Skip to main content
Top
Published in: BMC Cancer 1/2023

Open Access 01-12-2023 | Hepatocellular Carcinoma | Research

Saroglitazar suppresses the hepatocellular carcinoma induced by intraperitoneal injection of diethylnitrosamine in C57BL/6 mice fed on choline deficient, l-amino acid- defined, high-fat diet

Authors: Suresh R. Giri, Bibhuti Bhoi, Chitrang Trivedi, Akshyaya Rath, Rohan Rathod, Anish Sharma, Ramchandra Ranvir, Shekhar Kadam, Kailash Ingale, Hiren Patel, Abraham Nyska, Mukul R. Jain

Published in: BMC Cancer | Issue 1/2023

Login to get access

Abstract

Background

Saroglitazar is a novel PPAR-α/γ agonist with predominant PPAR-α activity. In various preclinical models, saroglitazar has been shown to prevent & reverse symptoms of NASH. In view of these observations, and the fact that NASH is a progressive disease leading to HCC, we hypothesized that saroglitazar may prevent the development of HCC in rodents.

Methods

HCC was induced in C57BL/6 mice by a single intraperitoneal injection of 25 mg/kg diethylnitrosamine (DEN) at the age of 4 weeks and then feeding the animal a choline-deficient, L-amino acid- defined, high-fat diet (CDAHFD) for the entire study duration. Eight weeks after initiation of CDAHFD, saroglitazar (1 and 3 mg/kg) treatment was started and continued for another 27 weeks.

Results

Saroglitazar treatment significantly reduced the liver injury markers (serum ALT and AST), reversed hepatic steatosis and decreased the levels of pro-inflammatory cytokines like TNF-α in liver. It also resulted in a marked increase in serum adiponectin and osteopontin levels. All disease control animals showed hepatic tumors, which was absent in saroglitazar (3 mg/kg)- treatment group indicating 100% prevention of hepatic tumorigenesis. This is the first study demonstrating a potent PPARα agonist causing suppression of liver tumors in rodents, perhaps due to a strong anti-NASH activity of Saroglitazar that overrides its rodent-specific peroxisome proliferation activity.

