Pharmacometabolomics Reveals Irinotecan Mechanism of Action in Cancer Patients

The purpose of this study was to identify early circulating metabolite changes implicated in the mechanism of action of irinotecan, a DNA topoisomerase I inhibitor, in cancer patients. A liquid chromatography–tandem mass spectrometry–based targeted metabolomic platform capable of measuring 254 endogenous metabolites was applied to profile circulating metabolites in plasma samples collected pre‐ and post‐irinotecan treatment from 13 cancer patients. To gain further mechanistic insights, metabolic profiling was also performed for the culture medium of human primary hepatocytes (HepatoCells) and 2 cancer cell lines on exposure to SN‐38 (an active metabolite of irinotecan). Intracellular reactive oxygen species (ROS) was detected by dihydroethidium assay. Irinotecan induced a global metabolic change in patient plasma, as represented by elevations of circulating purine/pyrimidine nucleobases, acylcarnitines, and specific amino acid metabolites. The plasma metabolic signature was well replicated in HepatoCells medium on SN‐38 exposure, whereas in cancer cell medium SN‐38 induced accumulation of pyrimidine/purine nucleosides and nucleobases while having no impact on acylcarnitines and amino acid metabolites. SN‐38 induced ROS in HepatoCells, but not in cancer cells. Distinct metabolite signatures of SN‐38 exposure in HepatoCells medium and cancer cell medium revealed different mechanisms of drug action on hepatocytes and cancer cells. Elevations in circulating purine/pyrimidine nucleobases may stem from nucleotide degradation following irinotecan‐induced DNA double‐strand breaks. Accumulations of circulating acylcarnitines and specific amino acid metabolites may reflect, at least in part, irinotecan‐induced mitochondrial dysfunction and oxidative stress in the liver. The plasma metabolic signature of irinotecan exposure provides early insights into irinotecan mechanism of action in patients.

[1]  C. Sander,et al.  Mitochondrial respiratory gene expression is suppressed in many cancers , 2016, eLife.

[2]  E. Sausville,et al.  Phase I Safety, Pharmacokinetic, and Pharmacodynamic Study of the Poly(ADP-ribose) Polymerase (PARP) Inhibitor Veliparib (ABT-888) in Combination with Irinotecan in Patients with Advanced Solid Tumors , 2016, Clinical Cancer Research.

[3]  G. Guo,et al.  Mechanistic review of drug-induced steatohepatitis. , 2015, Toxicology and applied pharmacology.

[4]  M. Uysal,et al.  Effect of rosiglitazone on asymmetric dimethylarginine metabolism in thioacetamide-induced acute liver injury. , 2015, Pathophysiology : the official journal of the International Society for Pathophysiology.

[5]  Martin L. Miller,et al.  Mitochondrial DNA copy number variation across human cancers , 2015, bioRxiv.

[6]  R. Weinshilboum,et al.  Pharmacometabolomics: Implications for Clinical Pharmacology and Systems Pharmacology , 2014, Clinical pharmacology and therapeutics.

[7]  Mitchell R. McGill,et al.  Circulating acylcarnitines as biomarkers of mitochondrial dysfunction after acetaminophen overdose in mice and humans , 2014, Archives of Toxicology.

[8]  D. Harrison,et al.  Methods for detection of mitochondrial and cellular reactive oxygen species. , 2014, Antioxidants & redox signaling.

[9]  D. Palmer,et al.  Chemotherapy induced hepatotoxicity in metastatic colorectal cancer: a review of mechanisms and outcomes. , 2013, Critical reviews in oncology/hematology.

[10]  S. Bhattacharyya,et al.  Acylcarnitine Profiles in Acetaminophen Toxicity in the Mouse: Comparison to Toxicity, Metabolism and Hepatocyte Regeneration , 2013, Metabolites.

[11]  O. Fiehn,et al.  Purine Pathway Implicated in Mechanism of Resistance to Aspirin Therapy: Pharmacometabolomics-Informed-Pharmacogenomics , 2013, Clinical pharmacology and therapeutics.

[12]  Xiangmei Wu,et al.  Oxidative stress induces DNA demethylation and histone acetylation in SH-SY5Y cells: potential epigenetic mechanisms in gene transcription in Aβ production , 2013, Neurobiology of Aging.

