A Canadian Study of Cisplatin Metabolomics and Nephrotoxicity (ACCENT): A Clinical Research Protocol

Anshika Jain, Ryan Huang, Jasmine Lee, Natasha Jawa, Yong Jin Lim, Mike Guron, Sharon Abish, Paul C Boutros, Michael Brudno, Bruce Carleton, Geoffrey D E Cuvelier, Lakshman Gunaratnam, Cheryl Ho, Khosrow Adeli, Sara Kuruvilla, Giles Lajoie, Geoffrey Liu, Paul C Nathan, Shahrad Rod Rassekh, Michael Rieder, Sushrut S Waikar, Stephen A Welch, Matthew A Weir, Eric Winquist, David S Wishart, Alexandra P Zorzi, Tom Blydt-Hansen, Michael Zappitelli, Bradley Urquhart, Anshika Jain, Ryan Huang, Jasmine Lee, Natasha Jawa, Yong Jin Lim, Mike Guron, Sharon Abish, Paul C Boutros, Michael Brudno, Bruce Carleton, Geoffrey D E Cuvelier, Lakshman Gunaratnam, Cheryl Ho, Khosrow Adeli, Sara Kuruvilla, Giles Lajoie, Geoffrey Liu, Paul C Nathan, Shahrad Rod Rassekh, Michael Rieder, Sushrut S Waikar, Stephen A Welch, Matthew A Weir, Eric Winquist, David S Wishart, Alexandra P Zorzi, Tom Blydt-Hansen, Michael Zappitelli, Bradley Urquhart

Abstract

Background: Cisplatin, a chemotherapy used to treat solid tumors, causes acute kidney injury (AKI), a known risk factor for chronic kidney disease and mortality. AKI diagnosis relies on biomarkers which are only measurable after kidney damage has occurred and functional impairment is apparent; this prevents timely AKI diagnosis and treatment. Metabolomics seeks to identify metabolite patterns involved in cell tissue metabolism related to disease or patient factors. The A Canadian study of Cisplatin mEtabolomics and NephroToxicity (ACCENT) team was established to harness the power of metabolomics to identify novel biomarkers that predict risk and discriminate for presence of cisplatin nephrotoxicity, so that early intervention strategies to mitigate onset and severity of AKI can be implemented.

Objective: Describe the design and methods of the ACCENT study which aims to identify and validate metabolomic profiles in urine and serum associated with risk for cisplatin-mediated nephrotoxicity in children and adults.

Design: Observational prospective cohort study.

Setting: Six Canadian oncology centers (3 pediatric, 1 adult and 2 both).

Patients: Three hundred adults and 300 children planned to receive cisplatin therapy.

Measurements: During two cisplatin infusion cycles, serum and urine will be measured for creatinine and electrolytes to ascertain AKI. Many patient and disease variables will be collected prospectively at baseline and throughout therapy. Metabolomic analyses of serum and urine will be done using mass spectrometry. An untargeted metabolomics approach will be used to analyze serum and urine samples before and after cisplatin infusions to identify candidate biomarkers of cisplatin AKI. Candidate metabolites will be validated using an independent cohort.

Methods: Patients will be recruited before their first cycle of cisplatin. Blood and urine will be collected at specified time points before and after cisplatin during the first infusion and an infusion later during cancer treatment. The primary outcome is AKI, defined using a traditional serum creatinine-based definition and an electrolyte abnormality-based definition. Chart review 3 months after cisplatin therapy end will be conducted to document kidney health and survival.

Limitations: It may not be possible to adjust for all measured and unmeasured confounders when evaluating prediction of AKI using metabolite profiles. Collection of data across multiple sites will be a challenge.

Conclusions: ACCENT is the largest study of children and adults treated with cisplatin and aims to reimagine the current model for AKI diagnoses using metabolomics. The identification of biomarkers predicting and detecting AKI in children and adults treated with cisplatin can greatly inform future clinical investigations and practices.

