351
|
Karthika C, Hari B, Rahman MH, Akter R, Najda A, Albadrani GM, Sayed AA, Akhtar MF, Abdel-Daim MM. Multiple strategies with the synergistic approach for addressing colorectal cancer. Biomed Pharmacother 2021; 140:111704. [PMID: 34082400 DOI: 10.1016/j.biopha.2021.111704] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer treatment is improving widely over time, but finding a proper defender to beat them seems like a distant dream. The quest for identification and discovery of drugs with an effective action is still a vital work. The role of a membrane protein called P-glycoprotein, which functions as garbage chute that efflux the waste, xenobiotics, and toxins out of the cancer cells acts as a major reason behind the therapeutic failure of most chemotherapeutic drugs. In this review, we mainly focused on a multiple strategies by employing 5-Fluorouracil, curcumin, and lipids in Nano formulation for the possible treatment of colorectal cancer and its metastasis. Eventually, multidrug resistance and angiogenesis can be altered and it would be helpful in colorectal cancer targeting.We have depicted the possible way for the depletion of colorectal cancer cells without disturbing the normal cells. The concept of focusing on multiple pathways for marking the colorectal cancer cells could help in activating one among the pathways if the other one fails. The activity of the 5-Fluorouracil can be enhanced with the help of curcumin which acts as a chemosensitizer, chemotherapeutic agent, and even for altering the resistance. As we eat to survive, so do the cancer cells. The cancer cells utilize the energy source to stay alive and survive. Fatty acids can be used as the energy source and this concept can be employed for targeting the colorectal cancer cells and also for altering the resistant part.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty 643001, Tamil Nadu, India
| | - Balaji Hari
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty 643001, Tamil Nadu, India
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh.
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka 1100, Bangladesh
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland.
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Amany A Sayed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Pakistan
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
352
|
Gao Y, Men K, Pan C, Li J, Wu J, Chen X, Lei S, Gao X, Duan X. Functionalized DMP-039 Hybrid Nanoparticle as a Novel mRNA Vector for Efficient Cancer Suicide Gene Therapy. Int J Nanomedicine 2021; 16:5211-5232. [PMID: 34366664 PMCID: PMC8335320 DOI: 10.2147/ijn.s319092] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/07/2021] [Indexed: 02/05/2023] Open
Abstract
Background Gene therapy has emerged as a new strategy for cancer therapy. As an alternative nucleic acid material, messenger ribonucleic acid (mRNA) is being increasingly utilized in cancer gene therapy. However, unfulfilled requirements and a lack of ideal mRNA delivery vectors persist. Methods We developed an advanced mRNA delivery system, DMP-039, by fusing a cell-penetrating peptide, cRGD-R9, and a cationic nano-sized DMP backbone together. The DMP gene vector backbone was synthesized by the self-assembly of DOTAP lipid and mPEG-PCL polymer. Introduction of the cRGD-R9 peptide onto the DMP backbone was performed to elevate the mRNA delivery capacity, which resulted in a peptide-functionalized hybrid delivery system. Results The average size of the synthesized DMP-039 was 268.9 ± 12.4 nm (PDI = 0.382), with a potential of 17.4 ± 0.5 mV. The synthesized DMP-039 hybrid nanoparticles exhibited high mRNA delivery efficiency through multiple mechanisms during transmembrane transportation. By loading the encoding mRNA from the suicide gene Bim, a locally administered mBim/DMP-039 complex strongly inhibited growth in two colon cancer models. Moreover, intravenous administration of the mBim/DMP-039 complex efficiently suppressed C26 pulmonary metastatic tumor progression with high safety. The in vivo distribution, degradation, and excretion were also investigated in detail. Conclusion Our results suggest that the DMP-039 peptide-functionalized hybrid nanoparticle is an advanced candidate for mRNA-based suicide gene therapy.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Congbin Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Jingmei Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Xiaohua Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Xiang Gao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| |
Collapse
|
353
|
Nam JH, Lee KH, Lim YJ. Examination of Entire Gastrointestinal Tract: A Perspective of Mouth to Anus (M2A) Capsule Endoscopy. Diagnostics (Basel) 2021; 11:diagnostics11081367. [PMID: 34441301 PMCID: PMC8394372 DOI: 10.3390/diagnostics11081367] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Capsule endoscopy (CE) is the only non-invasive diagnostic tool that enables the direct visualization of the gastrointestinal (GI) tract. Even though CE was initially developed for small-bowel investigation, its clinical application is expanding, and technological advances continue. The final iteration of CE will be a mouth to anus (M2A) capsule that investigates the entire GI tract by the ingestion of a single capsule. This narrative review describes the current developmental status of CE and discusses the possibility of realizing an M2A capsule and what needs to be overcome in the future.
Collapse
Affiliation(s)
- Ji Hyung Nam
- Division of Gastroenterology, Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang 10326, Korea;
| | - Kwang Hoon Lee
- Division of Rheumatology, Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang 10326, Korea;
| | - Yun Jeong Lim
- Division of Gastroenterology, Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang 10326, Korea;
- Correspondence: ; Tel.: +82-31-961-7133
| |
Collapse
|
354
|
Huang RL, Liu Q, Wang YX, Zou JY, Hu LF, Wang W, Huang YH, Wang YZ, Zeng B, Zeng X, Zeng Y. Awareness, attitude and barriers of colorectal cancer screening among high-risk populations in China: a cross-sectional study. BMJ Open 2021; 11:e045168. [PMID: 34253663 PMCID: PMC8276297 DOI: 10.1136/bmjopen-2020-045168] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To assess the awareness, attitude and barriers of colorectal cancer screening among high-risk populations in China. DESIGN A cross-sectional study was employed. SETTING This study was conducted in nine hospitals in Hunan province, China. PARTICIPANTS Individuals with a high-risk for colorectal cancer were interviewed using a pretested structured questionnaire. PRIMARY AND SECONDARY OUTCOME MEASURES Knowledge, attitude towards colorectal cancer screening, sociodemographic factors associated with screening knowledge and behaviour and barriers of colorectal cancer screening. RESULTS This study included 684 participants. The mean knowledge score was 11.86/24 (SD 4.84). But over 70% of them held a positive attitude towards screening. Only 13.3% had undergone colorectal cancer screening. Independent factors related to knowledge were education level of college or above, working as a white collar, higher income, having health insurance, having seen a doctor in the past year and with a high perceived risk (p<0.05). Factors independently associated with screening behaviour included personal history of colorectal disease, having seen a doctor in the past year, previous discussion of colorectal cancer screening, high perceived risk and better knowledge (p<0.05). Main reasons for not undergoing screening were no symptoms or discomfort (71.1%), never having thought of the disease or screening (67.4%) and no doctor advised me (29.8%). CONCLUSION In China, the majority of high-risk people had deficient knowledge and had never undergone colorectal cancer screening. But most of them held a positive attitude towards the benefits of colorectal cancer screening. This has promising implications to design targeted educational campaigns and establish screening programmes to improve colorectal cancer awareness and screening participation. Healthcare professionals should advise high-risk individuals to participate in screening and inform them about cancer risk.
Collapse
Affiliation(s)
- Ruo-Lin Huang
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| | - Qi Liu
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
- School of Nursing, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Ying-Xin Wang
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| | - Jin-Yu Zou
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| | - Li-Feng Hu
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| | - Wen Wang
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| | - Ying-Hui Huang
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| | - Yi-Zhuo Wang
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| | - Bo Zeng
- Hengyang No.8 High School, Hengyang, China
| | - Xi Zeng
- Cancer Research Institute, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, University of South China, Hengyang, Hunan, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China
| | - Ying Zeng
- Department of International and Humanistic Nursing, School of Nursing, University of South China, Hengyang, Hunan, China
| |
Collapse
|
355
|
Feitosa MR, Parra RS, Freitas LFD, Camargo HPD, Rocha JJRD, Féres O. TEACHING BASIC COLONOSCOPY SKILLS: QUALITY AND SAFETY STANDARDS CAN BE FULFILLED IN AN OUTPATIENT UNIVERSITY CENTER. ARQUIVOS DE GASTROENTEROLOGIA 2021; 58:384-389. [PMID: 34705975 DOI: 10.1590/s0004-2803.202100000-64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recent studies have shown that endoscopy fellows can perform colonoscopy effectively and safely. However, little is known about the performance of surgical residents without prior knowledge of endoscopic techniques. OBJECTIVE To assess whether quality indicators were met at an outpatient endoscopy center and whether surgical residents, without prior upper or lower endoscopy skills, could perform colonoscopy adequately. METHODS A prospective non-randomized cohort study was undertaken. All exams were performed either by assistant physicians or by residents. Quality measures were compared between those groups. RESULTS A total of 2720 colonoscopies were analyzed. In the resident group, we observed older patients (57.7±12.7 years vs 51.5±14.5 years, P<0.001), a higher prevalence of screening colonoscopies (52% vs 39.4%, P<0.001) and a higher prevalence of colorectal cancer (6.4% vs 1.8%, P<0.001). The cecal intubation rate was higher in the attending group (99.9% vs 89.3%; P<0.001). The polyp detection rate was 40.8%, and no differences were observed between the studied groups. The residents had a higher rate of perforation in all exams (0.4% vs 0%; P=0.02). Postpolypectomy bleeding and 7-day readmission rates were the same (0.2%). All readmissions in 7 days occurred due to low digestive bleeding, and none required intervention. CONCLUSION Quality indicators were met at a university outpatient endoscopy center; however, medical residents achieved lower rates of cecal intubation and higher rates of perforation than the attending physicians.
Collapse
Affiliation(s)
- Marley Ribeiro Feitosa
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Departamento de Cirurgia e Anatomia, Ribeirão Preto, SP, Brasil
| | - Rogério Serafim Parra
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Departamento de Cirurgia e Anatomia, Ribeirão Preto, SP, Brasil
| | - Lucas Fernandes de Freitas
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Departamento de Cirurgia e Anatomia, Ribeirão Preto, SP, Brasil
| | - Hugo Parra de Camargo
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Departamento de Cirurgia e Anatomia, Ribeirão Preto, SP, Brasil
| | - José Joaquim Ribeiro da Rocha
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Departamento de Cirurgia e Anatomia, Ribeirão Preto, SP, Brasil
| | - Omar Féres
- Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Departamento de Cirurgia e Anatomia, Ribeirão Preto, SP, Brasil
| |
Collapse
|
356
|
He Q, Li Z, Lei X, Zou Q, Yu H, Ding Y, Xu G, Zhu W. The underlying molecular mechanisms and prognostic factors of RNA binding protein in colorectal cancer: a study based on multiple online databases. Cancer Cell Int 2021; 21:325. [PMID: 34193169 PMCID: PMC8244213 DOI: 10.1186/s12935-021-02031-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/19/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND RNA binding protein (RBP) is an active factor involved in the occurrence and development of colorectal cancer (CRC). Therefore, the potential mechanism of RBP in CRC needs to be clarified by dry-lab analyses or wet-lab experiments. METHODS The differential RBP gene obtained from the GEPIA 2 (Gene Expression Profiling Interactive Analysis 2) were performed functional enrichment analysis. Then, the alternative splicing (AS) events related to survival were acquired by univariate regression analysis, and the correlation between RBP and AS was analyzed by R software. The online databases were conducted to analyze the mutation and methylation of RBPs in CRC. Moreover, 5 key RBP signatures were obtained through univariate and multivariate Cox regression analysis and established as RBP prognosis model. Subsequently, the above model was verified through another randomized group of TCGA CRC cohorts. Finally, multiple online databases and qRT-PCR analysis were carried to further confirm the expression of the above 5 RBP signatures in CRC. RESULTS Through a comprehensive bioinformatics analysis, it was revealed that RBPs had genetic and epigenetic changes in CRC. We obtained 300 differentially expressed RBPs in CRC samples. The functional analysis suggested that they mainly participated in spliceosome. Then, a regulatory network for RBP was established to participate in AS and DDX39B was detected to act as a potentially essential factor in the regulation of AS in CRC. Our analysis discovered that 11 differentially expressed RBPs with a mutation frequency higher than 5%. Furthermore, we found that 10 differentially expressed RBPs had methylation sites related to the prognosis of CRC, and a prognostic model was constructed by the 5 RBP signatures. In another randomized group of TCGA CRC cohorts, the prognostic performance of the 5 RBP signatures was verified. CONCLUSION The potential mechanisms that regulate the aberrant expression of RBPs in the development of CRC was explored, a network that regulated AS was established, and the RBP-related prognosis model was constructed and verified, which could improve the individualized prognosis prediction of CRC.