Conclusion

The data reveals potential of saroglitazar for chemoprevention of hepatocellular carcinoma in patients with NAFLD/NASH.
Appendix
Available only for authorised users
Literature
3.
go back to reference Burt AD, Lackner C, Tiniakos DG. Diagnosis and assessment of NAFLD: definitions and histopathological classification. Semin Liver Dis. 2015;35(3):207–20.CrossRef Burt AD, Lackner C, Tiniakos DG. Diagnosis and assessment of NAFLD: definitions and histopathological classification. Semin Liver Dis. 2015;35(3):207–20.CrossRef
4.
go back to reference Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications - a review. Nutr J. 2014;13(1):1–10.CrossRef Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications - a review. Nutr J. 2014;13(1):1–10.CrossRef
5.
go back to reference Pai V, Paneerselvam A, Mukhopadhyay S, Bhansali A, Kamath D, Shankar V, et al. A Multicenter , Prospective , Randomized , Double-blind Study to Evaluate the Safety and Efficacy of Saroglitazar 2 and 4 mg Compared to Pioglitazone 45 mg in Diabetic Dyslipidemia ( PRESS V ). J Diabetes Sci Technol. 2014;8(1):132–41.CrossRef Pai V, Paneerselvam A, Mukhopadhyay S, Bhansali A, Kamath D, Shankar V, et al. A Multicenter , Prospective , Randomized , Double-blind Study to Evaluate the Safety and Efficacy of Saroglitazar 2 and 4 mg Compared to Pioglitazone 45 mg in Diabetic Dyslipidemia ( PRESS V ). J Diabetes Sci Technol. 2014;8(1):132–41.CrossRef
6.
go back to reference Kumar DP, Ca R, Marioneaux J, Santhekadur PK, Bhat M, Alonso C, et al. The PPAR α / γ agonist Saroglitazar improves insulin resistance and Steatohepatitis in a diet induced animal model of nonalcoholic fatty liver disease. Sci Rep. 2020;10(1):1–14. Kumar DP, Ca R, Marioneaux J, Santhekadur PK, Bhat M, Alonso C, et al. The PPAR α / γ agonist Saroglitazar improves insulin resistance and Steatohepatitis in a diet induced animal model of nonalcoholic fatty liver disease. Sci Rep. 2020;10(1):1–14.
7.
go back to reference Jain MR, Giri SR, Bhoi B, Trivedi C, Rath A, Rathod R, et al. Dual PPARα/γ agonist saroglitazar improves liver histopathology and biochemistry in experimental NASH models. Liver Int. 2018;38(6):1084–94.CrossRef Jain MR, Giri SR, Bhoi B, Trivedi C, Rath A, Rathod R, et al. Dual PPARα/γ agonist saroglitazar improves liver histopathology and biochemistry in experimental NASH models. Liver Int. 2018;38(6):1084–94.CrossRef
8.
go back to reference Jani RH, Pai V, Jha P, Jariwala G, Mukhopadhyay S, Bhansali A, et al. Study to evaluate the safety and efficacy of Saroglitazar 2 and 4 mg compared with placebo in type 2 diabetes mellitus patients having hypertriglyceridemia not controlled with atorvastatin therapy (Press VI) VI. Press Diabetes Tech Ther. 2014;16(2):1–9. Jani RH, Pai V, Jha P, Jariwala G, Mukhopadhyay S, Bhansali A, et al. Study to evaluate the safety and efficacy of Saroglitazar 2 and 4 mg compared with placebo in type 2 diabetes mellitus patients having hypertriglyceridemia not controlled with atorvastatin therapy (Press VI) VI. Press Diabetes Tech Ther. 2014;16(2):1–9.
10.
go back to reference Samer Gawrieh NPC. LO10: a phase 2, prospective, multicenter, double-blind, randomized study of Saroglitazar magnesium 1 mg, 2 mg or 4 mg versus placebo in patients with nonalcoholic fatty liver disease and/or nonalcoholic Steatohepatitis (evidences IV). Hepatology. 2019;70(6):1477A–501A. Samer Gawrieh NPC. LO10: a phase 2, prospective, multicenter, double-blind, randomized study of Saroglitazar magnesium 1 mg, 2 mg or 4 mg versus placebo in patients with nonalcoholic fatty liver disease and/or nonalcoholic Steatohepatitis (evidences IV). Hepatology. 2019;70(6):1477A–501A.
11.
go back to reference Rubenstrunk A, Hanf R, Hum DW, Fruchart JC, Staels B. Safety issues and prospects for future generations of PPAR modulators. Biochim Biophys Acta - Mol Cell Biol Lipids. 2007;1771(8):1065–81.CrossRef Rubenstrunk A, Hanf R, Hum DW, Fruchart JC, Staels B. Safety issues and prospects for future generations of PPAR modulators. Biochim Biophys Acta - Mol Cell Biol Lipids. 2007;1771(8):1065–81.CrossRef
12.
go back to reference Reddy JK, Chu R. Peroxisome proliferator-induced pleiotropic responses: pursuit of a phenomenon: Annals of the New York Academy of Sciences; 1996. Reddy JK, Chu R. Peroxisome proliferator-induced pleiotropic responses: pursuit of a phenomenon: Annals of the New York Academy of Sciences; 1996.
13.
