Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Breast Cancer ; 28(1): 99-109, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32683606

ABSTRACT

BACKGROUND: Neratinib is a potent irreversible pan-ErbB tyrosine kinase inhibitor, approved by the FDA for extended adjuvant treatment of HER2-positive breast cancer. Diarrhea is the most frequently observed adverse event with tyrosine kinase inhibitor therapy. In this study, we developed a reproducible model for neratinib-induced diarrhea in male and female rats. METHODS: At first, male rats were treated with neratinib at 15, 30 or 50 mg/kg or vehicle control via oral gavage for 28 days (total n = 12). Secondly, we compared outcomes of male (n = 7) and female (n = 8) rats, treated with 50 mg/kg neratinib. RESULTS: Rats treated with a 50 mg/kg daily dose of neratinib had a reproducible and clinically relevant level of diarrhea and therefore was confirmed as an appropriate dose. Male rats treated with neratinib had significant changes to their gut microbiome. This included neratinib-induced increases in Ruminococcaceae (P = 0.0023) and Oscillospira (P = 0.026), and decreases in Blautia (P = 0.0002). On average, female rats experienced more significant neratinib-induced diarrhea (mean grade 1.526) compared with male rats (mean grade 1.182) (P < 0.0001). Neratinib caused a reduction in percentage weight gain after 28 days of treatment in females (P = 0.0018) compared with vehicle controls. Females and males both showed instances of villus atrophy and fusion, most severely in the distal ileum. Serum neratinib concentration was higher in female rats compared to male rats (P = 0.043). CONCLUSIONS: A reproducible diarrhea model was developed in both female and male rats, which indicated that diarrhea pathogenesis is multifactorial, including anatomical disruption particularly evident in the distal ileum, and alterations in microbial composition.


Subject(s)
Diarrhea/chemically induced , Gastrointestinal Microbiome/drug effects , Protein Kinase Inhibitors/adverse effects , Quinolines/adverse effects , Receptor, ErbB-2/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Diarrhea/blood , Diarrhea/microbiology , Diarrhea/pathology , Disease Models, Animal , Female , Humans , Ileum/drug effects , Ileum/microbiology , Ileum/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Rats , Sex Factors
2.
Cancer Chemother Pharmacol ; 83(3): 531-543, 2019 03.
Article in English | MEDLINE | ID: mdl-30535958

ABSTRACT

PURPOSE: Neratinib is an irreversible pan-ErbB tyrosine kinase inhibitor used for the extended adjuvant treatment of early-stage HER2-positive breast cancer. Its use is associated with the development of severe diarrhea in up to 40% of patients in the absence of proactive management. We previously developed a rat model of neratinib-induced diarrhea and found inflammation and anatomical disruption in the ileum and colon. Here we tested whether anti-diarrheal interventions, budesonide and colesevelam, can reduce neratinib-induced diarrhea and intestinal pathology. METHODS: Rats were treated with 50 mg/kg neratinib via oral gavage for 14 or 28 days (total n = 64). Body weight and diarrhea severity were recorded daily. Apoptosis was measured using immunohistochemistry for caspase-3. Inflammation was measured via a multiplex cytokine/chemokine assay. ErbB levels were measured using PCR and Western Blot. RESULTS: Budesonide co-treatment caused rats to gain significantly less weight than neratinib alone from day 4 of treatment (P = 0.0418). Budesonide (P = 0.027) and colesevelam (P = 0.033) each reduced the amount of days with moderate diarrhea compared to neratinib alone. In the proximal colon, rats treated with neratinib had higher levels of apoptosis compared to controls (P = 0.0035). Budesonide reduced histopathological injury in the proximal (P = 0.0401) and distal colon (P = 0.027) and increased anti-inflammatory IL-4 tissue concentration (ileum; P = 0.0026, colon; P = 0.031) compared to rats treated with neratinib alone. In the distal ileum, while budesonide decreased ErbB1 mRNA expression compared to controls (P = 0.018) (PCR), an increase in total ErbB1 protein was detected (P = 0.0021) (Western Blot). CONCLUSION: Both budesonide and colesevelam show potential as effective interventions against neratinib-induced diarrhea.


Subject(s)
Budesonide/therapeutic use , Colesevelam Hydrochloride/therapeutic use , Diarrhea/drug therapy , Protein Kinase Inhibitors/adverse effects , Quinolines/adverse effects , Animals , Diarrhea/chemically induced , Diarrhea/diagnosis , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Male , Rats , Rats, Wistar , Receptor, ErbB-2/antagonists & inhibitors , Severity of Illness Index , Time Factors , Treatment Outcome
3.
Chemotherapy ; 63(5): 284-292, 2018.
Article in English | MEDLINE | ID: mdl-30731451

ABSTRACT

BACKGROUND: The common cytotoxic mechanisms that underpin chemoefficacy and toxicity have hampered efforts to deliver effective supportive care interventions, particularly for gastrointestinal (GI) toxicity. Matrix metalloproteinases (MMPs) have been implicated in both tumor growth and GI toxicity, and as such MMP inhibitors present as a novel therapeutic avenue to simultaneously enhance treatment efficacy and reduce toxicity. OBJECTIVES: The aim of this study was to determine the efficacy of an MMP-9/12 inhibitor, AZD3342, on tumor growth and GI toxicity in a rat model. METHODS: Female tumor-bearing Dark Agouti rats (n = 90) were divided into 4 groups: vehicle control; methotrexate (MTX); AZD3342, and MTX + AZD3342. Tumors were measured daily (for 5 days) using digital calipers. GI toxicity was assessed using well-established clinical markers (diarrhea/weight loss), histopathological analysis, and functional assessment of intestinal barrier permeability. RESULTS: AZD3342 delayed the onset of severe diarrhea by 1 day (vs. MTX) but was unable to improve the overall severity of diarrhea. No changes were detected in tissue morphology or intestinal barrier function. AZD3342 alone suppressed tumor growth (p = 0.003 vs. vehicle) but did not enhance the efficacy of MTX. CONCLUSIONS: This study showed partial efficacy of AZD3342 in reducing tumor growth and delaying the onset of severe diarrhea caused by MTX in rats. We suggest further studies be undertaken targeting appropriate scheduling of AZD3342 as well as investigating different cytotoxic therapies that strongly activate MMP signaling.


