1
|
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
|
2
|
Ofori EK, Nketiah-Dwomo I, Tagoe EA, Amponsah SK, Adams I, Nyarko ENY, Amanquah SD. Comparative Determination of Glomerular Filtration Rate Estimation Formulae in Type 2 Diabetic Patients: An Observational Study. BIOMED RESEARCH INTERNATIONAL 2024; 2024:9532236. [PMID: 38903148 PMCID: PMC11189678 DOI: 10.1155/2024/9532236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/02/2024] [Accepted: 05/02/2024] [Indexed: 06/22/2024]
Abstract
Assessing glomerular filtration rate (GFR) involves collecting timed urine samples for 24 hours, requiring significant time and resources in the clinical setting. Using predictive GFR formulae to assess renal function may be a better alternative. Our goal was to determine which predictive GFR formula had the highest level of concordance with the GFR that has been measured in a resource-poor setting. This is an observational study. We selected fifty (50) individuals diagnosed with type 2 diabetes (T2DM) in Kumasi, Ghana. The sociodemographic and clinical characteristics were obtained using a structured questionnaire. Urine was obtained from each subject over 24 hours. The levels of glucose (FBG) and creatinine in patients' blood, as well as the levels of creatinine in their urine, were measured after the patients had fasted overnight. Participants had a mean age of 57.4 ± 10.7 (years), BMI of 27.8 ± 4.1 (kg/m2), FBG of 9.0 ± 3.1 (mmol/L), and creatinine concentrations of 95.6 ± 29.1 (μmol/L). A Krouwer plot was used to compare the measured GFR with three formulae: Chronic Kidney Disease Epidemiology (CKD-EPI), Modification of Diet in Renal Disease (MDRD), and Cockroft-Gault (CG) for GFR prediction. Among the 3 estimates, CG showed nonsignificance (p > 0.05) with the measured GFR. The primary finding was that the GFR calculated using the CG formula was not different from the GFR measured, suggesting that CG is the most appropriate alternative GFR estimate among a cross-section of T2DM patients in Ghana.
Collapse
Affiliation(s)
| | | | | | | | - Ismaila Adams
- Department of Medical PharmacologyU.G.M.S.University of Ghana, Accra, Ghana
| | | | | |
Collapse
|
3
|
Wicha SG, Wansing EMA, Dadkhah A, Ayuk FA, Kröger NM, Langebrake C. Chimeric antigen receptor T-cell therapy and fludarabine: precision dosing imperatives. Blood Adv 2024; 8:797-798. [PMID: 38191740 PMCID: PMC10847728 DOI: 10.1182/bloodadvances.2023012068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/15/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024] Open
Affiliation(s)
- Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Eva M. A. Wansing
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
- University Medical Center Hamburg-Eppendorf, Hospital Pharmacy, Hamburg, Germany
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Adrin Dadkhah
- University Medical Center Hamburg-Eppendorf, Hospital Pharmacy, Hamburg, Germany
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francis A. Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicolaus M. Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Langebrake
- University Medical Center Hamburg-Eppendorf, Hospital Pharmacy, Hamburg, Germany
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Praiss AM, Miller A, Smith J, Lichtman SM, Bookman M, Aghajanian C, Sabbatini P, Backes F, Cohn DE, Argenta P, Friedlander M, Goodheart MJ, Mutch DG, Gershenson DM, Tewari KS, Wenham RM, Wahner Hendrickson AE, Lee RB, Gray H, Secord AA, Van Le L, O'Cearbhaill RE. Carboplatin dosing in the treatment of ovarian cancer: An NRG oncology group study. Gynecol Oncol 2023; 174:213-223. [PMID: 37229879 PMCID: PMC10330633 DOI: 10.1016/j.ygyno.2023.05.013] [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: 02/24/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE To determine the effects of using National Comprehensive Cancer Network (NCCN) guidelines to estimate renal function on carboplatin dosing and explore adverse effects associated with a more accurate estimation of lower creatinine clearance (CrCl). METHODS Retrospective data were obtained for 3830 of 4312 patients treated on GOG182 (NCT00011986)-a phase III trial of platinum-based chemotherapy for advanced-stage ovarian cancer. Carboplatin dose per patient on GOG182 was determined using the Jelliffe formula. We recalculated CrCl to determine dosing using Modification of Diet in Renal Disease (MDRD) and Cockcroft-Gault (with/without NCCN recommended modifications) formulas. Associations between baseline CrCl and toxicity were described using the area under the receiver operating characteristic curve (AUC). Sensitivity and positive predictive values described the model's ability to discriminate between subjects with/without the adverse event. RESULTS AUC statistics (range, 0.52-0.64) showed log(CrClJelliffe) was not a good predictor of grade ≥3 adverse events (anemia, thrombocytopenia, febrile neutropenia, auditory, renal, metabolic, neurologic). Of 3830 patients, 628 (16%) had CrCl <60 mL/min. Positive predictive values for adverse events ranged from 1.8%-15%. Using the Cockcroft-Gault, Cockcroft-Gault with NCCN modifications, and MDRD (instead of Jelliffe) formulas to estimate renal function resulted in a >10% decrease in carboplatin dosing in 16%, 32%, and 5.2% of patients, respectively, and a >10% increase in carboplatin dosing in 41%, 9.6% and 12% of patients, respectively. CONCLUSION The formula used to estimate CrCl affects carboplatin dosing. Estimated CrCl <60 mL/min (by Jelliffe) did not accurately predict adverse events. Efforts continue to better predict renal function. Endorsing National Cancer Institute initiatives to broaden study eligibility, our data do not support a minimum threshold CrCl <60 mL/min as an exclusion criterion from clinical trials.
Collapse
Affiliation(s)
- Aaron M Praiss
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| | - Austin Miller
- NRG Oncology Statistics and Data Center, Roswell Park Cancer Institute, Buffalo, NY, United States of America.
| | - Judith Smith
- McGovern Medical School, The University of Texas Health Science Center, Houston, TX, United States of America.
| | - Stuart M Lichtman
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America.
| | - Michael Bookman
- Department of Medical Oncology, Kaiser-Permanente Northern California, San Francisco, CA, United States of America.
| | - Carol Aghajanian
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America.
| | - Paul Sabbatini
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America.
| | - Floor Backes
- Department of Oncology, James Cancer Center, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States of America.
| | - David E Cohn
- Department of Oncology, James Cancer Center, Ohio State University Comprehensive Cancer Center, Columbus, OH, United States of America.
| | - Peter Argenta
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, MN, United States of America.
| | - Michael Friedlander
- Department of Medical Oncology, Prince of Wales Hospital and Prince of Wales Clinical School, UNSW, Sydney, New South Wales, Australia.
| | - Michael J Goodheart
- Gynecologic Oncology, University of Iowa Hospitals, Iowa City, IA, United States of America.
| | - David G Mutch
- Gynecologic Oncology, Washington University, St. Louis, MO, United States of America.
| | - David M Gershenson
- Gynecologic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States of America.
| | - Krishnansu S Tewari
- Gynecologic Oncology, University of California, Irvine Medical Center, Orange, CA, USA.
| | - Robert M Wenham
- Gynecologic Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, United States of America.
| | | | - Roger B Lee
- Gynecologic Oncology, Tacoma General Hospital, Tacoma, WA, United States of America
| | - Heidi Gray
- Gynecologic Oncology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America.
| | - Angeles Alvarez Secord
- Gynecologic Oncology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, United States of America.
| | - Linda Van Le
- Gynecologic Oncology, University of North Carolina, United States of America.
| | - Roisin E O'Cearbhaill
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America.
| |
Collapse
|
5
|
Telles JP, Diegues MS, Migotto KC, de Souza Borges O, Reghini R, Gavazza BV, Pinto L, Caruso P, E Silva ILF, Schmidt S, de Lima Moreira F. Failure to predict amikacin elimination in critically ill patients with cancer based on the estimated glomerular filtration rate: applying PBPK approach in a therapeutic drug monitoring study. Eur J Clin Pharmacol 2023:10.1007/s00228-023-03516-1. [PMID: 37256410 DOI: 10.1007/s00228-023-03516-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/22/2023] [Indexed: 06/01/2023]
Abstract
PURPOSE The aim of this work was to integrate the Therapeutic Drug Monitoring (TDM) with the model-informed precision dosing (MIPD) approach, using Physiologically-based Pharmacokinetic/Pharmacodynamic (PBPK/PD) modelling and simulation, to explore the relationship between amikacin exposure and estimated glomerular filtration rate (GFR) in critically ill patients with cancer. METHODS In the TDM study, samples from 51 critically-ill patients with cancer treated with amikacin were analysed. Patients were stratified according to renal function based on GFR status. A full-body PBPK model with 12 organs model was developed using Simcyp V. 21, including steady-state volume of distribution of 0.21 L/kg and renal clearance of 6.9 L/h in healthy adults. PK parameters evaluated were within the 2-fold error range. RESULTS During the validation step, predicted vs observed amikacin clearance values after single infusion dose in patients with normal renal function, mild and moderate renal impairment were 7.6 vs 8.1 L/h (7.5 mg/kg dose); 3.8 vs 4.5 L/h (1500 mg dose) and 2.2 vs 3.1 L/h (25 mg/kg dose), respectively. However, predicted vs observed amikacin clearance after a single dose infusion of 1400 mg in critically-ill patients with cancer were 1.46 vs 1.63 (P = 0.6406) L/h (severe), 2.83 vs 1.08 (P < 0.05) L/h (moderate), 4.23 vs 2.49 (P = 0.0625) L/h (mild) and 7.41 vs 3.36 (P < 0.05) L/h (normal renal function). CONCLUSION This study demonstrated that estimated GFR did not predict amikacin elimination in critically-ill patients with cancer. Further studies are necessary to find amikacin PK covariates to optimize the pharmacotherapy in this population. Therefore, TDM of amikacin is imperative in cancer patients.
