Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Toxicol Mech Methods ; 34(5): 572-583, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38390772

ABSTRACT

Irinotecan-induced severe diarrhea (IISD) not only limits irinotecan's application but also significantly affects patients' quality of life. However, existing animal models often inadequately represent the dynamics of IISD development, progression, and resolution across multiple chemotherapy cycles, yielding non-reproducible and highly variable response with limited clinical translation. Our studies aim to establish a reproducible and validated IISD model that better mimics the pathophysiology progression observed in patients, enhancing translational potential. We investigated the impact of dosing regimens (including different dose, infusion time, and two cycles of irinotecan administration), sex, age, tumor-bearing conditions, and irinotecan formulation on the IISD incidence and severity in mice and rats. Lastly, we investigated above factors' impact on pharmacokinetics of irinotecan, intestinal injury, and carboxylesterase activities. In summary, we successfully established a standard model establishment procedure for an optimized IISD model with highly reproducible severe diarrhea incidence rate (100%) and a low mortality rate (11%) in F344 rats. Additionally, the rats tolerated at least two cycles of irinotecan chemotherapy treatment. In contrast, the mouse model exhibited suboptimal IISD incidence rates (60%) and an extremely high mortality rate (100%). Notably, dosing regimen, age and tumor-bearing conditions of animals emerged as critical factors in IISD model establishment. In conclusion, our rat IISD model proves superior in mimicking pathophysiology progression and characteristics of IISD in patients, which stands as an effective tool for mechanism and efficacy studies in future chemotherapy-induced gut toxicity research.


Subject(s)
Diarrhea , Disease Models, Animal , Irinotecan , Rats, Inbred F344 , Irinotecan/toxicity , Animals , Diarrhea/chemically induced , Male , Female , Mice , Rats , Severity of Illness Index , Dose-Response Relationship, Drug , Humans , Reproducibility of Results
2.
Sci Rep ; 13(1): 6528, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37085612

ABSTRACT

Irinotecan is a topoisomerase I inhibitor which has been widely used to combat several solid tumors, whereas irinotecan therapy can induce liver injury. Liver injury generally leads to tissue hypoxia, and hypoxia-inducible factor-1α (HIF-1α), a pivotal transcription factor, mediates adaptive pathophysiological responses to lower oxygen condition. Previous studies have reported a relationship between HIF-1α and autophagy, and autophagy impairment is a common characteristic in a variety of diseases. Here, irinotecan (50 mg/kg) was employed on mice, and HepG2 and L-02 cells were cultured with irinotecan (10, 20 and 40 µM). In vivo study, we found that irinotecan treatment increased final liver index, serum aminotransferase level and hepatic lipid accumulation. Impaired autophagic flux and activation of HIF-1α/BNIP3 pathway were also demonstrated in the liver of irinotecan-treated mice. Moreover, irinotecan treatment significantly deteriorated hepatic oxidative stress, evidenced by increased MDA and ROS contents, as well as decreased GSH-Px, SOD and CAT contents. Interestingly, protein levels of NLRP3, cleaved-caspase 1 and IL-1ß were enhanced in the liver of mice injected with irinotecan. In vitro study, irinotecan-treated HepG2 and L-02 cells also showed impaired autophagic flux, while HIF-1α inhibition efficaciously removed the accumulated autophagosomes induced by irinotecan. Additionally, irinotecan treatment aggravated lipid accumulation in HepG2 and L-02 cells, and HIF-1α inhibition reversed the effect of irinotecan. Furthermore, HIF-1α inhibition weakened irinotecan-induced NLRP3 inflammasome activation in HepG2 cells. Taken together, our results suggest that irinotecan induces liver injury by orchestrating autophagy via HIF-1α/BNIP3 pathway, and HIF-1α inhibition could alleviate irinotecan-induced lipid accumulation in HepG2 and L-02 cells, which will provide a new clue and direction for the prevention of side effects of clinical chemotherapy drugs.


Subject(s)
Autophagy , Chemical and Drug Induced Liver Injury, Chronic , Hypoxia-Inducible Factor 1, alpha Subunit , Lipid Metabolism , Animals , Mice , Autophagy/genetics , Chemical and Drug Induced Liver Injury, Chronic/genetics , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Irinotecan/toxicity , Lipid Metabolism/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein
3.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36834959