Trial registration: ClinicalTrials.gov NCT04442516.

Keywords: acute kidney injury; cisplatin nephrotoxicity; cohort study; metabolomics; pediatrics.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

© The Author(s) 2021.

Figures

Figure 1.
Figure 1.
Steering committee, participating centers, and collaborators. Note. The figure demonstrates the individuals involved in each section of the project and their locations. UBC, University of British Columbia; UWO, University of Western Ontario; UofA, University of Alberta; UofM, University of Manitoba; PMH, Princess Margaret Hospital; BU, Boston University. This figure was created with BioRender.com.
Figure 2.
Figure 2.
Schematic of proposed proximal tubule cisplatin toxicity. Note. GST = glutathione S-transferase; GGT = γ glutamyl transpeptidase; APN = aminopeptidase.,,, This figure was created with BioRender.com.
Figure 3.
Figure 3.
Overview of experimental design. Note. DNA is captured only once at the first cisplatin cycle. Both blood and urine are collected 6 times during the study (3 collection time points for each of 2 cisplatin cycles).
Figure 4.
Figure 4.
Sequential overview of the data analysis process. Note. Encompasses data collection, data processing, and multivariate analysis.
Figure 5.
Figure 5.
Timeline of progress to date. Note. Description of progress as of March 2021. This figure was created with BioRender.com.