Collapse
Affiliation(s)
- João Paulo Telles
- Department of Infectious Diseases, AC Camargo Cancer Center, Professor Antonio Prudente Street, N. 211, São Paulo-SP, 01509-001, Brazil.
| | | | | | | | - Rodrigo Reghini
- Department of Infectious Diseases, AC Camargo Cancer Center, Professor Antonio Prudente Street, N. 211, São Paulo-SP, 01509-001, Brazil
| | - Brenda Vianna Gavazza
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro-RJ, Brazil
| | - Leonardo Pinto
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto-MG, Brazil
| | - Pedro Caruso
- Department of Intensive Care Unit, AC Camargo Cancer Center, São Paulo-SP, Brazil
| | - Ivan Leonardo França E Silva
- Department of Infectious Diseases, AC Camargo Cancer Center, Professor Antonio Prudente Street, N. 211, São Paulo-SP, 01509-001, Brazil
| | - Stephan Schmidt
- Department of Pharmaceutics Lake Nona, University of Florida, Orlando-FL, USA
| | | |
Collapse
|
6
|
Drug dosing in cancer patients with decreased kidney function: A practical approach. Cancer Treat Rev 2020; 93:102139. [PMID: 33370636 DOI: 10.1016/j.ctrv.2020.102139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/06/2020] [Indexed: 10/22/2022]
Abstract
Correct drug dosing of anticancer agents is essential to obtain optimal outcomes. Overdosing will result in increased toxicity, treatment interruption and possible cessation of anticancer treatment. Underdosing may result in suboptimal anti-cancer effects and may increase the risk of cancer-related mortality. As it is practical nor feasible to perform therapeutic drug monitoring for all anti-cancer drugs, kidney function is used to guide drug dosing for those drugs whose primary mode of excretion is through the kidney. However, it is not well-established what method should be utilized to measure or estimate kidney function and the choice of method does influence treatment decisions regarding eligibility for anti-cancer drugs and their dose. In this review, we will provide an overview regarding the importance of drug dosing, the preferred method to determine kidney function and a practical approach to drug dosing of anticancer drugs.
Collapse
|
7
|
Tsang C, Akbari A, Frechette D, Brown PA. Accurate determination of glomerular filtration rate in adults for carboplatin dosing: Moving beyond Cockcroft and Gault. J Oncol Pharm Pract 2020; 27:368-375. [PMID: 33297846 DOI: 10.1177/1078155220978446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Carboplatin is a cytotoxic chemotherapy drug developed in the 1980s which is still widely used today across various tumour types. Despite its common application, there remains a significant controversy and practice variation on its unique method of dosing by area under the curve (AUC). One potential reason for this variability stems from the reliance of using an estimated glomerular filtration rate (eGFR) as an extrapolation of the measured GFR (mGFR) which the commonly used Calvert equation was originally validated for. This review takes a novel and collaborative nephro-oncology approach to highlight the historical evolution of carboplatin dosing, methods for estimating GFR and its relative performance in the application of carboplatin dosing for adult patients. DATA SOURCES We reviewed all pertinent publications comparing carboplatin AUC-based dosing in adult patients based on the various methods of GFR measurements or estimations in order to provide a comprehensive description of each method's advantages and risks. DATA SUMMARY AND CONCLUSIONS The Cockcroft-Gault equation has been widely studied but newer eGFR equations, such as the CKD-Epidemiology Collaboration (CKD-EPI) or Janowitz-Williams equation have outperformed the Cockcroft-Gault in recent studies.
Collapse
Affiliation(s)
- Corey Tsang
- Department of Pharmacy, The Ottawa Hospital, Ottawa, ON, Canada
| | - Ayub Akbari
- Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, ON, Canada.,Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Dominque Frechette
- Department of Medicine, Centre Integré des Services Sociaux de l'Outaouais, Gatineau, Québec, Canada
| | - Pierre Antoine Brown
- Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, ON, Canada.,Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
8
|
Carboplatin dose capping affects pCR rate in HER2-positive breast cancer patients treated with neoadjuvant Docetaxel, Carboplatin, Trastuzumab, Pertuzumab (TCHP). Breast Cancer Res Treat 2020; 184:481-489. [PMID: 32860550 PMCID: PMC7599187 DOI: 10.1007/s10549-020-05868-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/06/2020] [Indexed: 10/26/2022]
Abstract
PURPOSE Estimated glomerular filtration rate (eGFR) is commonly used to calculate carboplatin doses and capping the eGFR may be used to reduce the risk of excessive dosing and toxicity. We sought to retrospectively examine the impact of our carboplatin guidelines on pathological complete response rates (pCR) and toxicity in women with HER2+ breast cancer receiving neoadjuvant docetaxel, carboplatin, trastuzumab and pertuzumab (TCHP). METHODS The delivered area under the curve (dAUC) was calculated [(actual carboplatin dose at cycle 1 ÷ dose calculated with uncapped/unbanded eGFR) × 6] and dichotomized at the median value. The impact of this and other clinical factors on pCR rate, dose intensity (DI) and toxicity was assessed. RESULTS 124 eligible patients were identified of whom 63.7% (79/124) achieved pCR. The median dAUC at cycle 1 was 5.75 mg × ml/min. Those with lower dAUC were more frequently younger and overweight/obese. Patients with lower dAUC had significantly inferior pCR rates of 54.8% (34/62) vs 72.6% (45/62), respectively (p = 0.040). Similar results were seen in the ER+ subgroup; 45.2% (19/42) vs 68.3% (28/41), p = 0.037%, whereas no significant difference was seen among ER- patients; 75.0% (15/20) vs 81.0% (17/21), p = 0.72. DI and toxicity were comparable between the two dAUC groups. CONCLUSIONS The overall pCR rate was high in patients with HER2+ breast cancer receiving the TCHP regimen; however, carboplatin dose capping resulted in inferior pCR rates, particularly in the ER+ subgroup. To ensure optimal dosing, isotopic measurement of renal function is warranted in patients who would otherwise have their eGFR and dose capped.
Collapse
|
9
|
Carboplatin dosing in the era of IDMS-creatinine; the Cockroft-Gault formula no longer provides a sufficiently accurate estimate of glomerular filtration rate for routine use in clinical care. Gynecol Oncol 2020; 157:793-798. [DOI: 10.1016/j.ygyno.2020.03.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/15/2020] [Indexed: 11/20/2022]
|
10
|
A physiologically based pharmacokinetic - pharmacodynamic modelling approach to predict incidence of neutropenia as a result of drug-drug interactions of paclitaxel in cancer patients. Eur J Pharm Sci 2020; 150:105355. [PMID: 32438273 DOI: 10.1016/j.ejps.2020.105355] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/21/2020] [Accepted: 04/17/2020] [Indexed: 12/24/2022]
Abstract
Paclitaxel is the backbone of standard chemotherapeutic regimens used in a number of malignancies and is frequently given with concomitant medications. Newly developed oncolytic agents, including tyrosine kinase inhibitors are often shown to be CYP3A4 and P-gp inhibitors. The aim of this study was to develop a PBPK model for intravenously administered paclitaxel in order to predict the incidence of neutropenia and to estimate the DDI potential as a victim drug. The dose-dependent effects on paclitaxel plasma protein binding, volume of distribution and drug clearance were considered for dose levels of 80 mg/m2, 135 mg/m2 and 175 mg/m2. A pharmacodynamics model that incorporate the impact of paclitaxel on the neutrophil was developed. The relative metabolic clearance via CYP3A4 and CYP2C8, the renal clearance as well as P-gp mediated biliary clearance were incorporated in the model in order to assess the neutropenia in the presence of DDI. The developed PBPK-PD model was able to recover the drop in neutrophils observed after the administration of 175mg/m2 of paclitaxel over a 3-h duration. The mean nadir observed was 1.9 × 109 neutrophils/L and was attained after 10 days of treatment, and a fraction of 47% of the population was predicted to have at some point a neutropenia including 12% with severe neutropenia. In the case of concomitant administration of ketoconazole, 39% of the population was predicted to suffer from severe neutropenia. In summary, PBPK-PD modeling allows a priori prediction of DDIs and safety events involving complex combination therapies which are often utilized in an oncology setting.