ABSTRACT

Cancer treatment evokes impediments to liver metabolism that culminate in fatty liver. This study determined hepatic fatty acid composition and expression of genes and mediators involved in lipid metabolism following chemotherapy treatment. Female rats bearing the Ward colon tumor were administered Irinotecan (CPT-11) +5-fluorouracil (5-FU) and maintained on a control diet or a diet containing eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA) (2.3 g/100 g fish oil). Healthy animals provided with a control diet served as a reference group. Livers were collected one week after chemotherapy. Triacylglycerol (TG), phospholipid (PL), ten lipid metabolism genes, leptin, and IL-4 were measured. Chemotherapy increased TG content and reduced EPA content in the liver. Expression of SCD1 was upregulated by chemotherapy, while dietary fish oil downregulated its expression. Dietary fish oil down-regulated expression of the fatty acid synthesis gene FASN, while restoring the long chain fatty acid converting genes FADS2 and ELOVL2, and genes involved in mitochondrial ß-oxidation (CPT1α) and lipid transport (MTTP1), to values similar to reference animals. Neither leptin nor IL-4 were affected by chemotherapy or diet. Depletion of EPA is associated with pathways evoking enhanced TG accumulation in the liver. Restoring EPA through diet may pose a dietary strategy to attenuate chemotherapy-associated impediments in liver fatty acid metabolism.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Eicosapentaenoic Acid , Fish Oils , Neoplasms , Stearoyl-CoA Desaturase , Animals , Female , Rats , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/metabolism , Fatty Acids/metabolism , Fish Oils/pharmacology , Interleukin-4/metabolism , Leptin/metabolism , Liver/metabolism , Neoplasms/metabolism , Stearoyl-CoA Desaturase/metabolism , Triglycerides/metabolism , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Fatty Liver/chemically induced , Fatty Liver/metabolism , Irinotecan/adverse effects , Irinotecan/toxicity , Fluorouracil/adverse effects , Fluorouracil/toxicity
4.
Immunol Invest ; 51(6): 1756-1771, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35152824

ABSTRACT

Cancer chemotherapy and radiotherapy may result in mucositis characterized by stem cell damage and inflammation in the gastrointestinal tract. The molecular mechanisms underlying this pathology remain unknown. Based on the assumption that mitochondrial CPG-DNA (mtDNA) released and sensed by TLR9 could underlie mucositis pathology, we analyzed the mtDNA levels in sera as well as inflammatory and disease parameters in the small intestine from wild-type (WT) and TLR9-deficient mice (TLR9-/-) in an experimental model of intestinal mucositis induced by irinotecan. Additionally, we verified the ability of WT and TLR9-/- macrophages to respond to CpG-DNA in vitro. WT mice injected with irinotecan presented a progressive increase in mtDNA in the serum along with increased hematocrit, shortening of small intestine length, reduction of intestinal villus:crypt ratio and increased influx of neutrophils, which were followed by higher expression of Nlrp3 and Casp1 mRNA and increased IL-1ß levels in the ileum when compared to vehicle-injected mice. TLR9-deficient mice were protected in all these parameters when compared to WT mice. Furthermore, TLR9 was required for the production of IL-1ß and NO after macrophage stimulation with CpG-DNA. Overall, our findings show that the amount of circulating free CpG-DNA is increased upon chemotherapy and that TLR9 activation is important for NLRP3 inflammasome transcription and further IL-1ß release, playing a central role in the development of irinotecan-induced intestinal mucositis. We suggest that TLR9 antagonism may be a new therapeutic strategy for limiting irinotecan-induced intestinal inflammation.


Subject(s)
Mucositis , Animals , DNA, Mitochondrial/genetics , Inflammation/metabolism , Irinotecan/toxicity , Ligands , Mice , Mice, Knockout , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
5.
Drug Metab Dispos ; 50(1): 33-42, 2022 01.
Article in English | MEDLINE | ID: mdl-34697081

ABSTRACT

The human UDP-glucuronosyltransferases (UGTs) represent an important family of drug-metabolizing enzymes, with UGT1A1 targeting the conjugation and detoxification of many exogenous substances, including pharmaceutical drugs. In this study we generated humanized UGT1A1 mice expressing the human UGT1A1 gene in either liver (hUGT1A1HEP ) or intestine (hUGT1A1GI ), enabling experiments to examine tissue-specific properties of UGT1A1-specific glucuronidation. Hepatic and intestinal tissue-specific expression and function of UGT1A1 were demonstrated. Although the liver is considered a major organ for detoxification, intestinal UGT1A1 is an important contributor for drug clearance. Mice were challenged with irinotecan (CPT-11), a prodrug hydrolyzed by carboxylesterases to form the active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38) and detoxified by UGT1A1. Humanized UGT1A1HEP mice that have no intestinal UGT1A1 displayed a greater lethality rate when exposed to CPT-11 than hUGT1A1GI mice. When exposed to a low dose of CPT-11 (10 mg/kg), hUGT1A1HEP mice displayed greater intestinal inflammatory (IL-1ß and IL-6) insult in addition to p53-triggered apoptotic responses. In vitro studies with intestinal crypt organoids exposed to CPT-11 confirmed the results observed in vivo and indicated that CPT-11 impacts stemness, apoptosis, and endoplasmic reticulum (ER) stress in organoids deficient in UGT1A1. When we examined the induction of ER stress in organoids with thapsigargin, an inhibitor of sarco/endoplasmic reticulum Ca2+ ATPase, apoptosis and the caspase surge that occurred in hUGT1A1HEP mice were blocked in hUGT1A1GI organoids. This study reveals the importance of intestinal UGT1A1 in preventing inflammation, apoptosis, and loss of stemness capacity upon systemic challenge with an important chemotherapeutic agent. SIGNIFICANCE STATEMENT: Hepatic and intestinal UGT1A1 play a key role in the metabolism and detoxification of endogenous and exogenous compounds. The use of tissue-specific humanized models expressing UGT1A1 in liver or intestine has confirmed the relevance of the intestinal tract in the detoxification of irinotecan. Mechanistic studies using intestinal organoids highlighted the importance of UGT1A1 in reducing inflammation, apoptosis, and loss of stemness. These new models provide valuable tools for studying tissue-specific glucuronidation of substances that are metabolized by human UGT1A1.