[13]  Peter D. Karp,et al.  Metabolomics Reveals Amino Acids Contribute to Variation in Response to Simvastatin Treatment , 2012, PloS one.

[14]  C. Martel,et al.  Mitochondrial Roles and Cytoprotection in Chronic Liver Injury , 2012, Biochemistry research international.

[15]  J. Asara,et al.  A positive/negative ion–switching, targeted mass spectrometry–based metabolomics platform for bodily fluids, cells, and fresh and fixed tissue , 2012, Nature Protocols.

[16]  P. Giriwono,et al.  Fermented Barley Extract Supplementation Maintained Antioxidative Defense Suppressing Lipopolysaccharide-Induced Inflammatory Liver Injury in Rats , 2011, Bioscience, biotechnology, and biochemistry.

[17]  Junxiang Zhang,et al.  Amine metabolomics of hyperglycemic endothelial cells using capillary LC-MS with isobaric tagging. , 2011, Journal of proteome research.

[18]  S. Stürzenbaum,et al.  C. elegans metallothioneins: response to and defence against ROS toxicity. , 2011, Molecular bioSystems.

[19]  B. Fromenty,et al.  Drug-induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and deleterious consequences for the liver. , 2011, Journal of hepatology.

[20]  S. Clarke,et al.  Pharmacometabonomic Profiling as a Predictor of Toxicity in Patients with Inoperable Colorectal Cancer Treated with Capecitabine , 2011, Clinical Cancer Research.

[21]  F. Wright,et al.  Use of Pharmaco‐Metabonomics for Early Prediction of Acetaminophen‐Induced Hepatotoxicity in Humans , 2010, Clinical pharmacology and therapeutics.

[22]  S-D Kim,et al.  An Integrative Approach for Identifying a Metabolic Phenotype Predictive of Individualized Pharmacokinetics of Tacrolimus , 2010, Clinical pharmacology and therapeutics.

[23]  G. Van den Berghe,et al.  The effect of rosiglitazone on asymmetric dimethylarginine (ADMA) in critically ill patients. , 2009, Pharmacological research.

[24]  A. Zhu,et al.  Hepatic toxicities associated with the use of preoperative systemic therapy in patients with metastatic colorectal adenocarcinoma to the liver. , 2009, The oncologist.

[25]  John C Lindon,et al.  Pharmacometabonomic identification of a significant host-microbiome metabolic interaction affecting human drug metabolism , 2009, Proceedings of the National Academy of Sciences.

[26]  G. Labbe,et al.  Drug‐induced liver injury through mitochondrial dysfunction: mechanisms and detection during preclinical safety studies , 2008, Fundamental & clinical pharmacology.

[27]  C. Punt,et al.  Capecitabine and irinotecan as first-line treatment of advanced colorectal cancer. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  V. Monnier,et al.  Aging, Diabetes, and Renal Failure Catalyze the Oxidation of Lysyl Residues to 2‐Aminoadipic Acid in Human Skin Collagen , 2008, Annals of the New York Academy of Sciences.

[29]  J. Sastre,et al.  Mitochondrial involvement in non-alcoholic steatohepatitis. , 2008, Molecular aspects of medicine.

[30]  R. Weinshilboum,et al.  Metabolomics: a global biochemical approach to drug response and disease. , 2008, Annual review of pharmacology and toxicology.

[31]  V. Monnier,et al.  2-aminoadipic acid is a marker of protein carbonyl oxidation in the aging human skin: effects of diabetes, renal failure and sepsis. , 2007, The Biochemical journal.

[32]  Roger Williams,et al.  Inflammation is an important determinant of levels of the endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA) in acute liver failure , 2007, Liver transplantation : official publication of the American Association for the Study of Liver Diseases and the International Liver Transplantation Society.

[33]  Roger Williams,et al.  Increasing dimethylarginine levels are associated with adverse clinical outcome in severe alcoholic hepatitis , 2007, Hepatology.

[34]  T. Pawlik,et al.  Chemotherapy regimen predicts steatohepatitis and an increase in 90-day mortality after surgery for hepatic colorectal metastases. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[35]  Xiao-hong Zhang,et al.  Lysophosphatidylcholine-induced elevation of asymmetric dimethylarginine level by the NADPH oxidase pathway in endothelial cells. , 2006, Vascular pharmacology.

