Tighiouart
- Institute for Clinical Research and Health Policy Studies, Biostatistics, Epidemiology, and Research Design (BERD) Center, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Shiyuan Miao
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Paul Mathew
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Emmanuel A Burdmann
- LIM 12, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, BR
| |
Collapse
|
29
|
Bonnet M, Jouinot A, Boudou-Rouquette P, Seif V, Villeminey C, Arrondeau J, Vidal M, Batista R, Wislez M, Blanchet B, Goldwasser F, Thomas-Schoemann A. Predictive factors associated with pemetrexed acute toxicity. Eur J Clin Pharmacol 2023; 79:635-641. [PMID: 36951965 DOI: 10.1007/s00228-023-03478-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/13/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE Pemetrexed has shown efficacy as monotherapy or in combination with platinum salts in the treatment of non-small cell lung cancer and mesothelioma. However, severe hematological toxicities induced by pemetrexed-based chemotherapy have been observed. Some studies have suggested that drug interactions may be associated with pemetrexed toxicity. The objective of this study was to determine predictive factors, including drug interactions, associated with pemetrexed toxicity. METHODS This retrospective open monocentric study included patients consecutively treated with pemetrexed after a multidisciplinary risk assessment. Patients who experienced toxicity of grade 3 or 4 according to the Common Terminology Criteria for Adverse Events v5.0, or a grade 2 leading to a change in management, during the first four courses of pemetrexed, were assigned to the early limiting toxicities (ELT) group. Univariate and multivariable logistic regression models were used to test the association variables with the occurrence of ELT. RESULTS Seventy-four patients were included in this study (median age: 67 years, with non-small cell lung cancer adenocarcinoma (88%), mesothelioma (7%), or others (5%). Thirty-six patients (49%) were assigned to the ELT group (27 grades 3 and 4; 9 grade 2 with management modification). Three baseline factors were associated with pemetrexed ELT in univariate and multivariate analysis: cystatin clearance (p = 0.0135), albumin level (p = 0.0333), and proton pump inhibitors use (p = 0.035). CONCLUSION To conclude, ELT induced by pemetrexed-based treatments occur frequently in cancer patients in a real-world setting. A pretherapeutic assessment before pemetrexed initiation should include three major checkpoints: use of proton pump inhibitors, sarcopenia, and denutrition evaluation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michel Vidal
- Pharmacocinétique et Pharmacochimie, Hôpital Cochin, Paris, France
- UMR8038, CNRS, U.1268 INSERM, Faculty of Pharmacy, University Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | | | | | - Benoit Blanchet
- Pharmacocinétique et Pharmacochimie, Hôpital Cochin, Paris, France
| | | | - Audrey Thomas-Schoemann
- Pharmacie, Hôpital Cochin, Paris, France.
- Oncologie, Hôpital Cochin, Paris, France.
- UMR8038, CNRS, U.1268 INSERM, Faculty of Pharmacy, University Paris Descartes, PRES Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
30
|
Min B, Yun SR, Yoon SH, Kim JD, Hwang WJ, Hwang WM, Park Y. Comparison of the association intensity of creatinine and cystatin C with hyperphosphatemia and hyperparathyroidism in patients with chronic kidney disease. Sci Rep 2023; 13:3855. [PMID: 36890290 PMCID: PMC9995313 DOI: 10.1038/s41598-023-31048-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/06/2023] [Indexed: 03/10/2023] Open
Abstract
Herein, we compared the association intensity of estimated glomerular filtration rate (eGFR) equations using creatinine (Cr) or cystatin C (CysC) with hyperphosphatemia and secondary hyperparathyroidism occurrence, which reflect the physiological changes occurring during chronic kidney disease (CKD) progression. This study included 639 patients treated between January 2019 and February 2022. The patients were divided into low- and high-difference groups based on the median value of the difference between the Cr-based eGFR (eGFRCr) and CysC-based eGFR (eGFRCysC). Sociodemographic and laboratory factors underlying a high difference between eGFRCr and eGFRCysC were analyzed. The association intensity of eGFRCr, eGFRCysC and both Cr- and CysC-based eGFR (eGFRCr-CysC) was compared using the area under the receiver operating characteristic curve (AuROC) values for hyperphosphatemia and hyperparathyroidism occurrence in the overall cohort and the low- and high-difference groups. Age > 70 years and CKD grade 3 based on eGFRCr were significant factors affecting the high differences. eGFRCysC and eGFRCr-CysC showed higher AuROC values than that of eGFRCr, especially in the high-difference group and in patients with CKD grade 3. Our results show that CysC should be evaluated in patients with significant factors, including age > 70 years and CKD grade 3, to accurately assess kidney function to better determine the physiological changes in CKD progression and predict prognosis accurately.
