1
|
Ni Y, Li R, Shen X, Yi D, Ren Y, Wang F, Geng Y, You Q. Diaphorobacter nitroreducens synergize with oxaliplatin to reduce tumor burden in mice with lung adenocarcinoma. mSystems 2024; 9:e0132323. [PMID: 38483163 PMCID: PMC11019951 DOI: 10.1128/msystems.01323-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024] Open
Abstract
Lung adenocarcinoma (LADC) is the most common lung cancer and the leading cause of cancer-related deaths globally. Accumulating evidence suggests that the gut microbiota regulates the host response to chemotherapeutic drugs and can be targeted to reduce the toxicity of current chemotherapeutic agents. However, the effect of Diaphorobacter nitroreducens synergized with oxaliplatin on the gut microbiota and their impact on LADC have never been explored. This study aimed to evaluate the anti-cancer effects of D. nitroreducens, oxaliplatin, and their combined treatment on tumor growth in tumor-bearing mice. The composition of gut microbiota and the immune infiltration of tumors were evaluated by using 16S rRNA gene high-throughput sequencing and immunofluorescence, respectively. The inhibitory effect of the combination treatment with D. nitroreducens and oxaliplatin was significantly stronger than that of oxaliplatin alone in tumor-bearing mice. Furthermore, we observed that the combination treatment significantly increased the relative abundance of Lactobacillus and Akkermansia in the gut microbiota. Meanwhile, the combination treatment significantly increased the proportions of macrophage but decreased the proportion of regulatory T cells in the LADC tumor tissues of mice. These findings underscored the relationship between D. nitroreducens and the gut microbiota-immune cell-LADC axis, highlighting potential therapeutic avenues for LADC treatment. IMPORTANCE Oxaliplatin is widely used as an effective chemotherapeutic agent in cancer treatment, but its side effects and response rate still need to be improved. Conventional probiotics potentially benefit cancer chemotherapy by regulating gut microbiota and tumor immune infiltration. This study was novel in reporting a more significant inhibitory effect of Diaphorobacter nitroreducens on lung adenocarcinoma (LADC) cells compared with common traditional probiotics and validating its potential as an adjuvant therapy for LADC chemotherapy in mice. This study investigated the impact of D. nitroreducens combined with oxaliplatin on the gut microbiota and immune infiltration of tumors as a potential mechanism to improve anticancer effects.
Collapse
Affiliation(s)
- Yalan Ni
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Rui Li
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoyu Shen
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Deli Yi
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yilin Ren
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Fudong Wang
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Geng
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, China
| | - Qingjun You
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
2
|
Sun K, Wang X, Zhang H, Lin G, Jiang R. Management and Mechanisms of Diarrhea Induced by Tyrosine Kinase Inhibitors in Human Epidermal Growth Factor Receptor-2-Positive Breast Cancer. Cancer Control 2024; 31:10732748241278039. [PMID: 39159918 PMCID: PMC11334140 DOI: 10.1177/10732748241278039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/20/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Breast cancer has the highest incidence among female malignancies, significantly impacting women's health. Recently, numerous HER2-targeted therapies have achieved excellent clinical outcomes. Currently, anti-HER2 drugs are divided into three main categories: monoclonal antibodies, small-molecule tyrosine kinase inhibitors, and antibody-coupled drugs (ADCs). The main toxic side effects of small molecule TKI-based therapy are diarrhea, hand-foot syndrome, rash, nausea, and vomiting. Diarrhea is a potential predictor of tumor response, affecting up to 95% of cancer patients treated with TKIs. Severe gastrointestinal toxicity can result in the need for dose reductions and treatment interruptions. This not only compromises the efficacy of TKIs but also deteriorates human nutrition and quality of life. The majority of individuals develop diarrhea within 7 days of starting treatment, with approximately 30% developing grade 3 or higher diarrhea within 2-3 days of starting treatment. The severity of diarrhea typically correlates with the dosage of most TKIs. Current prevention and management strategies are primarily empirical, focusing on symptom alleviation rather than addressing the toxicological mechanisms underlying TKI-induced diarrhea. Consequently, anti-diarrheal drugs are often less effective in managing this condition in cancer patients receiving TKIs. Moreover, our understanding of the toxicological mechanisms responsible for such diarrhea remains limited, underscoring the urgent need to identify these mechanisms in order to develop effective anti-diarrheal medications tailored to this specific context. This review aims to elucidate management approaches and mechanisms for diarrhea induced by TKIs during HER2-positive breast cance.
Collapse
Affiliation(s)
- Kena Sun
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Xiaojia Wang
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Huanping Zhang
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Guang Lin
- Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Ruiyuan Jiang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
3
|
Danan CH, Naughton KE, Hayer KE, Vellappan S, McMillan EA, Zhou Y, Matsuda R, Nettleford SK, Katada K, Parham LR, Ma X, Chowdhury A, Wilkins BJ, Shah P, Weitzman MD, Hamilton KE. Intestinal transit-amplifying cells require METTL3 for growth factor signaling and cell survival. JCI Insight 2023; 8:e171657. [PMID: 37883185 PMCID: PMC10795831 DOI: 10.1172/jci.insight.171657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 10/25/2023] [Indexed: 10/27/2023] Open
Abstract
Intestinal epithelial transit-amplifying cells are essential stem progenitors required for intestinal homeostasis, but their rapid proliferation renders them vulnerable to DNA damage from radiation and chemotherapy. Despite these cells' critical roles in intestinal homeostasis and disease, few studies have described genes that are essential to transit-amplifying cell function. We report that RNA methyltransferase-like 3 (METTL3) is required for survival of transit-amplifying cells in the murine small intestine. Transit-amplifying cell death after METTL3 deletion was associated with crypt and villus atrophy, loss of absorptive enterocytes, and uniform wasting and death in METTL3-depleted mice. Sequencing of polysome-bound and methylated RNAs in enteroids and in vivo demonstrated decreased translation of hundreds of methylated transcripts after METTL3 deletion, particularly transcripts involved in growth factor signal transduction such as Kras. Further investigation verified a relationship between METTL3 and Kras methylation and protein levels in vivo. Our study identifies METTL3 as an essential factor supporting the homeostasis of small intestinal tissue via direct maintenance of transit-amplifying cell survival. We highlight the crucial role of RNA modifications in regulating growth factor signaling in the intestine with important implications for both homeostatic tissue renewal and epithelial regeneration.
Collapse
Affiliation(s)
- Charles H. Danan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
- Medical Scientist Training Program, Perelman School of Medicine; and
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kaitlyn E. Naughton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
| | - Katharina E. Hayer
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Protective Immunity, Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine; University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sangeevan Vellappan
- Waksman Institute of Microbiology and
- Department of Genetics, Rutgers University, Piscataway, New Jersey, USA
- Human Genetics Institute of New Jersey, Piscataway, New Jersey, USA
| | - Emily A. McMillan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
| | - Yusen Zhou
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Rina Matsuda
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Pathobiology, School of Veterinary Medicine, and
| | - Shaneice K. Nettleford
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
| | - Kay Katada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Louis R. Parham
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xianghui Ma
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
| | - Afrah Chowdhury
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
| | - Benjamin J. Wilkins
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine; University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Premal Shah
- Department of Genetics, Rutgers University, Piscataway, New Jersey, USA
- Human Genetics Institute of New Jersey, Piscataway, New Jersey, USA
| | - Matthew D. Weitzman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Protective Immunity, Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine; University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathryn E. Hamilton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Perelman School of Medicine
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Chen GQ, Nan Y, Huang SC, Ning N, Du YH, Lu DD, Yang YT, Meng FD, Yuan L. Research progress of ginger in the treatment of gastrointestinal tumors. World J Gastrointest Oncol 2023; 15:1835-1851. [DOI: 10.4251/wjgo.v15.i11.1835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer seriously endangers human health. Gastrointestinal cancer is the most common and major malignant tumor, and its morbidity and mortality are gradually increasing. Although there are effective treatments such as radiotherapy and chemotherapy for gastrointestinal tumors, they are often accompanied by serious side effects. According to the traditional Chinese medicine and food homology theory, many materials are both food and medicine. Moreover, food is just as capable of preventing and treating diseases as medicine. Medicine and food homologous herbs not only have excellent pharmacological effects and activities but also have few side effects. As a typical medicinal herb with both medicinal and edible uses, some components of ginger have been shown to have good efficacy and safety against cancer. A mass of evidence has also shown that ginger has anti-tumor effects on digestive tract cancers (such as gastric cancer, colorectal cancer, liver cancer, laryngeal cancer, and pancreatic cancer) through a variety of pathways. The aim of this study is to investigate the mechanisms of action of the main components of ginger and their potential clinical applications in treating gastrointestinal tumors.
Collapse
Affiliation(s)
- Guo-Qing Chen
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shi-Cong Huang
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Na Ning
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yu-Hua Du
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Dou-Dou Lu
- School of Clinical Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ya-Ting Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Fan-Di Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
5
|
Mekhael M, Larsen HM, Lauritzen MB, Thorlacius-Ussing O, Laurberg S, Krogh K, Drewes AM, Christensen P, Juul T. Bowel dysfunction following pelvic organ cancer: a prospective study on the treatment effect in nurse-led late sequelae clinics. Acta Oncol 2023; 62:70-79. [PMID: 36757368 DOI: 10.1080/0284186x.2023.2168214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Bowel dysfunction following treatment of pelvic organ cancer is prevalent and impacts the quality of life (QoL). The present study aimed to evaluate the feasibility and effects of treating bowel dysfunction in two nurse-led late sequelae clinics. MATERIAL AND METHODS Treatment effects were monitored prospectively by patient-reported outcome measures collected at baseline and discharge. Change in bowel function was evaluated by 15 bowel symptoms, the St. Mark's Incontinence Score, the Patients Assessment of Constipation-Symptoms (PAC-SYM) score and self-rated bowel function. QoL was evaluated by the EuroQol 5-dimension 5-level (EQ-5D-5L) utility score and by measuring the impact of bowel function on QoL. RESULTS From June 2018 to December 2021, 380 cancer survivors (46% rectal, 15% gynaecological, 13% anal, 12% colon, 12% prostate, and 2% other cancers) completed a baseline questionnaire and started treatment for bowel dysfunction. At referral, 96% of patients were multisymptomatic. The most frequent symptoms were faecal urgency (95%), fragmented defaecation (93%), emptying difficulties (92%), flatus/faecal incontinence (flatus 89%, liquid 59%, solid 33%), and obstructed defaecation (79%). In total, 169 patients were discharged from the clinics in the follow-up period. At discharge, 69% received conservative treatment only and 24% also received transanal irrigation; 4% were surgically treated; 3% discontinued treatment. Improvements were seen in all 15 bowel symptoms (p < 0.001), the mean St. Mark's Incontinence Score (12.0 to 9.9, p < 0.001), the mean PAC-SYM score (1.04 to 0.84, p < 0.001) and the mean EQ-5D-5L utility score (0.78 to 0.84, p < 0.001). Self-rated bowel function improved in 56% (p < 0.001) of cases and the impact of bowel function on QoL improved in 46% (p < 0.001). CONCLUSION Treatment of bowel dysfunction in nurse-led late sequelae clinics is feasible and significantly improved bowel function and QoL.
