Elsevier

Chemico-Biological Interactions

Volume 224, 5 December 2014, Pages 1-12
Chemico-Biological Interactions

Carcinogenicity in rats of the SGLT2 inhibitor canagliflozin

https://doi.org/10.1016/j.cbi.2014.09.018Get rights and content

Highlights

Abstract

The carcinogenicity potential of canagliflozin, an inhibitor of SGLT2, was evaluated in a 2-year rat study (10, 30, and 100 mg/kg). Rats showed an increase in pheochromocytomas, renal tubular tumors, and testicular Leydig cell tumors. Systemic exposure multiples at the highest dose relative to the maximum clinical dose were 12- to 21-fold. Pheochromocytomas and renal tubular tumors were noted in both sexes at 100 mg/kg. Leydig cell tumors were observed in males in all dose groups and were associated with increased luteinizing hormone levels. Hyperplasia was increased in the adrenal medulla at 100 mg/kg, but only a limited increase in simple tubular hyperplasia was observed in the kidney of males at 100 mg/kg. Hyperostosis occurred and was accompanied by substantial effects on calcium metabolism, including increased urinary calcium excretion and decreased levels of calcium regulating hormones (1,25-dihydroxyvitamin D and parathyroid hormone). A separate study with radiolabeled calcium confirmed that increased urinary calcium excretion was mediated via increased calcium absorption from the gastrointestinal tract. It was hypothesized that, at high doses, canagliflozin might have inhibited glucose absorption in the intestine via SGLT1 inhibition that resulted in glucose malabsorption, which increased calcium absorption by stimulating colonic glucose fermentation and reducing intestinal pH. Pheochromocytomas and adrenal medullary hyperplasia were attributed to altered calcium homeostasis, which have a known relationship in the rat. In conclusion, Leydig cell tumors were associated with increased luteinizing hormone levels and pheochromocytomas were most likely related to glucose malabsorption and altered calcium homeostasis. Renal tubular tumors may also have been linked to glucose malabsorption.

Introduction

SGLT2, a high-capacity, low-affinity glucose transporter, is expressed primarily in the proximal renal tubule and is responsible for most glucose reabsorption in the kidneys [1]. The transport of glucose is against its concentration gradient, coupled to the active transport of sodium. Inhibition of SGLT2 decreases glucose reabsorption in the renal tubule, increases urinary glucose excretion, and thereby reduces plasma glucose [1]. Phlorizin, which is a nonselective inhibitor of SGLT1 (another isoform which is predominantly present in the gastrointestinal tract) and SGLT2, reduces plasma glucose in preclinical models of type 2 diabetes mellitus (T2DM) [2]. Due to its nonselective nature, with activity on SGLT1, SGLT2, and glucose transporters and its poor pharmaceutical properties, phlorizin is unsuitable for clinical development [3]. A number of selective, metabolically stable, SGLT2 inhibitors have been discovered and are in clinical development to treat T2DM [4]. Canagliflozin was the first SGLT2 inhibitor approved in the United States for the treatment of T2DM.

Canagliflozin is a potent inhibitor of SGLT2 activity and has low potency SGLT1 inhibitory activity. Canagliflozin has been shown to increase urinary glucose excretion and decrease plasma glucose in animal models and in humans with T2DM [5], [6]. In repeated-dose toxicity studies with canagliflozin in mice up to 3 months, rats up to 6 months, and dogs up to 12 months, the compound was well tolerated and preneoplastic lesions were not observed [7], [8]. In these studies, there were substantial effects on urinalysis parameters that were considered to be related to the mode of action. These effects included increases in urinary volume, glucose, and electrolytes. Rats appeared to be the most sensitive species, with additional effects on calcium homeostasis and bone that were not observed in mice or dogs.

This report describes the preclinical assessment of the carcinogenicity potential of canagliflozin that was conducted as part of the development program to support world-wide registration. Where applicable, the results of the 3-month toxicity study in rats will be used for interpretation of findings in the carcinogenicity study. To assist interpretation of marked changes in calcium homeostasis (i.e., increased urinary excretion of calcium and increased trabecular bone formation) in rats treated with canagliflozin, a radiolabeled calcium absorption study was conducted in rats and the results are discussed in this article. In addition, possible mechanisms for induction of tumors in rats by canagliflozin and its clinical relevance are discussed in this article. Additional investigative studies that support the proposed mechanism of tumor formation are reported in a companion paper [9].

Section snippets

Test substance

Canagliflozin (CAS# 842133-18-0) with assay purity of 98.0% was formulated as an aqueous suspension containing 0.5% w/v hydroxypropyl methylcellulose in water. Formulations were stored refrigerated (2–8 °C) and in the dark. The concentrations, homogeneity, and stability of the formulations were checked during the studies using liquid chromatography and were confirmed to be within acceptance criteria.

Animals

Studies were performed with Sprague-Dawley rats purchased from Charles River (Kingston, NY and

Body weight, food consumption, and clinical observations

Dose-related decreases were observed in body weight and body weight gain throughout the study. Decreases were especially pronounced in the high-dose group, with terminal body weights and body weight gains of 0.79× and 0.70× for males and 0.77× and 0.67× for females, respectively, compared to vehicle control rats that received 0.5% w/v hydroxypropyl methylcellulose without canagliflozin. Food consumption was increased in a generally dose-related manner during the entire study. In males/females at

Discussion

The carcinogenicity profile of canagliflozin showed treatment-related tumors in three target organs in rats. These consisted of RTTs, pheochromocytomas, and LCTs. Canagliflozin is not genotoxic based on a battery of in vitro (Ames, mouse lymphoma) and in vivo (micronucleus, Comet) tests [7], [8]. Therefore, the tumors are considered to occur by a non-genetic mechanism.