Collapse
|
11
|
Sprangers B, Abudayyeh A, Latcha S, Perazella MA, Jhaveri KD. How to determine kidney function in cancer patients? Eur J Cancer 2020; 132:141-149. [PMID: 32361629 DOI: 10.1016/j.ejca.2020.03.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/29/2020] [Indexed: 12/18/2022]
Abstract
A precise and efficient method for estimating kidney function in cancer patients is important to determine their eligibility for clinical trials and surgery and to allow for appropriate dose adjustment of anti-cancer drugs, especially toxic drugs with a narrow therapeutic index. Since direct measurement of glomerular filtration rate (GFR) is cumbersome, several formulae have been developed to estimate kidney function. Most of these are based on serum creatinine concentration. Though the CKD-EPI formula is recognised as being the most accurate, there is an ongoing debate on which is the optimal formula for cancer patients. In this review, we provide an overview of different GFR estimating equations for kidney function and the advantages and disadvantages of each method and compare their performance in cancer patients. We discuss the importance of body surface area-indexing and propose a framework for evaluating kidney function in cancer patients.
Collapse
Affiliation(s)
- Ben Sprangers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Belgium; Division of Nephrology, University Hospitals Leuven, Both in Leuven, Belgium.
| | - Ala Abudayyeh
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheron Latcha
- Renal Division, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark A Perazella
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, USA; Veterans Affairs Medical Center, West Haven, CT, USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| |
Collapse
|
12
|
McMahon BA, Rosner MH. GFR Measurement and Chemotherapy Dosing in Patients with Kidney Disease and Cancer. KIDNEY360 2020; 1:141-150. [PMID: 35372903 PMCID: PMC8809099 DOI: 10.34067/kid.0000952019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chemotherapeutic agents require precise dosing to ensure optimal efficacy and minimize complications. For those agents that are removed from the body by the kidney, accurate knowledge of GFR is critical. In addition, GFR needs to be determined rapidly, easily, and, if possible, with little additional cost. The ability to easily measure GFR also allows for rapid detection of nephrotoxicity. Current methodologies include direct clearance measurement of an indicator substance or estimation of creatinine clearance or GFR through regression equations that use a serum marker, such as creatinine or cystatin C. These methodologies all have shortfalls and limitations, some of which are specific to the patient with cancer. Newer methodologies that directly measure GFR are in clinical trials and offer the ability to rapidly and noninvasively provide accurate estimates of drug clearance as well as detection of nephrotoxicity. These methods offer the opportunity to refine drug dosing and improve outcomes.
Collapse
Affiliation(s)
- Blaithin A. McMahon
- Division of Nephrology, Medical University of South Carolina, Charleston, South Carolina; and
| | - Mitchell H. Rosner
- Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
13
|
Sprangers B, Jhaveri KD, Perazella MA. Improving Cancer Care for Patients With Chronic Kidney Disease. J Clin Oncol 2020; 38:188-192. [DOI: 10.1200/jco.19.02138] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ben Sprangers
- Department of Microbiology, Immunology, and Transplantation, Laboratory of Molecular Immunology, Rega Institute, Katholieke Universiteit Leuven, and Division of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Kenar D. Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY
| | - Mark A. Perazella
- Section of Nephrology, Yale University School of Medicine, New Haven, CT
- Veterans Affairs Medical Center, West Haven, CT
| |
Collapse
|
14
|
Summary of the International Conference on Onco-Nephrology: an emerging field in medicine. Kidney Int 2019; 96:555-567. [DOI: 10.1016/j.kint.2019.04.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 01/10/2023]
|
15
|
Garner AE, Barnfield MC, Waller ML, Hall GD, Bosomworth MP. Comparing glomerular filtration rate equations and the impact of different creatinine assays on the assessment of renal function in cancer patients. Ann Clin Biochem 2019; 56:266-274. [PMID: 30791693 DOI: 10.1177/0004563218822667] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Equations to estimate glomerular filtration rate based on serum creatinine are commonly used in cancer patients to assess renal function. However, there is uncertainty regarding which equation is most appropriate for this population and the impact of different creatinine assays. METHODS Measured isotopic glomerular filtration rate results from 120 oncology patients were used to evaluate and compare all four versions of the Wright equation, Cockcroft and Gault, Modification of Diet in Renal Disease (MDRD), Chronic Kidney Disease Epidemiology Collaboration and the Janowitz and Williams formula; using eight different creatinine assays (five Jaffe, three enzymatic). RESULTS The enzymatic version of the Wright equation without creatine kinase performed better than the other versions for all eight creatinine assays. However, MDRD and Janowitz and Williams gave the best overall performance in this patient population. Performance was highly dependent on the creatinine assay used, for example, the percentage of results within 30% of the isotopic glomerular filtration rate (P30) ranged from 90.8% to 60.8% for MDRD. CONCLUSION The performance of any equation to estimate glomerular filtration rate is highly dependent on the creatinine assay used. Oncology units should assess the performance of glomerular filtration rate equations using their laboratory creatinine assay to determine whether they can be used safely and effectively in cancer patients.
Collapse
Affiliation(s)
- Ashley E Garner
- 1 Department of Blood Sciences, Leeds General Infirmary, Old Medical School, Leeds, UK
| | - Mark C Barnfield
- 2 The Department of Medical Physics & Engineering, St. James's University Hospital, Leeds, UK
| | - Michael L Waller
- 2 The Department of Medical Physics & Engineering, St. James's University Hospital, Leeds, UK
| | - Geoff D Hall
- 3 Cancer Research UK Clinical Cancer Centre in Leeds, St. James's University Hospital, Leeds, UK
| | - Mike P Bosomworth
- 1 Department of Blood Sciences, Leeds General Infirmary, Old Medical School, Leeds, UK
| |
Collapse
|
16
|
Morrow A, Garland C, Yang F, De Luna M, Herrington JD. Analysis of carboplatin dosing in patients with a glomerular filtration rate greater than 125 mL/min: To cap or not to cap? A retrospective analysis and review. J Oncol Pharm Pract 2018; 25:1651-1657. [DOI: 10.1177/1078155218805136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The use of the Calvert formula to calculate carboplatin doses allows clinicians to achieve the appropriate carboplatin area under the concentration (AUC) curve. Thrombocytopenia is the dose limiting toxicity of carboplatin and optimizing AUC minimizes the risk of thrombocytopenia. Carboplatin clearance directly correlates with glomerular filtration rate (GFR) and, therefore, an accurate estimation of the renal function is needed. The Calvert formula was designed using the GFR measured by 51Cr-EDTA; however, many clinicians substitute estimated creatinine clearance (CrCl) as calculated by the Cockcroft–Gault (C–G) equation. The potential for overestimating AUC occurs when clinicians substitute actual weight in obese patients or use a low serum creatinine when calculating C–G estimated CrCl. In 2010, the National Cancer Institute recommended the GFR value within the Calvert formula should not exceed 125 mL/min, thereby capping the carboplatin dose. However, there are studies demonstrating that certain patients’ actual GFR values do exceed 125 mL/min. Therefore, capping the carboplatin dose in these patients may lead to underestimating the carboplatin AUC. A single-center, retrospective study was performed to evaluate the change in platelet count pre- and post-carboplatin exposure in patients with C–G estimated CrCl greater than 125 mL/min receiving capped versus uncapped carboplatin doses. A review of carboplatin dosing strategies is also presented. This study indicated there was a larger mean difference in pre- and post-platelet count in patients receiving uncapped carboplatin compared to patients receiving capped carboplatin with no differences in toxicities. Dose capping this patient population will likely lead to a lower AUC rather than the intended AUC target, which could ultimately lead to substandard outcomes.
Collapse
Affiliation(s)
- Amy Morrow
- Baylor Scott & White Medical Center, Temple, TX, USA
- The University of Texas at Austin College of Pharmacy, Austin, TX, USA
| | - Campbell Garland
- Baylor Scott & White Medical Center, Temple, TX, USA
- Texas A&M College of Medicine, Temple, TX, USA
| | - Fei Yang
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Mike De Luna
- Baylor Scott & White Medical Center, Temple, TX, USA
| | - Jon D Herrington
- Baylor Scott & White Medical Center, Temple, TX, USA
- The University of Texas at Austin College of Pharmacy, Austin, TX, USA
- Texas A&M College of Medicine, Temple, TX, USA
| |
Collapse
|
17
|
Cuesta-Grueso C, Burgos-San José A, Cajaraville-Ordoñana G, Poquet-Jornet JE, Mangues-Bafalluy I, Díaz-Carrasco MS. Variability of carboplatin dose calculation methods in Spain. J Oncol Pharm Pract 2018; 25:1551-1557. [PMID: 30176785 DOI: 10.1177/1078155218796912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To describe and analyze the variability in carboplatin dosing strategies in Spanish hospitals. METHODS We designed a questionnaire consisting of 19 multiple-choice items structured in two sections (hospital characteristics and carboplatin dosing data). The questionnaire was sent by e-mail to all the oncology pharmacists included in the register of the Spanish Oncology Pharmacy Group (GEDEFO), and we analyzed the completed questionnaires. RESULTS Response rate was 33.5% from a total of 185 pharmacy services invited to take part in the survey. All hospitals used the Calvert formula to calculate carboplatin dose with glomerular filtration rate estimated by a formula, most commonly the Cockcroft-Gault equation (80.7%). Carboplatin doses were capped in most hospitals (91.9%): 54.8% capped creatinine clearance at 125 mL/min, 11.3% capped serum creatinine, and 19.3% capped both creatinine clearance and serum creatinine. Serum creatinine cut-off values ranged from 0.36 mg/dL to 1 mg/dL. The most commonly used body weight was actual body weight for underweight, normal weight, and overweight patients. The use of adjusted ideal body weight increased in obese and especially in morbidly obese patients. CONCLUSION The results from this survey show the variability that exists in carboplatin dose calculation methods among Spanish hospitals and the need to continue investigating to find the optimum dose calculation method and unify criteria to avoid differences between sites that can affect effectiveness and toxicity of carboplatin-containing treatments.