Collapse
Affiliation(s)
- Mira Mekhael
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark
| | - Helene M Larsen
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark.,Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark.,Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Michael B Lauritzen
- Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark.,Department of Gastrointestinal Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Ole Thorlacius-Ussing
- Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark.,Department of Gastrointestinal Surgery, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Søren Laurberg
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark.,Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark
| | - Klaus Krogh
- Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark.,Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Asbjørn M Drewes
- Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark.,Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Peter Christensen
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark.,Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark
| | - Therese Juul
- Department of Surgery, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Danish Cancer Society Centre for Research on Survivorship and Late Adverse Effects after Cancer in the Pelvic Organs, Aarhus and Aalborg University Hospitals, Aarhus and Aalborg, Denmark
| |
Collapse
|
6
|
Lai J, Jiang F, Zhuo X, Xu X, Liu L, Yin K, Wang J, Zhao J, Xu W, Liu H, Wang X, Jiang W, Wang K, Yang S, Guo H, Qi F, Yuan X, Lin X, Fu G. Effects of Shenling Baizhu powder on pyrotinib-induced diarrhea: analysis of gut microbiota, metabonomics, and network pharmacology. Chin Med 2022; 17:140. [PMID: 36528679 PMCID: PMC9759852 DOI: 10.1186/s13020-022-00696-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Shenling Baizhu Powder (SBP) is a traditional Chinese medicine (TCM) prescription, which has the good efficacy on gastrointestinal toxicity. In this study, we used gut microbiota analysis, metabonomics and network pharmacology to investigate the therapeutic effect of SBP on pyrotinib-induced diarrhea. METHODS 24 Rats were randomly divided into 4 groups: control group, SBP group (3.6 g/kg /bid SBP for 10 days), pyrotinib model group (80 mg/kg/qd pyrotinib) and pyrotinib + SBP treatment group. A 16S rRNA sequencing was used to detect the microbiome of rat fecal bowel. Metabolic profiles were collected by non-targeted metabolomics and key metabolic pathways were identified using MetaboAnalyst 5.0. The antitumor effect of SBP on cells treated with pyrotinib was measured using a CCK-8 assay. Network pharmacology was used to predict the target and action pathway of SBP in treating pyrotinib-related diarrhea. RESULTS In vivo study indicated that SBP could significantly alleviate pyrotinib-induced diarrhea, reaching a therapeutic effect of 66.7%. SBP could regulate pyrotinib-induced microbiota disorder. LEfSe research revealed that the SBP could potentially decrease the relative abundance of Escherichia, Helicobacter and Enterobacteriaceae and increase the relative abundance of Lachnospiraceae, Bacilli, Lactobacillales etc. In addition, 25-Hydroxycholesterol, Guanidinosuccinic acid, 5-Hydroxyindolepyruvate and cAMP were selected as potential biomarkers of SBP for pyrotinib-induced diarrhea. Moreover, Spearman's analysis showed a correlation between gut microbiota and metabolite: the decreased 25-hydroxycholesterol in the pyrotinib + SBP treatment group was negatively correlated with Lachnospiraceae while positively correlated with Escherichia and Helicobacter. Meanwhile, SBP did not affect the inhibitory effect of pyrotinib on BT-474 cells and Calu-3 cells in vitro. Also, the network analysis further revealed that SBP treated pyrotinib-induced diarrhea through multiple pathways, including inflammatory bowel disease, IL-17 signaling pathway, pathogenic Escherichia coli infection and cAMP signaling pathway. CONCLUSIONS SBP could effectively relieve pyrotinib-induced diarrhea, revealing that intestinal flora and its metabolites may be involved in this process.
Collapse
Affiliation(s)
- Jingjiang Lai
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250002, People's Republic of China
| | - Fengxian Jiang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250002, People's Republic of China
| | - Xiaoli Zhuo
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
- The Clinical Medical College, Shandong First Medical University (Shandong Academy of Medicine), Jinan, 250117, People's Republic of China
| | - Xiaoying Xu
- Department of Pathology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
| | - Lei Liu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
- The Clinical Medical College, Shandong First Medical University (Shandong Academy of Medicine), Jinan, 250117, People's Republic of China
| | - Ke Yin
- Department of Pathology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
| | - Jingliang Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250002, People's Republic of China
| | - Jing Zhao
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
- The Clinical Medical College, Shandong First Medical University (Shandong Academy of Medicine), Jinan, 250117, People's Republic of China
| | - Wei Xu
- Department of Oncology, Shandong Provincial Hospital Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
| | - Hongjing Liu
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250002, People's Republic of China
| | - Xuan Wang
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250002, People's Republic of China
| | - Wen Jiang
- Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Ke Wang
- Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Shuping Yang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Honglin Guo
- Department of Central Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
| | - Fanghua Qi
- Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Xiaotian Yuan
- Laboratory Animal Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Guobin Fu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China.
- Department of Oncology, Shandong Provincial Hospital Cheeloo College of Medicine, Shandong University, Jinan, 250021, People's Republic of China.
| |
Collapse
|
7
|
Kaur H, Fernández JR, Locher JL, Demark-Wahnefried W. Rural and Urban Differences in Vegetable and Fruit Consumption Among Older Cancer Survivors in the Deep South: An Exploratory Cross-Sectional Study. J Acad Nutr Diet 2022; 122:1717-1724.e4. [PMID: 35017097 PMCID: PMC9271124 DOI: 10.1016/j.jand.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/21/2021] [Accepted: 01/06/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Cancer survivors, especially those who are older, experience increased comorbidity and risk for secondary cancers. A varied dietary pattern, rich in vegetables and fruit (V&F) is recommended to improve health. However, V&F intake may differ by rural versus urban status. OBJECTIVE To assess differences in V&F consumption among older cancer survivors residing in urban- and rural-designated areas, and explore whether differences exist by sex, race, and cancer type. DESIGN This was a cross-sectional secondary analysis. PARTICIPANTS/SETTING Screening data from the Harvest for Health trial were obtained from October 2016 to November 2019 on 731 Medicare-eligible cancer survivors across Alabama. MAIN OUTCOME MEASURES V&F consumption was measured by 2-items from the Eating at America's Table NCI Dietary Screener. Rural and urban residence was coded at the zip-code level using the USDA's Rural-Urban Commuting Area (RUCA) coding schema using five different classifications (A-E). Sex, race and cancer-type were dichotomized as male/female, Non-Hispanic White (NHW)/Non-Hispanic Black (NHB) and gastro-intestinal/other cancers, respectively. STATISTICAL ANALYSES Kruskal Wallis rank sum and post-hoc tests were performed to detect differences in V&F consumption (α<0.05). RESULTS The study sample was largely female (66.2%), NHW (78.1%), of mean age 70 years and reported an average V&F intake of 1.47 cups/day. V&F consumption of cancer survivors living in isolated, small, rural towns was roughly half that consumed by survivors living elsewhere; thus, statistically significant rural-urban differences were found in models that accounted specifically for this subgroup, i.e., RUCA categorizations A and E.V&F consumption also was significantly lower in NHB (1.32 ± 0.98 cups/day) than NHW survivors (1.51 ± 1.10 cups/day) (p=0.0456); however, no statistically significant differences were detected by sex and cancer type. CONCLUSION Analyses that address the variability within "rural" designated areas are important in future studies. Moreover, a greater understanding is needed of factors that adversely affect V&F consumption of those most vulnerable, i.e., older NHB cancer survivors as well as those living in isolated, small, rural towns to best target future interventions.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition Sciences, University of Alabama at Birmingham (UAB), 1675 University Boulevard, Webb Nutrition Sciences Bldg., Room 604A Birmingham, AL 35294-3360.
| | - José R Fernández
- Professor and Vice Chair for Education, Department of Nutrition Sciences, UAB, 1675 University Boulevard, Webb Nutrition Sciences Bldg., Room 522 Birmingham, AL 35294-3360
| | - Julie L Locher
- Professor Emerita, Department of Medicine, UAB, CH19-Room 218F; Birmingham, Alabama 35294-2041
| | - Wendy Demark-Wahnefried
- Associate Director for Cancer Prevention and Control for the O'Neal Comprehensive Cancer Center at UAB, Professor and Webb Endowed Chair of Nutrition Sciences, American Cancer Society Clinical Research Professor, Department of Nutrition Sciences, UAB, 1675 University Boulevard, Webb Nutrition Sciences Bldg., Room 650 Birmingham, AL 35294-3360
| |
Collapse
|
8
|
Secombe KR, Van Sebille YZA, Mayo BJ, Coller JK, Gibson RJ, Bowen JM. Diarrhea Induced by Small Molecule Tyrosine Kinase Inhibitors Compared With Chemotherapy: Potential Role of the Microbiome. Integr Cancer Ther 2021; 19:1534735420928493. [PMID: 32493068 PMCID: PMC7273583 DOI: 10.1177/1534735420928493] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Small molecule receptor tyrosine kinase inhibitors (SM-TKIs) are among a group of
targeted cancer therapies, intended to be more specific to cancer cells compared
with treatments, such as chemotherapy, hence reducing adverse events.
Unfortunately, many patients report high levels of diarrhea, the pathogenesis of
which remains under investigation. In this article, we compare the current state
of knowledge of the pathogenesis of chemotherapy-induced diarrhea (CID) in
comparison to SM-TKI–induced diarrhea, and investigate how a similar research
approach in both areas may be beneficial. To this end, we review evidence that
both treatment modalities may interact with the gut microbiome, and as such the
microbiome should be investigated for its ability to reduce the risk of
diarrhea.
Collapse
Affiliation(s)
- Kate R Secombe
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Ysabella Z A Van Sebille
- UniSA Online, Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Bronwen J Mayo
- Division of Health Sciences, University of South Australia, South Australia, Australia
| | - Janet K Coller
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Rachel J Gibson
- School of Allied Health Science and Practice, University of Adelaide, South Australia, Australia
| | - Joanne M Bowen
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
9
|
Nexoe AB, Pedersen AA, von Huth S, Sorensen GL, Holmskov U, Jiang PP, Detlefsen S, Husby S, Rathe M. No effect of deleted in malignant brain tumors 1 deficiency on chemotherapy induced murine intestinal mucositis. Sci Rep 2021; 11:14687. [PMID: 34282203 PMCID: PMC8289998 DOI: 10.1038/s41598-021-94076-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Mucositis is a serious adverse effect of chemotherapeutic treatment. During intestinal mucositis, the mucosal barrier is compromised, increasing the risk of severe infections. Mucositis necessitates dose reduction or pauses in treatment, which affect the outcome of the treatment. Deleted in malignant brain tumors 1 (DMBT1) is a secreted scavenger protein with effects on innate immunity and epithelial regeneration. We have previously shown that jejunal DMBT1 expression is increased in piglets during chemotherapeutic treatment. We hypothesized that DMBT1 ameliorates doxorubicin-induced mucositis. Individually-caged Dmbt1+/+ (WT) and Dmbt1-/- (KO) female mouse littermates received intraperitoneal injections of either doxorubicin or saline. They were euthanized after three (D3) or seven days (D7). Weight loss was monitored every day, and serum citrulline levels were measured at termination. Intestinal tissue was analyzed for the expression of DMBT1 and proinflammatory cytokines (IL-1β, IL-6, and TNF). Specimens from the small intestines and colon were scored for inflammation and epithelial and mucosal architecture changes. We detected no effect of DMBT1 on weight loss, serum citrulline levels, expression of proinflammatory cytokines, or histologic damage. We detected a significant increase in crypt depth in WT mice compared to that in KO mice on D3. In conclusion, DMBT1 does not affect doxorubicin-induced mucositis in mice.
Collapse
Affiliation(s)
- Anders B Nexoe
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
- Department of Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Andreas A Pedersen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Sebastian von Huth
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
| | - Grith L Sorensen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Uffe Holmskov
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Ping-Ping Jiang
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Steffen Husby
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Mathias Rathe
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense C, Denmark.