Subject(s)
Glucuronosyltransferase/metabolism , Intestines/metabolism , Irinotecan/toxicity , Animals , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Enteritis/chemically induced , Enteritis/pathology , Glucuronosyltransferase/genetics , Humans , Intestines/enzymology , Intestines/pathology , Liver/enzymology , Male , Mice , Mice, Transgenic , Microsomes, Liver , Stem Cells
6.
Arch Toxicol ; 96(2): 499-510, 2022 02.
Article in English | MEDLINE | ID: mdl-34654938

ABSTRACT

The small intestine plays a critical role in the absorption and metabolism of orally administered drugs. Therefore, a model capable of evaluating drug absorption and metabolism in the small intestine would be useful for drug discovery. Patients with genotype UGT1A1*6 (exon 1, 211G > A) treated with the antineoplastic drug SN-38 have been reported to exhibit decreased glucuronide conjugation and increased incidence of intestinal toxicity and its severe side effects, including severe diarrhea. To ensure the safety of drugs, we must develop a drug metabolism and toxicity evaluation model which considers UGT1A1*6. In this study, we generated CYP3A4·POR·UGT1A1 KI- and CYP3A4·POR·UGT1A1*6 KI-Caco-2 cells for pharmaceutical research using a PITCh system. The CYP3A4·POR·UGT1A1 KI-Caco-2 cells were shown to express functional CYP3A4 and UGT1A1. The CYP3A4·POR·UGT1A1*6 KI-Caco-2 cells were sensitive to SN-38-induced intestinal toxicity. We thus succeeded in generating CYP3A4·POR·UGT1A1 KI- and CYP3A4·POR·UGT1A1*6 KI-Caco-2 cells, which can be used in pharmaceutical research. We also developed an intestinal epithelial cell model of patients with UGT1A1*6 and showed that it was useful as a tool for drug discovery.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Glucuronosyltransferase/genetics , Intestinal Mucosa/enzymology , Intestine, Small/enzymology , Antineoplastic Agents/toxicity , Caco-2 Cells/enzymology , Drug Discovery/methods , Genotype , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestine, Small/cytology , Intestine, Small/drug effects , Irinotecan/toxicity
7.
Chem Biol Interact ; 351: 109712, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34699766

ABSTRACT

Irinotecan (CPT-11) is one of the main agents used to treat colorectal cancer; unfortunately, it is associated with increased intestinal mucositis developing. Luteolin has been shown to prevent damage induced by this chemotherapeutic in mice; thus, in this research, we have investigated luteolin's action mechanism in human intestinal epithelial cells. The potential of luteolin in reducing inflammation and oxidative stress induced by irinotecan in Caco-2 cells was evaluated by PCR through mRNA expression of inflammatory and oxidative genes and by ELISA at the protein level. To assess whether luteolin's ability to control irinotecan-induced damage occurs in a PPARγ dependent manner, experiments were performed on PPARγ downregulated cells. Irinotecan downregulated PPARγ expression and upregulated inflammatory and oxidative genes, while luteolin upregulated PPARγ, HO-1, SOD and decreased expression of IL-1ß and iNOS. Interestingly, when the cells were co-stimulated with luteolin and irinotecan, the flavonoid reversed the inflammation and oxidative imbalance evoked by the chemotherapeutic. However, when these experiments were performed in cells downregulated for PPARγ, luteolin lost the capacity to increase PPARγ and reverse the effect of irinotecan in all tested genes, except by IL-1ß. The present study showed that the protective effect of luteolin against irinotecan is PPARγ dependent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Irinotecan/toxicity , Luteolin/pharmacology , PPAR gamma/metabolism , Caco-2 Cells , Down-Regulation/drug effects , Heme Oxygenase-1/metabolism , Humans , Interleukin-1beta/metabolism , Nitric Oxide Synthase Type II/metabolism , Superoxide Dismutase/metabolism , Up-Regulation/drug effects
8.
Eur Rev Med Pharmacol Sci ; 25(23): 7440-7448, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34919246

ABSTRACT

OBJECTIVE: Irinotecan (IR/CPT-11) is a semisynthetic, water-soluble derivative of the alkaloid camptothecin. It is a topoisomerase I group antineoplastic drug commonly used for the treatment of many cancer types, although it has side effects in tissues such as the testis. Curcumin (CRC) is a polyphenol compound produced from the Indian saffron root; it is used as food colouring and food flavouring. This study examined the testis-specific side effects of IR and the ability of CRC to protect against these side effects. MATERIALS AND METHODS: Forty male Sprague-Dawley rats were used in our study (n = 10). The rats were randomly divided into the following four groups: control, IR, IR + CRC, and CRC. IR 10 mg/kg/day was administered intraperitoneally and CRC 100 mg/kg was administered orally. Blood and testicular samples were collected from rats in all four groups on day 30 after drug administration. Histological, biochemical, and spermatological analyses were conducted. RESULTS: Testis tissue and blood samples were collected from the four groups. Tissue samples from the control and CRC groups demonstrated normal histological appearance on light microscopy. The IR group exhibited the following findings: vascular congestion in the tunica albuginea layer; tubular degeneration and vascular congestion in the interstitial area; oedema, vacuolisation, and luminised cells in the seminiferous tubule; and cells that temporarily stopped dividing at any stage of division in the seminiferous tubule epithelium. In the IR+CRC group, histopathological damage was significantly reduced by CRC treatment. Biochemical analysis showed that the level of thiobarbituric acid reactive substance (TBARS) was significantly increased in the IR group, compared with the other groups. CRC treatment significantly decreased this IR-mediated increase in TBARS level, and the TBARS level in the IR + CRC group approached the level observed in the control group. IR treatment caused significant decreases in glutathione (GSH), superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) levels. However, CRC administration tended to ameliorate the decreases in GSH, SOD, CAT, and GPx levels. CONCLUSIONS: In this study, IR had some toxic effects in rat testis tissue; these effects were ameliorated by CRC treatment. Further studies are warranted to confirm our results.