Basophilic RTTs (adenomas and carcinomas) were noted in both sexes at 100 mg/kg. The 2 RTTs (1 benign, 1 malignant) in males at 30

Conclusion

In conclusion, the carcinogenicity potential of canagliflozin was investigated in a 2-year study in rats. Three types of treatment-related tumors (LCTs, pheochromocytomas, and RTTs) were observed. LCTs were attributed to increased LH, an established mechanism in rats. Pheochromocytomas were attributed primarily to altered calcium homeostasis following increased calcium absorption secondary to carbohydrate malabsorption. RTTs may also have been linked to carbohydrate malabsorption, and

Funding

This work was supported by Janssen Research & Development, LLC and Janssen Research & Development, a Division of Janssen Pharmaceutica NV. The authors are all employees and, as such, designed the studies, participated in data collection and analysis, made the decision to publish, and prepared the manuscript.

Conflict of Interest

The authors declare that there are no conflicts of interest.

Transparency Document

.

Acknowledgements

The authors are grateful to Dr. Richard Sharpe (University of Edinburgh) for advice on the mechanistic studies related to LCTs and Dr. Terry Nett (Colorado State University) for assistance with the mechanistic studies and for the conduct of assays for testosterone and LH. The authors are grateful to Dr. Gordon Hard and Dr. Jerry Hardisty (Experimental Pathology Laboratories) for assistance with microscopic evaluation of the kidneys. The authors thank Verna Hillsamer (Janssen Research &

References (41)

  • J. Rosenstock et al.

    Dose-ranging effects of canagliflozin, a sodium-glucose cotransporter 2 inhibitor, as add-on to metformin in subjects with type 2 diabetes

    Diabetes Care

    (2012)
  • U.S. Food and Drug Administration, Center for Drug Evaluation and Research. Pharmacology/Toxicology Review(s) of...
  • European Medicines Agency, Committee for Medicinal Products for Human Use. Marketing Authorisation Application...
  • J. Gabrielsson et al.

    Chapter 3.7.2: non-compartmental analysis

  • S. Siegel et al.

    Nonparametric Statistics for the Behavioral Sciences (International Edition)

    (1988)
  • R. Peto et al.

    Guidelines for simple, sensitive significance tests for carcinogenic effects in long-term animal experiments

    IARC Monogr. Eval. Carcinog. Risk Chem. Hum. Suppl.

    (1980)
  • L.A. Hothorn et al.

    Proof of hazard and proof of safety in toxicological studies using simultaneous confidence intervals for differences and ratios to control

    J. Biopharm. Stat.

    (2008)
  • G.P. Bailey et al.

    Preclinical development of a pharmaceutical product for children

  • T. Zoetis et al.

    Species comparison of anatomical and functional renal development

    Birth Defects Res. B Dev. Reprod. Toxicol.

    (2003)
  • O.J. Dunn

    Multiple comparisons sing rank sums

    Technometrics

    (1964)
  • Cited by (41)

    • Effectiveness in the inhibition of dapagliflozin and canagliflozin on M-type K<sup>+</sup> current and α-methylglucoside-induced current in pituitary tumor (GH<inf>3</inf>) and pheochromocytoma PC12 cells

      2020, European Journal of Pharmacology
      Citation Excerpt :

      One may speculate that the IK(M) detected in GH3 cells be distinguishable from that existing in different types of endocrine or neuroendocrine cells. An earlier report showed that the administration of CANA, inhibitor of SGLT2, could increase the incidence of pheochromocytomas in rats (De Jonghe et al., 2014). The possible effect of DAPA on IK(M) in pheochromocytoma PC12 cells were thus further investigated.

    • Diabetes and cancer: Pathophysiological fundamentals of a ‘dangerous affair’

      2018, Diabetes Research and Clinical Practice
      Citation Excerpt :

      Canagliflozin was shown to be not genotoxic, and in carcinogenicity studies in mice and dogs no pre-neoplastic lesions were detected. Although a 2-year carcinogenicity study in rats showed a higher incidence of pheochromocytomas, Leydig cells tumors and renal tubule tumors, these were attributed to specific features of metabolism regarding carbohydrate malabsorption and calcium homeostasis in rodents and thus not relevant to humans [94]. In a meta-analysis of 46 randomized controlled studies, SGLT2 inhibitors showed no association with overall cancer (OR 1.14, CI-95% 0.96–1.36), even though the statistical analysis for each type of malignancy suggested that the risk of bladder cancer might be increased (OR 3.87, CI-95% 1.48–10.08), especially with empagliflozin (OR 4.49, CI-95% 1.21–16.73) [95].

    • Renal tubular and adrenal medullary tumors in the 2-year rat study with canagliflozin confirmed to be secondary to carbohydrate (glucose) malabsorption in the 15-month mechanistic rat study

      2017, Chemico-Biological Interactions
      Citation Excerpt :

      In rats at high doses canagliflozin also inhibits the absorption of glucose from the small intestine via the inhibition of SGLT1. Canagliflozin was not genotoxic in a battery of in vitro assays (Ames, mouse lymphoma) and in vivo studies (micronucleus test and comet assay) [1]. In a 2-year rat carcinogenicity study, tumors were observed in the kidney, adrenal glands, and testes [1].

    View all citing articles on Scopus
    View full text