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
10
|
Nardini P, Pini A, Bessard A, Duchalais E, Niccolai E, Neunlist M, Vannucchi MG. GLP-2 Prevents Neuronal and Glial Changes in the Distal Colon of Mice Chronically Treated with Cisplatin. Int J Mol Sci 2020; 21:ijms21228875. [PMID: 33238628 PMCID: PMC7700273 DOI: 10.3390/ijms21228875] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/19/2020] [Accepted: 11/21/2020] [Indexed: 12/20/2022] Open
Abstract
Cisplatin is a chemotherapeutic agent widely used for the treatment of solid cancers. Its administration is commonly associated with acute and chronic gastrointestinal dysfunctions, likely related to mucosal and enteric nervous system (ENS) injuries, respectively. Glucagon-like peptide-2 (GLP-2) is a pleiotropic hormone exerting trophic/reparative activities on the intestine, via antiapoptotic and pro-proliferating pathways, to guarantee mucosal integrity, energy absorption and motility. Further, it possesses anti-inflammatory properties. Presently, cisplatin acute and chronic damages and GLP-2 protective effects were investigated in the mouse distal colon using histological, immunohistochemical and biochemical techniques. The mice received cisplatin and the degradation-resistant GLP-2 analog ([Gly2]GLP-2) for 4 weeks. Cisplatin-treated mice showed mucosal damage, inflammation, IL-1β and IL-10 increase; decreased number of total neurons, ChAT- and nNOS-immunoreactive (IR) neurons; loss of SOX-10-IR cells and reduced expression of GFAP- and S100β-glial markers in the myenteric plexus. [Gly2]GLP-2 co-treatment partially prevented mucosal damage and counteracted the increase in cytokines and the loss of nNOS-IR and SOX-10-IR cells but not that of ChAT-IR neurons. Our data demonstrate that cisplatin causes mucosal injuries, neuropathy and gliopathy and that [Gly2]GLP-2 prevents these injuries, partially reducing mucosal inflammation and inducing ENS remodeling. Hence, this analog could represent an effective strategy to overcome colonic injures induced by cisplatin.
Collapse
Affiliation(s)
- Patrizia Nardini
- Histology and Embryology Research Unit, Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (A.P.); (E.N.)
| | - Alessandro Pini
- Histology and Embryology Research Unit, Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (A.P.); (E.N.)
| | - Anne Bessard
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, University of Nantes, 44035 Nantes, France; (A.B.); (E.D.); (M.N.)
| | - Emilie Duchalais
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, University of Nantes, 44035 Nantes, France; (A.B.); (E.D.); (M.N.)
| | - Elena Niccolai
- Histology and Embryology Research Unit, Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (A.P.); (E.N.)
| | - Michel Neunlist
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, University of Nantes, 44035 Nantes, France; (A.B.); (E.D.); (M.N.)
| | - Maria Giuliana Vannucchi
- Histology and Embryology Research Unit, Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (A.P.); (E.N.)
- Correspondence: ; Tel.: +39-055-275-8152
| |
Collapse
|
11
|
Zhou W, Chen K, Lu Q, Luo Y, Zhang C, Zheng Y, Zhuo Z, Guo K, Wang J, Chen H, Sha W. The Protective Effect of Rosavin from Rhodiola rosea on Radiation-Induced Intestinal Injury. Chem Biodivers 2020; 17:e2000652. [PMID: 33089958 DOI: 10.1002/cbdv.202000652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/20/2020] [Indexed: 02/05/2023]
Abstract
Bioactive constituents from Rhodiola rosea L. (RRL) exhibit multiple pharmacological effects on diverse diseases. However, whether they are suitable for the treatment of radiation-induced intestinal injury (RIII) remains unclear. This study aims to investigate their roles and mechanisms in the RIII rat model. The radioprotective effects of the four bioactive constituents of RRL (salidroside, herbacetin, rosavin and arbutin) were evaluated by the cell viability of irradiated IEC-6 cells. Intestinal tissues were collected for histological analysis, localized inflammation and oxidative stress assessments. Our work showed that salidroside, rosavin and arbutin improved the cell viability of the irradiated IEC-6 cells, with the highest improvement in 12.5 μM rosavin group. The rosavin treatment significantly improved survival rate and intestinal damage in irradiated rats by modulating the inflammatory response and oxidative stress. Our work indicated that rosavin may be the optimal constituent of RRL for RIII treatment, providing an attractive candidate for radioprotection.
Collapse
Affiliation(s)
- Weijie Zhou
- School of Medicine, South China University of Technology, Guangzhou, 510030, P. R. China.,Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Kequan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510030, P. R. China
| | - Quan Lu
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Yujun Luo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Chen Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Yue Zheng
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Kehang Guo
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Jinghua Wang
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| | - Weihong Sha
- School of Medicine, South China University of Technology, Guangzhou, 510030, P. R. China.,Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, P. R. China
| |
Collapse
|
12
|
An overview of acute gastrointestinal side effects of systemic anti-cancer therapy and their management. Best Pract Res Clin Gastroenterol 2020; 48-49:101691. [PMID: 33317796 DOI: 10.1016/j.bpg.2020.101691] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/06/2020] [Indexed: 01/31/2023]
Abstract
Treatment-related acute gastrointestinal toxicities are a common and often debilitating hurdle encountered in the treatment of cancer patients. While the introduction of targeted therapies such as tyrosine kinase inhibitors has led to improvements in survival outcomes, their use has also been complicated by a high frequency of clinically important adverse effects. Gastrointestinal toxicities such as nausea, vomiting, diarrhoea and hepatotoxicity represent potentially serious adverse events that may necessitate dose reductions, treatment interruptions and cessation of treatment. An improved knowledge of the incidence, pathophysiology, management and prophylaxis of these toxicities is crucial in order to reduce patient morbidity and mortality. In this review, we discuss the main gastrointestinal toxicities associated with chemotherapy and targeted therapies in oncology, outlining their incidence, pathophysiology and expert management guidelines.
Collapse
|
13
|
Chan YT, Cheung F, Zhang C, Fu B, Tan HY, Norimoto H, Wang N, Feng Y. Ancient Chinese Medicine Herbal Formula Huanglian Jiedu Decoction as a Neoadjuvant Treatment of Chemotherapy by Improving Diarrhea and Tumor Response. Front Pharmacol 2020; 11:252. [PMID: 32210825 PMCID: PMC7076183 DOI: 10.3389/fphar.2020.00252] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/24/2020] [Indexed: 12/25/2022] Open
Abstract
Background Diarrhea is a major gastrointestinal complication in cancer patients receiving chemotherapy. Prognosis and treatment of chemotherapy-induced diarrhea (CID) remain unsatisfactory. This study aims to explore the potential of an ancient Chinese Medicine herbal formula Huanglian Jiedu Decoction (HLJDD) as an adjuvant treatment on CID. Method HLJDD extract was prepared by GMP manufacturing standard with quality and stability being checked. 5-fluorouracil (5-Fu) and irinotecan (CPT-11)-induced diarrhea model in mice was established and pre-, co- and post-treatment of HLJDD was implemented. Mechanism of action was explored by detecting related protein expression. In addition, the effect of HLJDD on diarrhea and tumor response induced by clinical regimens FOLFOX and FOLFIRI was measured in murine orthotopic colorectal cancer model. Results HLJDD exhibited consistency in quality and stability after 24-month storage. Pre-treatment of HLJDD, but not co-treatment or post-treatment, could significantly improve the diarrhea score, body weight loss and intestinal damage in 5-Fu- and CPT-11-treated mice. Pre-treatment of HLJDD reduced cell apoptosis in the intestine of chemotherapy-treated mice, and promoted renewal of intestinal cell wall. CD44 was predicted as the potential target of HLJDD-containing compounds in CID. HLJDD pre-treatment induced presentation of CD44-postive cells in the intestine of chemotherapy-treated mice, and initiated expression of stemness-associated genes. Transcriptional products of the downstream Wnt signaling of CD44 were elevated. Furthermore, pre-treatment of HLJDD could significantly improve the tumor response of clinical chemotherapy regimens FOLFOX and FOLFIRI in orthotopic colorectal cancer, and reduce diarrhea and intestinal damage. Conclusion: Our study suggests the potential of HLJDD as a neoadjuvant treatment of chemotherapy by reducing diarrhea and improving tumor response.
Collapse
Affiliation(s)
- Yau-Tuen Chan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Fan Cheung
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Bowen Fu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hor-Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | | | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
14
|
Koch C, Reitz C, Schreckenbach T, Eichler K, Filmann N, Al-Batran SE, Götze T, Zeuzem S, Bechstein WO, Kraus T, Bojunga J, Düx M, Trojan J, Blumenstein I. Sarcopenia as a prognostic factor for survival in patients with locally advanced gastroesophageal adenocarcinoma. PLoS One 2019; 14:e0223613. [PMID: 31639132 PMCID: PMC6805048 DOI: 10.1371/journal.pone.0223613] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/24/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND AIMS Patients with gastric cancer often show signs of malnutrition. We sought to evaluate the influence of sarcopenia in patients with locally advanced, not metastasized, gastric or gastro-esophageal junction (GEJ) cancer undergoing curative treatment (perioperative chemotherapy and surgery) on morbidity and mortality in order to identify patients in need for nutritional intervention. PATIENTS AND METHODS Two-centre study, conducted in the Frankfurt University Clinic and Krankenhaus Nordwest (Frankfurt) as part of the University Cancer Center Frankfurt (UCT). 47/83 patients were treated in the FLOT trial (NCT01216644). Patients´ charts were reviewed for clinical data. Two consecutive CT scans were retrospectively analyzed to determine the degree of sarcopenia. Survival was calculated using the Kaplan-Meier method, multivariate analysis was performed using the Cox regression. RESULTS 60 patients (72.3%) were male and 23 (27.7%) female. 45 patients (54.2%) had GEJ type 1-3 and 38 (45.8%) gastric tumors, respectively. Sarcopenic patients were significantly older than non-sarcopenic patients (mean age 65.1 years vs. 59.5 years, p = 0.042), terminated the chemotherapy significantly earlier (50% vs. 22.6%, p = 0.037) and showed higher Clavien-Dindo scores, indicating more severe perioperative complications (score ≥3 43.3 vs. 17.0%, p = 0.019). Sarcopenic patients had a significantly shorter survival than non-sarcopenic patients (139.6 ± 19.5 [95% CI, 101.3-177.9] vs. 206.7 ± 13.8 [95% CI, 179.5-233.8] weeks, p = 0.004). Multivariate Cox regression analysis showed that, besides UICC stage, sarcopenia significantly influenced survival. CONCLUSION Sarcopenia is present in a large proportion of patients with locally advanced gastric or GEJ cancer and significantly influences tolerability of chemotherapy, surgical complications and survival.