Subject(s)
Curcumin/pharmacology , Irinotecan/toxicity , Protective Agents/pharmacology , Testis/drug effects , Animals , Male , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism , Topoisomerase I Inhibitors/toxicity
9.
Chem Biol Interact ; 349: 109672, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34560068

ABSTRACT

Homogentisic acid (HGA) is the most abundant phenolic compound in strawberry tree (Arbutus unedo L.) honey and an intermediate in the metabolism of phenylalanine and tyrosine. Since HGA exerts its dual nature (pro-oxidant and antioxidant), which depends on the concentration and cell type, the aim of study was to determine whether HGA possess cytoprotective effects and could counteract the cyto- and genotoxic effects of the antineoplastic drug irinotecan (IRI). Tested concentrations corresponded to HGA content in average daily dose of strawberry tree honey as well as five- and ten-fold higher concentrations. Cyto- and genoprotective effects were tested on human peripheral blood lymphocytes using chromosomal aberrations assay and cytokinesis-block micronucleus cytome assay. HGA, even at concentrations 10-fold higher than the one present in the daily amount of consumed strawberry tree honey, posed a non-significant cytotoxic threat to lymphocytes, had a negligible potential for causing cytogenetic damage in treated cells, and did not significantly impair their proliferation. Results of the chromosomal aberration assay and CBMN Cyt assay also showed that HGA efficiently counteracted the detrimental cytogenetic effects of IRI in vitro. The finding on cyto- and genoprotective effects of HGA merits further research in order to better explain the safety profile of this compound and to assess its potency for the development of novel nutraceutical products.


Subject(s)
Homogentisic Acid/pharmacology , Irinotecan/toxicity , Lymphocytes/drug effects , Topoisomerase I Inhibitors/toxicity , Chromosome Aberrations , Humans , In Vitro Techniques
10.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34360955

ABSTRACT

Novel nontoxic derivatives of SN38 with favorable antineoplastic properties were characterized in water solution using NMR. The phenomena observed by NMR were linked to basic pharmacological properties, such as solubility, bioavailability, chemical and stereochemical stability, and binding to natural DNA oligomers through the terminal G-C base pair, which is commonly considered a biological target of Topo I inhibitors. Compound 1, with bulky substituents at both C5(R) and C20(S) on the same side of a camptothecin core, manifests self-association, whereas diastereomers 2, with bulky C5(S) and C20(S) substituents are mostly monomeric in solution. The stereogenic center at C5 is stable in water solution at pH 5-6. The compound with an (N-azetidinyl)methyl substituent at C9 can undergo the retro Mannich reaction after a prolonged time in water solution. Both diastereomers exhibit different abilities in terms of binding to DNA oligomers: compound 1 is strongly bound, whereas the binding of compound 2 is rather weak. Molecular modeling produced results consistent with NMR experiments. These complementary data allow linking of the observed phenomena in NMR experiments to basic preliminary information on the pharmacodynamic character of compounds and are essential for planning further development research.


Subject(s)
Antineoplastic Agents/chemistry , DNA/chemistry , Irinotecan/analogs & derivatives , Molecular Docking Simulation , Topoisomerase I Inhibitors/chemistry , Antineoplastic Agents/toxicity , Irinotecan/toxicity , Topoisomerase I Inhibitors/toxicity
11.
Br J Pharmacol ; 178(20): 4193-4209, 2021 10.
Article in English | MEDLINE | ID: mdl-34216140

ABSTRACT

BACKGROUND AND PURPOSE: Severe diarrhoea, a common gastrointestinal manifestation of anticancer treatment with irinotecan, might involve single nucleotide polymorphisms (SNPs) of toll-like receptors (TLRs), described as critical bacterial sensors in the gut. Here, colorectal cancer patients carrying missense TLR4 A896G (rs4986790) or C1,196T (rs4986791) SNPs and Tlr4 knockout (Tlr4-/-) mice were given irinotecan to investigate the severity of the induced diarrhoea. EXPERIMENTAL APPROACH: Forty-six patients treated with irinotecan-based regimens had diarrhoea severity analysed according to TLR4 genotypes. In the experimental setting, wild-type (WT) or Tlr4-/- mice were given irinotecan (45 or 75 mg·kg-1 , i.p.) or saline (3 ml·kg-1 ). Diarrhoea severity was evaluated by measuring intestinal injury and inflammatory markers expression after animals were killed. KEY RESULTS: All patients with TLR4 SNPs chemotherapy-treated presented diarrhoea, whereas gastrointestinal toxicity was observed in 50% of the wild homozygous individuals. Mice injected with irinotecan presented systemic bacterial translocation and increased TLR4 immunostaining in the intestine. In line with the clinical findings, Tlr4 gene deficiency enhanced irinotecan-related diarrhoea and TLR9 expression in mice. An increased myeloperoxidase activity and Il-18 expression along with IL-10 decreased production in Tlr4-/- mice also indicated an intensified intestinal damage and inflammatory response. CONCLUSION AND IMPLICATIONS: TLR4 deficiency upregulates TLR9 expression and enhances intestinal damage and the severity of late-onset diarrhoea during irinotecan-based treatment. Identifying patients genetically predisposed to chemotherapy-associated diarrhoea is a strategy toward precision medicine.