Collapse
Affiliation(s)
- Qinglian He
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Ziqi Li
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Xue Lei
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Qian Zou
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China
| | - Haibing Yu
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Yuanlin Ding
- School of Public Health, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Institute of Clinical Laboratory, Guangdong Medical University, Dongguan, 523808, Guangdong Province, China
| | - Wei Zhu
- Department of Pathology, Guangdong Medical University, No.1 Xincheng Road, Dongguan, 523808, Guangdong Province, China.
| |
Collapse
|
357
|
Chen S, Ben S, Xin J, Li S, Zheng R, Wang H, Fan L, Du M, Zhang Z, Wang M. The biogenesis and biological function of PIWI-interacting RNA in cancer. J Hematol Oncol 2021; 14:93. [PMID: 34118972 PMCID: PMC8199808 DOI: 10.1186/s13045-021-01104-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Small non-coding RNAs (ncRNAs) are vital regulators of biological activities, and aberrant levels of small ncRNAs are commonly found in precancerous lesions and cancer. PIWI-interacting RNAs (piRNAs) are a novel type of small ncRNA initially discovered in germ cells that have a specific length (24-31 nucleotides), bind to PIWI proteins, and show 2'-O-methyl modification at the 3'-end. Numerous studies have revealed that piRNAs can play important roles in tumorigenesis via multiple biological regulatory mechanisms, including silencing transcriptional and posttranscriptional gene processes and accelerating multiprotein interactions. piRNAs are emerging players in the malignant transformation of normal cells and participate in the regulation of cancer hallmarks. Most of the specific cancer hallmarks regulated by piRNAs are involved in sustaining proliferative signaling, resistance to cell death or apoptosis, and activation of invasion and metastasis. Additionally, piRNAs have been used as biomarkers for cancer diagnosis and prognosis and have great potential for clinical utility. However, research on the underlying mechanisms of piRNAs in cancer is limited. Here, we systematically reviewed recent advances in the biogenesis and biological functions of piRNAs and relevant bioinformatics databases with the aim of providing insights into cancer diagnosis and clinical applications. We also focused on some cancer hallmarks rarely reported to be related to piRNAs, which can promote in-depth research of piRNAs in molecular biology and facilitate their clinical translation into cancer treatment.
Collapse
Affiliation(s)
- Silu Chen
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, People's Republic of China.,Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shuai Ben
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Junyi Xin
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shuwei Li
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rui Zheng
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hao Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lulu Fan
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Mulong Du
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, People's Republic of China. .,Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China. .,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China. .,Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
358
|
Thabet NA, El-Khouly D, Sayed-Ahmed MM, Omran MM. Thymoquinone chemosensitizes human colorectal cancer cells to imatinib via uptake/efflux genes modulation. Clin Exp Pharmacol Physiol 2021; 48:911-920. [PMID: 33783002 DOI: 10.1111/1440-1681.13476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
Abstract
Imatinib (IM) is a pharmaceutical drug that inhibits tyrosine kinase enzymes that are responsible for the activation of many proteins by signal transduction cascades as c-Abl, c-Kit and the platelet-derived growth factor (PDGF) receptor. Thymoquinone (TQ) is an active constituent of Nigella sativa seeds. Thymoquinone benefits are attributed to its medicinal uses as antioxidant, anticancer and antimicrobial agent. This study aimed to investigate the impact of using TQ with IM in the HCT116 human colorectal cancer cell line model. The HCT116 cells were treated with IM or/and TQ in non-constant ratios, in which the fixed concentrations of TQ (5, 10 or 20 µmol/L) were co-treated with various concentrations of IM (7.5-120 µmol/L) for 24, 48 and 72 hours. Imatinib-TQ interaction was analysed using CompuSyn software. The IC50 values for IM were 105, 72 μmol/L after 48 and 72 hours, respectively, and were significantly reduced to 7.3, 7 and 5.5 μmol/L after combination with TQ (10 μmol/L) and to 5.8, 5.6 and 4.6 μmol/L after combination with TQ (20 μmol/L) to 24, 48 and 72 hours, respectively. The combination index (CI) and dose reduction index (DRI) values indicate a significant synergism in HCT-116 cells at different treatment time points. Thymoquinone significantly enhances the cellular uptake of IM in HCT116 cells in a time and concentration-dependent manner. A significant downregulation in ATP-binding cassette (ABC) subfamily B member 1 (ABCB1), ABC subfamily G member 2 (ABCG2) and human organic cation transporter 1 (hOCT1) genes was observed in the cells exposed to IM+TQ combination as compared to IM alone, which resulted in a substantial elevation in uptake/efflux ratio in combination group. In conclusion, TQ potentiates IM efficacy on HCT116 cells via uptake/efflux genes modulation.
Collapse
Affiliation(s)
- Nadia A Thabet
- Pharmacology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Dalia El-Khouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Mohamed M Sayed-Ahmed
- Pharmacology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mervat M Omran
- Pharmacology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
359
|
Frissora KD, Biernacki PJ, Walloch J. Increasing Awareness of Screening Methods for Colorectal Cancer Improves Outcomes. J Nurse Pract 2021. [DOI: 10.1016/j.nurpra.2021.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
360
|
Li J, Tian L, Jing Z, Guo Z, Nan P, Liu F, Zou S, Yang L, Xie X, Zhu Y, Zhao Y, Sun W, Sun Y, Zhao X. Cytoplasmic RAD23B interacts with CORO1C to synergistically promote colorectal cancer progression and metastasis. Cancer Lett 2021; 516:13-27. [PMID: 34062216 DOI: 10.1016/j.canlet.2021.05.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
Colorectal cancers (CRCs) are characterized by diffuse infiltration of tumor cells into the regional lymph nodes and metastasis to distant organs, and its highly invasive nature contributes to disease recurrence and poor outcomes. The molecular mechanisms underlying CRC cell invasion remain incompletely understood. Here, we identified the upregulation of DNA damage repair-related protein RAD23B in CRC cells and tissues and showed that it associates with coronin 1C or coronin 3 (CORO1C) to facilitate invasion. We found that knockdown of RAD23B expression significantly inhibited the proliferation, invasion, and migration abilities of CRC cells both in vitro and in vivo, and suppressed the talin1/2/integrin/FAK/RhoA/Rac1/CORO1C signaling pathways. Interestingly, RAD23B interacted and co-localized with CORO1C, and CORO1C aggregated toward the margin of cancer cells in both CRC cells and tissues when RAD23B overexpressed. Mechanistically, overexpression of RAD23B and/or CORO1C further increased invadopodia formation and matrix degradation in SW480 and HCT8 CRC cells. Conversely, silencing of RAD23B expression suppressed tumorigenesis and liver metastasis in xenotransplant murine models. Furthermore, we found that RAD23B was significantly overexpressed in tumor tissues (n = 720) compared to adjacent non-tumor tissues (n = 694) of patients with CRC. Finally, we identified a strong correlation between higher levels of cytoplasmic expression of RAD23B, and poor prognosis and liver metastasis in CRC patients. Taken together, our data highlight a novel RAD23B-CORO1C signaling axis in CRC cell invasion and metastasis that may be of clinical significance.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lusong Tian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zongpan Jing
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhengguang Guo
- Core Facility of Instruments, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Peng Nan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuangmei Zou
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lijun Yang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiufeng Xie
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ying Zhu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Sun
- Core Facility of Instruments, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yulin Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xiaohang Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
361
|
Sun Z, Xia W, Lyu Y, Song Y, Wang M, Zhang R, Sui G, Li Z, Song L, Wu C, Liew CC, Yu L, Cheng G, Cheng C. Immune-related gene expression signatures in colorectal cancer. Oncol Lett 2021; 22:543. [PMID: 34079596 PMCID: PMC8157333 DOI: 10.3892/ol.2021.12804] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/11/2021] [Indexed: 12/24/2022] Open
Abstract
The immune system is crucial in regulating colorectal cancer (CRC) tumorigenesis. Identification of immune-related transcriptomic signatures derived from the peripheral blood of patients with CRC would provide insights into CRC pathogenesis, and suggest novel clues to potential immunotherapy strategies for the disease. The present study collected blood samples from 59 patients with CRC and 62 healthy control patients and performed whole blood gene expression profiling using microarray hybridization. Immune-related gene expression signatures for CRC were identified from immune gene datasets, and an algorithmic predictive model was constructed for distinguishing CRC from controls. Model performance was characterized using an area under the receiver operating characteristic curve (ROC AUC). Functional categories for CRC-specific gene expression signatures were determined using gene set enrichment analyses. A Kaplan-Meier plotter survival analysis was also performed for CRC-specific immune genes in order to characterize the association between gene expression and CRC prognosis. The present study identified five CRC-specific immune genes [protein phosphatase 3 regulatory subunit Bα (PPP3R1), amyloid β precursor protein, cathepsin H, proteasome activator subunit 4 and DEAD-Box Helicase 3 X-Linked]. A predictive model based on this five-gene panel showed good discriminatory power (independent test set sensitivity, 83.3%; specificity, 94.7%, accuracy, 89.2%; ROC AUC, 0.96). The candidate genes were involved in pathways associated with ‘adaptive immune responses’, ‘innate immune responses’ and ‘cytokine signaling’. The survival analysis found that a high level of PPP3R1 expression was associated with a poor CRC prognosis. The present study identified five CRC-specific immune genes that were potential diagnostic biomarkers for CRC. The biological function analysis indicated a close association between CRC pathogenesis and the immune system, and may reveal more information about the immunogenic and pathogenic mechanisms driving CRC in the future. Overall, the association between PPP3R1 expression and survival of patients with CRC revealed potential new targets for CRC immunotherapy.