Collapse
Affiliation(s)
- Christine Koch
- Department of Gastroenterology, Hepatology, Endocrinology and Nutrition, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Cornelius Reitz
- Department of Gastroenterology, Hepatology, Endocrinology and Nutrition, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Teresa Schreckenbach
- Department of General and Visceral Surgery, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Katrin Eichler
- Institute for Diagnostic and Interventional Radiology, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Natalie Filmann
- Institute for Biostatistics and Mathematical Modelling, Goethe University, Frankfurt am Main, Germany
| | - Salah-Eddin Al-Batran
- Institute for Clinical Research, (IKF), Hospital Nordwest, Frankfurt am Main, Germany
| | - Thorsten Götze
- Institute for Clinical Research, (IKF), Hospital Nordwest, Frankfurt am Main, Germany
| | - Stefan Zeuzem
- Department of Gastroenterology, Hepatology, Endocrinology and Nutrition, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Wolf Otto Bechstein
- Department of General and Visceral Surgery, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Thomas Kraus
- Central Institute for Radiology and Neuroradiology, Nordwest Hospital, Frankfurt am Main, Germany
| | - Jörg Bojunga
- Department of Gastroenterology, Hepatology, Endocrinology and Nutrition, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Markus Düx
- Department of General and Viseral Surgery, Hospital Nordwest, Frankfurt am Main, Germany
| | - Jörg Trojan
- Department of Gastroenterology, Hepatology, Endocrinology and Nutrition, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Irina Blumenstein
- Department of Gastroenterology, Hepatology, Endocrinology and Nutrition, University Hospital, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
15
|
Zhang T, Shi L, Xu Y, Li Y, Li S, Guan B, Qi Z, Zhang Y, Liu L. Purified PEGylated human glucagon-like peptide-2 reduces the severity of irradiation-induced acute radiation enteritis in rats. JOURNAL OF RADIATION RESEARCH 2019; 60:7-16. [PMID: 30247656 PMCID: PMC6373673 DOI: 10.1093/jrr/rry076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/08/2018] [Indexed: 06/08/2023]
Abstract
Radiation-induced acute intestinal injury after abdominal and pelvic irradiation is a common and serious problem in the clinical setting. Glucagon-like peptide-2 (GLP-2), a 33-amino acid peptide, exerts diverse effects related to the regulation of gastrointestinal growth and function. However, GLP-2 is relatively unstable in vivo. The aim of the present study was to improve GLP-2 stability in vivo and to evaluate its therapeutic effect on acute radiation enteritis. We generated long-lasting intestinal protection peptides by conjugating human GLP-2 (hGLP-2) peptides to polyethyleneglycol (PEG) to produce mPEGylation hGLP-2 (Mono-PEG-hGLP-2) through an enzymatic site-specific transglutamination reaction. Mono-PEG-hGLP-2 synthesized under optimal reaction conditions and separated by one-step ion-exchange chromatography was found to be resistant to degradation in vitro. Pretreatment with Mono-PEG-hGLP-2 reduced the severity of radiation-induced intestinal injury, oxidative stress, and the expression of NF-κB in rats with irradiation-induced acute radiation enteritis. The enhanced biological potency of Mono-PEG-hGLP-2 highlights its potential as a therapeutic agent for intestinal diseases.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Lei Shi
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Yuan Xu
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Yang Li
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Shicao Li
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Bo Guan
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Zhihua Qi
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Ye Zhang
- Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| | - Linna Liu
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, PR China
| |
Collapse
|
16
|
Secombe KR, Coller JK, Gibson RJ, Wardill HR, Bowen JM. The bidirectional interaction of the gut microbiome and the innate immune system: Implications for chemotherapy‐induced gastrointestinal toxicity. Int J Cancer 2018; 144:2365-2376. [DOI: 10.1002/ijc.31836] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/14/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Kate R. Secombe
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
| | - Janet K. Coller
- Cancer Treatment Toxicities Group, Discipline of PharmacologyAdelaide Medical School, University of Adelaide Adelaide South Australia Australia
| | - Rachel J. Gibson
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
- Division of Health SciencesUniversity of South Australia Adelaide South Australia Australia
| | - Hannah R. Wardill
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
- Department of Pediatric Oncology/Hematology, University of Groningen, Beatrix Children's HospitalUniversity Medical Center Groningen Groningen The Netherlands
| | - Joanne M. Bowen
- Cancer Treatment Toxicities Group, Discipline of Physiology, Adelaide Medical SchoolUniversity of Adelaide Adelaide South Australia Australia
| |
Collapse
|
17
|
Stojanovska V, McQuade RM, Fraser S, Prakash M, Gondalia S, Stavely R, Palombo E, Apostolopoulos V, Sakkal S, Nurgali K. Oxaliplatin-induced changes in microbiota, TLR4+ cells and enhanced HMGB1 expression in the murine colon. PLoS One 2018; 13:e0198359. [PMID: 29894476 PMCID: PMC5997344 DOI: 10.1371/journal.pone.0198359] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 05/17/2018] [Indexed: 02/07/2023] Open
Abstract
Oxaliplatin is a platinum-based chemotherapeutic used for cancer treatment. Its use associates with peripheral neuropathies and chronic gastrointestinal side-effects. Oxaliplatin induces immunogenic cell death by provoking the presentation of damage associated molecular patterns. The damage associated molecular patterns high-mobility group box 1 (HMGB1) protein exerts pro-inflammatory cytokine-like activity and binds to toll-like receptors (namely TLR4). Gastrointestinal microbiota may influence chemotherapeutic efficacy and contribute to local and systemic inflammation. We studied effects of oxaliplatin treatment on 1) TLR4 and high-mobility group box 1 expression within the colon; 2) gastrointestinal microbiota composition; 3) inflammation within the colon; 4) changes in Peyer's patches and mesenteric lymph nodes immune populations in mice. TLR4+ cells displayed pseudopodia-like extensions characteristic of antigen sampling co-localised with high-mobility group box 1 -overexpressing cells in the colonic lamina propria from oxaliplatin-treated animals. Oxaliplatin treatment caused significant reduction in Parabacteroides and Prevotella1, but increase in Prevotella2 and Odoribacter bacteria at the genus level. Downregulation of pro-inflammatory cytokines and chemokines in colon samples, a reduction in macrophages and dendritic cells in mesenteric lymph nodes were found after oxaliplatin treatment. In conclusion, oxaliplatin treatment caused morphological changes in TLR4+ cells, increase in gram-negative microbiota and enhanced HMGB1 expression associated with immunosuppression in the colon.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Rachel M. McQuade
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Sarah Fraser
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Monica Prakash
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Shakuntla Gondalia
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, Melbourne, Victoria, Australia
| | - Rhian Stavely
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Enzo Palombo
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Melbourne, Victoria, Australia
| | - Vasso Apostolopoulos
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Samy Sakkal
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
- Department of Medicine Western Health, The University of Melbourne, Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Stojanovska V, McQuade RM, Miller S, Nurgali K. Effects of Oxaliplatin Treatment on the Myenteric Plexus Innervation and Glia in the Murine Distal Colon. J Histochem Cytochem 2018; 66:723-736. [PMID: 29741434 DOI: 10.1369/0022155418774755] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Oxaliplatin (platinum-based chemotherapeutic agent) is a first-line treatment of colorectal malignancies; its use associates with peripheral neuropathies and gastrointestinal side effects. These gastrointestinal dysfunctions might be due to toxic effects of oxaliplatin on the intestinal innervation and glia. Male Balb/c mice received intraperitoneal injections of sterile water or oxaliplatin (3 mg/kg/d) triweekly for 2 weeks. Colon tissues were collected for immunohistochemical assessment at day 14. The density of sensory, adrenergic, and cholinergic nerve fibers labeled with calcitonin gene-related peptide (CGRP), tyrosine hydroxylase (TH), and vesicular acetylcholine transporter (VAChT), respectively, was assessed within the myenteric plexus of the distal colon. The number and proportion of excitatory neurons immunoreactive (IR) against choline acetyltransferase (ChAT) were counted, and the density of glial subpopulations was determined by using antibodies specific for glial fibrillary acidic protein (GFAP) and s100β protein. Oxaliplatin treatment induced significant reduction of sensory and adrenergic innervations, as well as the total number and proportion of ChAT-IR neurons, and GFAP-IR glia, but increased s100β expression within the myenteric plexus of the distal colon. Treatment with oxaliplatin significantly alters nerve fibers and glial cells in the colonic myenteric plexus, which could contribute to long-term gastrointestinal side effects following chemotherapeutic treatment.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Rachel M McQuade
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Sarah Miller
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia.,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Ng HS, Koczwara B, Roder D, Vitry A. Changes in the prevalence of comorbidity in the Australian population with cancer, 2007-2014. Cancer Epidemiol 2018; 54:56-62. [PMID: 29597133 DOI: 10.1016/j.canep.2018.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Coexistence of multiple chronic diseases is highly prevalent among the cancer population. This study aims to assess changes in the prevalence of chronic conditions among the population with cancer compared to the Australian general population between 2007 and 2014. METHODS Data from three successive National Health Surveys conducted by the Australian Bureau of Statistics between 2007 and 2014 were utilized. Comparisons were made between the samples of the Australian population aged 25 years and above with a history of cancer and those respondents who did not report having had a cancer using logistics regression models. RESULTS People with a history of cancer had significantly higher odds of reporting non-infectious comorbidity compared to the non-cancer groups across the three surveys. There were no significant changes in the prevalence of diseases affecting circulatory, musculoskeletal, digestive, nervous system, blood and blood forming organs, eye, skin and infectious and parasitic diseases over time among the population with cancer. The prevalence of mental and behavioural problems, endocrine, nutritional and metabolic diseases, and diseases of respiratory and genitourinary system has increased over time among the cancer survivors. CONCLUSION Comorbidity is more prevalent among the cancer population than the general population without cancer. The prevalence of comorbidity was fairly stable for most but not all comorbidities in the population with cancer over the eight-year study period. Further studies on the impacts of coordinated care models for the management of multi-morbidity experienced by cancer survivors that align with the 'National Strategic Framework for Chronic Conditions' are needed.
Collapse
Affiliation(s)
- Huah Shin Ng
- School of Pharmacy and Medical Sciences, University of South Australia, Australia.
| | - Bogda Koczwara
- Flinders Centre for Innovation in Cancer, Flinders University, Australia
| | - David Roder
- Cancer Epidemiology and Population Health, Centre of Population Health Research, School of Health Sciences, University of South Australia, Australia
| | - Agnes Vitry
- School of Pharmacy and Medical Sciences, University of South Australia, Australia
| |
Collapse
|
20
|
Bonnesen TG, Winther JF, Andersen KK, Asdahl PH, de Fine Licht S, Gudmundsdottir T, Sällfors Holmqvist A, Madanat-Harjuoja LM, Tryggvadottir L, Wesenberg F, Heilmann C, Olsen JH, Hasle H. Liver diseases in Adult Life after Childhood Cancer in Scandinavia (ALiCCS): A population-based cohort study of 32,839 one-year survivors. Int J Cancer 2017; 142:702-708. [PMID: 29023764 DOI: 10.1002/ijc.31093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 09/15/2017] [Accepted: 09/20/2017] [Indexed: 12/25/2022]
Abstract
Information on late onset liver complications after childhood cancer is scarce. To ensure an appropriate follow-up of childhood cancer survivors and reducing late liver complications, the need for comprehensive and accurate information is presented. We evaluate the risk of liver diseases in a large childhood cancer survivor cohort. We included all 1-year survivors of childhood cancer treated in the five Nordic countries. A Cox proportional hazards model was used to estimate hospitalisation rate (hazard) ratios (HRs) for each liver outcome according to type of cancer. We used the risk among survivors of central nervous system tumour as internal reference. With a median follow-up time of 10 years, 659 (2%) survivors had been hospitalised at least once for a liver disease. The risk for hospitalisation for any liver disease was high after hepatic tumour (HR = 6.9) and leukaemia (HR = 1.7). The Danish sub-cohort of leukaemia treated with haematopoietic stem cell transplantation had a substantially higher risk for hospitalisation for all liver diseases combined (HR = 3.8). Viral hepatitis accounted for 286 of 659 hospitalisations corresponding to 43% of all survivors hospitalised for liver disease. The 20-year cumulative risk of viral hepatitis was 1.8% for survivors diagnosed with cancer before 1990 but only 0.3% for those diagnosed after 1990. The risk of liver disease was low but significantly increased among survivors of hepatic tumours and leukaemia. Further studies with focus on the different treatment modalities are needed to further strengthen the prevention of treatment-induced late liver complications.