Subject(s)
Diarrhea , Irinotecan , Mucositis , Toll-Like Receptor 4 , Toll-Like Receptor 9 , Animals , Diarrhea/chemically induced , Diarrhea/genetics , Humans , Irinotecan/toxicity , Mice , Mucositis/chemically induced , Mucositis/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics
12.
Drug Metab Dispos ; 49(8): 683-693, 2021 08.
Article in English | MEDLINE | ID: mdl-34074730

ABSTRACT

The anticancer drug irinotecan shows serious dose-limiting gastrointestinal toxicity regardless of intravenous dosing. Although enzymes and transporters involved in irinotecan disposition are known, quantitative contributions of these mechanisms in complex in vivo disposition of irinotecan are poorly understood. We explained intestinal disposition and toxicity of irinotecan by integrating 1) in vitro metabolism and transport data of irinotecan and its metabolites, 2) ex vivo gut microbial activation of the toxic metabolite SN-38, and 3) the tissue protein abundance data of enzymes and transporters relevant to irinotecan and its metabolites. Integration of in vitro kinetics data with the tissue enzyme and transporter abundance predicted that carboxylesterase (CES)-mediated hydrolysis of irinotecan is the rate-limiting process in the liver, where the toxic metabolite formed is rapidly deactivated by glucuronidation. In contrast, the poor SN-38 glucuronidation rate as compared with its efficient formation by CES2 in the enterocytes is the key mechanism of the intestinal accumulation of the toxic metabolite. The biliary efflux and organic anion transporting polypeptide-2B1-mediated enterocyte uptake can also synergize buildup of SN-38 in the enterocytes, whereas intestinal P-glycoprotein likely facilitates SN-38 detoxification in the enterocytes. The higher SN-38 concentration in the intestine can be further nourished by ß-d-glucuronidases. Understanding the quantitative significance of the key metabolism and transport processes of irinotecan and its metabolites can be leveraged to alleviate its intestinal side effects. Further, the proteomics-informed quantitative approach to determine intracellular disposition can be extended to determine susceptibility of cancer cells over normal cells for precision irinotecan therapy. SIGNIFICANCE STATEMENT: This work provides a deeper insight into the quantitative relevance of irinotecan hydrolysis (activation), conjugation (deactivation), and deconjugation (reactivation) by human or gut microbial enzymes or transporters. The results of this study explain the characteristic intestinal exposure and toxicity of irinotecan. The quantitative tissue-specific in vitro to in vivo extrapolation approach presented in this study can be extended to cancer cells.


Subject(s)
Gastrointestinal Microbiome/drug effects , Hepatobiliary Elimination , Inactivation, Metabolic/drug effects , Irinotecan , Organic Anion Transporters/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Carboxylesterase/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glucuronidase/metabolism , Hepatobiliary Elimination/drug effects , Hepatobiliary Elimination/physiology , Humans , Irinotecan/analogs & derivatives , Irinotecan/pharmacokinetics , Irinotecan/toxicity , Liver/enzymology , Topoisomerase I Inhibitors/pharmacokinetics , Topoisomerase I Inhibitors/toxicity
13.
Arch Oral Biol ; 126: 105135, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33930649

ABSTRACT

OBJECTIVE: To investigate the ability of a green tea extract and epigallocatechin-3-gallate (EGCG) to protect oral epithelial cells against the deleterious effects of the chemotherapeutic agent irinotecan, with respect to cytotoxicity; reactive oxygen species (ROS) generation; cytokine and matrix metalloproteinase (MMP) production; and cell proliferation and migration. METHODS: The B11 oral keratinocyte and GMSM-K oral epithelial cell lines were used in this study. Cell viability was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay. A fluorometric assay was used to quantify ROS production. Cell proliferation was assessed using a fluorescent cell tracker dye, while a migration assay kit was used to monitor cell migration. Cytokine and MMP secretion was quantified by an enzyme-linked immunosorbent assay. RESULTS: The green tea extract and EGCG reduced the cytotoxicity of irinotecan toward oral keratinocyte and epithelial cell lines. Irinotecan-induced intracellular ROS generation by oral keratinocytes was reduced by the green tea extract and EGCG. Irinotecan negatively affected the proliferation and migration of oral keratinocytes in a dose-dependent manner. However, these effects were not neutralized by the green tea extract, while EGCG showed a trend to attenuate the irinotecan-induced decrease in cell migration. The green tea extract and EGCG also had a dose-dependent inhibitory effect on irinotecan-induced secretion of interleukin-6 and interleukin-8 by oral epithelial cells. Lastly, the irinotecan-induced decrease in the secretion of MMP-2 and MMP-9 by oral epithelial cells was partially restored by the green tea extract and EGCG. CONCLUSIONS: The green tea extract and EGCG, through anti-cytotoxic, anti-oxidative, and anti-inflammatory properties, may protect the oral mucosa against the deleterious effects of the chemotherapeutic agent irinotecan and may be of interest for treating oral mucositis.