Collapse
Affiliation(s)
- Zhenqing Sun
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Wei Xia
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Yali Lyu
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Yanan Song
- Department of Nuclear Medicine, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Min Wang
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Ruirui Zhang
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Guode Sui
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhenlu Li
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Li Song
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Changliang Wu
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Choong-Chin Liew
- Golden Health Diagnostics Inc., Yan Cheng, Jiangsu 224000, P.R. China.,Department of Clinical Pathology and Laboratory Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lei Yu
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| | - Guang Cheng
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Changming Cheng
- R&D Department, Huaxia Bangfu Technology Incorporated, Beijing 100000, P.R. China
| |
Collapse
|
362
|
Bibault JE, Chang DT, Xing L. Development and validation of a model to predict survival in colorectal cancer using a gradient-boosted machine. Gut 2021; 70:884-889. [PMID: 32887732 DOI: 10.1136/gutjnl-2020-321799] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The success of treatment planning relies critically on our ability to predict the potential benefit of a therapy. In colorectal cancer (CRC), several nomograms are available to predict different outcomes based on the use of tumour specific features. Our objective is to provide an accurate and explainable prediction of the risk to die within 10 years after CRC diagnosis, by incorporating the tumour features and the patient medical and demographic information. DESIGN In the prostate, lung, colorectal and ovarian cancer screening (PLCO) Trial, participants (n=154 900) were randomised to screening with flexible sigmoidoscopy, with a repeat screening at 3 or 5 years, or to usual care. We selected patients who were diagnosed with CRC during the follow-up to train a gradient-boosted model to predict the risk to die within 10 years after CRC diagnosis. Using Shapley values, we determined the 20 most relevant features and provided explanation to prediction. RESULTS During the follow-up, 2359 patients were diagnosed with CRC. Median follow-up was 16.8 years (14.4-18.9) for mortality. In total, 686 patients (29%) died from CRC during the follow-up. The dataset was randomly split into a training (n=1887) and a testing (n=472) dataset. The area under the receiver operating characteristic was 0.84 (±0.04) and accuracy was 0.83 (±0.04) with a 0.5 classification threshold. The model is available online for research use. CONCLUSIONS We trained and validated a model with prospective data from a large multicentre cohort of patients. The model has high predictive performances at the individual scale. It could be used to discuss treatment strategies.
Collapse
Affiliation(s)
| | - Daniel T Chang
- Radiation Oncology, Stanford Medicine, Stanford, California, USA
| | - Lei Xing
- Radiation Oncology, Stanford Medicine, Stanford, California, USA
| |
Collapse
|
363
|
Joseph DF, Li E, Stanley III SL, Zhu YC, Li XN, Yang J, Ottaviano LF, Bucobo JC, Buscaglia JM, Miller JD, Veluvolu R, Follen M, Grossman EB. Impact of type 2 diabetes on adenoma detection in screening colonoscopies performed in disparate populations. World J Clin Cases 2021; 9:2433-2445. [PMID: 33889609 PMCID: PMC8040183 DOI: 10.12998/wjcc.v9.i11.2433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/23/2020] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The Black/African Ancestry (AA) population has a higher prevalence of type 2 diabetes mellitus (T2DM) and a higher incidence and mortality rate for colorectal cancer (CRC) than all other races in the United States. T2DM has been shown to increase adenoma risk in predominantly white/European ancestry (EA) populations, but the effect of T2DM on adenoma risk in Black/AA individuals is less clear. We hypothesize that T2DM has a significant effect on adenoma risk in a predominantly Black/AA population.
AIM To investigate the effect of T2DM and race on the adenoma detection rate (ADR) in screening colonoscopies in two disparate populations.
METHODS A retrospective cohort study was conducted on ADR during index screening colonoscopies (age 45-75) performed at an urban public hospital serving a predominantly Black/AA population (92%) (2017-2018, n = 1606). Clinical metadata collected included basic demographics, insurance, body mass index (BMI), family history of CRC, smoking, diabetes diagnosis, and aspirin use. This dataset was combined with a recently reported parallel retrospective cohort data set collected at a suburban university hospital serving a predominantly White/EA population (87%) (2012-2015, n = 2882).
RESULTS The ADR was higher in T2DM patients than in patients without T2DM or prediabetes (35.2% vs 27.9%, P = 0.0166, n = 981) at the urban public hospital. Multivariable analysis of the combined datasets showed that T2DM [odds ratio (OR) = 1.29, 95% confidence interval (CI): 1.08-1.55, P = 0.0049], smoking (current vs never OR = 1.47, 95%CI: 1.18-1.82, current vs past OR = 1.32, 95%CI: 1.02-1.70, P = 0.0026), older age (OR = 1.05 per year, 95%CI: 1.04-1.06, P < 0.0001), higher BMI (OR = 1.02 per unit, 95%CI: 1.01-1.03, P = 0.0003), and male sex (OR = 1.87, 95%CI: 1.62-2.15, P < 0.0001) were associated with increased ADR in the combined datasets, but race, aspirin use and insurance were not.
CONCLUSION T2DM, but not race, is significantly associated with increased ADR on index screening colonoscopy while controlling for other factors.
Collapse
Affiliation(s)
- Dimitri F Joseph
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Ellen Li
- Department of Medicine, Division of Gastroenterology, Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Samuel L Stanley III
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600, United States
| | - Yi-Cong Zhu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600, United States
| | - Xiao-Ning Li
- Department of Biostatistics and Bioinformatics Shared Resource, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794-3600, United States
| | - Jie Yang
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600, United States
- Department of Family, Population, and Preventative Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8461, United States
| | - Lorenzo F Ottaviano
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Juan Carlos Bucobo
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Jonathan M Buscaglia
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Joshua D Miller
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Rajesh Veluvolu
- Department of Medicine, NYC Health and Hospitals/Kings County, Brooklyn, NY 11203, United States
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Michele Follen
- Department of Obstetrics and Gynecology, NYC Health and Hospitals/Kings County, Brooklyn, NY 11203, United States
| | - Evan B Grossman
- Department of Medicine, NYC Health and Hospitals/Kings County, Brooklyn, NY 11203, United States
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| |
Collapse
|
364
|
Zhang K, Zhou X, Wang J, Zhou Y, Qi W, Chen H, Nie S, Xie M. Dendrobium officinale polysaccharide triggers mitochondrial disorder to induce colon cancer cell death via ROS-AMPK-autophagy pathway. Carbohydr Polym 2021; 264:118018. [PMID: 33910741 DOI: 10.1016/j.carbpol.2021.118018] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
The homeostasis between mitochondrial function and autophagy is crucial to the physiological activity of cancer cells, and its mechanism is conducive to the development of anti-tumor drugs. Here, we aimed to explore the effect and mechanism of Dendrobium officinale polysaccharide (DOP) on colon cancer cell line CT26. Our data showed that DOP significantly inhibited the proliferation of CT26 cells and elevated autophagy level. Moreover, DOP disrupted mitochondrial function through increasing reactive oxygen species (ROS) and reducing mitochondrial membrane potential (MMP), thereby impairing ATP biosynthesis, which activated AMPK/mTOR autophagy signaling. Intriguingly, the further experiments demonstrated that DOP-induced cytotoxicity, excessive autophagy and mitochondrial dysfunction were reversed after CT26 cells pretreated with antioxidant (N-acetyl-l-cysteine). Herein, these findings implied that DOP-induced mitochondrial dysfunction and cytotoxic autophagy repressed the propagation of CT26 cells via ROS-ATP-AMPK signaling, providing a new opinion for the study of antineoplastic drugs.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Xingtao Zhou
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China.
| | - Junqiao Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Yujia Zhou
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Wucheng Qi
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Haihong Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, 235 Nanjing East Road, Nanchang, Jiangxi, 330047, China.
| |
Collapse
|
365
|
Akimoto N, Ugai T, Zhong R, Hamada T, Fujiyoshi K, Giannakis M, Wu K, Cao Y, Ng K, Ogino S. Rising incidence of early-onset colorectal cancer - a call to action. Nat Rev Clin Oncol 2021; 18:230-243. [PMID: 33219329 PMCID: PMC7994182 DOI: 10.1038/s41571-020-00445-1] [Citation(s) in RCA: 374] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
The incidence of early-onset colorectal cancer (CRC), which occurs in individuals <50 years of age, has been increasing worldwide and particularly in high-income countries. The reasons for this increase remain unknown but plausible hypotheses include greater exposure to potential risk factors, such as a Western-style diet, obesity, physical inactivity and antibiotic use, especially during the early prenatal to adolescent periods of life. These exposures can not only cause genetic and epigenetic alterations in colorectal epithelial cells but also affect the gut microbiota and host immunity. Early-onset CRCs have differential clinical, pathological and molecular features compared with later-onset CRCs. Certain existing resources can be utilized to elucidate the aetiology of early-onset CRC and inform the development of effective prevention, early detection and therapeutic strategies; however, additional life-course cohort studies spanning childhood and young adulthood, integrated with prospective biospecimen collections, omics biomarker analyses and a molecular pathological epidemiology approach, are needed to better understand and manage this disease entity. In this Perspective, we summarize our current understanding of early-onset CRC and discuss how we should strategize future research to improve its prevention and clinical management.
Collapse
Affiliation(s)
- Naohiko Akimoto
- Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Gastroenterology, Nippon Medical School, Graduate School of Medicine, Tokyo, Japan
| | - Tomotaka Ugai
- Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rong Zhong
- Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Tsuyoshi Hamada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenji Fujiyoshi
- Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Surgery, Kurume University, Kurume, Japan
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kana Wu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yin Cao
- Division of Public Health Sciences, Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuji Ogino
- Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, MA, USA.
| |
Collapse
|
366
|
Silva CMS, Barros-Filho MC, Wong DVT, Mello JBH, Nobre LMS, Wanderley CWS, Lucetti LT, Muniz HA, Paiva IKD, Kuasne H, Ferreira DPP, Cunha MPSS, Hirth CG, Silva PGB, Sant’Ana RO, Souza MHLP, Quetz JS, Rogatto SR, Lima-Junior RCP. Circulating let-7e-5p, miR-106a-5p, miR-28-3p, and miR-542-5p as a Promising microRNA Signature for the Detection of Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13071493. [PMID: 33804927 PMCID: PMC8037203 DOI: 10.3390/cancers13071493] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The detection of early-stage colorectal cancer increases the chance to prevent tumor progression and death by the disease. Colonoscopy is one sensitive screening test to detect malignant or potentially malignant lesions in the intestines. However, it has some disadvantages, including sedation requirements, increased risk of colon perforation, and bleeding. Circulating microRNAs (miRNAs) in plasma or serum from cancer patients have been investigated and described as potential diagnostic or prognostic markers. We conducted an miRNAs screening test in plasma samples from colorectal cancer patients and subjects without cancer, aiming to identify markers for the early detection of the disease. We identified and validated four miRNAs capable of distinguishing cancer from non-cancer cases. Our non-invasive diagnostic biomarkers presented high performance and are easily applicable to clinical practice. Abstract Colorectal cancer (CRC) is a disease with high incidence and mortality. Colonoscopy is a gold standard among tests used for CRC traceability. However, serious complications, such as colon perforation, may occur. Non-invasive diagnostic procedures are an unmet need. We aimed to identify a plasma microRNA (miRNA) signature for CRC detection. Plasma samples were obtained from subjects (n = 109) at different stages of colorectal carcinogenesis. The patients were stratified into a non-cancer (27 healthy volunteers, 17 patients with hyperplastic polyps, 24 with adenomas), and a cancer group (20 CRC and 21 metastatic CRC). miRNAs (381) were screened by TaqMan Low-Density Array. A classifier based on four differentially expressed miRNAs (miR-28-3p, let-7e-5p, miR-106a-5p, and miR-542-5p) was able to discriminate cancer versus non-cancer cases. The overexpression of these miRNAs was confirmed by RT-qPCR, and a cross-study validation step was implemented using eight data series retrieved from Gene Expression Omnibus (GEO). In addition, another external data validation using CRC surgical specimens from The Cancer Genome Atlas (TCGA) was carried out. The predictive model’s performance in the validation set was 76.5% accuracy, 59.4% sensitivity, and 86.8% specificity (area under the curve, AUC = 0.716). The employment of our model in the independent publicly available datasets confirmed a good discrimination performance in five of eight datasets (median AUC = 0.823). Applying this algorithm to the TCGA cohort, we found 99.5% accuracy, 99.7% sensitivity, and 90.9% specificity (AUC = 0.998) when the model was applied to solid colorectal tissues. Overall, we suggest a novel signature of four circulating miRNAs, i.e., miR-28-3p, let-7e-5p, miR-106a-5p, and miR-542-5p, as a predictive tool for the detection of CRC.