References

    1. Society CC. Chemotherapy for oropharyngeal cancer. . Accessed October 27, 2021.
    1. Yao X, Panichpisal K, Kurtzman N, Nugent K. Cisplatin nephrotoxicity: a review. Am J Med Sci. 2007;334:115-124.
    1. Fujieda M, Matsunaga A, Hayashi A, Tauchi H, Chayama K, Sekine T. Children’s toxicology from bench to bed-Drug-induced Renal Injury (2): nephrotoxiciy induced by cisplatin and ifosfamide in children. J Toxicol Sci. 2009;34:SP251-SP257.
    1. McMahon KR, Rassekh SR, Schultz KR, et al.. Epidemiologic characteristics of acute kidney injury during cisplatin infusions in children treated for cancer. JAMA Netw Open. 2020;3:e203639.
    1. Sahni V, Choudhury D, Ahmed Z. Chemotherapy-associated renal dysfunction. Nat Rev Nephrol. 2009;5:450-462.
    1. Coca SG, Singanamala S, Parikh CR. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int. 2012;81:442-448.
    1. Mammen C, al Abbas A, Skippen P, et al.. Long-term risk of CKD in children surviving episodes of acute kidney injury in the intensive care unit: a prospective cohort study. Am J Kidney Dis. 2012;59:523-530.
    1. Chawla LS, Eggers PW, Star RA, Kimmel PL. Acute kidney injury and chronic kidney disease as interconnected syndromes. N Engl J Med. 2014;371:58-66.
    1. Greenberg JH, Coca S, Parikh CR. Long-term risk of chronic kidney disease and mortality in children after acute kidney injury: a systematic review. BMC Nephrol. 2014;15:184.
    1. Zappitelli M, Moffett BS, Hyder A, Goldstein SL. Acute kidney injury in non-critically ill children treated with aminoglycoside antibiotics in a tertiary healthcare centre: a retrospective cohort study. Nephrol Dial Transplant. 2011;26:144-150.
    1. Alkandari O, Eddington KA, Hyder A, et al.. Acute kidney injury is an independent risk factor for pediatric intensive care unit mortality, longer length of stay and prolonged mechanical ventilation in critically ill children: a two-center retrospective cohort study. Crit Care. 2011;15:1-12.
    1. Zappitelli M, Bernier P-L, Saczkowski RS, et al.. A small post-operative rise in serum creatinine predicts acute kidney injury in children undergoing cardiac surgery. Kidney Int. 2009;76:885-892.
    1. Li S, Krawczeski CD, Zappitelli M, et al.. Incidence, risk factors, and outcomes of acute kidney injury after pediatric cardiac surgery–a prospective multicenter study. Crit Care Med. 2011;39:1493-1499.
    1. Odutayo A, Wong CX, Farkouh M, et al.. AKI and long-term risk for cardiovascular events and mortality. J Am Soc Nephrol. 2017;28:377-387.
    1. McMahon KR, Rod Rassekh S, Schultz KR, et al.. Design and methods of the pan-Canadian applying biomarkers to minimize long-term effects of childhood/adolescent cancer treatment (ABLE) nephrotoxicity study: a prospective observational cohort study. Can J Kidney Health Dis. 2017;4:2054358117690338.
    1. American Society of Nephrology. American society of nephrology renal research report. J Am Soc Nephrol. 2005;16:1886-1903.
    1. Al-Ismaili Z, Palijan A, Zappitelli M. Biomarkers of acute kidney injury in children: discovery, evaluation, and clinical application. Pediatr Nephrol. 2011;26:29-40.
    1. Ferguson MA, Vaidya VS, Bonventre JV. Biomarkers of nephrotoxic acute kidney injury. Toxicology. 2008;245:182-193.
    1. Ariceta G, Rodriguez Soriano J, Vallo A, Navajas A. Acute and chronic effects of cisplatin therapy on renal magnesium homeostasis. Med Pediatr Oncol. 1997;28:35-40.
    1. Goren MP, Wright RK, Horowitz ME. Cumulative renal tubular damage associated with cisplatin nephrotoxicity. Cancer Chemother Pharmacol. 1986;18:69-73.
    1. İçli F, Karaoǧuz H, Dinçol D, et al.. Severe vascular toxicity associated with cisplatin-based chemotherapy. Cancer. 1993;72:587-593.
    1. de Jongh FE, van Veen RN, Veltman SJ, et al.. Weekly high-dose cisplatin is a feasible treatment option: analysis on prognostic factors for toxicity in 400 patients. Br J Cancer. 2003;88:1199-1206.