Subject(s)
Catechin , Tea , Catechin/analogs & derivatives , Catechin/pharmacology , Epithelial Cells , Irinotecan/toxicity , Plant Extracts/pharmacology
14.
J Ethnopharmacol ; 266: 113421, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33022337

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Irinotecan (CPT-11) is a valuable chemotherapeutic compound, but its use is associated with severe diarrhea in some patients. The CPT-11 prodrug is converted into the active 7-ethyl-10-hydroxycamptothecin (SN-38) metabolite, which can then be retained for extended periods in the intestine, leading to the onset of diarrhea and related symptoms. Banxia Xiexin Decoction (BXD) is commonly employed for the treatment of gastroenteritis in traditional Chinese medicine (TCM), and in clinical settings, it is used to prevent diarrhea in patients undergoing CPT-11 treatment. To date, however, there have been no studies specifically examining which components of BXD can alleviate the gastrointestinal symptoms associated with CPT-11 administration. AIM: This study aimed to identify the main herbal components of BXD associated with protection against CPT-11-induced intestinal toxicity in a murine model system. MATERIALS AND METHODS: SN-38 levels were measured by UPLC-ESI-MS/MS in samples collected from mice subjected to CPT-11-induced diarrhea that had been administered BXD or different components thereof. Pearson correlation and Grey relational analyses were then used to explore spectrum-effect relationships between reductions in intestinal SN-38 levels and specific chemical fingerprints in samples from mice administered particular combinations of BXD component herbs. RESULTS: We found that different herbal combinations were associated with significant differences in intestinal SN-38 reductions in treated mice. Our spectrum-effect analysis revealed that BXD components including chrysin 6-C-arabinoside-8-C-glucoside, coptisine, hydroxyl oroxylin A 7-O-glucuronide (hydroxyl wogonoside), baicalin, an isomer of 5,6,7-trihydroxyl-flavanone-7-O-glucuronide, berberine, palmatine, and chrysin-7-O-glucuronide were all directly linked with reductions in intestinal SN-38 levels. We therefore speculate that these compounds are the primary bioactive components of BXD, suggesting that they offer protection against CPT-11-induced diarrhea. CONCLUSION: By utilizing UPLC to analyze SN-38 levels in mice treated with a variety of herbal combinations, we were able to effectively explore BXD spectrum-effect relationships and to thereby establish the components of this medicinal preparation that were bioactive and capable of preventing CPT-11-induced diarrhea in mice. This and similar spectrum-effect studies represent a robust means of exploring the mechanistic basis for the pharmacological activity of TCM preparations.


Subject(s)
Diarrhea/prevention & control , Drugs, Chinese Herbal/pharmacology , Intestinal Diseases/prevention & control , Irinotecan/toxicity , Animals , Chromatography, High Pressure Liquid , Diarrhea/chemically induced , Drugs, Chinese Herbal/chemistry , Female , Intestinal Diseases/chemically induced , Mice , Mice, Inbred ICR , Tandem Mass Spectrometry , Topoisomerase I Inhibitors/toxicity
15.
Cancer Chemother Pharmacol ; 87(3): 327-336, 2021 03.
Article in English | MEDLINE | ID: mdl-33130913

ABSTRACT

PURPOSE: Intestinal mucositis is an important adverse effect of antineoplastic therapy, which remains without adequate treatment. The present study aimed to carry out a complete evaluation of the histopathological changes during irinotecan-induced intestinal mucositis, using the protocol most found in the pharmacological reports nowadays to better understand irinotecan toxicity and support future studies on drug discovery. METHODS: Intestinal mucositis was induced by treating swiss mice for 4 days with irinotecan (75 mg/kg, i.p.). After 72 h post irinotecan, the mice were sacrificed and the small intestine and colon were excised to performed histological analysis by stained tissue with hematoxylin/eosin (H&E). RESULTS: Histoarchitecture loss, villus/crypt ratio reduction, atrophy of the muscular layer, hypertrophy in the submucosal and mucous layers, ruptures in the epithelium, as well as extent cellular infiltrate and presence of micro abscesses and the fusion of the crypts were observed in the histological analysis. Moreover, duodenum and colon had increased intraepithelial lymphocytes and mitotic figures. However, submucosal ganglia were decreased in the duodenum and increased in the colon. CONCLUSIONS: The data obtained in the present study provides new evidence that irinotecan-induced intestinal mucositis highly affects small intestine and colon, further contributing to establish criteria in light of the histopathological changes induced by irinotecan during intestinal mucositis and facilitating inter-study comparisons.