Collapse
Affiliation(s)
- Camila Meirelles S. Silva
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil; (L.M.S.N.); (C.W.S.W.); (L.T.L.); (H.A.M.); (I.K.D.P.)
- Correspondence: (C.M.S.S.); (S.R.R.); (R.C.P.L.-J.); Tel.: +55-85-3366-8585 (C.M.S.S. & R.C.P.L.-J.); +45-7940-6669 (S.R.R.)
| | - Mateus C. Barros-Filho
- International Research Center—CIPE, A.C. Camargo Cancer Center, Sao Paulo 01525-001, Brazil; (M.C.B.-F.); (H.K.)
- Department of Head and Neck Surgery, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo/LIM-28-São Paulo, Sao Paulo 05403-000, Brazil
| | - Deysi Viviana T. Wong
- Department of Pathology and Forensic Medicine, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-160, Brazil; or
- Haroldo Juaçaba Hospital—Cancer Institute of Ceará, Fortaleza 60430-230, Brazil; (M.P.S.S.C.); (C.G.H.); (P.G.B.S.); (R.O.S.); (J.S.Q.)
| | - Julia Bette H. Mello
- Molecular Carcinogenesis Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20230-240, Brazil;
| | - Livia Maria S. Nobre
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil; (L.M.S.N.); (C.W.S.W.); (L.T.L.); (H.A.M.); (I.K.D.P.)
| | - Carlos Wagner S. Wanderley
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil; (L.M.S.N.); (C.W.S.W.); (L.T.L.); (H.A.M.); (I.K.D.P.)
| | - Larisse T. Lucetti
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil; (L.M.S.N.); (C.W.S.W.); (L.T.L.); (H.A.M.); (I.K.D.P.)
| | - Heitor A. Muniz
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil; (L.M.S.N.); (C.W.S.W.); (L.T.L.); (H.A.M.); (I.K.D.P.)
| | - Igor Kenned D. Paiva
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil; (L.M.S.N.); (C.W.S.W.); (L.T.L.); (H.A.M.); (I.K.D.P.)
| | - Hellen Kuasne
- International Research Center—CIPE, A.C. Camargo Cancer Center, Sao Paulo 01525-001, Brazil; (M.C.B.-F.); (H.K.)
| | | | - Maria Perpétuo S. S. Cunha
- Haroldo Juaçaba Hospital—Cancer Institute of Ceará, Fortaleza 60430-230, Brazil; (M.P.S.S.C.); (C.G.H.); (P.G.B.S.); (R.O.S.); (J.S.Q.)
| | - Carlos G. Hirth
- Haroldo Juaçaba Hospital—Cancer Institute of Ceará, Fortaleza 60430-230, Brazil; (M.P.S.S.C.); (C.G.H.); (P.G.B.S.); (R.O.S.); (J.S.Q.)
| | - Paulo Goberlânio B. Silva
- Haroldo Juaçaba Hospital—Cancer Institute of Ceará, Fortaleza 60430-230, Brazil; (M.P.S.S.C.); (C.G.H.); (P.G.B.S.); (R.O.S.); (J.S.Q.)
| | - Rosane O. Sant’Ana
- Haroldo Juaçaba Hospital—Cancer Institute of Ceará, Fortaleza 60430-230, Brazil; (M.P.S.S.C.); (C.G.H.); (P.G.B.S.); (R.O.S.); (J.S.Q.)
- School of Medicine, University of Fortaleza, Fortaleza 60811-905, Brazil
| | | | - Josiane S. Quetz
- Haroldo Juaçaba Hospital—Cancer Institute of Ceará, Fortaleza 60430-230, Brazil; (M.P.S.S.C.); (C.G.H.); (P.G.B.S.); (R.O.S.); (J.S.Q.)
| | - Silvia R. Rogatto
- Department of Clinical Genetics, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Institute of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
- Danish Colorectal Cancer Center South, 7100 Vejle, Denmark
- Correspondence: (C.M.S.S.); (S.R.R.); (R.C.P.L.-J.); Tel.: +55-85-3366-8585 (C.M.S.S. & R.C.P.L.-J.); +45-7940-6669 (S.R.R.)
| | - Roberto César P. Lima-Junior
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza 60430-270, Brazil; (L.M.S.N.); (C.W.S.W.); (L.T.L.); (H.A.M.); (I.K.D.P.)
- Correspondence: (C.M.S.S.); (S.R.R.); (R.C.P.L.-J.); Tel.: +55-85-3366-8585 (C.M.S.S. & R.C.P.L.-J.); +45-7940-6669 (S.R.R.)
| |
Collapse
|
367
|
Yen SY, Huang HE, Lien GS, Liu CW, Chu CF, Huang WM, Suk FM. Automatic lumen detection and magnetic alignment control for magnetic-assisted capsule colonoscope system optimization. Sci Rep 2021; 11:6460. [PMID: 33742067 PMCID: PMC7979719 DOI: 10.1038/s41598-021-86101-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
We developed a magnetic-assisted capsule colonoscope system with integration of computer vision-based object detection and an alignment control scheme. Two convolutional neural network models A and B for lumen identification were trained on an endoscopic dataset of 9080 images. In the lumen alignment experiment, models C and D used a simulated dataset of 8414 images. The models were evaluated using validation indexes for recall (R), precision (P), mean average precision (mAP), and F1 score. Predictive performance was evaluated with the area under the P-R curve. Adjustments of pitch and yaw angles and alignment control time were analyzed in the alignment experiment. Model D had the best predictive performance. Its R, P, mAP, and F1 score were 0.964, 0.961, 0.961, and 0.963, respectively, when the area of overlap/area of union was at 0.3. In the lumen alignment experiment, the mean degrees of adjustment for yaw and pitch in 160 trials were 21.70° and 13.78°, respectively. Mean alignment control time was 0.902 s. Finally, we compared the cecal intubation time between semi-automated and manual navigation in 20 trials. The average cecal intubation time of manual navigation and semi-automated navigation were 9 min 28.41 s and 7 min 23.61 s, respectively. The automatic lumen detection model, which was trained using a deep learning algorithm, demonstrated high performance in each validation index.
Collapse
Affiliation(s)
- Sheng-Yang Yen
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Hao-En Huang
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Gi-Shih Lien
- Division of Gastroenterology, Department of Internal Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, No. 111, Section 3, Xing Long Road, Taipei, 116, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Wen Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Chia-Feng Chu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Wei-Ming Huang
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan
| | - Fat-Moon Suk
- Division of Gastroenterology, Department of Internal Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, No. 111, Section 3, Xing Long Road, Taipei, 116, Taiwan. .,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
368
|
Herring E, Tremblay É, McFadden N, Kanaoka S, Beaulieu JF. Multitarget Stool mRNA Test for Detecting Colorectal Cancer Lesions Including Advanced Adenomas. Cancers (Basel) 2021; 13:1228. [PMID: 33799738 PMCID: PMC7998137 DOI: 10.3390/cancers13061228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Current approved non-invasive screening methods for colorectal cancer (CRC) include FIT and DNA-FIT testing, but their efficacy for detecting precancerous lesions that are susceptible to progressing to CRC such as advanced adenomas (AA) remains limited, thus requiring further options to improve the detection of CRC lesions at earlier stages. One of these is host mRNA stool testing. The aims of the present study were to identify specific stool mRNA targets that can predict AA and to investigate their stability under a clinical-like setting. A panel of mRNA targets was tested on stool samples obtained from 102 patients including 78 CRC stage I-III and 24 AA as well as 32 healthy controls. Area under the receiver operating characteristic (ROC) curves were calculated to establish sensitivities and specificities for individual and combined targets. Stability experiments were performed on freshly obtained specimens. Six of the tested targets were found to be specifically increased in the stools of patients with CRC and three in the stools of both AA and CRC patients. After optimization for the choice of the 5 best markers for AA and CRC, ROC curve analysis revealed overall sensitivities of 75% and 89% for AA and CRC, respectively, for a ≥95% specificity, and up to 75% and 95% for AA and CRC, respectively, when combined with the FIT score. Targets were found to be stable in the stools up to 3 days at room temperature. In conclusion, these studies show that the detection of host mRNA in the stools is a valid approach for the screening of colorectal cancerous lesions at all stages and is applicable to a clinical-like setup.
Collapse
Affiliation(s)
- Elizabeth Herring
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (E.H.); (É.T.)
- Centre de Recherche du Centre Hospitalier, Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Éric Tremblay
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (E.H.); (É.T.)
- Centre de Recherche du Centre Hospitalier, Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Nathalie McFadden
- Centre de Recherche du Centre Hospitalier, Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Department of Surgery, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Shigeru Kanaoka
- Department of Gastroenterology, Hamamatsu Medical Center, Naka-ku, Hamamatsu 432-8580, Japan;
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; (E.H.); (É.T.)
- Centre de Recherche du Centre Hospitalier, Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| |
Collapse
|
369
|
Wu X, Zhang Y, Hu T, He X, Zou Y, Deng Q, Ke J, Lian L, He X, Zhao D, Cai X, Chen Z, Wu X, Fan JB, Gao F, Lan P. A novel cell-free DNA methylation-based model improves the early detection of colorectal cancer. Mol Oncol 2021; 15:2702-2714. [PMID: 33694305 PMCID: PMC8486566 DOI: 10.1002/1878-0261.12942] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/21/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Screening for early‐stage disease is vital for reducing colorectal cancer (CRC)‐related mortality. Methylation of circulating tumor DNA has been previously used for various types of cancer screening. A novel cell‐free DNA (cfDNA) methylation‐based model which can improve the early detection of CRC is warranted. For our study, we collected 313 tissue and 577 plasma samples from patients with CRC, advanced adenoma (AA), non‐AA and healthy controls. After quality control, 187 tissue DNA samples (91 non‐malignant tissue from CRC patients, 26 AA and 70 CRC) and 489 plasma cfDNA samples were selected for targeted DNA methylation sequencing. We further developed a cfDNA methylation model based on 11 methylation biomarkers for CRC detection in the training cohort (area under curve [AUC] = 0.90 (0.85–0.94]) and verified the model in the validation cohort (AUC = 0.92 [0.88–0.96]). The cfDNA methylation model robustly detected patients pre‐diagnosed with early‐stage CRC (AUC = 0.90 [0.86–0.95]) or AA (AUC = 0.85 [0.78–0.91]). Here we established and validated a non‐invasive cfDNA methylation model based on 11 DNA methylation biomarkers for the detection of early‐stage CRC and AA. The utilization of the model in clinical practice may contribute to the early diagnosis of CRC.
Collapse
Affiliation(s)
- Xianrui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yunfeng Zhang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Tuo Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaowen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yifeng Zou
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiling Deng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jia Ke
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Lian
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaosheng He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dezhi Zhao
- AnchorDx Medical Co. Ltd, Guangzhou, China
| | - Xuyu Cai
- AnchorDx Medical Co. Ltd, Guangzhou, China
| | - Zhiwei Chen
- AnchorDx Medical Co. Ltd, Guangzhou, China.,AnchorDx, Inc., Fremont, CA, USA
| | - Xiaojian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian-Bing Fan
- AnchorDx Medical Co. Ltd, Guangzhou, China.,Department of Pathology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Feng Gao
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
370
|
Jiang TY, Ma JJ, Zheng MH. Controversies and consensus in transanal total mesorectal excision (taTME): Is it a valid choice for rectal cancer? J Surg Oncol 2021; 123 Suppl 1:S59-S64. [PMID: 33650698 DOI: 10.1002/jso.26340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023]
Abstract
Transanal total mesorectal excision (taTME) is a novel approach to radical surgery for low rectal cancer. taTME is associated with the benefits of a higher rate of free distal resection margins (DRM) under direct visualization, better visualization of the mesorectal plane, and the feasibility of overcoming the restriction of the distal pelvis. Thus, it is increasingly used globally. In this review, we investigated whether taTME yields better short- and long-term outcomes than laparoscopic TME.