Collapse
Affiliation(s)
- Trine Gade Bonnesen
- Department of Paediatrics, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Jeanette F Winther
- Danish Cancer Society Research Center, Strandboulevarden 49, Copenhagen, 2100, Denmark
| | - Klaus K Andersen
- Danish Cancer Society Research Center, Strandboulevarden 49, Copenhagen, 2100, Denmark
| | - Peter H Asdahl
- Department of Paediatrics, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Sofie de Fine Licht
- Danish Cancer Society Research Center, Strandboulevarden 49, Copenhagen, 2100, Denmark
| | | | - Anna Sällfors Holmqvist
- Department of Clinical Sciences, Paediatric Oncology and Haematology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Laura-Maria Madanat-Harjuoja
- Finnish Cancer Registry, Pieni Roobertinkatu 9, Helsinki, 00130, Finland.,Department of Paediatrics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Laufey Tryggvadottir
- Faculty of Medicine, Laeknagardur, University of Iceland, Reykjavik, Iceland.,The Icelandic Cancer Registry, Skogarhlid 9, PO Box 5420, Reykjavik, 125, Iceland
| | - Finn Wesenberg
- Cancer Registry of Norway, P.O. Box 5313, Majorstuen, Oslo, Norway.,Department of Paediatrics, Oslo University Hospital, Postboks 4950 Nydalen, 0424, Oslo.,Medical Faculty, University of Oslo, P.O box 1078, Blindern, 0316, Oslo
| | - Carsten Heilmann
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark
| | - Jørgen H Olsen
- Danish Cancer Society Research Center, Strandboulevarden 49, Copenhagen, 2100, Denmark
| | - Henrik Hasle
- Department of Paediatrics, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | | |
Collapse
|
21
|
Gangadharan A, Choi SE, Hassan A, Ayoub NM, Durante G, Balwani S, Kim YH, Pecora A, Goy A, Suh KS. Protein calorie malnutrition, nutritional intervention and personalized cancer care. Oncotarget 2017; 8:24009-24030. [PMID: 28177923 PMCID: PMC5410360 DOI: 10.18632/oncotarget.15103] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/23/2017] [Indexed: 12/27/2022] Open
Abstract
Cancer patients often experience weight loss caused by protein calorie malnutrition (PCM) during the course of the disease or treatment. PCM is expressed as severe if the patient has two or more of the following characteristics: obvious significant muscle wasting, loss of subcutaneous fat; nutritional intake of <50% of recommended intake for 2 weeks or more; bedridden or otherwise significantly reduced functional capacity; weight loss of >2% in 1 week, 5% in 1 month, or 7.5% in 3 months. Cancer anorexia-cachexia syndrome (CACS) is a multifactorial condition of advanced PCM associated with underlying illness (in this case cancer) and is characterized by loss of muscle with or without loss of fat mass. Cachexia is defined as weight loss of more than 5% of body weight in 12 months or less in the presence of chronic disease. Hence with a chronic illness on board even a small amount of weight loss can open the door to cachexia. These nutritional challenges can lead to severe morbidity and mortality in cancer patients. In the clinic, the application of personalized medicine and the ability to withstand the toxic effects of anti-cancer therapies can be optimized when the patient is in nutritional homeostasis and is free of anorexia and cachexia. Routine assessment of nutritional status and appropriate intervention are essential components of the effort to alleviate effects of malnutrition on quality of life and survival of patients.
Collapse
Affiliation(s)
- Anju Gangadharan
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Sung Eun Choi
- Department of Family, Nutrition, and Exercise Sciences, Queens College, The City University of New York, Flushing, NY, USA
| | - Ahmed Hassan
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Gina Durante
- Department of Clinical Nutrition, Baystate Medical Center, Springfield, MA, USA
| | - Sakshi Balwani
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Young Hee Kim
- Department of Clinical Nutrition, Baystate Medical Center, Springfield, MA, USA
| | - Andrew Pecora
- Clinical Divisions, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Andre Goy
- Clinical Divisions, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - K Stephen Suh
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| |
Collapse
|
22
|
|
23
|
Uranga JA, García-Martínez JM, García-Jiménez C, Vera G, Martín-Fontelles MI, Abalo R. Alterations in the small intestinal wall and motor function after repeated cisplatin in rat. Neurogastroenterol Motil 2017; 29. [PMID: 28261911 DOI: 10.1111/nmo.13047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/12/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Gastrointestinal adverse effects occurring during cancer chemotherapy are well known and feared; those persisting once treatment has finished are relatively unknown. We characterized the alterations occurring in the rat small intestine, after repeated treatment with cisplatin. METHODS Male Wistar rats received saline or cisplatin (2 mg kg-1 week-1 , for 5 weeks, ip). Gastric motor function was studied non-invasively throughout treatment (W1-W5) and 1 week after treatment finalization (W6). During W6, upper gastrointestinal motility was also invasively studied and small intestinal samples were collected for histopathological and molecular studies. Structural alterations in the small intestinal wall, mucosa, submucosa, muscle layers, and lymphocytic nodules were histologically studied. Periodic acid-Schiff staining and immunohistochemistry for Ki-67, chromogranin A, and neuronal-specific enolase were used to detect secretory, proliferating, endocrine and neural cells, respectively. The expression of different markers in the tunica muscularis was analyzed by RT/qPCR. KEY RESULTS Repeated cisplatin induced motility alterations during and after treatment. After treatment (W6), the small intestinal wall showed histopathological alterations in most parameters measured, including a reduction in the thickness of circular and longitudinal muscle layers. Expression of c-KIT (for interstitial cells of Cajal), nNOS (for inhibitory motor neurons), pChAT, and cChAT (for excitatory motor neurons) increased significantly (although both ChATs to a lesser extent). CONCLUSIONS & INFERENCES Repeated cisplatin induces relatively long-lasting gut dysmotility in rat associated with important histopathological and molecular alterations in the small intestinal wall. In cancer survivors, the possible chemotherapy-induced histopathological, molecular, and functional intestinal sequelae should be evaluated.
Collapse
Affiliation(s)
- J A Uranga
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain
| | - J M García-Martínez
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo de Compuestos químicos y materiales nanoestructurados con aplicaciones Avanzadas (QUINANOAP), Madrid, Spain
| | - C García-Jiménez
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo de Compuestos químicos y materiales nanoestructurados con aplicaciones Avanzadas (QUINANOAP), Madrid, Spain
| | - G Vera
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Madrid, Spain
| | - M I Martín-Fontelles
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Madrid, Spain
| | - R Abalo
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Madrid, Spain
| |
Collapse
|
24
|
Scarpignato C, Gatta L, Zullo A, Blandizzi C. Effective and safe proton pump inhibitor therapy in acid-related diseases - A position paper addressing benefits and potential harms of acid suppression. BMC Med 2016; 14:179. [PMID: 27825371 PMCID: PMC5101793 DOI: 10.1186/s12916-016-0718-z] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 10/14/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The introduction of proton pump inhibitors (PPIs) into clinical practice has revolutionized the management of acid-related diseases. Studies in primary care and emergency settings suggest that PPIs are frequently prescribed for inappropriate indications or for indications where their use offers little benefit. Inappropriate PPI use is a matter of great concern, especially in the elderly, who are often affected by multiple comorbidities and are taking multiple medications, and are thus at an increased risk of long-term PPI-related adverse outcomes as well as drug-to-drug interactions. Herein, we aim to review the current literature on PPI use and develop a position paper addressing the benefits and potential harms of acid suppression with the purpose of providing evidence-based guidelines on the appropriate use of these medications. METHODS The topics, identified by a Scientific Committee, were assigned to experts selected by three Italian Scientific Societies, who independently performed a systematic search of the relevant literature using Medline/PubMed, Embase, and the Cochrane databases. Search outputs were distilled, paying more attention to systematic reviews and meta-analyses (where available) representing the best evidence. The draft prepared on each topic was circulated amongst all the members of the Scientific Committee. Each expert then provided her/his input to the writing, suggesting changes and the inclusion of new material and/or additional relevant references. The global recommendations were then thoroughly discussed in a specific meeting, refined with regard to both content and wording, and approved to obtain a summary of current evidence. RESULTS Twenty-five years after their introduction into clinical practice, PPIs remain the mainstay of the treatment of acid-related diseases, where their use in gastroesophageal reflux disease, eosinophilic esophagitis, Helicobacter pylori infection, peptic ulcer disease and bleeding as well as, and Zollinger-Ellison syndrome is appropriate. Prevention of gastroduodenal mucosal lesions (and symptoms) in patients taking non-steroidal anti-inflammatory drugs (NSAIDs) or antiplatelet therapies and carrying gastrointestinal risk factors also represents an appropriate indication. On the contrary, steroid use does not need any gastroprotection, unless combined with NSAID therapy. In dyspeptic patients with persisting symptoms, despite successful H. pylori eradication, short-term PPI treatment could be attempted. Finally, addition of PPIs to pancreatic enzyme replacement therapy in patients with refractory steatorrhea may be worthwhile. CONCLUSIONS Overall, PPIs are irreplaceable drugs in the management of acid-related diseases. However, PPI treatment, as any kind of drug therapy, is not without risk of adverse effects. The overall benefits of therapy and improvement in quality of life significantly outweigh potential harms in most patients, but those without clear clinical indication are only exposed to the risks of PPI prescription. Adhering with evidence-based guidelines represents the only rational approach to effective and safe PPI therapy. Please see related Commentary: doi: 10.1186/s12916-016-0724-1 .
Collapse
Affiliation(s)
- Carmelo Scarpignato
- Clinical Pharmacology & Digestive Pathophysiology Unit, Department of Clinical & Experimental Medicine, University of Parma, Maggiore University Hospital, Cattani Pavillon, I-43125, Parma, Italy.
| | - Luigi Gatta
- Clinical Pharmacology & Digestive Pathophysiology Unit, Department of Clinical & Experimental Medicine, University of Parma, Maggiore University Hospital, Cattani Pavillon, I-43125, Parma, Italy
- Gastroenterology & Endoscopy Unit, Versilia Hospital, Azienda USL Toscana Nord Ovest, Lido di Camaiore, Italy
| | - Angelo Zullo
- Division of Gastroenterology & Digestive Endoscopy, Nuovo Regina Elena Hospital, Rome, Italy
| | - Corrado Blandizzi
- Division of Pharmacology, Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
25
|
Robinson AM, Stojanovska V, Rahman AA, McQuade RM, Senior PV, Nurgali K. Effects of Oxaliplatin Treatment on the Enteric Glial Cells and Neurons in the Mouse Ileum. J Histochem Cytochem 2016; 64:530-45. [PMID: 27389702 PMCID: PMC5006136 DOI: 10.1369/0022155416656842] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/06/2016] [Indexed: 12/17/2022] Open
Abstract
Oxaliplatin, currently used for treatment of colorectal and other cancers, causes severe gastrointestinal side effects, including nausea, vomiting, diarrhea, and constipation that are attributed to mucosal damage. However, delayed onset and long-term persistence of these side effects suggest that damage to the enteric nervous system (ENS) regulating physiological function of the gastrointestinal tract may also occur. The ENS comprises myenteric and submucosal neurons and enteric glial cells (EGCs). This study aimed to investigate the effects of oxaliplatin treatment on enteric neurons and EGCs within the mouse ileum. BALB/c mice received repeated intraperitoneal injections of oxaliplatin (3 mg/kg, 3 injections/week). Tissues were collected 3, 7, 14, and 21 days from the commencement of treatment. Decreases in glial fibrillary acidic protein-immunoreactive (IR) EGCs and protein gene product 9.5/β-Tubulin III-IR neurons as well as increase in s100β-IR EGCs after chronic oxaliplatin administration were observed in both the myenteric and submucosal plexi. Changes in EGCs were further observed in cross-sections of the ileum at day 14 and confirmed by Western blotting. Alterations in EGCs correlated with loss of myenteric and submucosal neurons in the ileum from oxaliplatin-treated mice. These changes to the ENS may contribute to the mechanisms underlying gastrointestinal side effects associated with oxaliplatin treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Kulmira Nurgali
- Kulmira Nurgali, Western Centre for Health Research & Education, Sunshine Hospital, 176 Furlong Road, St Albans, VIC 3021, Australia.
| |
Collapse
|
26
|
Mashtoub S, Lampton LS, Eden GL, Cheah KY, Lymn KA, Bajic JE, Howarth GS. Emu Oil Combined with Lyprinol™ Reduces Small Intestinal Damage in a Rat Model of Chemotherapy-Induced Mucositis. Nutr Cancer 2016; 68:1171-80. [DOI: 10.1080/01635581.2016.1208829] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
27
|
Riyaz A, Nazir S, Khushtar M, Mishra A, Jahan Y, Ahmad A. Effect of Nigella sativa L. seed extract on cisplatin-induced delay in gastric emptying in Sprague-Dawley rats. Nat Prod Res 2016; 31:588-592. [PMID: 27348571 DOI: 10.1080/14786419.2016.1201670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aim of this study was focused on investigating the possible protective effect of Nigella sativa L. seed extract against cisplatin-induced delay in gastric emptying, in a rat model. Twenty-five male Sprague-Dawley rats were divided into five equal groups as follows: Group I or control group, Group II (cisplatin 10 mg/kg, i.p at day 5), Group III (N. sativa L. 250 mg/kg for 5 days + cisplatin 10 mg/kg, i.p on day 5), Group IV (N. sativa L. 500 mg/kg for 5 days + cisplatin 10 mg/kg, i.p on day 5) and Group V (ondansetron 3 mg/kg/day, per os + cisplatin 10 mg/kg, i.p on day 5). Phenol red meal was adopted to estimate gastric emptying in different groups of the rats. Gastric emptying was significantly increased (p < 0.01) in N. sativa L. seed extract-pretreated rats (Group III and Group IV) when compared to cisplatin treatment alone (Group II). However, ondansetron produced significantly (p < 0.01) better reversal than N. sativa L. seed extract.