Subject(s)
Intestinal Mucosa/drug effects , Irinotecan/toxicity , Mucositis/chemically induced , Topoisomerase I Inhibitors/toxicity , Animals , Colon/drug effects , Colon/pathology , Female , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/pathology , Irinotecan/administration & dosage , Mice , Mucositis/pathology , Topoisomerase I Inhibitors/administration & dosage
16.
Br J Pharmacol ; 177(18): 4193-4208, 2020 09.
Article in English | MEDLINE | ID: mdl-32548889

ABSTRACT

BACKGROUND AND PURPOSE: Irinotecan, used in colorectal cancer therapy, is metabolized by glucuronidation involving different UDP-glucuronosyltransferase (UGT)1A isoforms leading to facilitated elimination from the body. Individuals homozygous for the genetic variants UGT1A1*28 (Gilbert syndrome) and UGT1A7*3 are more susceptible to irinotecan side effects, severe diarrhoea and leukopenia. The aim of this study was to investigate the protective effects and active constituents of coffee during irinotecan therapy using humanized transgenic (htg)UGT1A-WT and htgUGT1A-SNP (carry UGT1A1*28 and UGT1A7*3 polymorphisms) mice. EXPERIMENTAL APPROACH: HtgUGT1A mice were pretreated with coffee or caffeic acid (CA) + caffeic acid phenylethyl ester (CAPE) and injected with irinotecan. The effects of coffee and CA + CAPE were investigated using reporter gene assays, immunoblot, TaqMan-PCR, siRNA analyses and blood counts. KEY RESULTS: Only the combination of the two coffee ingredients, CA and CAPE, mediates protective effects of coffee in a model of irinotecan toxicity by activation of UGT1A genes. Coffee and CA + CAPE significantly increased UGT1A expression and activity along with SN-38 glucuronide excretion in irinotecan-injected htgUGT1A mice, resulting in significant improvement of leukopenia, intestinal oxidative stress and inflammation. CONCLUSION AND IMPLICATIONS: In this study, we identify the compounds responsible for mediating the previously reported coffee-induced activation of UGT1A gene expression. CA and CAPE represent key factors for the protective properties of coffee which are capable of reducing irinotecan toxicity, exerting antioxidant and protective effects. Provided that CA + CAPE do not affect irinotecan efficacy, they might represent a novel strategy for the treatment of irinotecan toxicity.


Subject(s)
Caffeic Acids , Coffee , Irinotecan , Leukopenia , Oxidative Stress , Animals , Caffeic Acids/pharmacology , Camptothecin/toxicity , Esters , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Irinotecan/toxicity , Leukopenia/chemically induced , Leukopenia/prevention & control , Mice
17.
J Cell Physiol ; 235(1): 194-209, 2020 01.
Article in English | MEDLINE | ID: mdl-31219187

ABSTRACT

The transformation abilities of CD44s and CD44v6 in normal intestinal epithelial cells have not yet been reported. Herein, we established both CD44s and CD44v6 overexpressing stable clones from rat IEC-6 cells and demonstrated that the CD44v6 clones had higher saturation density and anchorage independence. Additionally, CD44v6 clones were more resistant to oxaliplatin and irinotecan which might be attributed to a significantly increased B-cell lymphoma 2 level and a reduced DNA damage response in these cells. Moreover, c-Met and vascular endothelial growth factor receptor 2 signalings were involved in modulating the saturation density in CD44v6 clones. Interestingly, higher activation of both AKT and extracellular-signal-regulated kinase (ERK) were detected in CD44v6 clones which might account in part for the cell density-independent nuclear localization of Yes-associated protein (YAP). To no surprise, increases of both saturation density and anchorage independence in CD44v6 clones were markedly diminished by PI3K, AKT, MEK, and ERK inhibitors as well as YAP knockdown. By contrast, overexpression of a constitutively active YAP robustly increased the aforementioned phenotypes in IEC-6 cells. Collectively, our results suggest that upregulation of CD44v6, but not CD44s, induces the transformation of normal intestinal epithelial cells possibly via activating the c-Met/AKT/YAP pathway which might also explain the important role of CD44v6 in the initiation of various carcinomas.


Subject(s)
Cell Transformation, Neoplastic/pathology , Hyaluronan Receptors/genetics , Animals , Antineoplastic Agents/toxicity , Cell Line , Cell Transformation, Neoplastic/genetics , Epithelial Cells , Gene Expression Regulation/drug effects , Humans , Irinotecan/toxicity , Oxaliplatin/toxicity , Protein Isoforms , Rats , Topoisomerase I Inhibitors/toxicity
18.
Pathol Oncol Res ; 26(2): 955-965, 2020 Apr.
Article in English | MEDLINE | ID: mdl-30919275