Collapse
|
18
|
Graciano Vera N, Pino Villarreal L, Ureña Vargas J. Carboplatin Dosing Accuracy by Estimation of Glomerular Filtration versus Creatinuria in Cancer Patients. Chemotherapy 2018; 63:137-142. [DOI: 10.1159/000488538] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/17/2018] [Indexed: 11/19/2022]
Abstract
Background: The glomerular filtration rate (GFR) is essential for calculating the dose and the monitoring of carboplatin. Although GFR measurement (mGFR) by external markers is ideal, in most cases these are not employed; the most used method is GFR estimation (eGFR) by formulae, hence the need to identify the formula with the best performance. Methods: Patients admitted between 2011 and 2017 and diagnosed with ovarian, endometrial, lung, esophageal, or testicular cancer were assessed retrospectively. The accuracy of the carboplatin dose calculated by creatinine concordance and by the Cockroft-Gault (CG), CKD-EPI, MDRD, Wright, and Jelliffe formulae was assessed using the intraclass correlation coefficient. Results: Fifty-six medical histories were analyzed. The best accuracy was observed between the Wright formula (i.e., 0.71) and the dose calculated based on the 24-h creatinine clearance. Stratification by CKD was made in depurations < 60 mL/min, where the Jelliffe value was excellent (i.e., 0.75). In depurations ≥60 mL/min, CKD-EPI was the best formula, with an accuracy of 0.65. CG was the formula with the worst performance in calculating the dose and glomerular filtration, losing its usefulness with very low filtrations. Conclusions: GFR formulae and calculation of the carboplatin dose have good accuracy with the GFR obtained based on the 24-h creatinine clearance, with the Wright formula being the one with best performance and CG the one with worst performance.
Collapse
|
19
|
Fehr M, Maranta AF, Reichegger H, Gillessen S, Cathomas R. Carboplatin dose based on actual renal function: no excess of acute haematotoxicity in adjuvant treatment in seminoma stage I. ESMO Open 2018. [PMID: 29531843 PMCID: PMC5844370 DOI: 10.1136/esmoopen-2018-000320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Introduction The practice of carboplatin dosing is not concordant among different centres and oncologists. Some clinical guidelines recommend capping of the carboplatin dose at, for example, creatinine-clearance (Crea-Cl) of 125 mL/min because of concerns of excessive toxicity. Clinical data to support such recommendations are lacking, especially in patients with seminoma. Methods This is a retrospective analysis of acute haematotoxicity of patients with stage I seminoma treated with adjuvant carboplatin area under the curve (AUC) 7 in routine practice in two Swiss centres in 2005–2015, and a comparison of incidence and grade (according to Common Terminology Criteria for Adverse Events v4.0) of haematological adverse events (hAEs) in patients with Crea-Cl <125 mL/min vs >125 mL/min without dose capping. Results 74 patients with 229 documented measurements were included (median 3/patient). A total of 151 hAEs occurred. Platelet nadir occurred earlier than median white cell/neutrophil count (median day 15 vs day 22; P<0.0001). The majority of hAEs were mild, with more than 80% being of grade 1. Only two (2.7%) clinically relevant hAEs necessitating subsequent interventions occurred (one patient received platelet transfusion, one patient with febrile neutropaenia). Haematological toxicities were not statistically different in patients dosed with Crea-Cl >125 mL/min versus those with Crea-Cl <125 mL/min. No hAEs other than grade 1 occurred before day 10 and after day 24. Conclusions Toxicity after single-dose carboplatin AUC 7 is generally mild. No excess of toxicity occurs in patients with high Crea-Cl above 125 mL/min, and therefore dose capping is not routinely necessary. In addition, this study provides a rationale for efficient use of healthcare services without compromising patients’ safety.
Collapse
Affiliation(s)
- Martin Fehr
- Clinic for Medical Oncology and Haematology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | | | - Hermann Reichegger
- Clinic for Medical Oncology and Haematology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Silke Gillessen
- Clinic for Medical Oncology and Haematology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Richard Cathomas
- Department of Oncology/Haematology, Cantonal Hospital Grisons, Chur, Switzerland
| |
Collapse
|
20
|
Beumer JH, Inker LA, Levey AS. Improving Carboplatin Dosing Based on Estimated GFR. Am J Kidney Dis 2018; 71:163-165. [PMID: 29217306 PMCID: PMC6317965 DOI: 10.1053/j.ajkd.2017.10.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/09/2017] [Indexed: 01/30/2023]
Affiliation(s)
- Jan H Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA; Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA; Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Lesley A Inker
- Division of Nephrology, Tufts Medical Center, Boston, MA
| | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, MA.
| |
Collapse
|
21
|
Horie S, Oya M, Nangaku M, Yasuda Y, Komatsu Y, Yanagita M, Kitagawa Y, Kuwano H, Nishiyama H, Ishioka C, Takaishi H, Shimodaira H, Mogi A, Ando Y, Matsumoto K, Kadowaki D, Muto S. Guidelines for treatment of renal injury during cancer chemotherapy 2016. Clin Exp Nephrol 2018; 22:210-244. [PMID: 28856465 PMCID: PMC5805816 DOI: 10.1007/s10157-017-1448-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Department of Advanced Informatics for Genetic Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yoshinari Yasuda
- Department of CKD Initiatives/Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Komatsu
- Division of Nephrology, Department of Medicine, St. Luke's International Hospital, Tokyo, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Kuwano
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Chikashi Ishioka
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University, Miyagi, Japan
| | - Hiromasa Takaishi
- Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Hideki Shimodaira
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University, Miyagi, Japan
| | - Akira Mogi
- Department of General Surgical Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Aichi, Japan
| | - Koji Matsumoto
- Division of Medical Oncology, Hyogo Cancer Center, Hyogo, Japan
| | - Daisuke Kadowaki
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Muto
- Department of Urology, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Department of Advanced Informatics for Genetic Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Torres da Costa e Silva V, Costalonga EC, Coelho FO, Caires RA, Burdmann EA. Assessment of Kidney Function in Patients With Cancer. Adv Chronic Kidney Dis 2018; 25:49-56. [PMID: 29499887 DOI: 10.1053/j.ackd.2017.10.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 12/14/2022]
Abstract
Cancer patients are living longer. The sequelae of cancer treatment and the role of comorbid conditions present before the diagnosis, such as CKD, have been increasingly recognized. The interface between CKD and cancer is multifaceted. CKD is frequently observed in patients with cancer, and cancer treatment contributes to CKD development and progression. In addition, CKD has been recognized as an important risk factor for cancer development and reduced specific cancer survival. In this context, an accurate evaluation of the glomerular filtration rate (GFR) during oncologic treatment is pivotal and is used to define surgery strategies, program prophylactic management of contrasted examinations, make decisions on cisplatin eligibility, and adjust drug prescriptions, particularly chemotherapy agents. Although the most commonly used equations to estimate GFR based on serum creatinine levels in clinical practice (Cockcroft-Gault, Modification of Diet in Renal Disease Study, and CKD Epidemiology Collaboration equations) have not been validated in patients with cancer in large prospective studies, there is increasingly evidence supporting the use of CKD Epidemiology Collaboration equation to assess the GFR in patients with cancer, including for the use of chemotherapy prescriptions. Many patients with cancer may have changes in nutrition status and clearance measurements such as exogenous filtration markers might be extremely useful when clinical decisions differ depending on the GFR level. Future perspectives include the advent of new serum GFR biomarkers such as cystatin C, beta-trace protein, and beta-2 microglobulin as well as the GFR assessment by measuring total kidney parenchymal volume through image examinations.
Collapse
|
23
|
Delanaye P, Guerber F, Scheen A, Ellam T, Bouquegneau A, Guergour D, Mariat C, Pottel H. Discrepancies between the Cockcroft-Gault and Chronic Kidney Disease Epidemiology (CKD-EPI) Equations: Implications for Refining Drug Dosage Adjustment Strategies. Clin Pharmacokinet 2017; 56:193-205. [PMID: 27417226 DOI: 10.1007/s40262-016-0434-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The dosages of many medications require adjustment for renal function. There is debate regarding which equation, the Chronic Kidney Disease Epidemiology (CKD-EPI) equation vs. the Cockcroft-Gault (CG) equation, should be recommended to estimate glomerular filtration rate. METHODS We used a mathematical simulation to determine how patient characteristics influence discrepancies between equations and analyzed clinical data to demonstrate the frequency of such discrepancies in clinical practice. In the simulation, the modifiable variables were sex, age, serum creatinine, and weight. We considered estimated glomerular filtration rate results in mL/min, deindexed for body surface area, because absolute excretory function (rather than per 1.73 m2 body surface area) determines the rate of filtration of a drug at a given plasma concentration. An absolute and relative difference of maximum (±) 10 mL/min and 10 %, respectively, were considered concordant. Clinical data for patients aged over 60 years (n = 9091) were available from one hospital and 25 private laboratories. RESULTS In the simulation, differences between the two equations were found to be influenced by each variable but age and weight had the biggest effect. Clinical sample data demonstrated concordance between CKD-EPI and CG results in 4080 patients (45 %). The majority of discordant results reflected a CG result lower than the CKD-EPI equation. With aging, the CG result became progressively lower than the CKD-EPI result. When weight increased, the opposite occurred. DISCUSSION The choice of equation for excretory function adjustment of drug dosage will have different implications for patients of different ages and body habitus. CONCLUSIONS The optimum equation for drug dosage adjustment should be defined with consideration of individual patient characteristics.