    1. Skinner R, Pearson ADJ, English MW, et al.. Cisplatin dose rate as a risk factor for nephrotoxicity in children. Br J Cancer. 1998;77:1677-1682.
    1. Xu EY, Perlina A, Vu H, et al.. Integrated pathway analysis of rat urine metabolic profiles and kidney transcriptomic profiles to elucidate the systems toxicology of model nephrotoxicants. Chem Res Toxicol. 2008;21:1548-1561.
    1. Hu S, Leblanc AF, Gibson AA, et al.. Identification of OAT1/OAT3 as contributors to cisplatin toxicity. Clin Transl Sci. 2017;10:412-420.
    1. Wen X, Buckley B, McCandlish E, et al.. Transgenic expression of the human MRP2 transporter reduces cisplatin accumulation and nephrotoxicity in Mrp2-null mice. Am J Pathol. 2014;184:1299-1308.
    1. Filipski KK, Loos WJ, Verweij J, Sparreboom A. Interaction of Cisplatin with the human organic cation transporter 2. Clin Cancer Res. 2008;14:3875-3880.
    1. Filipski KK, Mathijssen RH, Mikkelsen TS, Schinkel AH, Sparreboom A. Contribution of organic cation transporter 2 (OCT2) to cisplatin-induced nephrotoxicity. Clin Pharmacol Ther. 2009;86:396-402.
    1. Pabla N, Murphy RF, Liu K, Dong Z. The copper transporter Ctr1 contributes to cisplatin uptake by renal tubular cells during cisplatin nephrotoxicity. Am J Physiol Renal Physiol. 2009;296:F505-F511.
    1. Sprowl JA, van Doorn L, Hu S, et al.. Conjunctive therapy of cisplatin with the OCT2 inhibitor cimetidine: influence on antitumor efficacy and systemic clearance. Clin Pharmacol Ther. 2013;94:585-592.
    1. Townsend DM, Deng M, Zhang L, Lapus MG, Hanigan MH. Metabolism of cisplatin to a nephrotoxin in proximal tubule cells. J Am Soc Nephrol. 2003;14:1-10.
    1. Townsend DM, Marto JA, Deng M, Macdonald TJ, Hanigan MH. High pressure liquid chromatography and mass spectrometry characterization of the nephrotoxic biotransformation products of cisplatin. Drug Metab Dispos. 2003;31:705-713.
    1. Karasawa T, Steyger PS. An integrated view of cisplatin-induced nephrotoxicity and ototoxicity. Toxicol Lett. 2015;237:219-227.
    1. Nakamura T, Yonezawa A, Hashimoto S, Katsura T, Inui KI. Disruption of multidrug and toxin extrusion MATE1 potentiates cisplatin-induced nephrotoxicity. Biochem Pharmacol. 2010;80:1762-1767.
    1. Ohtsuki S, Asaba H, Takanaga H, et al.. Role of blood–brain barrier organic anion transporter 3 (OAT3) in the efflux of indoxyl sulfate, a uremic toxin: its involvement in neurotransmitter metabolite clearance from the brain. J Neurochem. 2002;83:57-66.
    1. Wikoff WR, Nagle MA, Kouznetsova VL, Tsigelny IF, Nigam SK. Untargeted metabolomics identifies enterobiome metabolites and putative uremic toxins as substrates of organic anion transporter 1 (Oat1). J Proteome Res. 2011;10:2842-2851.
    1. Motojima M, Hosokawa A, Yamato H, Muraki T, Yoshioka T. Uraemic toxins induce proximal tubular injury via organic anion transporter 1-mediated uptake. Br J Pharmacol. 2002;135:555-563.
    1. Miyamoto Y, Watanabe H, Noguchi T, et al.. Organic anion transporters play an important role in the uptake of p-cresyl sulfate, a uremic toxin, in the kidney. Nephrol Dial Transplant. 2011;26:2498-2502.
    1. Teft WA, Morse BL, Leake BF, et al.. Identification and characterization of trimethylamine-N-oxide uptake and efflux transporters. Mol Pharm. 2017;14:310-318.
    1. Pariyani R, Ismail IS, Azam A, et al.. Urinary metabolic profiling of cisplatin nephrotoxicity and nephroprotective effects of Orthosiphon stamineus leaves elucidated by 1H NMR spectroscopy. J Pharm Biomed Anal. 2017;135:20-30.
    1. Portilla D, Li S, Nagothu KK, et al.. Metabolomic study of cisplatin-induced nephrotoxicity. Kidney Int. 2006;69:2194-2204.
    1. Gu L, Shi H, Zhang R, et al.. Simultaneous determination of five specific and sensitive nephrotoxicity biomarkers in serum and urine samples of four drug-induced kidney injury models. J Chromatogr Sci. 2016;55:60-68.