go back to reference Peters JM, Cattley RC, Gonzalez FJ. Role of PPARα in the mechanism of action of the nongenotoxic carcinogen and peroxisome proliferator Wy-14,643. Carcinogenesis. 1997;18(11):2029–33.CrossRef Peters JM, Cattley RC, Gonzalez FJ. Role of PPARα in the mechanism of action of the nongenotoxic carcinogen and peroxisome proliferator Wy-14,643. Carcinogenesis. 1997;18(11):2029–33.CrossRef
14.
go back to reference Cool B, Zinker B, Chiou W, Kifle L, Cao N, Perham M, et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006;3(6):403–16.CrossRef Cool B, Zinker B, Chiou W, Kifle L, Cao N, Perham M, et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006;3(6):403–16.CrossRef
15.
go back to reference Kittel B, Ruehl-Fehlert C, Morawietz G, Klapwijk J, Elwell MR, Lenz B, et al. Revised guides for organ sampling and trimming in rats and mice - part 2: a joint publication of the RITA and NACAD groups. Exp Toxicol Pathol. 2004;55(6):413–31.CrossRef Kittel B, Ruehl-Fehlert C, Morawietz G, Klapwijk J, Elwell MR, Lenz B, et al. Revised guides for organ sampling and trimming in rats and mice - part 2: a joint publication of the RITA and NACAD groups. Exp Toxicol Pathol. 2004;55(6):413–31.CrossRef
16.
go back to reference Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology. 2005;41(6):1313–21.CrossRef Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology. 2005;41(6):1313–21.CrossRef
17.
go back to reference Thoolen B, Maronpot RR, Harada T, Nyska A, Rousseaux C, Nolte T, et al. Proliferative and nonproliferative lesions of the rat and mouse hepatobiliary system. Toxicol Pathol. 2010;38(7 SUPPL):5–81.CrossRef Thoolen B, Maronpot RR, Harada T, Nyska A, Rousseaux C, Nolte T, et al. Proliferative and nonproliferative lesions of the rat and mouse hepatobiliary system. Toxicol Pathol. 2010;38(7 SUPPL):5–81.CrossRef
19.
go back to reference Carcinoma H, Nakagawa H, Hayata Y, Kawamura S, Yamada T, Fujiwara N. Lipid Metabolic Reprogramming in 2018;10–15. Carcinoma H, Nakagawa H, Hayata Y, Kawamura S, Yamada T, Fujiwara N. Lipid Metabolic Reprogramming in 2018;10–15.
20.
go back to reference Chatterjee S, Majumder A, Ray S. Observational Study of Effects of Saroglitazar on Glycaemic and Lipid Parameters on Indian Patients with Type 2 diabetes. Sci Rep. 2015;5:1–5.CrossRef Chatterjee S, Majumder A, Ray S. Observational Study of Effects of Saroglitazar on Glycaemic and Lipid Parameters on Indian Patients with Type 2 diabetes. Sci Rep. 2015;5:1–5.CrossRef
21.
go back to reference Caffrey R, Marioneaux J, Santhekadur P, Bedossa P, Philip B, Jain M, et al. Saroglitazar Treatment Prevents NASH, Eliminates Hepatocyte Ballooning , and Significantly Improves Serum LFTs , Lipids and Insulin Resistance in DIAMOND ™ Mice Compared to Pioglitazone Benchmark. J Hepatol. 2017;68(1):S578–9. Caffrey R, Marioneaux J, Santhekadur P, Bedossa P, Philip B, Jain M, et al. Saroglitazar Treatment Prevents NASH, Eliminates Hepatocyte Ballooning , and Significantly Improves Serum LFTs , Lipids and Insulin Resistance in DIAMOND ™ Mice Compared to Pioglitazone Benchmark. J Hepatol. 2017;68(1):S578–9.
22.
go back to reference Sklavos A, Poutahidis T, Giakoustidis A, Makedou K, Angelopoulou K, Hardas A, et al. Effects of wnt-1 blockade in DEN-induced hepatocellular adenomas of mice. Oncol Lett. 2018;15(1):1211–9. Sklavos A, Poutahidis T, Giakoustidis A, Makedou K, Angelopoulou K, Hardas A, et al. Effects of wnt-1 blockade in DEN-induced hepatocellular adenomas of mice. Oncol Lett. 2018;15(1):1211–9.
23.
go back to reference Lee JS, Chu IS, Mikaelyan A, Calvisi DF, Heo J, Reddy JK, et al. Application of comparative functional genomics to identify best-fit mouse models to study human cancer. Nat Genet. 2004;36(12):1306–11.CrossRef Lee JS, Chu IS, Mikaelyan A, Calvisi DF, Heo J, Reddy JK, et al. Application of comparative functional genomics to identify best-fit mouse models to study human cancer. Nat Genet. 2004;36(12):1306–11.CrossRef
24.
go back to reference Matsumoto M, Hada N, Sakamaki Y, Uno A, Shiga T, Tanaka C, et al. An improved mouse model that rapidly develops fibrosis in non-alcoholic steatohepatitis. Int J Exp Pathol. 2013;94(2):93–103.CrossRef Matsumoto M, Hada N, Sakamaki Y, Uno A, Shiga T, Tanaka C, et al. An improved mouse model that rapidly develops fibrosis in non-alcoholic steatohepatitis. Int J Exp Pathol. 2013;94(2):93–103.CrossRef
25.
go back to reference Jain MR, Giri SR, Trivedi C, Bhoi B, Rath A, Vanage G, et al. Saroglitazar, a novel PPARα/γ agonist with predominant PPARα activity, shows lipid-lowering and insulin-sensitizing effects in preclinical models. Pharmacol Res Perspect. 2015;3(3 e00136):1–14. Jain MR, Giri SR, Trivedi C, Bhoi B, Rath A, Vanage G, et al. Saroglitazar, a novel PPARα/γ agonist with predominant PPARα activity, shows lipid-lowering and insulin-sensitizing effects in preclinical models. Pharmacol Res Perspect. 2015;3(3 e00136):1–14.