Collapse
Affiliation(s)
- Pierre Delanaye
- Division of Nephrology, Dialysis and Transplantation, CHU Sart Tilman, University of Liège (ULg-CHU), 4000, Liège, Belgium.
| | | | - André Scheen
- Division of Clinical Pharmacology, Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
| | - Timothy Ellam
- Sheffield Kidney Institute, Northern General Hospital and Department of Infection, Immunity and Cardiovascular Science, University of Sheffield, Sheffield, UK
| | - Antoine Bouquegneau
- Division of Nephrology, Dialysis and Transplantation, CHU Sart Tilman, University of Liège (ULg-CHU), 4000, Liège, Belgium
| | - Dorra Guergour
- Biochemistry Laboratory, Grenoble University Hospital, Grenoble, France
| | - Christophe Mariat
- Division of Nephrology, Dialysis, Transplantation and Hypertension, CHU Hôpital Nord, University Jean Monnet, PRES Université de LYON, Saint-Etienne, France
| | - Hans Pottel
- Department of Public Health and Primary Care, KU, Leuven Kulak, Kortrijk, Belgium
| |
Collapse
|
24
|
Janowitz T, Williams EH, Marshall A, Ainsworth N, Thomas PB, Sammut SJ, Shepherd S, White J, Mark PB, Lynch AG, Jodrell DI, Tavaré S, Earl H. New Model for Estimating Glomerular Filtration Rate in Patients With Cancer. J Clin Oncol 2017; 35:2798-2805. [PMID: 28686534 PMCID: PMC5562175 DOI: 10.1200/jco.2017.72.7578] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The glomerular filtration rate (GFR) is essential for carboplatin chemotherapy dosing; however, the best method to estimate GFR in patients with cancer is unknown. We identify the most accurate and least biased method. Methods We obtained data on age, sex, height, weight, serum creatinine concentrations, and results for GFR from chromium-51 (51Cr) EDTA excretion measurements (51Cr-EDTA GFR) from white patients ≥ 18 years of age with histologically confirmed cancer diagnoses at the Cambridge University Hospital NHS Trust, United Kingdom. We developed a new multivariable linear model for GFR using statistical regression analysis. 51Cr-EDTA GFR was compared with the estimated GFR (eGFR) from seven published models and our new model, using the statistics root-mean-squared-error (RMSE) and median residual and on an internal and external validation data set. We performed a comparison of carboplatin dosing accuracy on the basis of an absolute percentage error > 20%. Results Between August 2006 and January 2013, data from 2,471 patients were obtained. The new model improved the eGFR accuracy (RMSE, 15.00 mL/min; 95% CI, 14.12 to 16.00 mL/min) compared with all published models. Body surface area (BSA)-adjusted chronic kidney disease epidemiology (CKD-EPI) was the most accurate published model for eGFR (RMSE, 16.30 mL/min; 95% CI, 15.34 to 17.38 mL/min) for the internal validation set. Importantly, the new model reduced the fraction of patients with a carboplatin dose absolute percentage error > 20% to 14.17% in contrast to 18.62% for the BSA-adjusted CKD-EPI and 25.51% for the Cockcroft-Gault formula. The results were externally validated. Conclusion In a large data set from patients with cancer, BSA-adjusted CKD-EPI is the most accurate published model to predict GFR. The new model improves this estimation and may present a new standard of care.
Collapse
Affiliation(s)
- Tobias Janowitz
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Edward H. Williams
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrea Marshall
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicola Ainsworth
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Peter B. Thomas
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Stephen J. Sammut
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Scott Shepherd
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jeff White
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Patrick B. Mark
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andy G. Lynch
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Duncan I. Jodrell
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Simon Tavaré
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Helena Earl
- Tobias Janowitz, Edward H. Williams, Stephen J. Sammut, Andy G. Lynch, Duncan I. Jodrell, Simon Tavaré, and Helena Earl, Cancer Research UK Cambridge Institute, Tobias Janowitz, Peter B. Thomas, and Duncan I. Jodrell, University of Cambridge, Addenbrooke’s Hospital, Cambridge; Andrea Marshall, University of Warwick, Coventry; Nicola Ainsworth, Queen Elizabeth Hospital, King’s Lynn; Scott Shepherd, Royal Marsden Hospital, London; Jeff White, NHS Greater Glasgow and Clyde; and Patrick B. Mark, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
25
|
Peterson LL, Hurria A, Feng T, Mohile SG, Owusu C, Klepin HD, Gross CP, Lichtman SM, Gajra A, Glezerman I, Katheria V, Zavala L, Smith DD, Sun CL, Tew WP. Association between renal function and chemotherapy-related toxicity in older adults with cancer. J Geriatr Oncol 2016; 8:96-101. [PMID: 27856262 DOI: 10.1016/j.jgo.2016.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 10/14/2016] [Accepted: 10/31/2016] [Indexed: 12/27/2022]
Abstract
PURPOSE To evaluate the association between renal function (RF) and chemotherapy-related toxicity (CRT) in older adults with cancer and to compare the effect of different RF formulas and body weight measurements on this association. METHODS This is a secondary analysis of data from a prospective multicenter study of patients ≥ age 65 who were starting a new chemotherapy regimen. RF was estimated with 4 formulas (modified Jelliffe [Jelliffe], Cockcroft-Gault [CG], Wright, and Modification of Diet in Renal Disease [MDRD]), using actual, ideal and adjusted body weights for 492 patients. The association between baseline RF and grade 3-5 CRT was evaluated by unconditional logistic regression. RESULTS As a continuous variable, decreased creatinine clearance (CrCl) calculated by CG with actual body weight was associated with increased odds of CRT (OR 1.12, P<0.01; 95% CI 1.04-1.20) indicating that on average for every 10mL/min decrease in CrCl the odds of CRT increased by 12%. Very low RF (in the lowest 10%) with all formulas (CG, Jelliffe, Wright and MDRD) was associated with increased odds for CRT. This association is independent of the type of chemotherapy received (those requiring dose adjustment for renal function vs not). Neither primary dose reduction nor chemotherapy duration was associated with CRT. Serum creatinine alone was not associated with increased odds of CRT (OR 0.67, P=0.15). CONCLUSIONS Decreased RF is associated with increased odds of CRT and should be considered when assessing risk of CRT in older adults with cancer. Serum creatinine alone is not adequate for risk assessment.
Collapse
Affiliation(s)
| | - Arti Hurria
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Tao Feng
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | | | - Cynthia Owusu
- Case Western Reserve University, Cleveland, OH, United States
| | - Heidi D Klepin
- Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Cary P Gross
- Yale Comprehensive Cancer Center, New Haven, CT, United States
| | | | - Ajeet Gajra
- State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY, United States
| | | | - Vani Katheria
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Laura Zavala
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - David D Smith
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Can-Lan Sun
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - William P Tew
- Memorial Sloan-Kettering Cancer Center, United States
| |
Collapse
|
26
|
Pharmacokinetics of Selected Anticancer Drugs in Elderly Cancer Patients: Focus on Breast Cancer. Cancers (Basel) 2016; 8:cancers8010006. [PMID: 26729170 PMCID: PMC4728453 DOI: 10.3390/cancers8010006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/30/2015] [Accepted: 12/29/2015] [Indexed: 01/07/2023] Open
Abstract
Background: Elderly patients receiving anticancer drugs may have an increased risk to develop treatment-related toxicities compared to their younger peers. However, a potential pharmacokinetic (PK) basis for this increased risk has not consistently been established yet. Therefore, the objective of this study was to systematically review the influence of age on the PK of anticancer agents frequently administered to elderly breast cancer patients. Methods: A literature search was performed using the PubMed electronic database, Summary of Product Characteristics (SmPC) and available drug approval reviews, as published by EMA and FDA. Publications that describe age-related PK profiles of selected anticancer drugs against breast cancer, excluding endocrine compounds, were selected and included. Results: This review presents an overview of the available data that describe the influence of increasing age on the PK of selected anticancer drugs used for the treatment of breast cancer. Conclusions: Selected published data revealed differences in the effect and magnitude of increasing age on the PK of several anticancer drugs. There may be clinically-relevant, age-related PK differences for anthracyclines and platina agents. In the majority of cases, age is not a good surrogate marker for anticancer drug PK, and the physiological state of the individual patient may better be approached by looking at organ function, Charlson Comorbidity Score or geriatric functional assessment.