[36]  D. Pessayre,et al.  Mitochondrial dysfunction in NASH: causes, consequences and possible means to prevent it. , 2006, Mitochondrion.

[37]  P. Vallance,et al.  Cardiovascular Biology of the Asymmetric Dimethylarginine:Dimethylarginine Dimethylaminohydrolase Pathway , 2004, Arteriosclerosis, thrombosis, and vascular biology.

[38]  D. Pessayre,et al.  The ins and outs of mitochondrial dysfunction in NASH. , 2004, Diabetes & metabolism.

[39]  D. Fitzpatrick,et al.  Tandem mass spectrometric determination of malonylcarnitine: diagnosis and neonatal screening of malonyl-CoA decarboxylase deficiency. , 2003, Clinical chemistry.

[40]  W. MacNee,et al.  Histone acetylation regulates epithelial IL-8 release mediated by oxidative stress from environmental particles. , 2003, American journal of physiology. Lung cellular and molecular physiology.

[41]  I. Adcock,et al.  The effect of oxidative stress on histone acetylation and IL-8 release. , 2003, Biochemical and biophysical research communications.

[42]  R. Nijveldt,et al.  The liver is an important organ in the metabolism of asymmetrical dimethylarginine (ADMA). , 2003, Clinical nutrition.

[43]  P. Vallance,et al.  S-nitrosylation of dimethylarginine dimethylaminohydrolase regulates enzyme activity: Further interactions between nitric oxide synthase and dimethylarginine dimethylaminohydrolase , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[44]  Dagmar Ringe,et al.  Human cystathionine beta-synthase is a heme sensor protein. Evidence that the redox sensor is heme and not the vicinal cysteines in the CXXC motif seen in the crystal structure of the truncated enzyme. , 2002, Biochemistry.

[45]  J. Verweij,et al.  Pharmacology of topoisomerase I inhibitors irinotecan (CPT-11) and topotecan. , 2002, Current cancer drug targets.

[46]  Pamela A. Silver,et al.  State of the Arg Protein Methylation at Arginine Comes of Age , 2001, Cell.

[47]  Janet Stone,et al.  Impaired fatty acid oxidation in propofol infusion syndrome , 2001, The Lancet.

[48]  E. Stadtman,et al.  Glutamic and aminoadipic semialdehydes are the main carbonyl products of metal-catalyzed oxidation of proteins. , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[49]  L. Saltz,et al.  Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. Irinotecan Study Group. , 2000, The New England journal of medicine.

[50]  E. Mosharov,et al.  The quantitatively important relationship between homocysteine metabolism and glutathione synthesis by the transsulfuration pathway and its regulation by redox changes. , 2000, Biochemistry.

[51]  R. Banerjee,et al.  Evidence for Heme-mediated Redox Regulation of Human Cystathionine β-Synthase Activity* , 1998, The Journal of Biological Chemistry.

[52]  Leroy F. Liu,et al.  Mechanism of Action of Camptothecin , 1996, Annals of the New York Academy of Sciences.

[53]  J. Barankiewicz,et al.  Metabolic consequences of DNA damage: alteration in purine metabolism following poly(ADP ribosyl)ation in human T-lymphoblasts. , 1987, Archives of biochemistry and biophysics.

[54]  M. Kimoto,et al.  Metabolism of NG,NG-and NG,N'G-dimethylarginine in rats. , 1987, Archives of biochemistry and biophysics.

[55]  H. Wolburg,et al.  Propofol Related Infusion Syndrome: Ultrastructural Evidence for a Mitochondrial Disorder , 2018, Critical care medicine.

[56]  R. Banerjee,et al.  Redox regulation and reaction mechanism of human cystathionine-beta-synthase: a PLP-dependent hemesensor protein. , 2005, Archives of biochemistry and biophysics.

[57]  Piero Rinaldo,et al.  Fatty acid oxidation disorders. , 2002, Annual review of physiology.

[58]  D. Pessayre,et al.  Inhibition of mitochondrial beta-oxidation as a mechanism of hepatotoxicity. , 1995, Pharmacology & therapeutics.

[59]  J. Weete Fatty Acid Metabolism , 1980 .