Subject(s)
Matrix Metalloproteinase Inhibitors/therapeutic use , Neoplasms/drug therapy , Organic Chemicals/therapeutic use , Animals , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Cell Line, Tumor , Diarrhea/etiology , Female , Humans , Intestinal Mucosa/pathology , Matrix Metalloproteinase Inhibitors/adverse effects , Matrix Metalloproteinases/chemistry , Matrix Metalloproteinases/metabolism , Methotrexate/adverse effects , Methotrexate/therapeutic use , Neoplasms/pathology , Organic Chemicals/adverse effects , Random Allocation , Rats , Transplantation, Heterologous
4.
Cancer Chemother Pharmacol ; 79(2): 431-434, 2017 02.
Article in English | MEDLINE | ID: mdl-28011980

ABSTRACT

PURPOSE: Irinotecan-induced gut toxicity is mediated in part by Toll-Like receptor 4 (TLR4) signalling. The primary purpose of this preclinical study was to determine whether blocking TLR4 signalling by administering (-)-naloxone, a TLR4 antagonist, would improve irinotecan-induced gut toxicity. Our secondary aim was to determine the impact of (-)-naloxone on tumour growth. METHODS: Female Dark Agouti (DA) tumour-bearing rats were randomly assigned to four treatments (n = 6 in each); control, (-)-naloxone (100 mg/kg oral gavage at -2, 24, 48, and 72 h), irinotecan (175 mg/kg intraperitoneal at 0 h), and (-)-naloxone and irinotecan. Body weight and tumour growth were measured daily, and diarrhoea incidence and severity were recorded 4× per day up to 72 h post-treatment. RESULTS: At 72 h, all rats that received irinotecan lost weight compared to controls (p = 0.03). In addition, rats that received (-)-naloxone and irinotecan lost significantly more weight compared to controls (p < 0.005) than irinotecan only compared to controls (p = 0.001). (-)-Naloxone did not attenuate irinotecan-induced severe diarrhoea at 48 and 72 h. Finally, (-)-naloxone caused increased tumour growth compared to control at 72 h (p < 0.05) and significantly reduced the efficacy of irinotecan (p = 0.001). CONCLUSIONS: (-)-Naloxone in our preclinical model was unable to block irinotecan-induced gut toxicity and decreased the efficacy of irinotecan. As (-)-naloxone-oxycodone combination is used for cancer pain, this may present a potential safety concern for patients receiving (-)-naloxone-oxycodone and irinotecan concurrently and requires further investigation.


Subject(s)
Camptothecin/analogs & derivatives , Naloxone/toxicity , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Camptothecin/toxicity , Cancer Pain/drug therapy , Diarrhea/chemically induced , Diarrhea/prevention & control , Female , Irinotecan , Naloxone/pharmacology , Rats , Toll-Like Receptor 4/physiology
5.
Cancer Biol Ther ; 15(2): 236-44, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316664

ABSTRACT

Chemotherapy for cancer causes significant gut toxicity, leading to severe clinical manifestations and an increased economic burden. Despite much research, many of the underlying mechanisms remain poorly understood hindering effective treatment options. Recently there has been renewed interest in the role tight junctions play in the pathogenesis of chemotherapy-induced gut toxicity. To delineate the underlying mechanisms of chemotherapy-induced gut toxicity, this study aimed to quantify the molecular changes in key tight junction proteins, ZO-1, claudin-1, and occludin, using a well-established preclinical model of gut toxicity. Female tumor-bearing dark agouti rats received irinotecan or vehicle control and were assessed for validated parameters of gut toxicity including diarrhea and weight loss. Rats were killed at 6, 24, 48, 72, 96, and 120 h post-chemotherapy. Tight junction protein and mRNA expression in the small and large intestines were assessed using semi-quantitative immunohistochemistry and RT-PCR. Significant changes in protein expression of tight junction proteins were seen in both the jejunum and colon, correlating with key histological changes and clinical features. mRNA levels of claudin-1 were significantly decreased early after irinotecan in the small and large intestines. ZO-1 and occludin mRNA levels remained stable across the time-course of gut toxicity. Findings strongly suggest irinotecan causes tight junction defects which lead to mucosal barrier dysfunction and the development of diarrhea. Detailed research is now warranted to investigate posttranslational regulation of tight junction proteins to delineate the underlying pathophysiology of gut toxicity and identify future therapeutic targets.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Camptothecin/analogs & derivatives , Intestinal Mucosa/drug effects , Intestine, Large/drug effects , Intestine, Small/drug effects , Tight Junctions/drug effects , Animals , Camptothecin/toxicity , Claudin-1/genetics , Claudin-1/metabolism , Diarrhea/chemically induced , Diarrhea/pathology , Female , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Large/metabolism , Intestine, Large/ultrastructure , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Irinotecan , Occludin/genetics , Occludin/metabolism , Rats , Tight Junctions/pathology , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...