Collapse
|
27
|
Front-line Treatment with Gemcitabine, Paclitaxel, and Doxorubicin for Patients With Unresectable or Metastatic Urothelial Cancer and Poor Renal Function: Final Results from a Phase II Study. Urology 2015; 89:83-9. [PMID: 26723185 DOI: 10.1016/j.urology.2015.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/01/2015] [Accepted: 12/03/2015] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To estimate the response rate of gemcitabine, paclitaxel, and doxorubicin in patients with advanced urothelial carcinoma, we conducted a phase II clinical trial. Patients with renal insufficiency cannot receive standard cisplatin-based chemotherapy for urothelial carcinoma, and carboplatin-based regimens have proved unsatisfactory. Secondary end points for this study included overall survival, safety of the regimen, and safety of same-day pegfilgrastim dosing. METHODS A two-stage design was chosen with target response rate of 40%. Key inclusion criteria were metastatic or unresectable urothelial carcinoma, no prior chemotherapy, glomerular filtration rate <60 mL/min, and no dialysis. Gemcitabine (900 mg/m(2)), paclitaxel (135 mg/m(2)), and doxorubicin (40 mg/m(2)) were administered on day 1 of each 14-day cycle. Pegfilgrastim was given with every cycle on either day 1 or optionally day 2. RESULTS Forty patients were enrolled and 39 were treated. Median age was 72 years (range 51-89). There were 7 complete and 15 partial responses, for a response rate of 56.4% (95% confidence interval, 39.6-72.2). Most cycles (82.8%) were given with same-day pegfilgrastim. Notable grade 3 and 4 nonhematologic toxicities were fatigue and mucositis (10.3% each). There were 4 episodes of neutropenic fever (4 of 198 cycles [2%]; 4 of 39 patients [10.3%]) and no treatment-related deaths. Median overall survival was 14.4 months. CONCLUSION The combination of gemcitabine, paclitaxel, and doxorubicin is effective first-line chemotherapy for patients with advanced urothelial carcinoma and renal insufficiency. Neutropenic prophylaxis was acceptable whether pegfilgrastim was given immediately or 24 hours after chemotherapy.
Collapse
|
28
|
Delanaye P, Flamant M, Cavalier É, Guerber F, Vallotton T, Moranne O, Pottel H, Boffa JJ, Mariat C. [Dosing adjustment and renal function: Which equation(s)?]. Nephrol Ther 2015; 12:18-31. [PMID: 26602880 DOI: 10.1016/j.nephro.2015.07.472] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/18/2022]
Abstract
While the CKD-EPI (for Chronic Kidney Disease Epidemiology) equation is now implemented worldwide, utilization of the Cockcroft formula is still advocated by some physicians for drug dosage adjustment. Justifications for this recommendation are that the Cockcroft formula was preferentially used to determine dose adjustments according to renal function during the development of many drugs, better predicts drugs-related adverse events and decreases the risk of drug overexposure in the elderly. In this opinion paper, we discuss the weaknesses of the rationale supporting the Cockcroft formula and endorse the French HAS (Haute Autorité de santé) recommendation regarding the preferential use of the CKD-EPI equation. When glomerular filtration rate (GFR) is estimated in order to adjust drug dosage, the CKD-EPI value should be re-expressed for the individual body surface area (BSA). Given the difficulty to accurately estimate GFR in the elderly and in individuals with extra-normal BSA, we recommend to prescribe in priority monitorable drugs in those populations or to determine their "true" GFR using a direct measurement method.
Collapse
Affiliation(s)
- Pierre Delanaye
- Service de néphrologie, dialyse et transplantation, CHU Sart-Tilman, université de Liège, 4000 Liège, Belgique
| | - Martin Flamant
- Service d'explorations fonctionnelles, hôpital Bichat, AP-HP, université Paris Diderot, Paris, France
| | - Étienne Cavalier
- Service de chimie clinique, CHU Sart-Tilman, université de Liège, 4000 Liège, Belgique
| | - Fabrice Guerber
- Laboratoire Oriade-Vizille, 75, chemin de la Terrasse, 38220 Vizille, France
| | - Thomas Vallotton
- Laboratoire Vialle, Bastia et Syndicat des jeunes biologistes médicaux, 20600 BastiaFrance
| | - Olivier Moranne
- EA 2415, biostatistique, épidémiologie et santé publique, institut universitaire de recherche clinique, université de Montpellier, 34093 Montpellier, France
| | - Hans Pottel
- Department of Primary Care and Public Health at Kulak, KU Leuven Kulak, 8500 Kortrijk, Belgique
| | - Jean-Jacques Boffa
- Inserm 1155, service de néphrologie et dialyse, hôpital Tenon, AP-HP, université Pierre-et-Marie-Curie, 75020 Paris, France
| | - Christophe Mariat
- Service de néphrologie, dialyse et transplantation, hôpital Nord, CHU de Saint-Étienne, université Jean-Monnet, 42055 Saint-Étienne, France.
| |
Collapse
|
29
|
Beumer JH, Ding F, Tawbi H, Lin Y, Viluh D, Chatterjee I, Rinker M, Chow SL, Ivy SP. Effect of Renal Dysfunction on Toxicity in Three Decades of Cancer Therapy Evaluation Program-Sponsored Single-Agent Phase I Studies. J Clin Oncol 2015; 34:110-6. [PMID: 26392101 DOI: 10.1200/jco.2014.59.7302] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Alterations in renal clearance of anticancer drugs can affect the occurrence of toxicities related to drug exposure. The National Cancer Institute and the US Food and Drug Administration (FDA) use different criteria to classify renal dysfunction. We examined those discrepancies and their potential association with the incidence of toxicities in patients enrolled onto Cancer Therapy Evaluation Program-sponsored single-agent phase I studies over three decades (1979 to 2010). METHODS Data to estimate creatinine clearance according to the Cockcroft-Gault and Jelliffe formulas were available from 10,236 patients, and data to estimate creatinine clearance according to the six- and four-variable Modification of Diet in Renal Disease formulas were available from a subset (n = 4,084). Patients were classified according to National Cancer Institute and FDA criteria, and the rates of clinically relevant toxicities were evaluated within groups and compared among groups. RESULTS Cockcroft-Gault estimated renal function improved over time, which may be attributed to an increase in weight of patients in the same time frame. Approximately 36% of patients enrolled onto phase I trials had mild renal dysfunction by FDA criteria. Relative to normal function, mild renal dysfunction was associated with a statistically significant but small increase in grade 3 or 4 nonhematologic toxicity and any relevant toxicities. CONCLUSION Patients with mild renal dysfunction by FDA criteria have routinely been enrolled onto phase I studies of antineoplastics without clinically meaningful increase in the risk of toxicity. In future oncology renal dysfunction trials based on the FDA classification, the FDA mild group may only need to be activated when the moderate and normal groups differ substantially in tolerability or pharmacokinetics.
Collapse
Affiliation(s)
- Jan H Beumer
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD.
| | - Fei Ding
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - Hussein Tawbi
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - Yan Lin
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - Diana Viluh
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - Indrani Chatterjee
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - Matthew Rinker
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - Selina L Chow
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| | - S Percy Ivy
- Jan H. Beumer, Fei Ding, Hussein Tawbi, Yan Lin, and Selina L. Chow, University of Pittsburgh Cancer Institute; Jan H. Beumer, University of Pittsburgh School of Pharmacy; Jan H. Beumer and Hussein Tawbi, University of Pittsburgh School of Medicine; Yan Lin, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA; Diana Viluh, Indrani Chatterjee, and Matthew Rinker, Theradex, Princeton, NJ; and S. Percy Ivy, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD
| |
Collapse
|
30
|
Dessein PH, Hsu HC, Tsang L, Millen AME, Woodiwiss AJ, Norton GR, Solomon A, Gonzalez-Gay MA. Kidney function, endothelial activation and atherosclerosis in black and white Africans with rheumatoid arthritis. PLoS One 2015; 10:e0121693. [PMID: 25806966 PMCID: PMC4373952 DOI: 10.1371/journal.pone.0121693] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 02/14/2015] [Indexed: 12/19/2022] Open
Abstract
Objective To determine whether kidney function independently relates to endothelial activation and ultrasound determined carotid atherosclerosis in black and white Africans with rheumatoid arthritis (RA). Methods We calculated the Jelliffe, 5 Cockcroft-Gault equations, Salazar-Corcoran, Modification of Diet in Renal Disease (MDRD) and Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) estimated glomerular filtration rate (EGFR) equations in 233 (112 black) RA patients. Results The CKD-EPI eGFR was <90 ml/min/1.73m2 in 49.1% and 30.6% of black and white patients, respectively (odds ratio (95% confidence interval) = 2.19 (1.28–3.75), p = 0.004). EGFRs were overall consistently associated with monocyte chemoattractant protein-1 and angiopoietin 2 concentrations in white patients, and with carotid intima-media thickness and plaque in black participants. Amongst black patients, plaque prevalence was 36.7% and the area under the curve (AUC) of the receiver operating characteristic (ROC) curve was not associated with plaque presence for the MDRD equation (p = 0.3), whereas the respective relationship was significant or borderline significant (p = 0.003 to 0.08) and of similar extent (p>0.1 for comparisons of AUC (SE)) for the other 8 equations. Based on optimal eGFR cutoff values with sensitivities and specificities ranging from 42 to 60% and 70 to 91% respectively, as determined in ROC curve analysis, a low eGFR increased the odds ratio for plaque 2.2 to 4.0 fold. Conclusion Reduced kidney function is independently associated with atherosclerosis and endothelial activation in black and white Africans with RA, respectively. CKD is highly prevalent in black Africans with RA. Apart from the MDRD, eGFR equations are useful in predicting carotid plaque presence, a coronary heart disease equivalent, amongst black African RA patients.