Collapse
Affiliation(s)
- Ambreena Riyaz
- a Faculty of Pharmacy , Integral University , Lucknow , India
| | - Shahid Nazir
- b School of Veterinary Medicine , Wollo University , Dessie , Ethiopia
| | | | - Anuradha Mishra
- a Faculty of Pharmacy , Integral University , Lucknow , India
| | - Yasmeen Jahan
- a Faculty of Pharmacy , Integral University , Lucknow , India
| | - Asad Ahmad
- a Faculty of Pharmacy , Integral University , Lucknow , India
| |
Collapse
|
28
|
Asdahl PH, Winther JF, Bonnesen TG, De Fine Licht S, Gudmundsdottir T, Holmqvist AS, Malila N, Tryggvadottir L, Wesenberg F, Dahlerup JF, Olsen JH, Hasle H. Gastrointestinal and liver disease in Adult Life After Childhood Cancer in Scandinavia: A population-based cohort study. Int J Cancer 2016; 139:1501-11. [PMID: 27194488 DOI: 10.1002/ijc.30198] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/25/2016] [Accepted: 05/04/2016] [Indexed: 01/01/2023]
Abstract
Survival after childhood cancer diagnosis has remarkably improved, but emerging evidence suggests that cancer-directed therapy may have adverse gastrointestinal late effects. We aimed to comprehensively assess the frequency of gastrointestinal and liver late effects among childhood cancer survivors and compare this frequency with the general population. Our population-based cohort study included all 1-year survivors of childhood and adolescent cancer in Denmark, Finland, Iceland, Norway and Sweden diagnosed from the 1940s and 1950s. Our outcomes of interest were hospitalization rates for gastrointestinal and liver diseases, which were ascertained from national patient registries. We calculated standardized hospitalization rate ratios (RRs) and absolute excess rates comparing hospitalizations of any gastrointestinal or liver disease and for specific disease entities between survivors and the general population. The study included 31,132 survivors and 207,041 comparison subjects. The median follow-up in the hospital registries were 10 years (range: 0-42) with 23% of the survivors being followed at least to the age of 40 years. Overall, survivors had a 60% relative excess of gastrointestinal or liver diseases [RR: 1.6, 95% confidence interval (CI): 1.6-1.7], which corresponds to an absolute excess of 360 (95% CI: 330-390) hospitalizations per 100,000 person-years. Survivors of hepatic tumors, neuroblastoma and leukemia had the highest excess of gastrointestinal and liver diseases. In addition, we observed a relative excess of several specific diseases such as esophageal stricture (RR: 13; 95% CI: 9.2-20) and liver cirrhosis (RR: 2.9; 95% CI: 2.0-4.1). Our findings provide useful information about the breadth and magnitude of late complications among childhood cancer survivors and can be used for generating hypotheses about potential exposures related to these gastrointestinal and liver late effects.
Collapse
Affiliation(s)
- Peter Haubjerg Asdahl
- Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Trine Gade Bonnesen
- Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | | | - Anna Sällfors Holmqvist
- Department of Clinical Sciences, Pediatric Oncology and Hematology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Nea Malila
- The Finnish Cancer Registry, Helsinki, Finland
| | - Laufey Tryggvadottir
- The Icelandic Cancer Registry, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Henrik Hasle
- Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
29
|
Saxena R, Rida PCG, Kucuk O, Aneja R. Ginger augmented chemotherapy: A novel multitarget nontoxic approach for cancer management. Mol Nutr Food Res 2016; 60:1364-73. [PMID: 26842968 DOI: 10.1002/mnfr.201500955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/11/2016] [Accepted: 01/12/2016] [Indexed: 01/22/2023]
Abstract
Cancer, referred to as the 'disease of civilization', continues to haunt humanity due to its dreadful manifestations and limited success of therapeutic interventions such as chemotherapy in curing the disease. Although effective, chemotherapy has repeatedly demonstrated inadequacy in disease management due to its debilitating side effects arising from its deleterious nonspecific effects on normal healthy cells. In addition, development of chemoresistance due to mono-targeting often results in cessation of chemotherapy. This urgently demands development and implementation of multitargeted alternative therapies with mild or no side effects. One extremely promising strategy that yet remains untapped in the clinic is augmenting chemotherapy with dietary phytochemicals or extracts. Ginger, depository of numerous bioactive molecules, not only targets cancer cells but can also mitigate chemotherapy-associated side effects. Consequently, combination therapy involving ginger extract and chemotherapeutic agents may offer the advantage of being efficacious with reduced toxicity. Here we discuss the remarkable and often overlooked potential of ginger extract to manage cancer, the possibility of developing ginger-based combinational therapies, and the major roadblocks along with strategies to overcome them in clinical translation of such inventions. We are optimistic that clinical implementation of such combination regimens would be a much sought after modality in cancer management.
Collapse
Affiliation(s)
- Roopali Saxena
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | | | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
30
|
Pini A, Garella R, Idrizaj E, Calosi L, Baccari MC, Vannucchi MG. Glucagon-like peptide 2 counteracts the mucosal damage and the neuropathy induced by chronic treatment with cisplatin in the mouse gastric fundus. Neurogastroenterol Motil 2016; 28:206-16. [PMID: 26547262 DOI: 10.1111/nmo.12712] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Glucagon-like peptide-2 (GLP-2) is a pleiotropic hormone synthesized and secreted by the enteroendocrine 'L' cells able to exert intestine-trophic and anti-inflammatory effects. The antineoplastic drug cisplatin causes gastrointestinal alterations with clinical symptoms (nausea and vomiting) that greatly affect the therapy compliance. Experimentally, it has been reported that chronic cisplatin treatment caused mucosal damage and enteric neuropathy in the rat colon. METHODS We investigated, through a combined immunohistochemical and functional approach, whether [Gly(2) ]GLP-2, a GLP-2 analog, was able to counteract the detrimental effects of long-term cisplatin administration in the mucosa and myenteric neurons of mouse gastric fundus. KEY RESULTS Morphological experiments showed a reduction in the epithelium thickness in cisplatin-treated mice, which was prevented by [Gly(2) ]GLP-2 co-treatment. Immunohistochemistry demonstrated that cisplatin caused a significant decrease in myenteric neurons, mainly those expressing neuronal nitric oxide synthase (nNOS), that was prevented by [Gly(2) ]GLP-2 co-treatment. In the functional experiments, [Gly(2) ]GLP-2 co-treatment counteracted the increase in amplitude of the neurally induced contractions observed in strips from cisplatin-treated animals. The NO synthesis inhibitor L-N(G) -nitro arginine caused an increase in amplitude of the contractile responses that was greater in preparations from cisplatin+[Gly(2) ]GLP-2 treated mice compared to the cisplatin-treated ones. CONCLUSIONS & INFERENCES The results demonstrate that in cisplatin long-term treated mice [Gly(2) ]GLP-2 is able to counteract both the mucosal gastric fundus damage, by preventing the epithelium thickness decrease, and the neuropathy, by protecting the nNOS neurons. Taken together, the present data suggest that [Gly(2) ]GLP-2 could represent an effective strategy to overcome the distressing gastrointestinal symptoms present during the anti-neoplastic therapy.
Collapse
Affiliation(s)
- A Pini
- Department of Experimental and Clinical Medicine, Histology and Embryology Research Unit, University of Florence, Florence, Italy
| | - R Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - E Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - L Calosi
- Department of Experimental and Clinical Medicine, Histology and Embryology Research Unit, University of Florence, Florence, Italy
| | - M C Baccari
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - M G Vannucchi
- Department of Experimental and Clinical Medicine, Histology and Embryology Research Unit, University of Florence, Florence, Italy
| |
Collapse
|
31
|
Mohaghegh F, Solhi H, Kazemifar AM. Silymarin (Milk Thistle) can revoke liver enzyme changes during chemotherapy of breast cancer with Taxanes. Eur J Integr Med 2015. [DOI: 10.1016/j.eujim.2015.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Kubota M, Shimizu M, Baba A, Ohno T, Kochi T, Shirakami Y, Moriwaki H. Combination of bevacizumab and acyclic retinoid inhibits the growth of hepatocellular carcinoma xenografts. J Nutr Sci Vitaminol (Tokyo) 2015; 60:357-62. [PMID: 25744425 DOI: 10.3177/jnsv.60.357] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The prognosis of patients with hepatocellular carcinoma (HCC) is poor and the development of effective treatments for this malignancy, including combination chemotherapy, is required. This study examined the possible combined inhibitory effects of bevacizumab, an anti-vascular endothelial growth factor monoclonal antibody, and acyclic retinoid (ACR), which can prevent the development of HCC, on the growth of Huh7 human HCC cells. Xenograft tumors were produced by subcutaneously injecting Huh7 cells into nude mice. Starting 1 wk after the tumor cell injection, the mice were treated with bevacizumab alone (5 mg/kg body weight, subcutaneous injection, twice a week), ACR alone (given in a diet containing 0.03%), or their combination for 6 wk, and the effects of these regimens on xenograft growth were examined. Combined treatment with bevacizumab plus ACR significantly suppressed the growth of Huh7 xenografts. The combination of these agents significantly inhibited the phosphorylation of the Akt protein in tumor tissues. With combination therapy, the population of Ki-67-positive cells in xenografts decreased, while that of TUNEL-positive cells increased. The combination of bevacizumab and ACR exerts growth-suppressing effects on HCC cells by inhibiting cell proliferation and inducing apoptosis. This combination might be an effective regimen for the treatment of HCC.