Collapse
Affiliation(s)
- Tian-Yu Jiang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jun-Jun Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min-Hua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
371
|
Gallardo-Gómez M, De Chiara L, Álvarez-Chaver P, Cubiella J. Colorectal cancer screening and diagnosis: omics-based technologies for development of a non-invasive blood-based method. Expert Rev Anticancer Ther 2021; 21:723-738. [PMID: 33507120 DOI: 10.1080/14737140.2021.1882858] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Colorectal cancer (CRC) is one of the most important health problems in the Western world. In order to reduce the burden of the disease, two strategies are proposed: screening and prompt detection in symptomatic patients. Although diagnosis and prevention are mainly based on colonoscopy, fecal hemoglobin detection has been widely implemented as a noninvasive strategy. Various studies aiming to discover blood-based biomarkers have recently emerged.Areas covered: The burgeoning omics field provides diverse high-throughput approaches for CRC blood-based biomarker discovery. In this review, we appraise the most robust and commonly used technologies within the fields of genomics, transcriptomics, epigenomics, proteomics, and metabolomics, together with their targeted validation approaches. We summarize the transference process from the discovery phase until clinical translation. Finally, we review the best candidate biomarkers and their potential clinical applicability.Expert opinion: Some available biomarkers are promising, especially in the field of epigenomics: DNA methylation and microRNA. Transference requires the joint collaboration of basic researchers, intellectual property experts, technology transfer officers and clinicians. Blood-based biomarkers will be selected not only based on their diagnostic accuracy and cost but also on their reliability, applicability to clinical analysis laboratories and their acceptance by the population.
Collapse
Affiliation(s)
- María Gallardo-Gómez
- Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain.,Biomedical Research Center (CINBIO), University of Vigo, Vigo, Spain
| | - Loretta De Chiara
- Department of Biochemistry, Genetics and Immunology, University of Vigo, Vigo, Spain.,Biomedical Research Center (CINBIO), University of Vigo, Vigo, Spain
| | - Paula Álvarez-Chaver
- Proteomics Unit, Service of Structural Determination, Proteomics and Genomics, Center for Scientific and Technological Research Support (CACTI), University of Vigo, Vigo, Spain
| | - Joaquin Cubiella
- Department of Gastroenterology, Hospital Universitario De Ourense, Ourense, Spain.,Instituto De Investigación Sanitaria Galicia Sur, Ourense, Spain.,Centro De Investigación Biomédica En Red Enfermedades Hepáticas Y Digestivas, Ourense, Spain
| |
Collapse
|
372
|
Han Y, Wang X, Mao E, Shen B, Huang L. lncRNA FLVCR1‑AS1 drives colorectal cancer progression via modulation of the miR‑381/RAP2A axis. Mol Med Rep 2021; 23:139. [PMID: 33313944 PMCID: PMC7751490 DOI: 10.3892/mmr.2020.11778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent types of cancer globally. Long non‑coding RNAs (lncRNAs) have been suggested to serve as vital regulators in CRC. lncRNA feline leukemia virus subgroup C receptor 1 antisense RNA 1 (FLVCR1‑AS1) is closely associated with the tumorigenesis of various types of cancer. The aim of the present study was to investigate the molecular mechanisms of lncRNA FLVCR1‑AS1 in CRC progression. The expression levels of FLVCR1‑AS1, microRNA (miR)‑381 and Ras‑related protein 2a (RAP2A) were measured by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). A Kaplan‑Meier analysis was performed to determine the overall survival rate of patients with CRC. Furthermore, cell viability, migration and invasion were assessed using Cell Counting Kit‑8 (CCK‑8) and Transwell assays. The interaction between genes was confirmed using dual‑luciferase reporter and pull‑down assays. The results demonstrated that FLVCR1‑AS1 was upregulated in CRC tissues and cells, and increased FLVCR1‑AS1 expression levels in patients with CRC were associated with poor prognosis. FLVCR1‑AS1 knockdown significantly attenuated the viability, migration and invasion ability of CRC cells. In addition, the results confirmed that FLVCR1‑AS1 directly binds with miR‑381‑3p, and that RAP2A is a direct target of miR‑381‑3p. The overexpression of FLVCR1‑AS1 increased RAP2A expression levels. Functional assays revealed that miR‑381 inhibitor or RAP2A overexpression attenuated the suppressive effects of FLVCR1‑AS1 silencing on CRC cell viability, migration and invasion. Overall, the findings of the current study suggest that FLVCR1‑AS1 promotes CRC progression via the miR‑381/RAP2A pathway. These findings may provide a novel approach for CRC treatment.
Collapse
Affiliation(s)
- Yi Han
- Department of Traumatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Xiaoyan Wang
- Department of Traumatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Enqiang Mao
- Department of Emergency, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Boyong Shen
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Liang Huang
- Department of Traumatology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| |
Collapse
|
373
|
Jin H, Wang J, Zhang C. The Value of Multi-targeted Fecal DNA Methylation Detection for Colorectal Cancer Screening in a Chinese Population. J Cancer 2021; 12:1644-1650. [PMID: 33613751 PMCID: PMC7890319 DOI: 10.7150/jca.47214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 12/11/2020] [Indexed: 12/23/2022] Open
Abstract
Objective: To design a multi-targeted fecal DNA methylation kit and explore its value for clinical application among Chinese people. Methods: Based on previous research, a multi-targeted fecal DNA methylation detection kit, using four genes, was designed and clinically validated. Results: The methylation PCR from 279 patients met the requirements for the detection criteria. When all four molecular markers were negative, the negative predictive value (NPV) for colorectal cancer was 100% and the NPV for colorectal polyps was 84.21%. When one molecular marker was positive, the sensitivity (Se) for colorectal cancer was 76.4%-90.3%, the specificity (Sp) was 68.3-93.4%, and the positive predictive value (PPV) for colorectal cancer was 54.5-85.5%, and the NPV was 87.0-95.0%. For colorectal polyps, the Se was 41.0-52.5%, Sp 69.5-91.5%, and the PPV for colorectal polyps was 41.0-70.3%, the NPV was 75.2-79.3%. When two molecular markers were positive, the Se for colorectal cancer was 52.6-73.7%, the Sp was 93.2-98.3%, the PPV for colorectal cancer was 84.6-96.2%, the NPV was 76.0-85.3%. For colorectal polyps, the Se was 25.9-40.7%, Sp was 93.2-98.3%, PPV for screening of colorectal polyps was 63.6-90.0%, and the NPV was 73.3-78.1%. When three molecular markers were positive, the Se for colorectal cancer was 31.6-52.6%, the Sp was 98.3-100.0%, the PPV for colorectal cancer was 94.4-100.0%, the NPV was 73.4-76.6%. For colorectal polyps, the Se was 14.8-25.9%, and Sp was 98.3-100.0%, the PPV for colorectal polyps was 85.7-100.0%, the NPV was 72.0-74.7%. When four molecular markers were positive, the Se for colorectal cancer was 31.6%, the Sp was 100.0%, and the colorectal cancer PPV was 100.0% and the NPV was 69.4%. For polyps, the Se was 14.8%, Sp was 100.0%, and PPV was 100.0% and the NPV was 72.0%. Conclusion: The multi-targeted fecal DNA methylation detection kit for colorectal cancer and polyps had the sensitivity and specificity to meet the requirements for screening of colorectal tumors, which is easy to operate, has stable results and important clinical value. Among the four molecular markers studied, when one marker was positive for DNA methylation, colonoscopy was required; as the number of positive methylation markers increased, the specificity for the diagnosis gradually increased as well.
Collapse
Affiliation(s)
- Heiying Jin
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, 23 Nanhu Road, Nanjing 210017, China
| | - Jun Wang
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, 23 Nanhu Road, Nanjing 210017, China
| | - Chunxia Zhang
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, 23 Nanhu Road, Nanjing 210017, China
| |
Collapse
|
374
|
Heisser T, Hoffmeister M, Brenner H. Effects of screening for colorectal cancer: Development, documentation and validation of a multistate Markov model. Int J Cancer 2021; 148:1973-1981. [PMID: 33320964 DOI: 10.1002/ijc.33437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/24/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022]
Abstract
Simulation models are a powerful tool to overcome gaps of evidence needed to inform medical decision-making. Here, we present development and application of COSIMO, a Markov-based Colorectal Cancer (CRC) Multi-state Simulation Model to simulate effects of CRC screening, along with a thorough assessment of the model's ability to reproduce real-life outcomes. Firstly, we provide a comprehensive documentation of COSIMO's development, structure and assumptions. Secondly, to assess the model's external validity, we compared model-derived cumulative incidence and prevalences of colorectal neoplasms to (a) results from KolosSal, a study in German screening colonoscopy participants, (b) registry-based estimates of CRC incidence in Germany, and (c) outcome patterns of randomized sigmoidoscopy screening studies. We found that (a) more than 90% of observed prevalences in the KolosSal study were within the 95% confidence intervals of the model-predicted neoplasm prevalences; (b) the 15-year cumulative CRC incidences estimated by simulations for the German population deviated by 0.0% to 0.2% units in men and 0.0% to 0.3% units in women when compared to corresponding registry-derived estimates; and (c) the time course of cumulative CRC incidence and mortality in the modeled intervention group and control group closely resembles the time course reported from sigmoidoscopy screening trials. Overall, COSIMO adequately predicted colorectal neoplasm prevalences and incidences in a German population for up to 25 years, with estimated patterns of the effect of screening colonoscopy resembling those seen in registry data and real-world studies. This suggests that the model may represent a valid tool to assess the comparative effectiveness of CRC screening strategies.
Collapse
Affiliation(s)
- Thomas Heisser
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
375
|
Abstract
PURPOSE OF REVIEW In the United States, only 67% of patients are up to date with colorectal cancer (CRC) screening. While colonoscopy is highly sensitive and specific for CRC and precursor lesion detection and removal, it is invasive, expensive and resource heavy. Hence, there is an unfulfilled need for multiple modality CRC screening that can improve current CRC screening rates and may be resource effective strategies when used in conjunction with a colonoscopy program. Our review highlights the complementary, often underutilized, noninvasive CRC screening methods with a focus on performance, risks, benefits, and recent updates. RECENT FINDINGS Studies demonstrate that fecal immunochemical testing (FIT) is superior to guaiac-based fecal occult blood tests for CRC screening. Studies show superiority of multitarget stool DNA test to FIT in sensitivity, though with concern for decreased specificity in setting of one-time tests. Technical advances continue to improve accuracy of colon capsule endoscopy. There are ongoing studies to characterize often difficult-to-detect high-risk lesions in computed tomography colonography. Septin 9 continues to have suboptimal accuracy for CRC screening, but has been shown to be associated with more advanced, invasive CRC stages. SUMMARY There are ongoing advances in noninvasive screening modalities for CRC; these should be considered as alternatives to colonoscopy in specific patient populations.