Collapse
Affiliation(s)
- Patrick H. Dessein
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- * E-mail:
| | - Hon-Chun Hsu
- Department of Nephrology, Milpark Hospital, Johannesburg, South Africa
| | - Linda Tsang
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aletta M. E. Millen
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Angela J. Woodiwiss
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gavin R. Norton
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ahmed Solomon
- Department of Rheumatology, Charlotte Maxeke Johannesburg Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Miguel A. Gonzalez-Gay
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Spain
| |
Collapse
|
31
|
Eppenga WL, Kramers C, Derijks HJ, Wensing M, Wetzels JFM, De Smet PAGM. Individualizing pharmacotherapy in patients with renal impairment: the validity of the Modification of Diet in Renal Disease formula in specific patient populations with a glomerular filtration rate below 60 ml/min. A systematic review. PLoS One 2015; 10:e0116403. [PMID: 25741695 PMCID: PMC4351004 DOI: 10.1371/journal.pone.0116403] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/09/2014] [Indexed: 01/20/2023] Open
Abstract
Background The Modification of Diet in Renal Disease (MDRD) formula is widely used in clinical practice to assess the correct drug dose. This formula is based on serum creatinine levels which might be influenced by chronic diseases itself or the effects of the chronic diseases. We conducted a systematic review to determine the validity of the MDRD formula in specific patient populations with renal impairment: elderly, hospitalized and obese patients, patients with cardiovascular disease, cancer, chronic respiratory diseases, diabetes mellitus, liver cirrhosis and human immunodeficiency virus. Methods and Findings We searched for articles in Pubmed published from January 1999 through January 2014. Selection criteria were (1) patients with a glomerular filtration rate (GFR) < 60 ml/min (/1.73m2), (2) MDRD formula compared with a gold standard and (3) statistical analysis focused on bias, precision and/or accuracy. Data extraction was done by the first author and checked by a second author. A bias of 20% or less, a precision of 30% or less and an accuracy expressed as P30% of 80% or higher were indicators of the validity of the MDRD formula. In total we included 27 studies. The number of patients included ranged from 8 to 1831. The gold standard and measurement method used varied across the studies. For none of the specific patient populations the studies provided sufficient evidence of validity of the MDRD formula regarding the three parameters. For patients with diabetes mellitus and liver cirrhosis, hospitalized patients and elderly with moderate to severe renal impairment we concluded that the MDRD formula is not valid. Limitations of the review are the lack of considering the method of measuring serum creatinine levels and the type of gold standard used. Conclusion In several specific patient populations with renal impairment the use of the MDRD formula is not valid or has uncertain validity.
Collapse
Affiliation(s)
- Willemijn L. Eppenga
- Radboud University Medical Center, Radboud Institute for Health Sciences, IQ Healthcare, Nijmegen, The Netherlands
- * E-mail:
| | - Cornelis Kramers
- Radboud University Medical Center, Department of Pharmacology and Toxicology, Nijmegen, The Netherlands
- Department of Clinical Pharmacy, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Hieronymus J. Derijks
- Hospital Pharmacy ‘ZANOB’, ‘s-Hertogenbosch, The Netherlands
- Division of Pharmacoepidemiology and Pharmacotherapy, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Michel Wensing
- Radboud University Medical Center, Radboud Institute for Health Sciences, IQ Healthcare, Nijmegen, The Netherlands
| | - Jack F. M. Wetzels
- Radboud University Medical Center, Department of Nephrology, Nijmegen, The Netherlands
| | - Peter A. G. M. De Smet
- Radboud University Medical Center, Radboud Institute for Health Sciences, IQ Healthcare, Nijmegen, The Netherlands
- Radboud University Medical Center, Department of Pharmacy, Nijmegen, The Netherlands
| |
Collapse
|
32
|
Chew-Harris JSC, Florkowski CM, George PM, Endre ZH. Comparative performances of the new chronic kidney disease epidemiology equations incorporating cystatin C for use in cancer patients. Asia Pac J Clin Oncol 2014; 11:142-51. [PMID: 25471594 DOI: 10.1111/ajco.12312] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2014] [Indexed: 01/28/2023]
Abstract
AIM In cancer patients receiving chemotherapy treatment, accurate assessment of kidney function is required. The aim of our study was to investigate whether the inclusion of cystatin C together with creatinine in prediction equations would improve the prediction of glomerular filtration rate (GFR). METHODS Plasma creatinine and cystatin C were analyzed in 155 patients (cancer, n = 80, kidney donors, n = 75) undergoing (99m) Technetium diethylenepentaacetic (Tc-DTPA) GFR clearance. Equations by the CKD-EPI (chronic kidney disease epidemiology) group (creatinine-, creatinine + cystatin C-, cystatin C-based, respectively) and Cockcroft-Gault were compared with Tc-DTPA GFR by difference plots, receiver operator characteristics curve analysis, root mean square error, chi-squared analysis and percentage concordance according to carboplatin dosage. Comparisons between two creatinine methodologies (enzymatic vs Jaffe) were also performed. RESULTS In the overall group, the combination creatinine and cystatin C equation had 69% of results within 20% of GFR (P20), a sensitivity of 86.3% and a specificity of 73.1% to detect reduced GFR at <90 mL/min/1.73 m(2), and a concordance of 78%. In contrast, the traditional Cockcroft-Gault equation had a P20 of 38.0%, with a large underestimation to predict GFR, thereby accounting for approximately 45% of dosing discordance. No obvious differences were obtained when comparing the performance of equations using the two creatinine methodologies. CONCLUSION The inclusion of cystatin C in the CKD-EPI equations improved the prediction of kidney function in the overall population, although probably not sufficiently for it to be favored over radioisotopic GFR for guiding chemotherapy. More research is warranted to further improve estimated GFR equations for these purposes.
Collapse
Affiliation(s)
- Janice S C Chew-Harris
- Clinical Biochemistry Unit, Canterbury Health Laboratories, Christchurch, New Zealand; Departments of Pathology and Medicine, University of Otago, Christchurch, New Zealand
| | | | | | | |
Collapse
|
33
|
Giglio D. A New Equation for Estimating Glomerular Filtration Rate in Cancer Patients. Chemotherapy 2014; 60:63-72. [DOI: 10.1159/000365724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/03/2014] [Indexed: 11/19/2022]
|
34
|
Ludwig H, Rauch E, Kuehr T, Adam Z, Weißmann A, Kasparu H, Autzinger EM, Heintel D, Greil R, Poenisch W, Müldür E, Zojer N. Lenalidomide and dexamethasone for acute light chain-induced renal failure: a phase II study. Haematologica 2014; 100:385-91. [PMID: 25398836 DOI: 10.3324/haematol.2014.115204] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We prospectively evaluated the activity and tolerance of lenalidomide-dexamethasone in 35 patients with acute light chain-induced renal failure. The lenalidomide dose was adapted to the estimated glomerular filtration rate and dexamethasone was given at high dose in cycle one and at low dose thereafter. Four patients died within the first two cycles, and five discontinued therapy leaving 26 patients for the per-protocol analysis. Responses were observed in 24/35 (68.6%) patients of the intent-to-treat population. Complete response was noted in seven patients (20%), very good partial response in three patients (8.6%), partial response in 14 patients (40%), and minimal response in one patient (2.9%). Renal response was observed in 16 (45.7%) patients: five (14.2%) achieved complete, four (11.4%) partial and seven (20%) minor renal responses. Five of 13 patients who were dialysis dependent at baseline became dialysis independent. The median time to myeloma and to renal response was 28 days for both parameters, while the median time to best myeloma and best renal response was 92 and 157 days, respectively. The median estimated glomerular filtration rate increased significantly in patients with partial response or better from 17.1 mL/min at baseline to 39.1 mL/min at best response (P=0.001). The median progression-free and overall survival was 5.5 and 21.8 months, respectively, in the intent-to-treat population and 12.1 and 31.4 months, respectively, in the per-protocol group. Infections, cardiotoxicity, anemia and thrombocytopenia were the most frequent toxicities. In conclusion, the lenalidomide-dexamethasone regimen achieved rapid and substantial myeloma and renal responses. The trial was registered under EUDRACT number 2008-006497-15.