Collapse
Affiliation(s)
- Masaya Kubota
- Department of Gastroenterology/Internal Medicine, Gifu University Graduate School of Medicine
| | | | | | | | | | | | | |
Collapse
|
33
|
Van Sebille YZA, Stansborough R, Wardill HR, Bateman E, Gibson RJ, Keefe DM. Management of Mucositis During Chemotherapy: From Pathophysiology to Pragmatic Therapeutics. Curr Oncol Rep 2015; 17:50. [DOI: 10.1007/s11912-015-0474-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Madenci AL, Fisher S, Diller LR, Goldsby RE, Leisenring WM, Oeffinger KC, Robison LL, Sklar CA, Stovall M, Weathers RE, Armstrong GT, Yasui Y, Weldon CB. Intestinal Obstruction in Survivors of Childhood Cancer: A Report From the Childhood Cancer Survivor Study. J Clin Oncol 2015; 33:2893-900. [PMID: 26261256 DOI: 10.1200/jco.2015.61.5070] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE For adult survivors of childhood cancer, knowledge about the long-term risk of intestinal obstruction from surgery, chemotherapy, and radiotherapy is limited. METHODS Intestinal obstruction requiring surgery (IOS) occurring 5 or more years after cancer diagnosis was evaluated in 12,316 5-year survivors in the Childhood Cancer Survivor Study (2,002 with and 10,314 without abdominopelvic tumors) and 4,023 sibling participants. Cumulative incidence of IOS was calculated with second malignant neoplasm, late recurrence, and death as competing risks. Using piecewise exponential models, we assessed the associations of clinical and demographic factors with rate of IOS. RESULTS Late IOS was reported by 165 survivors (median age at IOS, 19 years; range, 5 to 50 years; median time from diagnosis to IOS, 13 years) and 14 siblings. The cumulative incidence of late IOS at 35 years was 5.8% (95% CI, 4.4% to 7.3%) among survivors with abdominopelvic tumors, 1.0% (95% CI, 0.7% to 1.4%) among those without abdominopelvic tumors, and 0.3% (95% CI, 0.1% to 0.5%) among siblings. Among survivors, abdominopelvic tumor (adjusted rate ratio [ARR], 3.6; 95% CI, 1.9 to 6.8; P < .001) and abdominal/pelvic radiotherapy within 5 years of cancer diagnosis (ARR, 2.4; 95% CI, 1.6 to 3.7; P < .001) increased the rate of late IOS, adjusting for diagnosis year; sex; race/ethnicity; age at diagnosis; age during follow-up (as natural cubic spline); cancer type; and chemotherapy, radiotherapy, and surgery within 5 years of cancer diagnosis. Developing late IOS increased subsequent mortality among survivors (ARR, 1.8; 95% CI, 1.1 to 2.9; P = .016), adjusting for the same factors. CONCLUSION The long-term risk of IOS and its association with subsequent mortality underscore the need to promote awareness of this complication among patients and providers.
Collapse
Affiliation(s)
- Arin L Madenci
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stacey Fisher
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lisa R Diller
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert E Goldsby
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wendy M Leisenring
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kevin C Oeffinger
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Leslie L Robison
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Charles A Sklar
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Marilyn Stovall
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rita E Weathers
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gregory T Armstrong
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yutaka Yasui
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christopher B Weldon
- Arin L. Madenci and Christopher B. Weldon, Boston Children's Hospital and Harvard Medical School; Arin L. Madenci, Brigham and Women's Hospital and Harvard Medical School; Lisa R. Diller and Christopher B. Weldon, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Stacey Fisher and Yutaka Yasui, University of Alberta, Edmonton, Alberta, Canada; Robert E. Goldsby, School of Medicine, University of California, San Francisco, San Francisco, CA; Wendy M. Leisenring, Fred Hutchinson Cancer Research Center, Seattle, WA; Kevin C. Oeffinger and Charles A. Sklar, Memorial Sloan-Kettering Cancer Center, New York, NY; Leslie L. Robison and Gregory T. Armstrong, St Jude Children's Research Hospital, Memphis, TN; and Marilyn Stovall and Rita E. Weathers, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
35
|
ErbB small molecule tyrosine kinase inhibitor (TKI) induced diarrhoea: Chloride secretion as a mechanistic hypothesis. Cancer Treat Rev 2015; 41:646-52. [PMID: 26073491 DOI: 10.1016/j.ctrv.2015.05.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 12/27/2022]
Abstract
Diarrhoea is a common, debilitating and potentially life threatening toxicity of many cancer therapies. While the mechanisms of diarrhoea induced by traditional chemotherapy have been the focus of much research, the mechanism(s) of diarrhoea induced by small molecule ErbB TKI, have received relatively little attention. Given the increasing use of small molecule ErbB TKIs, identifying this mechanism is key to optimal cancer care. This paper critically reviews the literature and forms a hypothesis that diarrhoea induced by small molecule ErbB TKIs is driven by intestinal chloride secretion based on the negative regulation of chloride secretion by ErbB receptors being disrupted by tyrosine kinase inhibition.
Collapse
|
36
|
Fischer-Cartlidge EA. Assessment and management of gastrointestinal toxicities and lab abnormalities related to targeted therapy. Semin Oncol Nurs 2015; 30:183-9. [PMID: 25085030 DOI: 10.1016/j.soncn.2014.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To identify the most common gastrointestinal toxicities and laboratory abnormalities associated with targeted therapies, as well as the assessment and management necessary to minimize adverse events related to these side effects. DATA SOURCES Peer-reviewed articles and national guidelines for oncology practice. CONCLUSION Common toxicities of diarrhea, mucositis, and laboratory abnormalities are often associated with the use of targeted agents and require skilled assessment and early management interventions to prevent severe complications or treatment interruption. IMPLICATIONS FOR NURSING PRACTICE Emerging trends focused on targeted therapy increase the importance of the oncology nurse's role in assessment, education, and evidenced-based recommendations to meet patient needs.
Collapse
|
37
|
The age factor for mitoxantrone’s cardiotoxicity: Multiple doses render the adult mouse heart more susceptible to injury. Toxicology 2015; 329:106-19. [PMID: 25582955 DOI: 10.1016/j.tox.2015.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/04/2015] [Accepted: 01/08/2015] [Indexed: 11/30/2022]
|
38
|
Stojanovska V, Sakkal S, Nurgali K. Platinum-based chemotherapy: gastrointestinal immunomodulation and enteric nervous system toxicity. Am J Physiol Gastrointest Liver Physiol 2015; 308:G223-32. [PMID: 25501548 DOI: 10.1152/ajpgi.00212.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The efficacy of chemotherapeutic treatment of colorectal cancer is challenged by severe gastrointestinal side effects, which include nausea, vomiting, constipation, and diarrhea. These symptoms can persist long after the treatment has been ceased. An emerging concept is the ability of platinum-based drugs to stimulate immunity, which is in contrast to conventional chemotherapeutic agents that are immunosuppressive. Here, we review the immunomodulatory aspects of platinum-based anticancer chemotherapeutics and their impact on gastrointestinal innervation. Given the bidirectional communication between the enteric nervous system and gastrointestinal immune system; exploring the consequences of platinum-induced immunogenicity will facilitate better understanding of gut dysfunction caused by chemotherapeutic agents. We propose that the development of future successful chemotherapeutics should rely on targeting the mechanisms underlying long-term gastrointestinal side effects.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- College of Health and Biomedicine, Victoria University, Western Centre for Health, Research and Education, St Albans, Victoria, Australia
| | - Samy Sakkal
- College of Health and Biomedicine, Victoria University, Western Centre for Health, Research and Education, St Albans, Victoria, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Victoria University, Western Centre for Health, Research and Education, St Albans, Victoria, Australia
| |
Collapse
|
39
|
X-ray analysis of the effect of the 5-HT3 receptor antagonist granisetron on gastrointestinal motility in rats repeatedly treated with the antitumoral drug cisplatin. Exp Brain Res 2014; 232:2601-12. [DOI: 10.1007/s00221-014-3954-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
|
40
|
SalmanOgli A, Rostami A, Faranoush M, Dolatyari M, Rostami G. Enhancement of tumor smart-targeting efficiency based on optical communication between signaling and receiving nanoparticles (modeling and analysis). RSC Adv 2014. [DOI: 10.1039/c4ra02898h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
As far as we know, this is the first time optical communication between signaling and receiving nanoparticles has been analyzed and simulated for tumor smart-targeting.
Collapse
Affiliation(s)
- Ahmad SalmanOgli
- School of Engineering-Emerging Technologies
- University of Tabriz
- Tabriz, Iran
| | - Ali Rostami
- School of Engineering-Emerging Technologies
- University of Tabriz
- Tabriz, Iran
| | | | - Mahbobeh Dolatyari
- School of Engineering-Emerging Technologies
- University of Tabriz
- Tabriz, Iran
| | - Ghassem Rostami
- School of Engineering-Emerging Technologies
- University of Tabriz
- Tabriz, Iran
| |
Collapse
|
41
|
Nicolini A, Ferrari P, Masoni MC, Fini M, Pagani S, Giampietro O, Carpi A. Malnutrition, anorexia and cachexia in cancer patients: A mini-review on pathogenesis and treatment. Biomed Pharmacother 2013; 67:807-17. [DOI: 10.1016/j.biopha.2013.08.005] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/10/2013] [Indexed: 12/17/2022] Open
|
42
|
Wafai L, Taher M, Jovanovska V, Bornstein JC, Dass CR, Nurgali K. Effects of oxaliplatin on mouse myenteric neurons and colonic motility. Front Neurosci 2013; 7:30. [PMID: 23486839 PMCID: PMC3594784 DOI: 10.3389/fnins.2013.00030] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 02/22/2013] [Indexed: 11/13/2022] Open
Abstract
Oxaliplatin, an anti-cancer chemotherapeutic agent used for the treatment of colorectal cancer, commonly causes gastrointestinal side-effects such as constipation, diarrhoea, nausea, and vomiting. Damage to enteric neurons may underlie some of these gastrointestinal side-effects, as the enteric nervous system (ENS) controls functions of the bowel. In this study, neuronal loss and changes to the structure and immunoreactivity of myenteric neuronal nitric oxide synthase (nNOS) neurons were examined in colonic segments from mice following exposure to oxaliplatin ex vivo and following repeated intraperitoneal injections of oxaliplatin over 3 weeks in vivo, using immunohistochemistry and confocal microscopy. Significant morphological alterations and increases in the proportion of NOS-immunoreactive (IR) neurons were associated with both short-term oxaliplatin exposure and long-term oxaliplatin administration, confirming that oxaliplatin causes changes to the myenteric neurons. Long-term oxaliplatin administration induced substantial neuronal loss that was correlated with a reduction in both the frequency and propagation speed of colonic migrating motor complexes (CMMCs) in vitro. Similar changes probably produce some symptoms experienced by patients undergoing oxaliplatin treatment.
Collapse
Affiliation(s)
- Linah Wafai
- College of Health and Biomedicine, Victoria University Melbourne, VIC, Australia
| | | | | | | | | | | |
Collapse
|
43
|
Fang P, Hu JH, Cheng ZG, Liu ZF, Wang JL, Jiao SC. Efficacy and safety of bevacizumab for the treatment of advanced hepatocellular carcinoma: a systematic review of phase II trials. PLoS One 2012; 7:e49717. [PMID: 23284624 PMCID: PMC3526612 DOI: 10.1371/journal.pone.0049717] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 10/12/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common cancer associated with a poor prognosis. Bevacizumab is a monoclonal antibody that binds vascular endothelial growth factor, a mediator of tumor angiogenesis. Bevacizumab is currently under investigation as treatment for HCC. We performed a systematic review of the efficacy and safety of bevacizumab for the treatment of advanced HCC. METHODS PubMed, the Cochrane Library, and Google Scholar were searched using the terms "bevacizumab AND hepatocellular carcinoma AND (advanced OR unresectable)". Phase II trials of bevacizumab for the treatment of advanced HCC were included. Outcomes of interest included progression-free and overall survival (PFS and OS), tumor response, and toxicities. RESULTS A total of 26 records were identified. Of these, 18 were excluded. Hence, eight trials involving 300 patients were included. Bevacizumab was given as monotherapy (n = 1 trial) or in combination with erlotinib (n = 4 trials), capecitabine (n = 1 trial), capecitabine+oxaliplatin (n = 1 trial), or gemcitabine+oxaliplatin (n = 1 trial). Most trials (five of eight) reported median PFS and OS between 5.3 months and 9.0 months and 5.9 and 13.7 months, respectively. The disease control rate was consistent in five of eight trials, ranging from 51.1% to 76.9%. The response and partial response rates ranged from 0 to 23.7%, but were around 20% in four trials. Only one patient had a complete response. Frequently reported Grade 3/4 toxicities were increased aspartate transaminase/alanine transaminase (13%), fatigue (12%), hypertension (10%), diarrhea (8%), and neutropenia (5%). Thirty patients experienced gastrointestinal bleeding (grade 1/2 = 18, grade 3/4 = 12), typically due to esophageal varices. CONCLUSIONS Bevacizumab shows promise as an effective and tolerable treatment for advanced HCC. The reported efficacy of bevacizumab appears to compare favorably with that of sorafenib, the only currently approved treatment for unresectable HCC. Phase III trials are warranted to comprehensively examine the efficacy and safety of bevacizumab for treatment of advanced HCC.