Collapse
Affiliation(s)
- Susan Lou
- Division of Gastroenterology, Department of Medicine, University of Minnesota
| | - Aasma Shaukat
- Division of Gastroenterology, Department of Medicine, University of Minnesota
- Division of Gastroenterology, Department of Medicine, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota, USA
| |
Collapse
|
376
|
Improved Survival Outcome and Access to Cancer Screening from Hemorrhoid in Patients with Rectal Cancer. Gastroenterol Res Pract 2020; 2020:5045142. [PMID: 33381167 PMCID: PMC7749767 DOI: 10.1155/2020/5045142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 02/08/2023] Open
Abstract
Background The interventions for hemorrhoid increase access to rectal cancer screening and thus might reduce cancer death. We aimed to examine the impact of hemorrhoid on survival outcomes in rectal cancer. Methods We identified 510 patients with stage I to III rectal cancer from a prospectively collected database. Patients were divided into hemorrhoid and non-hemorrhoid group. The primary endpoints were disease-free survival (DFS) and overall survival (OS). Results Hemorrhoid group had significantly more stage I-II diseases in comparison to nonhemorrhoid group (71.1% vs. 55.9%, P = 0.049). The hemorrhoid group had significantly better DFS and OS compared to nonhemorrhoid group, the hazard ratios (HRs) of which were 0.39 (95% CI 0.17-0.88, P = 0.018) and 0.33 (95% CI 0.12-0.92, P = 0.034), respectively. Multivariate analysis revealed that hemorrhoid was independently associated with DFS [adjusted HR 0.43 (95% CI 0.17-0.95, P = 0.045)]. A nomogram for predicting DFS outcome was generated based on hemorrhoid history, with a concordance index of 0.71 (95% CI 0.66-0.75, P < 0.001). Conclusions There may exist a screening effect and survival benefit from hemorrhoid in rectal cancer, which supports the significance of rectal cancer screening in lowering its mortality.
Collapse
|
377
|
Cohen JD, Diergaarde B, Papadopoulos N, Kinzler KW, Schoen RE. Tumor DNA as a Cancer Biomarker through the Lens of Colorectal Neoplasia. Cancer Epidemiol Biomarkers Prev 2020; 29:2441-2453. [PMID: 33033144 PMCID: PMC7710619 DOI: 10.1158/1055-9965.epi-20-0549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/06/2020] [Accepted: 09/30/2020] [Indexed: 12/24/2022] Open
Abstract
Biomarkers have a wide range of applications in the clinical management of cancer, including screening and therapeutic management. Tumor DNA released from neoplastic cells has become a particularly active area of cancer biomarker development due to the critical role somatic alterations play in the pathophysiology of cancer and the ability to assess released tumor DNA in accessible clinical samples, in particular blood (i.e., liquid biopsy). Many of the early applications of tumor DNA as a biomarker were pioneered in colorectal cancer due to its well-defined genetics and common occurrence, the effectiveness of early detection, and the availability of effective therapeutic options. Herein, in the context of colorectal cancer, we describe how the intended clinical application dictates desired biomarker test performance, how features of tumor DNA provide unique challenges and opportunities for biomarker development, and conclude with specific examples of clinical application of tumor DNA as a biomarker with particular emphasis on early detection.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Joshua D Cohen
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brenda Diergaarde
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nickolas Papadopoulos
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kenneth W Kinzler
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert E Schoen
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
378
|
Bu F, Zhu X, Zhu J, Liu Z, Wu T, Luo C, Lin K, Huang J. Bioinformatics Analysis Identifies a Novel Role of GINS1 Gene in Colorectal Cancer. Cancer Manag Res 2020; 12:11677-11687. [PMID: 33235499 PMCID: PMC7680165 DOI: 10.2147/cmar.s279165] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most lethal malignancies and the incidence of CRC has been on the rise. Herein, we aimed to identify effective biomarkers for early diagnosis and treatment of colorectal cancer via bioinformatic tools. Methods To identify differentially expressed genes (DEGs) in CRC, we downloaded CRC gene expression data from GSE24514 and GSE110223 datasets in Gene Expression Omnibus (GEO) and employed R to analyze the data. We further performed functional enrichment analysis of the DEGs on the DAVID gene ontology analysis tool. STRING database and Cytoscape visualization tool were employed to construct a PPI (protein–protein interaction) network and establish intensive intervals in the network. Immunohistochemistry, qRT-PCR and Western blotting were performed to identify the expression level of GINS1 in CRC. In vitro and in vivo experiments were performed to assess the impact of GINS1 in the pathogenesis of CRC in terms of proliferation, migration and metastasis. Results Among the two datasets, 389 DEGs were identified and used to construct a PPI network. These genes were mainly involved in cell proliferation and cell cycle. Among them, 15 genes including GINS1 were found to be strongly associated with the PPI network. We further performed immunohistochemistry, qRT-PCR and Western blotting to identify that GINS1 expression was higher in CRC than in paired normal tissues. Moreover, in vitro and in vivo experiments demonstrated GINS1 could promote the proliferation, invasion and migration of colorectal cancer cells. Conclusions GINS1 could be considered as a potential biomarker for CRC patients.
Collapse
Affiliation(s)
- Fanqin Bu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China.,Jiangxi Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Xiaojian Zhu
- Research Center of The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Jinfeng Zhu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China.,Jiangxi Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Zitao Liu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China.,Jiangxi Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Ting Wu
- Department of Infectious Diseases of Guixi Traditional Chinese Medicine Hospital, Yingtan, People's Republic of China
| | - Chen Luo
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China.,Jiangxi Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Kang Lin
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China.,Jiangxi Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Jun Huang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| |
Collapse
|
379
|
Cancerona: Challenges of Cancer Management in Times of COVID-19 Pandemic. ACTA ACUST UNITED AC 2020; 2:2005-2014. [PMID: 33015555 PMCID: PMC7524598 DOI: 10.1007/s42399-020-00549-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has infected millions of people worldwide and emerged to be the biggest global health threat claiming hundreds of thousands of lives at exponential rates. The severity of the disease increases with old age and presence of underlying health conditions, such as cancer. Managing cancer patients under these circumstances is rather challenging, given their compromised immunity and the overwhelmed health care services by COVID-19 community transmission. Thus, it is prudent to establish common guidelines for the monitoring and treatment of cancer patients. In this review, we comprehensively investigate the various aspects of cancer care during the COVID-19 pandemic, discuss challenges faced while treating cancer patients, and propose potential approaches to manage COVID-19 among this vulnerable population. We also discuss molecular aberrations and genetic changes associated with cancer and their role in affecting the virus' infectivity and severity. Lastly, we shed light on therapeutic approaches that can encompass both diseases without compromising one over the other.
Collapse
|
380
|
Zhu Z, Hou Q, Guo H. NT5DC2 knockdown inhibits colorectal carcinoma progression by repressing metastasis, angiogenesis and tumor-associated macrophage recruitment: A mechanism involving VEGF signaling. Exp Cell Res 2020; 397:112311. [PMID: 32991874 DOI: 10.1016/j.yexcr.2020.112311] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed tumors among human worldwide. Angiogenesis and tumor-associated macrophage (TAM) recruitment are closely associated with CRC development. Nevertheless, the mechanisms revealing CRC progression are still not fully understood. 5'-Nucleotidase domain containing 2 (NT5DC2), a member of the NT5DC family, modulates various cellular events to mediate tumor growth, and thus serves as a disgnostic biomarker. Here, we explored the potential of NT5DC2 on tumor progression in CRC. We first found that NT5DC2 expression was significantly up-regulated in CRC tissues and cell lines. CRC patients with higher NT5DC2 expression showed poor overall survival. Furthermore, CRC cell lines stably transfected with shNT5DC2 lentivirus plasmids exhibited markedly reduced cell proliferation, migration and invasion compared with the negative control group. Hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGF-A) expression levels were remarkably reduced in CRC cells with NT5DC2 deletion, along with evidently reduced tube formation in the HUVECs cultured in the collected conditional medium. The expression levels of CC chemokine ligand 2 (CCL2) and its receptor CCR2 were found to be greatly down-regulated in CRC cells transfected with shNT5DC2. Moreover, NT5DC2 knockdown markedly suppressed the activation of protein kinase-B/nuclear transcription factor κB (AKT/NF-κB) signaling in CRC cells. Furthermore, we found that NT5DC2 deletion obviously reduced the TAM recruitments through suppressing CCL2/CCR2 and AKT/NF-κB signaling pathways. Intriguingly, our in vitro experiments demonstrated that VEGF reduction was necessary for shNT5DC2-inhibited cell proliferation, migration, invasion, angiogenesis and TAM recruitment. In vivo studies also confirmed that NT5DC2 knockdown effectively reduced the tumor growth and VEGF expression in a xonegraft mouse model with CRC. Lung metastasis of CRC cells was also hindered by NT5DC2 deletion in vivo. Collectively, our results indicated a previously unrecognized NT5DC2/VEGF/CCL2 axis involved in CRC development and metastasis.
Collapse
Affiliation(s)
- Zhenyu Zhu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qingsheng Hou
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Hongliang Guo
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
381
|
Gao Z, Cao C, Bao Y, Fan Y, Chen G, Fu P. Systematic Review and Meta-Analysis of Multitargeted Tyrosine Kinase Inhibitors in Patients With Intractable Metastatic Colorectal Cancer. Technol Cancer Res Treat 2020; 19:1533033820943241. [PMID: 32914703 PMCID: PMC7488883 DOI: 10.1177/1533033820943241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: The treatment options for intractable metastatic colorectal cancer include regorafenib, trifluridine/tipiracil, and fruquintinib. In this study, we aimed to conduct a network meta-analysis for comparing the efficacy of these agents. Methods: We searched the PubMed, EMBASE, Cochrane Central Register of Controlled Trials, and ClinicalTrials databases for relevant literature, up to February 2020. The data were collected from randomized controlled trials on regorafenib, trifluridine/tipiracil, or fruquintinib, administered to patients with metastatic colorectal cancer who failed on treatment with oxaliplatin, irinotecan, or fluoropyrimidine. The primary end points, namely, the overall survival and progression-free survival, were analyzed for subsequent network analysis using the Review Manager and Aggregate Data Drug Information System software for performing direct and indirect comparisons. Results: A total of 7 trials were analyzed in this study. Trifluridine/tipiracil and regorafenib proved to be superior to the placebo, with respect to the overall survival (odds ratio: 0.38, 95% confidence interval: 0.27-0.52 for trifluridine/tipiracil; odds ratio: 0.47, 95% confidence interval: 0.26-0.84 for regorafenib) and progression-free survival (odds ratio: 0.18, 95% confidence interval: 0.05-0.67 for trifluridine/tipiracil; odds ratio: 0.06, 95% confidence interval: 0.04-0.09 for regorafenib). Regorafenib (80 mg) was superior to the placebo in terms of the overall survival and progression-free survival and inferior to trifluridine/tipiracil and fruquintinib. Network analysis revealed that the efficacy of trifluridine/tipiracil and fruquintinib was fundamentally similar, and both the agents were superior to regorafenib. Conclusion: Regorafenib (80 mg) was superior to the placebo, but inferior to 160 mg regorafenib, trifluridine/tipiracil, and fruquintinib. This study further revealed that the efficiency of trifluridine/tipiracil and fruquintinib is identical, but their toxicity profiles are different.