Collapse
Affiliation(s)
- Byungju Min
- Division of Nephrology, Department of Internal Medicine, Konyang University Hospital, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea
| | - Sung-Ro Yun
- Division of Nephrology, Department of Internal Medicine, Konyang University Hospital, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea
| | - Se-Hee Yoon
- Division of Nephrology, Department of Internal Medicine, Konyang University Hospital, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea
| | - Jong-Dai Kim
- Division of Endocrinology, Department of Internal Medicine, Konyang University Hospital, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea
| | - Wan Jin Hwang
- Konyang University Myunggok Medical Research Institute, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea
| | - Won Min Hwang
- Division of Nephrology, Department of Internal Medicine, Konyang University Hospital, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea.
- Konyang University Myunggok Medical Research Institute, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea.
| | - Yohan Park
- Division of Nephrology, Department of Internal Medicine, Konyang University Hospital, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea.
- Konyang University Myunggok Medical Research Institute, Gwanjeodong-Ro 158, Seo-Gu, Daejeon, 35365, Republic of Korea.
| |
Collapse
|
31
|
Hanna PE, Wang Q, Strohbehn I, Moreno D, Harden D, Ouyang T, Katz-Agranov N, Seethapathy H, Reynolds KL, Gupta S, Leaf DE, Sise ME. Medication-related adverse events in patients with cancer and discrepancies in cystatin C- versus creatinine-based eGFR. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.18.23284656. [PMID: 36711583 PMCID: PMC9882433 DOI: 10.1101/2023.01.18.23284656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background Creatinine-based estimated glomerular filtration rate (eGFRCRE) may overestimate kidney function in patients with cancer. Cystatin C-based eGFR (eGFRCYS) is an alternative marker of kidney function. We investigated whether patients with an eGFR discrepancy, defined as eGFRCYS >30% lower than the concurrent eGFRCRE, had an increased risk of adverse events resulting from renally-cleared medications. Patients and Methods We conducted a cohort study of adult patients with cancer who had serum creatinine and cystatin C measured on the same day between May 2010 and January 2022 at two academic cancer centers in Boston, MA. The primary outcome was the incidence of each of the following medication-related adverse events: 1) supratherapeutic vancomycin levels (>30μg/mL); 2) trimethoprim-sulfamethoxazole-related hyperkalemia (>5.5mEq/L); 3) baclofen-induced neurotoxicity; and 4) supratherapeutic digoxin levels (>2.0ng/mL). Results 1988 patients with cancer had simultaneous eGFRCYS and eGFRCRE. The mean age was 66 years (SD±14), 965 (49%) were female, and 1555 (78%) were non-Hispanic white. eGFR discrepancy occurred in 579 patients (29%). Patients with eGFR discrepancy were more likely to experience medication-related adverse events compared to those without eGFR discrepancy: vancomycin levels >30μg/mL (24% vs. 10%, p=0.004), trimethoprim- sulfamethoxazole-related hyperkalemia (24% vs. 12%, p=0.013), baclofen-induced neurotoxicity (25% vs. 0%, p=0.13), and supratherapeutic digoxin levels (38% vs. 0%, p=0.03). The adjusted OR for vancomycin levels >30μg/mL was 2.30 (95% CI 1.05 - 5.51, p = 0.047). Conclusion Among patients with cancer with simultaneous assessment of eGFRCYS and eGFRCRE, medication-related adverse events occur more commonly in those with eGFR discrepancy. These findings underscore the importance of accurate assessment of kidney function and appropriate dosing of renally-cleared medications in patients with cancer.
Collapse
Affiliation(s)
- Paul E Hanna
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Qiyu Wang
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Ian Strohbehn
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Daiana Moreno
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Destiny Harden
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Tianqi Ouyang
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Nurit Katz-Agranov
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Harish Seethapathy
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Kerry L Reynolds
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Shruti Gupta
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
- Adult Survivorship Program, Dana-Farber Cancer Institute, Boston, MA
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Meghan E Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
32
|
Iversen E, Bengaard AK, Andersen A, Kallemose T, Damgaard M, Hornum M, Feldt-Rasmussen B, Andersen O, Houlind MB. Performance of the Cockcroft-Gault, Modification of Diet in Renal Disease, and new Chronic Kidney Disease Epidemiology Collaboration equations without race in older acute medical patients. Kidney Int 2022; 101:1087-1088. [DOI: 10.1016/j.kint.2022.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/24/2022]
|
33
|
The authors reply:. Kidney Int 2022; 101:1088-1089. [DOI: 10.1016/j.kint.2022.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022]
|