    1. Iwata K, Watanabe H, Morisaki T, et al.. Involvement of indoxyl sulfate in renal and central nervous system toxicities during cisplatin-induced acute renal failure. Pharm Res. 2007;24:662-671.
    1. Kusumoto M, Kamobayashi H, Sato D, et al.. Alleviation of cisplatin-induced acute kidney injury using phytochemical polyphenols is accompanied by reduced accumulation of indoxyl sulfate in rats. Clin Exp Nephrol. 2011;15:820-830.
    1. Uehara T, Horinouchi A, Morikawa Y, et al.. Identification of metabolomic biomarkers for drug-induced acute kidney injury in rats. J Appl Toxicol. 2014;34:1087-1095.
    1. Wang W, Hao G, Pan Y, et al.. Serum indoxyl sulfate is associated with mortality in hospital-acquired acute kidney injury: a prospective cohort study. BMC Nephrol. 2019;20:57.
    1. Mercier K, McRitchie S, Pathmasiri W, et al.. Preterm neonatal urinary renal developmental and acute kidney injury metabolomic profiling: an exploratory study. Pediatr Nephrol. 2017;32:151-161.
    1. Yu B, Zheng Y, Nettleton JA, Alexander D, Coresh J, Boerwinkle E. Serum metabolomic profiling and incident CKD among African Americans. Clin J Am Soc Nephrol. 2014;9:1410-1417.
    1. Silva RE, Baldim JL, Chagas-Paula DA, et al.. Predictive metabolomic signatures of end-stage renal disease: a multivariate analysis of population-based data. Biochimie. 2018;152:14-30.
    1. Velenosi TJ, Thomson BKA, Tonial NC, et al.. Untargeted metabolomics reveals N, N, N-trimethyl-L-alanyl-L-proline betaine (TMAP) as a novel biomarker of kidney function. Sci Rep. 2019;9:1-13.
    1. Wilmes A, Bielow C, Ranninger C, et al.. Mechanism of cisplatin proximal tubule toxicity revealed by integrating transcriptomics, proteomics, metabolomics and biokinetics. Toxicol In Vitro. 2015;30:117-127.
    1. Zsengellér ZK, Ellezian L, Brown D, et al.. Cisplatin nephrotoxicity involves mitochondrial injury with impaired tubular mitochondrial enzyme activity. J Histochem Cytochem. 2012;60:521-529.
    1. Moran SM, Myers BD. Course of acute renal failure studied by a model of creatinine kinetics. Kidney Int. 1985;27:928-937.
    1. Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney Injury Work Group. KDIGO clinical practice guideline for acute kidney injury. Kidney Int Suppl. 2012;2:1-138.
    1. Chawla LS, Kellum JA. Biomarkers are transforming our understanding of AKI. Nat Rev Nephrol. 2012;8:68-70.
    1. Ciarimboli G, Deuster D, Knief A, et al.. Organic cation transporter 2 mediates cisplatin-induced oto-and nephrotoxicity and is a target for protective interventions. Am J Pathol. 2010;176:1169-1180.
    1. Moneim LMA, Helmy MW, El-Abhar HS. Co-targeting of endothelin-A and vitamin D receptors: a novel strategy to ameliorate cisplatin-induced nephrotoxicity. Pharmacol Rep. 2019;71:917-925.
    1. Ilić S, Stojiljković N, Sokolović D, Jovanovic I, Stojanović N. Morphometric analysis of structural renal alterations and beneficial effects of aminoguanidine in acute kidney injury induced by cisplatin in rats. Can J Physiol Pharmacol. 2020;98:117-123.
    1. Li C, Li L, Yi Y, et al.. L-tetrahydropalmatine attenuates cisplatin-induced nephrotoxicity via selective inhibition of organic cation transporter 2 without impairing its antitumor efficacy. Biochem Pharmacol. 2020;177:114021.
    1. Hamano H, Ikeda Y, Goda M, et al.. Diphenhydramine may be a preventive medicine against cisplatin-induced kidney toxicity. Kidney Int. 2021;99:885-899.
    1. Karademir LD, Dogruel F, Kocyigit I, et al.. The efficacy of theophylline in preventing cisplatin-related nephrotoxicity in patients with cancer. Ren Fail. 2016;38:806-814.
    1. Kim JY, Jayne LA, Bai Y, et al.. Ribociclib mitigates cisplatin-associated kidney injury through retinoblastoma-1 dependent mechanisms. Biochem Pharmacol. 2020;177:113939.
    1. Okuda M, Saito H, Urakami Y, Takano M, Inui K. CDNA cloning and functional expression of a novel rat kidney organic cation transporter, OCT2. Biochem Biophys Res Commun. 1996;224:500-507.