Collapse
Affiliation(s)
- Zhenzhen Gao
- Department of General surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China.,Both the authors contributed equally to this work
| | - Chenxi Cao
- Department of Clinical Oncology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China.,Both the authors contributed equally to this work
| | - Yi Bao
- Department of General surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yaohua Fan
- Department of General surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Gang Chen
- Department of Orthopedic, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Peng Fu
- Department of Orthopedic, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China.,Department of Musculoskeletal Oncology, The Second Affiliated Hospital of Jiaxing University, Nanjing, China
| |
Collapse
|
382
|
Petersen MM, Ferm L, Kleif J, Piper TB, Rømer E, Christensen IJ, Nielsen HJ. Triage May Improve Selection to Colonoscopy and Reduce the Number of Unnecessary Colonoscopies. Cancers (Basel) 2020; 12:E2610. [PMID: 32932734 PMCID: PMC7563245 DOI: 10.3390/cancers12092610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 12/18/2022] Open
Abstract
Implementation of population screening for colorectal cancer by direct colonoscopy or follow-up colonoscopy after a positive fecal blood test has challenged the overall capacity of bowel examinations. Certain countries are facing serious colonoscopy capacity constraints, which have led to waiting lists and long time latency of follow-up examinations. Various options for improvement are considered, including increased cut-off values of the fecal blood tests. Results from major clinical studies of blood-based, cancer-associated biomarkers have, however, led to focus on a Triage concept for improved selection to colonoscopy. The Triage test may include subject age, concentration of hemoglobin in a feces test and a combination of certain blood-based cancer-associated biomarkers. Recent results have indicated that Triage may reduce the requirements for colonoscopy by around 30%. Such results may be advantageous for the capacity, the healthcare budgets and in particular, the subjects, who do not need an unnecessary, unpleasant and risk-associated bowel examination.
Collapse
Affiliation(s)
- Mathias M. Petersen
- Department of Surgical Gastroenterology, Hvidovre Hospital, 2650 Hvidovre, Denmark; (M.M.P.); (L.F.); (J.K.); (T.B.P.); (E.R.); (I.J.C.)
| | - Linnea Ferm
- Department of Surgical Gastroenterology, Hvidovre Hospital, 2650 Hvidovre, Denmark; (M.M.P.); (L.F.); (J.K.); (T.B.P.); (E.R.); (I.J.C.)
| | - Jakob Kleif
- Department of Surgical Gastroenterology, Hvidovre Hospital, 2650 Hvidovre, Denmark; (M.M.P.); (L.F.); (J.K.); (T.B.P.); (E.R.); (I.J.C.)
| | - Thomas B. Piper
- Department of Surgical Gastroenterology, Hvidovre Hospital, 2650 Hvidovre, Denmark; (M.M.P.); (L.F.); (J.K.); (T.B.P.); (E.R.); (I.J.C.)
| | - Eva Rømer
- Department of Surgical Gastroenterology, Hvidovre Hospital, 2650 Hvidovre, Denmark; (M.M.P.); (L.F.); (J.K.); (T.B.P.); (E.R.); (I.J.C.)
| | - Ib J. Christensen
- Department of Surgical Gastroenterology, Hvidovre Hospital, 2650 Hvidovre, Denmark; (M.M.P.); (L.F.); (J.K.); (T.B.P.); (E.R.); (I.J.C.)
| | - Hans J. Nielsen
- Department of Surgical Gastroenterology, Hvidovre Hospital, 2650 Hvidovre, Denmark; (M.M.P.); (L.F.); (J.K.); (T.B.P.); (E.R.); (I.J.C.)
- Institute of Clinical Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
383
|
Wang J, Yu S, Chen G, Kang M, Jin X, Huang Y, Lin L, Wu D, Wang L, Chen J. A novel prognostic signature of immune-related genes for patients with colorectal cancer. J Cell Mol Med 2020; 24:8491-8504. [PMID: 32564470 PMCID: PMC7412433 DOI: 10.1111/jcmm.15443] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers with an estimated 1.8 million new cases worldwide and associated with high mortality rates of 881 000 CRC-related deaths in 2018. Screening programs and new therapies have only marginally improved the survival of CRC patients. Immune-related genes (IRGs) have attracted attention in recent years as therapeutic targets. The aim of this study was to identify an immune-related prognostic signature for CRC. To this end, we combined gene expression and clinical data from the CRC data sets of The Cancer Genome Atlas (TCGA) into an integrated immune landscape profile. We identified a total of 476 IRGs that were differentially expressed in CRC vs normal tissues, of which 18 were survival related according to univariate Cox analysis. Stepwise multivariate Cox proportional hazards analysis established an immune-related prognostic signature consisting of SLC10A2, FGF2, CCL28, NDRG1, ESM1, UCN, UTS2 and TRDC. The predictive ability of this signature for 3- and 5-year overall survival was determined using receiver operating characteristics (ROC), and the respective areas under the curve (AUC) were 79.2% and 76.6%. The signature showed moderate predictive accuracy in the validation and GSE38832 data sets as well. Furthermore, the 8-IRG signature correlated significantly with tumour stage, invasion, lymph node metastasis and distant metastasis by univariate Cox analysis, and was established an independent prognostic factor by multivariate Cox regression analysis for CRC. Gene set enrichment analysis (GSEA) revealed a relationship between the IRG prognostic signature and various biological pathways. Focal adhesions and ECM-receptor interactions were positively correlated with the risk scores, while cytosolic DNA sensing and metabolism-related pathways were negatively correlated. Finally, the bioinformatics results were validated by real-time RT-qPCR. In conclusion, we identified and validated a novel, immune-related prognostic signature for patients with CRC, and this signature reflects the dysregulated tumour immune microenvironment and has a potential for better CRC patient management.
Collapse
Affiliation(s)
- Jun Wang
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Shaojun Yu
- Department of Surgical Oncologythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guofeng Chen
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Muxing Kang
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xiaoli Jin
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yi Huang
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lele Lin
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Dan Wu
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lie Wang
- Bone Marrow Transplantation Center of the First Affiliated HospitalInstitute of ImmunologyZhejiang University School of MedicineHangzhouChina
| | - Jian Chen
- Department of Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
384
|
Zhu Y, Yang T, Wu Q, Yang X, Hao J, Deng X, Yang S, Gu C, Wang Z. Diagnostic performance of various liquid biopsy methods in detecting colorectal cancer: A meta-analysis. Cancer Med 2020; 9:5699-5707. [PMID: 32628360 PMCID: PMC7433831 DOI: 10.1002/cam4.3276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/06/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Liquid biopsy is a promising method in detecting colorectal cancer (CRC). However, previous meta‐analyses only focused on the diagnostic performance of cell‐free DNA (cfDNA). Therefore, we firstly evaluated the overall performance of all liquid biopsy methods. The pooled sensitivities, specificities, diagnostic odds ratios, and area under curve (AUC) of summary receiver operating characteristic curve for all liquid biopsy methods, exosomes, circulating tumor cells (CTCs), and cfDNA were calculated, respectively. A total of 62 articles involving 18 739 individuals were included. Fifty‐one articles were about cfDNA, five articles were about CTCs, and six articles were about exosomes. The overall performance of all liquid biopsy methods had a pooled sensitivity, specificity, and AUC of 0.77 (95% confidence interval [CI] 0.76‐0.78), 0.89 (95% CI 0.88‐0.90), and 0.9004, respectively. The sensitivities were 0.82 (95% CI 0.79‐0.85), 0.76 (95% CI 0.72‐0.80), and 0.76 (95% CI 0.75‐0.77) for CTCs, exosomes, and cfDNA, respectively. The specificities were 0.97 (95% CI95% CI 0.95‐0.99), 0.92 (95% CI 0.89‐0.94), and 0.88 (95% CI 0.87‐0.89) for CTCs, exosomes, and cfDNA, respectively. The AUC were 0.9772, 0.9037, and 0.8963 for CTCs, exosomes, and cfDNA, respectively. The overall performance of all liquid biopsy methods had great diagnostic value in detecting CRC, regardless of subtypes. Among all liquid biopsy methods, CTCs showed the best diagnostic performance.
Collapse
Affiliation(s)
- Yuzhou Zhu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tinghan Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingbin Wu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xuyang Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jianqi Hao
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiangbing Deng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shuo Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chaoyang Gu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ziqiang Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
385
|
Parigi TL, Aghemo A. HCV screening: Moving from theory to practice. Liver Int 2020; 40:1538-1540. [PMID: 32573061 DOI: 10.1111/liv.14515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 02/13/2023]
Affiliation(s)
| | - Alessio Aghemo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Division of Internal Medicine and Hepatology, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| |
Collapse
|
386
|
Abstract
Cost-effectiveness analysis compares benefits and costs of different interventions to inform decision makers. Alternatives are compared based on an incremental cost-effectiveness ratio reported in terms of cost per quality-adjusted life-year gained. Multiple cost-effectiveness analyses of colorectal cancer (CRC) screening have been performed. Although regional epidemiology of CRC, relevant screening strategies, regional health system, and applicable medical costs in local currencies differ by country and region, several overarching points emerge from literature on cost-effectiveness of CRC screening. Cost-effectiveness analysis informs decisions in ongoing debates, including preferred age to begin average-risk CRC screening, and implementation of CRC screening tailored to predicted CRC risk.
Collapse
Affiliation(s)
- Uri Ladabaum
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, 430 Broadway Street, Pavilion C, 3rd Floor C-326, Redwood City, CA 94063-6341, USA.
| |
Collapse
|
387
|
Bednarz-Misa I, Fleszar MG, Zawadzki M, Kapturkiewicz B, Kubiak A, Neubauer K, Witkiewicz W, Krzystek-Korpacka M. L-Arginine/NO Pathway Metabolites in Colorectal Cancer: Relevance as Disease Biomarkers and Predictors of Adverse Clinical Outcomes Following Surgery. J Clin Med 2020; 9:jcm9061782. [PMID: 32521714 PMCID: PMC7355854 DOI: 10.3390/jcm9061782] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/30/2020] [Accepted: 06/05/2020] [Indexed: 12/24/2022] Open
Abstract
The L-Arginine/NO pathway is involved in carcinogenesis and immunity. Its diagnostic and prognostic value in colorectal cancer (CRC) was determined using tandem mass spectrometry in 199 individuals (137 with CRC) and, during a three-day follow up, in 60 patients undergoing colorectal surgery. Citrulline was decreased and asymmetric (ADMA) and symmetric (SDMA) dimethylarginines and dimethylamine (DMA) were increased in CRC. The DMA increase corresponded with CRC advancement while arginine, ADMA, and SDMA levels were higher in left-sided cancers. Arginine, citrulline, ADMA, and DMA dropped and SDMA increased post incision. Females experienced a more substantial drop in arginine. The arginine and ADMA dynamics depended on blood loss. The initial SDMA increase was higher in patients requiring transfusions. Postoperative dynamics in arginine and dimethylarginines differed in robot-assisted and open surgery. Concomitant SDMA, citrulline, and DMA quantification displayed a 92% accuracy in detecting CRC. Monitoring changes in arginine, ADMA, and SDMA in the early postoperative period predicted postoperative ileus with 84% and surgical site infections with 90% accuracy. Changes in ADMA predicted operative morbidity with 90% and anastomotic leakage with 77% accuracy. If positively validated, L-arginine/NO pathway metabolites may facilitate CRC screening and surveillance, support differential diagnosis, and assist in clinical decision-making regarding patients recovering from colorectal surgery.
Collapse
Affiliation(s)
- Iwona Bednarz-Misa
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Mariusz G Fleszar
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Marek Zawadzki
- Department of Oncological Surgery, Regional Specialist Hospital, 51-124 Wrocław, Poland
- Department of Physiotherapy, Wroclaw Medical University, 51-618 Wrocław, Poland
| | - Bartosz Kapturkiewicz
- First Department of Oncological Surgery of Lower Silesian Oncology Center, 53-413 Wrocław, Poland
| | - Agnieszka Kubiak
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Katarzyna Neubauer
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, 50-556 Wrocław, Poland
| | - Wojciech Witkiewicz
- Department of Oncological Surgery, Regional Specialist Hospital, 51-124 Wrocław, Poland
- Research and Development Centre at Regional Specialist Hospital, 51-124 Wrocław, Poland
| | | |
Collapse
|
388
|
Siskova A, Cervena K, Kral J, Hucl T, Vodicka P, Vymetalkova V. Colorectal Adenomas-Genetics and Searching for New Molecular Screening Biomarkers. Int J Mol Sci 2020; 21:ijms21093260. [PMID: 32380676 PMCID: PMC7247353 DOI: 10.3390/ijms21093260] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a malignant disease with an incidence of over 1.8 million new cases per year worldwide. CRC outcome is closely related to the respective stage of CRC and is more favorable at less advanced stages. Detection of early colorectal adenomas is the key to survival. In spite of implemented screening programs showing efficiency in the detection of early precancerous lesions and CRC in asymptomatic patients, a significant number of patients are still diagnosed in advanced stages. Research on CRC accomplished during the last decade has improved our understanding of the etiology and development of colorectal adenomas and revealed weaknesses in the general approach to their detection and elimination. Recent studies seek to find a reliable non-invasive biomarker detectable even in the blood. New candidate biomarkers could be selected on the basis of so-called liquid biopsy, such as long non-coding RNA, microRNA, circulating cell-free DNA, circulating tumor cells, and inflammatory factors released from the adenoma into circulation. In this work, we focused on both genetic and epigenetic changes associated with the development of colorectal adenomas into colorectal carcinoma and we also discuss new possible biomarkers that are detectable even in adenomas prior to cancer development.
Collapse
Affiliation(s)
- Anna Siskova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Videnska 1083, 14200 Prague, Czech Republic; (K.C.); (J.K.); (V.V.)
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
- Correspondence: (A.S.); (P.V.); Tel.: +420-241062251 (A.S.); +420-241062694 (P.V.)
| | - Klara Cervena
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Videnska 1083, 14200 Prague, Czech Republic; (K.C.); (J.K.); (V.V.)
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | - Jan Kral
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Videnska 1083, 14200 Prague, Czech Republic; (K.C.); (J.K.); (V.V.)
- Institute for Clinical and Experimental Medicine, Videnska 1958/9, 14021 Prague, Czech Republic;
| | - Tomas Hucl
- Institute for Clinical and Experimental Medicine, Videnska 1958/9, 14021 Prague, Czech Republic;
| | - Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Videnska 1083, 14200 Prague, Czech Republic; (K.C.); (J.K.); (V.V.)
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 32300 Pilsen, Czech Republic
- Correspondence: (A.S.); (P.V.); Tel.: +420-241062251 (A.S.); +420-241062694 (P.V.)
| | - Veronika Vymetalkova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Videnska 1083, 14200 Prague, Czech Republic; (K.C.); (J.K.); (V.V.)
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 32300 Pilsen, Czech Republic
| |
Collapse
|
389
|
Li Q, Gao HD, Liu CC, Zhang H, Li XH, Wu J, Zhang XK. Impact of cuff-assisted colonoscopy for adenoma detection: A protocol of systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e20243. [PMID: 32443361 PMCID: PMC7253660 DOI: 10.1097/md.0000000000020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND Previous studies have reported that cuff-assisted colonoscopy (CAC) can be used for detection of adenoma (DA). However, there are inconsistent results regarding the CAC for DA. Thus, this study will systematically explore the impact of CAC for DA. METHODS In order to retrieve potential eligible articles, this study will identify the following electronic databases from their inceptions to present: MEDLINE, EMBASE, Cochrane Library, PSYCINFO, Web of Science, Chinese Biomedical Literature Database, and China National Knowledge Infrastructure. All electronic databases will be searched without any language limitation. We will consider case-controlled studies that focused on exploring the impacts of CAC for DA. Two authors will perform study selection, information collection and risk of bias assessment, respectively. Any discrepancies between 2 authors will be resolved through discussion with a third author. RESULTS This study will summarize the most recent evidence to assess the impact of CAC for DA. CONCLUSION The findings of this study will provide evidence of CAC for DA in clinical practice. SYSTEMATIC REVIEW REGISTRATION INPLASY202040042.
Collapse
Affiliation(s)
- Qi Li
- Department of Endoscopy Center, Dashiqiao Central Hospital, Dashiqiao
| | - Hai-de Gao
- Department of Digestive Surgery, The Medical Group of Zhengzhou First People's Hospital, Zhengzhou, 450004, China
| | - Chun-Cheng Liu
- Department of Digestive Surgery, The Medical Group of Zhengzhou First People's Hospital, Zhengzhou, 450004, China
| | - Hao Zhang
- Department of Digestive Surgery, The Medical Group of Zhengzhou First People's Hospital, Zhengzhou, 450004, China
| | - Xun-Hai Li
- Department of Digestive Surgery, The Medical Group of Zhengzhou First People's Hospital, Zhengzhou, 450004, China
| | - Jia Wu
- Department of Digestive Surgery, The Medical Group of Zhengzhou First People's Hospital, Zhengzhou, 450004, China
| | - Xian-Kai Zhang
- Department of Digestive Surgery, The Medical Group of Zhengzhou First People's Hospital, Zhengzhou, 450004, China
| |
Collapse
|
390
|
Long noncoding RNA RHPN1-AS1 promotes colorectal cancer progression via targeting miR-7-5p/OGT axis. Cancer Cell Int 2020; 20:54. [PMID: 32099527 PMCID: PMC7029493 DOI: 10.1186/s12935-020-1110-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/16/2020] [Indexed: 02/08/2023] Open
Abstract
Background Rhophilin Rho GTPase binding protein 1 antisense RNA 1 (RHPN1-AS1) is a newly discovered oncogene in several diseases, such as breast cancer, non-small cell lung cancer and uveal melanoma. Nevertheless, its molecular role in colorectal cancer (CRC) remains unknown. This paper explored the role of RHPN1-AS1 in CRC progression. Methods qRT-PCR was used to detect relevant RNAs expression. CCK-8, EdU, flow cytometry, Transwell and western blot assays were performed to investigate the function of RHPN1-AS1 in CRC cells. Xenograft model was constructed to evaluate the effects of RHPN1-AS1 on tumor growth in vivo. Mechanical experiments were performed to investigate the relationship between relative genes. Results RHPN1-AS1 was significantly overexpressed in CRC cell lines. Knockdown of RHPN1-AS1 could inhibit cell proliferation, while stimulating cell apoptosis in vitro. Cell migration and invasion abilities were greatly suppressed after silencing RHPN1-AS1. Besides, signal transducer and activator of transcription 3 (STAT3) served as transcription factor of RHPN1-AS1. Moreover, miR-7-5p was identified as a target of RHPN1-AS1 and was negatively regulated by RHPN1-AS1 in CRC. MiR-7-5p inhibition rescued the oncogenic function of RHPN1-AS1. Additionally, O-GlcNAcylation transferase (OGT) was the downstream target of miR-7-5p. OGT overexpression could abrogate the anti-tumor effects of RHPN1-AS1 knockdown on CRC. Conclusion RHPN1-AS1 regulates CRC by mediating OGT through sponging miR-7-5p, suggesting that RHPN1-AS1 might be a potential therapeutic target for CRC.
Collapse
|
391
|
Loktionov A. Biomarkers for detecting colorectal cancer non-invasively: DNA, RNA or proteins? World J Gastrointest Oncol 2020; 12:124-148. [PMID: 32104546 PMCID: PMC7031146 DOI: 10.4251/wjgo.v12.i2.124] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/30/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a global problem affecting millions of people worldwide. This disease is unique because of its slow progress that makes it preventable and often curable. CRC symptoms usually emerge only at advanced stages of the disease, consequently its early detection can be achieved only through active population screening, which markedly reduces mortality due to this cancer. CRC screening tests that employ non-invasively detectable biomarkers are currently being actively developed and, in most cases, samples of either stool or blood are used. However, alternative biological substances that can be collected non-invasively (colorectal mucus, urine, saliva, exhaled air) have now emerged as new sources of diagnostic biomarkers. The main categories of currently explored CRC biomarkers are: (1) Proteins (comprising widely used haemoglobin); (2) DNA (including mutations and methylation markers); (3) RNA (in particular microRNAs); (4) Low molecular weight metabolites (comprising volatile organic compounds) detectable by metabolomic techniques; and (5) Shifts in gut microbiome composition. Numerous tests for early CRC detection employing such non-invasive biomarkers have been proposed and clinically studied. While some of these studies generated promising early results, very few of the proposed tests have been transformed into clinically validated diagnostic/screening techniques. Such DNA-based tests as Food and Drug Administration-approved multitarget stool test (marketed as Cologuard®) or blood test for methylated septin 9 (marketed as Epi proColon® 2.0 CE) show good diagnostic performance but remain too expensive and technically complex to become effective CRC screening tools. It can be concluded that, despite its deficiencies, the protein (haemoglobin) detection-based faecal immunochemical test (FIT) today presents the most cost-effective option for non-invasive CRC screening. The combination of non-invasive FIT and confirmatory invasive colonoscopy is the current strategy of choice for CRC screening. However, continuing intense research in the area promises the emergence of new superior non-invasive CRC screening tests that will allow the development of improved disease prevention strategies.
Collapse
|
392
|
Del Vecchio Blanco G, Calabrese E, Biancone L, Monteleone G, Paoluzi OA. The impact of COVID-19 pandemic in the colorectal cancer prevention. Int J Colorectal Dis 2020; 35:1951-1954. [PMID: 32500432 PMCID: PMC7271141 DOI: 10.1007/s00384-020-03635-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) has led to a policy of severe restrictions in almost all countries strongly involved by the pandemic. National Health System is among activities suffering from the COVID-19 and the lockdown. AIM To evaluate the impact of COVID-19 in colorectal cancer (CRC) prevention. METHODS We report the change in the hospital organization to meet the growing healthcare needs determined by COVID-19. The limitations of CRC prevention secondary to COVID-19 and their effects on the healthcare are analyzed considering the features of the CRC screening programs in the average-risk population and endoscopic surveillance in patients with inflammatory bowel diseases (IBD). RESULTS The interruption of CRC prevention may lead to a delayed diagnosis of CRC, possibly in a more advanced stage. The economic burden and the impact on workload for gastroenterologists, surgeons, and oncologists will be greater as long as the CRC prevention remains suspended. To respond to the increased demand for colonoscopy once COVID-19 will be under control, we should optimize the resources. It will be necessary to stratify the CRC risk and reach an order of priority. It should be implemented the number of health workers, equipment, and spaces dedicated to performing colonoscopy for screening purpose and in subjects with alarm symptoms in the shortest time. To this aim, the funds earmarked for healthcare should be increased. CONCLUSION The economic impact will be dramatic, but COVID-19 is the demonstration that healthcare has to be the primary goal of humans.
Collapse
Affiliation(s)
- Giovanna Del Vecchio Blanco
- grid.6530.00000 0001 2300 0941Gastroenterology Unit, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Emma Calabrese
- grid.6530.00000 0001 2300 0941Gastroenterology Unit, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Livia Biancone
- grid.6530.00000 0001 2300 0941Gastroenterology Unit, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Giovanni Monteleone
- grid.6530.00000 0001 2300 0941Gastroenterology Unit, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Omero Alessandro Paoluzi
- grid.6530.00000 0001 2300 0941Gastroenterology Unit, Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|