ABSTRACT

Irinotecan-induced mucositis is a major oncological problem. Goblet cells secrete mucus, protecting the intestinal mucosa, with secretion altered during mucositis. The enteric nervous system is involved in regulating gut motility and secretion. The aim of this study was to determine whether enteric neural cells and goblet cells are altered following irinotecan treatment. Tumour-bearing Dark Agouti rats were administered a single dose of 175 mg/kg of irinotecan intraperitoneally and 0.01 mg/kg atropine subcutaneously. Experimental and untreated control rats were killed at times 6, 24, 48, 72, 96 and 120 h after treatment. Jejunum and colon samples were formalin fixed. Haematoxylin and eosin staining, Alcian Blue-PAS staining, and immunohistochemistry with S-100 antibody (neural cell marker) were carried out. Statistical analyses were carried out using Kruskal-Wallis test with Dunns post test, Mann Whitney U test and nonlinear regression. Total goblet cells decreased at 72 h compared with controls in the colon (p < 0.05). The percentage of cavitated goblet cells decreased compared to all other time points at 120 h in the colon. The number of S-100 positive cells in the submucosal plexus decreased in the colon (p = 0.0046) and in the myenteric plexus of the jejunum and colon (p = 0.0058 and p = 0.0022, respectively), when comparing treated with control. Enteric ganglia in the myenteric plexus of the jejunum decreased at 24 h and 96 h. Irinotecan-induced mucositis is associated with increases in mucus secretion, and enteric neural cell change. These changes may contribute to the pathophysiology of mucositis through the dysregulation of neural signalling.


Subject(s)
Antineoplastic Agents/toxicity , Enteric Nervous System/drug effects , Goblet Cells/drug effects , Irinotecan/toxicity , Mucositis/chemically induced , Neurons/drug effects , Animals , Disease Models, Animal , Female , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mucositis/pathology , Neoplasms, Experimental/pathology , Rats
19.
Cancer Chemother Pharmacol ; 85(1): 195-204, 2020 01.
Article in English | MEDLINE | ID: mdl-31813002

ABSTRACT

Delayed diarrhea is a common side effect of irinotecan administration, leading to a reduction in dose and thus a delay in anticancer therapy. Ganciclovir (GCV), an antiretroviral drug, is used to treat cytomegalovirus (CMV) infection. It is unclear whether GCV exhibits protective effects against irinotecan-induced intestinal dysfunction. Intraperitoneal administration of irinotecan with or without GCV for 4 days induced intestinal toxicity in mice to analyze diarrhea; beta-glucuronidase (ß-GLU) activity; fecal occult blood; hepatic function in blood samples, histopathological changes; and NOD-like receptor 3 (NLRP3), toll-like receptor 4 (TLR4), high-mobility group box 1 protein (HMGB1), phosphorylated nuclear factor kappa B (p-NF-κB), occludin, and zonular occludens (ZO-1) expression in colonic and ileal tissues. In addition, an irinotecan-treated mouse model was constructed and analyzed based on survival time. Expression levels of NLRP3, TLR4, HMGB1, p-NF-κB, occludin, and ZO-1 in normal colonic epithelial cells (NCM460 cells) stimulated with SN-38 were analyzed. GCV treatment reduced various indicators of irinotecan-induced intestinal dysfunction, including delayed-onset diarrhea, pathomorphological changes indicating hepatotoxicity, and proteins related to the HMGB1/TLR4 pathway that induced inflammation; the results were increased body weight, food intake, and expression of the protective proteins occludin and ZO-1. No changes in ß-GLU activity were observed. Changes in the expression of proteins related to the HMGB1/TLR4 pathway, occludin, and ZO-1 in SN-38-stimulated NCM460 cells were similar to changes in these proteins in vivo. In addition, administration of GCV improved mouse survival, indicating that the drug had long-term efficacy. Furthermore, GCV + irinotecan did not decrease the anti-tumor effect of irinotecan in vivo. In summary, GCV had intestine-protective and anti-inflammatory properties that possibly reduced irinotecan-induced intestinal dysfunction.


Subject(s)
Colonic Neoplasms/drug therapy , Diarrhea/drug therapy , Ganciclovir/pharmacology , Inflammation/drug therapy , Intestinal Diseases/drug therapy , Irinotecan/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , Antiviral Agents/pharmacology , Apoptosis , Biomarkers , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Diarrhea/chemically induced , Diarrhea/metabolism , Diarrhea/pathology , Gene Expression Regulation , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Intestinal Diseases/chemically induced , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Topoisomerase I Inhibitors/toxicity , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
CPT Pharmacometrics Syst Pharmacol ; 8(12): 951-961, 2019 12.
Article in English | MEDLINE | ID: mdl-31671257

ABSTRACT

Gastrointestinal (GI)-related adverse events (AEs) are commonly observed in the clinic during cancer treatments. Citrulline is a potentially translatable biomarker of GI AEs. In this study, irinotecan-induced citrulline changes were studied for a range of doses and schedules in rats. A translational system toxicology model for GI AEs using citrulline was then developed based on new experimental data and parameters from a literature intestinal cell dynamic model. With the addition of feedback-development and tolerance-development mechanisms, the model well captured the plasma citrulline profiles after irinotecan treatment in rats. Subsequently, the model was translated to humans and predicted the observed GI AE dynamics in humans including dose-scheduling effect using the cytotoxic and feedback parameters estimated in rats with slight calibrations. This translational toxicology model could be used for other antineoplastic drugs to simulate various clinical dosing scenarios before human studies and mitigate potential GI AEs.


Subject(s)
Antineoplastic Agents/toxicity , Citrulline/blood , Gastrointestinal Tract/metabolism , Irinotecan/toxicity , Animals , Antineoplastic Agents/administration & dosage , Disease Models, Animal , Gastrointestinal Tract/drug effects , Humans , Irinotecan/administration & dosage , Rats , Toxicity Tests , Translational Research, Biomedical
SELECTION OF CITATIONS
SEARCH DETAIL
...