    1. Backshall A, Sharma R, Clarke SJ, Keun HC. Pharmacometabonomic profiling as a predictor of toxicity in patients with inoperable colorectal cancer treated with capecitabine. Clin Cancer Res. 2011;17:3019-3028.
    1. US Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. National Institutes of Health, National Cancer Institute. . Accessed October 27, 2021.
    1. Espandiari P, Rosenzweig B, Zhang J, et al.. Age-related differences in susceptibility to cisplatin-induced renal toxicity. J Appl Toxicol. 2010;30:172-182.
    1. Worley B, Powers R. Multivariate analysis in metabolomics. Curr Metabolomics. 2013;1:92-107.
    1. Nyamundanda G, Brennan L, Gormley IC. Probabilistic principal component analysis for metabolomic data. BMC Bioinformatics. 2010;11:571.
    1. Winnike JH, Li Z, Wright FA, Macdonald JM, O’Connell TM, Watkins PB. Use of pharmaco-metabonomics for early prediction of acetaminophen-induced hepatotoxicity in humans. Clin Pharmacol Ther. 2010;88:45-51.
    1. Rotroff DM, Oki NO, Liang X, et al.. Pharmacometabolomic assessment of metformin in non-diabetic, African Americans. Front Pharmacol. 2016;7:135.
    1. Blydt-Hansen TD, Sharma A, Gibson IW, et al.. Urinary metabolomics for noninvasive detection of antibody-mediated rejection in children after kidney transplantation. Transplantation. 2017;101:2553.
    1. Blydt Hansen TD, Sharma A, Gibson IW, Mandal R, Wishart DS. Urinary metabolomics for noninvasive detection of borderline and acute T cell–mediated rejection in children after kidney transplantation. Am J Transplant. 2014;14:2339-2349.
    1. Archdekin B, Sharma A, Gibson IW, Rush D, Wishart DS, Blydt-Hansen TD. Non-invasive differentiation of non-rejection kidney injury from acute rejection in pediatric renal transplant recipients. Pediatr Transplant. 2019;23:e13364.
    1. Shang X, Zhong X, Tian X. Metabolomics of papillary thyroid carcinoma tissues: potential biomarkers for diagnosis and promising targets for therapy. Tumor Biology. 2016;37:11163-11175.
    1. Zwollo P, Desiderio S. Specific recognition of the blk promoter by the B-lymphoid transcription factor B-cell-specific activator protein. J Biol Chem. 1994;269:15310-15317.
    1. Rombouts C, Hemeryck LY, van Hecke T, De Smet S, De Vos WH. Untargeted metabolomics of colonic digests reveals kynurenine pathway metabolites, dityrosine and 3-dehydroxycarnitine as red versus white meat discriminating metabolites. Sci Rep. 2017;7:1-13.
    1. Tan G, Zhao B, Li Y, et al.. Pharmacometabolomics identifies dodecanamide and leukotriene B4 dimethylamide as a predictor of chemosensitivity for patients with acute myeloid leukemia treated with cytarabine and anthracycline. Oncotarget. 2017;8:88697.
    1. Cajka T, Smilowitz JT, Fiehn O. Validating quantitative untargeted lipidomics across nine liquid chromatography-high-resolution mass spectrometry platforms. Anal Chem. 2017;89:12360-12368.
    1. Alonso A, Marsal S, Julià A. Analytical methods in untargeted metabolomics: state of the art in 2015. Front Bioeng Biotechnol. 2015;3:23.
    1. Cecatti JG, Souza RT, Sulek K, et al.. Use of metabolomics for the identification and validation of clinical biomarkers for preterm birth: preterm SAMBA. BMC Pregnancy Childbirth. 2016;16:1-9.
    1. Schrimpe-Rutledge AC, Codreanu SG, Sherrod SD, McLean JA. Untargeted metabolomics strategies—Challenges and emerging directions. J Am Soc Mass Spectrom. 2016;27:1897-1905.
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44:837-845.
    1. Pencina MJ, D’Agostino RB, Sr, Steyerberg EW. Extensions of net reclassification improvement calculations to measure usefulness of new biomarkers. Stat Med. 2011;30:11-21.
    1. Pencina MJ, D’Agostino RB, Sr, D’Agostino RB, Jr, Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med. 2008;27:157-172.

Source: PubMed

3
Suscribir