Collapse
Affiliation(s)
- Ping Fang
- Department of Oncology, Institute of Cancer, PLA General Hospital, Beijing, China.
| | | | | | | | | | | |
Collapse
|
44
|
Interventions for reducing diarrhoea in patients receiving chemotherapy for colorectal cancer. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2012. [DOI: 10.1002/14651858.cd009615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
45
|
Zhang LL, Li P, Li YM, Wang AQ. Preparation and characterization of magnetic alginate-chitosan hydrogel beads loaded matrine. Drug Dev Ind Pharm 2011; 38:872-82. [PMID: 22092063 DOI: 10.3109/03639045.2011.630397] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The aim of this study was to use alginate-chitosan (Alg-CS) hydrogel beads for developing an oral water-soluble drug delivery system, occupying pH-sensitive property and superparamagnetic. Matrine as a model drug was loaded in Alg-CS hydrogel beads to study the release character of the delivery system. The amount of matrine released from the beads was relatively low in pH 2.5 over 8 h (34.90%), but nearly all of the initial drug content was released in simulated intestinal fluid (SIF, pH 6.8) within 8 h. The results demonstrated that Alg-CS hydrogel beads possess unique pH-dependent swelling behaviors. In addition, the magnetic beads were characterized by Fourier transform infrared spectroscopy, scanning electron microscope, X-ray diffractometry and vibrating-sample magnetometry. Magnetometer measurements data suggested that Alg-CS beads also had superparamagnetic property as well as fast magnetic response. It can be expected that the beads can deliver and release encapsulated anticancer agent at the tumor by the weak magnetic field, and hence could be potential candidates as an orally administered drug delivery system.
Collapse
|
46
|
Emmink BL, Van Houdt WJ, Vries RG, Hoogwater FJH, Govaert KM, Verheem A, Nijkamp MW, Steller EJA, Jimenez CR, Clevers H, Borel Rinkes IHM, Kranenburg O. Differentiated human colorectal cancer cells protect tumor-initiating cells from irinotecan. Gastroenterology 2011; 141:269-78. [PMID: 21459094 DOI: 10.1053/j.gastro.2011.03.052] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 03/01/2011] [Accepted: 03/15/2011] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Stem cells of normal tissues have resistance mechanisms that allow them to survive genotoxic insults. The stem cell-like cells of tumors are defined by their tumor-initiating capacity and may have retained these resistance mechanisms, making them resistant to chemotherapy. We studied the relationship between resistance to the topoisomerase I inhibitor irinotecan and tumor-initiating potential in human colonosphere cultures and in mice with colorectal xenograft tumors. METHODS Colonosphere cultures were established from human colorectal tumor specimens obtained from patients who underwent colon or liver resection for primary or metastatic adenocarcinoma. Stem cell and differentiation markers were analyzed by immunoblotting and fluorescence-activated cell sorting. Clone- and tumor-initiating capacities were assessed by single-cell cloning and in immune-deficient mice. Sensitivity to irinotecan was assessed in vitro and in tumor-bearing mice. The relationship between drug resistance and tumor-initiating capacity was tested by fluorescence-activated cell sorting of colonosphere cells, based on expression of ABCB1 and aldehyde dehydrogenase (ALDH) activity. RESULTS Colonosphere cultures had a high capacity to initiate tumors in mice and were resistant to irinotecan. Inhibition of the drug-efflux pump ABCB1 by PSC-833 allowed irinotecan to eradicate tumor-initiating cells. However, ABCB1 was expressed only by a subpopulation of differentiated tumor cells that did not form clones or tumors. Conversely, tumor-initiating cells were ABCB1-negative and were identified by high ALDH activity. Tumorigenic ALDHhigh/ABCB1negative cells generated nontumorigenic ALDHlow/ABCB1positive daughter cells in vitro and in tumor xenografts. PSC-833 increased the antitumor efficacy of irinotecan in mice. CONCLUSIONS The resistance of colorectal tumors to irinotecan requires the cooperative action of tumor-initiating ALDHhigh/ABCB1negative cells and their differentiated, drug-expelling, ALDHlow/ABCB1positive daughter cells.
Collapse
Affiliation(s)
- Benjamin L Emmink
- Department of Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Goldsby R, Chen Y, Raber S, Li L, Diefenbach K, Shnorhavorian M, Kadan-Lottick N, Kastrinos F, Yasui Y, Stovall M, Oeffinger K, Sklar C, Armstrong GT, Robison LL, Diller L. Survivors of childhood cancer have increased risk of gastrointestinal complications later in life. Gastroenterology 2011; 140:1464-71.e1. [PMID: 21315721 PMCID: PMC3081911 DOI: 10.1053/j.gastro.2011.01.049] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/07/2011] [Accepted: 01/20/2011] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Children who receive cancer therapy experience numerous acute gastrointestinal (GI) toxicities. However, the long-term GI consequences have not been extensively studied. We evaluated the incidence of long-term GI outcomes and identified treatment-related risk factors. METHODS Upper GI, hepatic, and lower GI adverse outcomes were assessed in cases from participants in the Childhood Cancer Survivor Study, a study of 14,358 survivors of childhood cancer who were diagnosed between 1970 and 1986; data were compared with those from randomly selected siblings. The median age at cancer diagnosis was 6.8 years (range, 0-21.0 years), and the median age at outcome assessment was 23.2 years (5.6-48.9 years) for survivors and 26.6 years (1.8-56.2 years) for siblings. Rates of self-reported late GI complications (occurred 5 or more years after cancer diagnosis) were determined and associated with patient characteristics and cancer treatments, adjusting for age, sex, and race. RESULTS Compared with siblings, survivors had increased risk of late-onset complications of the upper GI tract (rate ratio [RR], 1.8; 95% confidence interval [CI], 1.6-2.0), liver (RR, 2.1; 95% CI, 1.8-2.5), and lower GI tract (RR, 1.9; 95% CI, 1.7-2.2). The RRs for requiring colostomy/ileostomy, liver biopsy, or developing cirrhosis were 5.6 (95% CI, 2.4-13.1), 24.1 (95% CI, 7.5-77.8), and 8.9 (95% CI, 2.0-40.0), respectively. Older age at diagnosis, intensified therapy, abdominal radiation, and abdominal surgery increased the risk of certain GI complications. CONCLUSIONS Individuals who received therapy for cancer during childhood have an increased risk of developing GI complications later in life.
Collapse
Affiliation(s)
- Robert Goldsby
- Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital, San Francisco, California, USA.
| | - Yan Chen
- Public Health Sciences, University of Alberta, Edmonton, AB
| | - Shannon Raber
- Pediatric Hematology/Oncology, UCSF Children’s Hospital, San Francisco, CA
| | - Linda Li
- Pediatric Hematology/Oncology, UCSF Children’s Hospital, San Francisco, CA
| | - Karen Diefenbach
- Pediatric Surgery, Yale University School of Medicine, New Haven, CT
| | | | | | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York , NY
| | - Yutaka Yasui
- Public Health Sciences, University of Alberta, Edmonton, AB
| | | | - Kevin Oeffinger
- Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Charles Sklar
- Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Gregory T. Armstrong
- Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| | - Leslie L. Robison
- Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN
| | - Lisa Diller
- Pediatric Oncology, Dana-Farber Cancer Institute/Children’s Hospital, Boston, MA
| |
Collapse
|
48
|
Lowe ME, Noll RB. Childhood cancer survival: a risk factor for GI disease. Gastroenterology 2011; 140:1383-6. [PMID: 21443888 DOI: 10.1053/j.gastro.2011.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
49
|
Scarberry KE, Mezencev R, McDonald JF. Targeted removal of migratory tumor cells by functionalized magnetic nanoparticles impedes metastasis and tumor progression. Nanomedicine (Lond) 2011; 6:69-78. [PMID: 21182419 DOI: 10.2217/nnm.10.103] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
AIMS To slow tumor progression by reducing migratory tumor cell burden using magnetic nanoparticles (MNPs) functionalized with ligands selective for malignant cell surface receptors. MATERIALS & METHODS Three groups of female C57BL/6 mice (control group I, control group II and experimental group) were intraperitoneally injected with a murine ovarian cancer cell line (ID8[VEGF160(+)/eGFP(+)]). Control group I received no intervention. MNPs were functionalized with ephrin-A1 mimetic peptides selective for the EphA2 receptor that is highly expressed by several cancers. Peritoneal fluids were removed by paracentesis from the experimental group and mixed with the functionalized MNPs. Magnetic filtration was used to remove particle/malignant cell conjugates and filtered peritoneal fluids were re-introduced intraperitoneally. Control group II received the same treatment as the experimental group without MNPs. RESULTS Experimental group tumor progression was 10.77-times slower than that of control group I. CONCLUSION Reduction of malignant cell titer significantly prolonged time to end point in a metastatic ovarian cancer model.
Collapse
Affiliation(s)
- Kenneth E Scarberry
- School of Biology & Petit Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | |
Collapse
|
50
|
Vera G, Castillo M, Cabezos PA, Chiarlone A, Martín MI, Gori A, Pasquinelli G, Barbara G, Stanghellini V, Corinaldesi R, De Giorgio R, Abalo R. Enteric neuropathy evoked by repeated cisplatin in the rat. Neurogastroenterol Motil 2011; 23:370-8, e162-3. [PMID: 21299719 DOI: 10.1111/j.1365-2982.2011.01674.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Acute administration of the antitumoral drug cisplatin can induce nausea/emesis and diarrhea. The long-term effects of cisplatin on gastrointestinal motility, particularly after repeated administration, are not well known. Because cisplatin is highly neurotoxic, myenteric neurons can be affected. Our aim was to study the prolonged effects of repeated cisplatin administration in a rat model, focusing on gastrointestinal motor function and myenteric neurons. METHODS Rats received saline or cisplatin (1 or 3 mg kg(-1), i.p.) once weekly for 5 weeks. One week after treatment, both upper gastrointestinal transit and colonic activity were evaluated, and tissue samples from ileum, colon and rectum were processed for histological analysis. Intestinal transit was measured invasively (charcoal method). Colonic activity was determined electromyographically. The gut wall structure was evaluated in sections using conventional histology and immunohistochemistry. Whole-mount preparations from the distal colon were labeled for different markers, including nitric oxide synthase (NOS) and calcitonin-gene related peptide (CGRP) to determine relative proportions of myenteric neurons vs the total neuronal population labeled with HuC/D. KEY RESULTS One week after repeated cisplatin exposure, the upper gastrointestinal transit rate and colonic activity were dose-dependently reduced. The number of NSE- or HuC/D-immunoreactive myenteric neurons per ganglion was decreased; the proportion of CGRP-immunoreactive neurons was decreased, whereas that of NOS-immunoreactive cells was increased. CONCLUSIONS & INFERENCES Chronic cisplatin may induce an enteric neuropathy characterized by changes in myenteric neurons associated with marked gastrointestinal motor dysfunction.
Collapse
Affiliation(s)
- G Vera
- Department of Pharmacology and Nutrition, Rey Juan Carlos University, Alcorcón, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|