Collapse
Affiliation(s)
- Heinz Ludwig
- Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Elisabeth Rauch
- Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Thomas Kuehr
- Department of Internal Medicine 4, Hospital Wels-Grieskirchen, Austria
| | | | | | - Hedwig Kasparu
- Department of Internal Medicine, Hospital Elisabethinen, Linz, Austria
| | | | - Daniel Heintel
- Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Richard Greil
- Department of Internal Medicine III, Hospital Salzburg, Austria
| | - Wolfram Poenisch
- Department of Hemato-Oncology, University Clinic Leipzig, Germany
| | - Ercan Müldür
- Department of Medicine I, Wilhelminenspital, Vienna, Austria
| | - Niklas Zojer
- Department of Medicine I, Wilhelminenspital, Vienna, Austria
| |
Collapse
|
35
|
Cathomas R, Klingbiel D, Geldart T, Mead G, Ellis S, Wheater M, Simmonds P, Nagaraj N, von Moos R, Fehr M. Relevant risk of carboplatin underdosing in cancer patients with normal renal function using estimated GFR: lessons from a stage I seminoma cohort. Ann Oncol 2014; 25:1591-7. [DOI: 10.1093/annonc/mdu129] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
36
|
Fehr M, Geldart T, Klingbiel D, Cathomas R. Measurement or estimation of glomerular filtration rate in seminoma patients: quite another cup of tea. Eur J Cancer 2014; 50:2176-7. [PMID: 24915777 DOI: 10.1016/j.ejca.2014.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 04/16/2014] [Indexed: 11/19/2022]
Affiliation(s)
- Martin Fehr
- Medical Oncology and Haematology, Kantonsspital St. Gallen, Switzerland.
| | - Tom Geldart
- Medical Oncology, Poole and Royal Bournemouth Hospitals, UK
| | - Dirk Klingbiel
- SAKK (Swiss Group for Clinical Cancer Research) Coordinating Centre Berne, Switzerland
| | - Richard Cathomas
- Oncology/Haematology, Kantonsspital Graubünden, Chur, Switzerland
| |
Collapse
|
37
|
Lauritsen J, Gundgaard MG, Mortensen MS, Oturai PS, Feldt-Rasmussen B, Daugaard G. Reliability of estimated glomerular filtration rate in patients treated with platinum containing therapy. Int J Cancer 2014; 135:1733-9. [DOI: 10.1002/ijc.28816] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 02/03/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Jakob Lauritsen
- Department of Oncology, Rigshospitalet; Copenhagen University Hospital; Copenhagen Denmark
| | - Maria G. Gundgaard
- Department of Oncology, Rigshospitalet; Copenhagen University Hospital; Copenhagen Denmark
- Survivorship, Danish Cancer Society Research Center; Copenhagen Denmark
| | - Mette S. Mortensen
- Department of Oncology, Rigshospitalet; Copenhagen University Hospital; Copenhagen Denmark
| | - Peter S. Oturai
- Department of Clinical Physiology; Nuclear Medicine and PET, Rigshospitalet; Copenhagen Denmark
| | - Bo Feldt-Rasmussen
- Department of Nephrology; Copenhagen University Hospital, Rigshospitalet; Copenhagen Denmark
| | - Gedske Daugaard
- Department of Oncology, Rigshospitalet; Copenhagen University Hospital; Copenhagen Denmark
| |
Collapse
|
38
|
Shepherd STC, Gillen G, Morrison P, Forte C, Macpherson IR, White JD, Mark PB. Performance of formulae based estimates of glomerular filtration rate for carboplatin dosing in stage 1 seminoma. Eur J Cancer 2014; 50:944-52. [PMID: 24445148 DOI: 10.1016/j.ejca.2013.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 12/19/2013] [Accepted: 12/25/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND Single cycle carboplatin, dosed by glomerular filtration rate (GFR), is standard adjuvant therapy for stage 1 seminoma. Accurate measurement of GFR is essential for correct dosing. Isotopic methods remain the gold standard for the determination of GFR. Formulae to estimate GFR have improved the assessment of renal function in non-oncological settings. We assessed the utility of these formulae for carboplatin dosing. METHODS We studied consecutive subjects receiving adjuvant carboplatin for stage 1 seminoma at our institution between 2007 and 2012. Subjects underwent 51Cr-ethylene diamine tetra-acetic acid (EDTA) measurement of GFR with carboplatin dose calculated using the Calvert formula. Theoretical carboplatin doses were calculated from estimated GFR using Chronic Kidney Disease-Epidemiology (CKD-EPI), Management of Diet in Renal Disease (MDRD) and Cockcroft-Gault (CG) formulae with additional correction for actual body surface area (BSA). Carboplatin doses calculated by formulae were compared with dose calculated by isotopic GFR; a difference <10% was considered acceptable. RESULTS 115 patients were identified. Mean isotopic GFR was 96.9 ml/min/1.73 m(2). CG and CKD-EPI tended to overestimate GFR whereas MDRD tended to underestimate GFR. The CKD-EPI formula had greatest accuracy. The CKD-EPI formula, corrected for actual BSA, performed best; 45.9% of patients received within 10% of correct carboplatin dose. Patients predicted as underdosed (13.5%) by CKD-EPI were more likely to be obese (p=0.013); there were no predictors of the 40.5% receiving an excess dose. CONCLUSIONS Our data support further evaluation of the CKD-EPI formula in this patient population but clinically significant variances in carboplatin dosing occur using non-isotopic methods of GFR estimation. Isotopic determination of GFR should remain the recommended standard for carboplatin dosing when accuracy is essential.
Collapse
Affiliation(s)
- Scott T C Shepherd
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, United Kingdom.
| | - Gerry Gillen
- Department of Nuclear Medicine, Gartnavel Hospital, Glasgow G12 0YN, United Kingdom
| | - Paula Morrison
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, United Kingdom
| | - Carla Forte
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, United Kingdom
| | - Iain R Macpherson
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, United Kingdom
| | - Jeff D White
- Beatson West of Scotland Cancer Centre, Glasgow G12 0YN, United Kingdom
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| |
Collapse
|
39
|
Dooley MJ, Poole SG, Rischin D. Dosing of cytotoxic chemotherapy: impact of renal function estimates on dose. Ann Oncol 2013; 24:2746-52. [PMID: 23928359 DOI: 10.1093/annonc/mdt300] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Oncology clinicians are now routinely provided with an estimated glomerular filtration rate on pathology reports whenever serum creatinine is requested. The utility of using this for the dose determination of renally excreted drugs compared with other existing methods is needed to inform practice. PATIENTS AND METHODS Renal function was determined by [Tc(99m)]DTPA clearance in adult patients presenting for chemotherapy. Renal function was calculated using the 4-variable Modification of Diet in Renal Disease (4v-MDRD), Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI), Cockcroft and Gault (CG), Wright and Martin formulae. Doses for renal excreted cytotoxic drugs, including carboplatin, were calculated. RESULTS The concordance of the renal function estimates according to the CKD classification with measured Tc(99m)DPTA clearance in 455 adults (median age 64.0 years: range 17-87 years) for the 4v-MDRD, CKD-EPI, CG, Martin and Wright formulae was 47.7%, 56.3%, 46.2%, 56.5% and 60.2%, respectively. Concordance for chemotherapy dose for these formulae was 89.0%, 89.5%, 85.1%, 89.9% and 89.9%, respectively. Concordance for carboplatin dose specifically was 66.4%, 71.4%, 64.0%, 73.8% and 73.2%. CONCLUSION All bedside formulae provide similar levels of concordance in dosage selection for the renal excreted chemotherapy drugs when compared with the use of a direct measure of renal function.
Collapse
Affiliation(s)
- M J Dooley
- Department of Pharmacy, Alfred Health, Melbourne
| | | | | |
Collapse
|
40
|
Blankenship P, Aubrey Waddell J. Survey of carboplatin dosing strategies in oncology practices. J Am Pharm Assoc (2003) 2013; 53:420-2. [DOI: 10.1331/japha.2013.12053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Do OH, Nguyen KT. The role of glycemia and blood pressure control on the rate of decline in glomerular filtration rate in Vietnamese type 2 diabetes patients. Int J Diabetes Dev Ctries 2013. [DOI: 10.1007/s13410-013-0112-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
42
|
Abstract
BACKGROUND Glomerular filtration rate (GFR) is used in the calculation of carboplatin dose. Glomerular filtration rate is measured using a radioisotope method (radionuclide GFR (rGFR)), however, estimation equations are available (estimated GFR (eGFR)). Our aim was to assess the accuracy of three eGFR equations and the subsequent carboplatin dose in an oncology population. PATIENTS AND METHODS Patients referred for an rGFR over a 3-year period were selected; eGFR was calculated using the Modification of Diet in Renal Disease (MDRD), Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) and Cockcroft-Gault (CG) equations. Carboplatin doses were calculated for those patients who had received carboplatin chemotherapy. Bias, precision and accuracy were examined. RESULTS Two hundred and eighty-eight studies met the inclusion/exclusion criteria. Paired t-tests showed significant differences for all three equations between rGFR and eGFR with biases of 12.3 (MDRD), 13.6 (CKD-EPI) and 7.7 ml min(-1) per 1.73 m(2) (CG). An overestimation in carboplatin dose was seen in 81%, 87% and 66% of studies using the MDRD, CKD-EPI and CG equations, respectively. CONCLUSION The MDRD and CKD-EPI equations performed poorly compared with the reference standard rGFR; the CG equation showed smaller bias and higher accuracy in our oncology population. On the basis of our results we recommend that the rGFR should be used for accurate carboplatin chemotherapy dosing and where unavailable the use of the CG equation is preferred.
Collapse
|