26.
go back to reference Strakova N, Ehrmann J, Bartos J, Malikova J, Dolezel J, Kolar Z. Peroxisome proliferator-activated receptors (PPAR) agonists affect cell viability, apoptosis and expression of cell cycle related proteins in cell lines of glial brain tumors. Neoplasma. 2005;52(2):126–36. Strakova N, Ehrmann J, Bartos J, Malikova J, Dolezel J, Kolar Z. Peroxisome proliferator-activated receptors (PPAR) agonists affect cell viability, apoptosis and expression of cell cycle related proteins in cell lines of glial brain tumors. Neoplasma. 2005;52(2):126–36.
27.
go back to reference Dongiovanni P, Valenti L. Peroxisome proliferator-activated receptor genetic polymorphisms and nonalcoholic fatty liver disease: Any role in disease susceptibility? PPAR Res. 2013;2013:452061.CrossRef Dongiovanni P, Valenti L. Peroxisome proliferator-activated receptor genetic polymorphisms and nonalcoholic fatty liver disease: Any role in disease susceptibility? PPAR Res. 2013;2013:452061.CrossRef
28.
go back to reference Rumi MAK, Ishihara S, Kazumori H, Kadowaki Y, Kinoshita Y. Can PPAR gamma ligands be used in cancer therapy? Curr Med Chem Anticancer Agents. 2004;4(6):465–77.CrossRef Rumi MAK, Ishihara S, Kazumori H, Kadowaki Y, Kinoshita Y. Can PPAR gamma ligands be used in cancer therapy? Curr Med Chem Anticancer Agents. 2004;4(6):465–77.CrossRef
29.
go back to reference Balfour JA, McTavish D, Heel RC. Fenofibrate: a review of its Pharmacodynamic and pharmacokinetic properties and therapeutic use in Dyslipidaemia. Drugs. 1990;40(2):260–90.CrossRef Balfour JA, McTavish D, Heel RC. Fenofibrate: a review of its Pharmacodynamic and pharmacokinetic properties and therapeutic use in Dyslipidaemia. Drugs. 1990;40(2):260–90.CrossRef
30.
go back to reference Peyrou M, Ramadori P, Bourgoin L, Foti M. PPARs in liver diseases and cancer: Epigenetic regulation by microRNAs. PPAR Res. 2012;2012(Article ID 757803):16. Peyrou M, Ramadori P, Bourgoin L, Foti M. PPARs in liver diseases and cancer: Epigenetic regulation by microRNAs. PPAR Res. 2012;2012(Article ID 757803):16.
31.
go back to reference Li X, Liu L, Li R, Wu A, Lu J, Wu Q, et al. Hepatic loss of lissencephaly 1 (lis1) induces fatty liver and accelerates liver tumorigenesis in mice. J Biol Chem. 2018;293(14):5160–71.CrossRef Li X, Liu L, Li R, Wu A, Lu J, Wu Q, et al. Hepatic loss of lissencephaly 1 (lis1) induces fatty liver and accelerates liver tumorigenesis in mice. J Biol Chem. 2018;293(14):5160–71.CrossRef
32.
go back to reference Shimizu M, Yasuda Y, Sakai H, Kubota M, Terakura D, Baba A, et al. Pitavastatin suppresses diethylnitrosamine-induced liver preneoplasms in male C57BL/KsJ-db/db obese mice. BMC Cancer. 2011;11:281.CrossRef Shimizu M, Yasuda Y, Sakai H, Kubota M, Terakura D, Baba A, et al. Pitavastatin suppresses diethylnitrosamine-induced liver preneoplasms in male C57BL/KsJ-db/db obese mice. BMC Cancer. 2011;11:281.CrossRef
33.
go back to reference He C, Fan X, Chen R, Liang B, Cao L, Guo Y, et al. Osteopontin is involved in estrogen-mediated protection against diethylnitrosamine-induced liver injury in mice. Food Chem Toxicol. 2012;50(8):2878–85.CrossRef He C, Fan X, Chen R, Liang B, Cao L, Guo Y, et al. Osteopontin is involved in estrogen-mediated protection against diethylnitrosamine-induced liver injury in mice. Food Chem Toxicol. 2012;50(8):2878–85.CrossRef
34.
go back to reference Fan X, He C, Jing W, Zhou X, Chen R, Cao L, et al. Intracellular osteopontin inhibits toll-like receptor signaling and impedes liver carcinogenesis. Cancer Res. 2015;75(1):86–97.CrossRef Fan X, He C, Jing W, Zhou X, Chen R, Cao L, et al. Intracellular osteopontin inhibits toll-like receptor signaling and impedes liver carcinogenesis. Cancer Res. 2015;75(1):86–97.CrossRef
Metadata
Title
Saroglitazar suppresses the hepatocellular carcinoma induced by intraperitoneal injection of diethylnitrosamine in C57BL/6 mice fed on choline deficient, l-amino acid- defined, high-fat diet
Authors
Suresh R. Giri
Bibhuti Bhoi
Chitrang Trivedi
Akshyaya Rath
Rohan Rathod
Anish Sharma
Ramchandra Ranvir
Shekhar Kadam
Kailash Ingale
Hiren Patel
Abraham Nyska
Mukul R. Jain
Publication date
01-12-2023
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2023
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10530-0

Other articles of this Issue 1/2023

BMC Cancer 1/2023 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine