1
|
Xu M, Yang JY, Meng T. Effectiveness of colonoscopy, immune fecal occult blood testing, and risk-graded screening strategies in colorectal cancer screening. World J Gastrointest Surg 2024; 16:2270-2280. [PMID: 39087098 PMCID: PMC11287692 DOI: 10.4240/wjgs.v16.i7.2270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignant tumors, and early screening is crucial to improving the survival rate of patients. The combination of colonoscopy and immune fecal occult blood detection has garnered significant attention as a novel method for CRC screening. Colonoscopy and fecal occult blood tests, when combined, can improve screening accuracy and early detection rates, thereby facilitating early intervention and treatment. However, certain risks and costs accompany it, making the establishment of a risk classification model crucial for accurate classification and management of screened subjects. AIM To evaluate the feasibility and effectiveness of colonoscopy, immune fecal occult blood test (FIT), and risk-graded screening strategies in CRC screening. METHODS Based on the randomized controlled trial of CRC screening in the population conducted by our hospital May 2020 to May 2023, participants who met the requirements were randomly assigned to a colonoscopy group, an FIT group, or a graded screening group at a ratio of 1:2:2 (after risk assessment, the high-risk group received colonoscopy, the low-risk group received an FIT test, and the FIT-positive group received colonoscopy). The three groups received CRC screening with different protocols, among which the colonoscopy group only received baseline screening, and the FIT group and the graded screening group received annual follow-up screening based on baseline screening. The primary outcome was the detection rate of advanced tumors, including CRC and advanced adenoma. The population participation rate, advanced tumor detection rate, and colonoscopy load of the three screening programs were compared. RESULTS A total of 19373 subjects who met the inclusion and exclusion criteria were enrolled, including 8082 males (41.7%) and 11291 females (58.3%). The mean age was 60.05 ± 6.5 years. Among them, 3883 patients were enrolled in the colonoscopy group, 7793 in the FIT group, and 7697 in the graded screening group. Two rounds of follow-up screening were completed in the FIT group and the graded screening group. The graded screening group (89.2%) and the colonoscopy group (42.3%) had the lowest overall screening participation rates, while the FIT group had the highest (99.3%). The results of the intentional analysis showed that the detection rate of advanced tumors in the colonoscopy group was greater than that of the FIT group [2.76% vs 2.17%, odds ratio (OR) = 1.30, 95% confidence interval (CI): 1.01-1.65, P = 0.037]. There was no significant difference in the detection rate of advanced tumors between the colonoscopy group and the graded screening group (2.76% vs 2.35%, OR = 1.9, 95%CI: 0.93-1.51, P = 0.156), as well as between the graded screening group and the FIT group (2.35% vs 2.17%, OR = 1.09%, 95%CI: 0.88-1.34, P = 0.440). The number of colonoscopy examinations required for each patient with advanced tumors was used as an index to evaluate the colonoscopy load during population screening. The graded screening group had the highest colonoscopy load (15.4 times), followed by the colonoscopy group (10.2 times), and the FIT group had the lowest (7.8 times). CONCLUSION A hierarchical screening strategy based on CRC risk assessment is feasible for screening for CRC in the population. It can be used as an effective supplement to traditional colonoscopy and FIT screening programs.
Collapse
Affiliation(s)
- Ming Xu
- Department of Colorectal Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Qingdao 266000, Shandong Province, China
| | - Jing-Yi Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Tao Meng
- Department of Colorectal Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Qingdao 266000, Shandong Province, China
| |
Collapse
|
2
|
Wang ZM, Pan SP, Zhang JJ, Zhou J. Prediction and analysis of albumin-bilirubin score combined with liver function index and carcinoembryonic antigen on liver metastasis of colorectal cancer. World J Gastrointest Surg 2024; 16:1670-1680. [PMID: 38983332 PMCID: PMC11230030 DOI: 10.4240/wjgs.v16.i6.1670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/13/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignant tumor, and liver metastasis is one of the main recurrence and metastasis modes that seriously affect patients' survival rate and quality of life. Indicators such as albumin bilirubin (ALBI) score, liver function index, and carcinoembryonic antigen (CEA) have shown some potential in the prediction of liver metastasis but have not been fully explored. AIM To evaluate its predictive value for liver metastasis of CRC by conducting the combined analysis of ALBI, liver function index, and CEA, and to provide a more accurate liver metastasis risk assessment tool for clinical treatment guidance. METHODS This study retrospectively analyzed the clinical data of patients with CRC who received surgical treatment in our hospital from January 2018 to July 2023 and were followed up for 24 months. According to the follow-up results, the enrolled patients were divided into a liver metastasis group and a nonliver metastasis group and randomly divided into a modeling group and a verification group at a ratio of 2:1. The risk factors for liver metastasis in patients with CRC were analyzed, a prediction model was constructed by least absolute shrinkage and selection operator (LASSO) logistic regression, internal validation was performed by the bootstrap method, the reliability of the prediction model was evaluated by subject-work characteristic curves, calibration curves, and clinical decision curves, and a column graph was drawn to show the prediction results. RESULTS Of 130 patients were enrolled in the modeling group and 65 patients were enrolled in the verification group out of the 195 patients with CRC who fulfilled the inclusion and exclusion criteria. Through LASSO regression variable screening and logistic regression analysis. The ALBI score, alanine aminotransferase (ALT), and CEA were found to be independent predictors of liver metastases in CRC patients [odds ratio (OR) = 8.062, 95% confidence interval (CI): 2.545-25.540], (OR = 1.037, 95%CI: 1.004-1.071) and (OR = 1.025, 95%CI: 1.008-1.043). The area under the receiver operating characteristic curve (AUC) for the combined prediction of CRLM in the modeling group was 0.921, with a sensitivity of 78.0% and a specificity of 95.0%. The H-index was 0.921, and the H-L fit curve had χ2 = 0.851, a P value of 0.654, and a slope of the calibration curve approaching 1. This indicates that the model is extremely accurate, and the clinical decision curve demonstrates that it can be applied effectively in the real world. We conducted internal verification of one thousand resamplings of the modeling group data using the bootstrap method. The AUC was 0.913, while the accuracy was 0.869 and the kappa consistency was 0.709. The combination prediction of liver metastasis in patients with CRC in the verification group had an AUC of 0.918, sensitivity of 85.0%, specificity of 95.6%, C-index of 0.918, and an H-L fitting curve with χ 2 = 0.586, P = 0.746. CONCLUSION The ALBI score, ALT level, and CEA level have a certain value in predicting liver metastasis in patients with CRC. These three criteria exhibit a high level of efficacy in forecasting liver metastases in patients diagnosed with CRC. The risk prediction model developed in this work shows great potential for practical application.
Collapse
Affiliation(s)
- Zhan-Mei Wang
- Department of Medical Oncology, Qilu Hospital (Qingdao), Cheeloo College Medicine, Shandong University, Qingdao 266000, Shandong Province, China
| | - Shu-Ping Pan
- Department of Gastroenterology, Feicheng People’s Hospital, Feicheng 271600, Shandong Province, China
| | - Jing-Jing Zhang
- Department of Anus and Intestine Surgery, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China
| | - Jun Zhou
- Department of Oncology, Qilu Hospital (Qingdao), Cheeloo College Medicine, Shandong University, Qingdao 266000, Shandong Province, China
| |
Collapse
|
3
|
McAuliffe T, Anderson JC, Larson RJ, Robertson DJ. Systematic scoping review: Use of the faecal immunochemical test residual buffer to enhance colorectal cancer screening. Aliment Pharmacol Ther 2024; 59:1033-1048. [PMID: 38534182 DOI: 10.1111/apt.17947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/07/2023] [Accepted: 03/03/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND The faecal immunochemical test (FIT) is an inexpensive and convenient modality to screen for colorectal cancer. However, its one-time sensitivity for detecting colorectal cancer and cancer precursors is limited. There is growing interest in using the non-haemoglobin contents of FIT residual buffer to enhance colonic neoplasia detection. AIM To establish from the literature a framework to catalogue candidate biomarkers within FIT residual buffer for non-invasive colorectal cancer screening. METHODS The search strategy evaluated PubMed, Scopus, Web of Science, Embase, and Google Scholar for publications through 25 October 2023, with search terms including FIT, buffer, OC-sensor, biomarkers, microbiome, microRNA (miR), colon, rectum, screening, neoplasm, and early detection. Studies employing home-based collection samples using quantitative FIT first processed for haemoglobin were included. One author reviewed all articles; a second author completed a 20% full-text audit to ensure adherence to eligibility criteria. RESULTS A broad search yielded 1669 studies and application of eligibility criteria identified 18 relevant studies. Multiple protein, DNA/RNA, and microbiome biomarkers (notably haptoglobin, miR-16, miR-27a-3p, miR-92a, miR-148a-3p, miR-223, miR-421, let-7b-5p, and Tyzzerella 4) were associated with colorectal neoplasia. Furthermore, studies highlighted the short-term stability of biomarkers for clinical use and long-term stability for research purposes. CONCLUSIONS This scoping review summarises the framework and progress of research on stability of biomarkers in FIT residual buffer and their associations with colorectal neoplasia to guide opportunities for further confirmatory studies to enhance colorectal cancer screening.
Collapse
Affiliation(s)
| | - Joseph C Anderson
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Department of Veterans Affairs Medical Center, White River Junction, Vermont, USA
| | - Robin J Larson
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- The Dartmouth Institute for Health Policy and Clinical Practice, Lebanon, New Hampshire, USA
| | - Douglas J Robertson
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
- Department of Veterans Affairs Medical Center, White River Junction, Vermont, USA
| |
Collapse
|
4
|
Chavez-Yenter D, Goodman MS, Chen Y, Chu X, Bradshaw RL, Lorenz Chambers R, Chan PA, Daly BM, Flynn M, Gammon A, Hess R, Kessler C, Kohlmann WK, Mann DM, Monahan R, Peel S, Kawamoto K, Del Fiol G, Sigireddi M, Buys SS, Ginsburg O, Kaphingst KA. Association of Disparities in Family History and Family Cancer History in the Electronic Health Record With Sex, Race, Hispanic or Latino Ethnicity, and Language Preference in 2 Large US Health Care Systems. JAMA Netw Open 2022; 5:e2234574. [PMID: 36194411 PMCID: PMC9533178 DOI: 10.1001/jamanetworkopen.2022.34574] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/12/2022] [Indexed: 11/14/2022] Open
Abstract
Importance Clinical decision support (CDS) algorithms are increasingly being implemented in health care systems to identify patients for specialty care. However, systematic differences in missingness of electronic health record (EHR) data may lead to disparities in identification by CDS algorithms. Objective To examine the availability and comprehensiveness of cancer family history information (FHI) in patients' EHRs by sex, race, Hispanic or Latino ethnicity, and language preference in 2 large health care systems in 2021. Design, Setting, and Participants This retrospective EHR quality improvement study used EHR data from 2 health care systems: University of Utah Health (UHealth) and NYU Langone Health (NYULH). Participants included patients aged 25 to 60 years who had a primary care appointment in the previous 3 years. Data were collected or abstracted from the EHR from December 10, 2020, to October 31, 2021, and analyzed from June 15 to October 31, 2021. Exposures Prior collection of cancer FHI in primary care settings. Main Outcomes and Measures Availability was defined as having any FHI and any cancer FHI in the EHR and was examined at the patient level. Comprehensiveness was defined as whether a cancer family history observation in the EHR specified the type of cancer diagnosed in a family member, the relationship of the family member to the patient, and the age at onset for the family member and was examined at the observation level. Results Among 144 484 patients in the UHealth system, 53.6% were women; 74.4% were non-Hispanic or non-Latino and 67.6% were White; and 83.0% had an English language preference. Among 377 621 patients in the NYULH system, 55.3% were women; 63.2% were non-Hispanic or non-Latino, and 55.3% were White; and 89.9% had an English language preference. Patients from historically medically undeserved groups-specifically, Black vs White patients (UHealth: 17.3% [95% CI, 16.1%-18.6%] vs 42.8% [95% CI, 42.5%-43.1%]; NYULH: 24.4% [95% CI, 24.0%-24.8%] vs 33.8% [95% CI, 33.6%-34.0%]), Hispanic or Latino vs non-Hispanic or non-Latino patients (UHealth: 27.2% [95% CI, 26.5%-27.8%] vs 40.2% [95% CI, 39.9%-40.5%]; NYULH: 24.4% [95% CI, 24.1%-24.7%] vs 31.6% [95% CI, 31.4%-31.8%]), Spanish-speaking vs English-speaking patients (UHealth: 18.4% [95% CI, 17.2%-19.1%] vs 40.0% [95% CI, 39.7%-40.3%]; NYULH: 15.1% [95% CI, 14.6%-15.6%] vs 31.1% [95% CI, 30.9%-31.2%), and men vs women (UHealth: 30.8% [95% CI, 30.4%-31.2%] vs 43.0% [95% CI, 42.6%-43.3%]; NYULH: 23.1% [95% CI, 22.9%-23.3%] vs 34.9% [95% CI, 34.7%-35.1%])-had significantly lower availability and comprehensiveness of cancer FHI (P < .001). Conclusions and Relevance These findings suggest that systematic differences in the availability and comprehensiveness of FHI in the EHR may introduce informative presence bias as inputs to CDS algorithms. The observed differences may also exacerbate disparities for medically underserved groups. System-, clinician-, and patient-level efforts are needed to improve the collection of FHI.
Collapse
Affiliation(s)
- Daniel Chavez-Yenter
- Huntsman Cancer Institute, University of Utah, Salt Lake City
- Department of Communication, University of Utah, Salt Lake City
| | - Melody S. Goodman
- School of Global Public Health, New York University, New York, New York
| | - Yuyu Chen
- School of Global Public Health, New York University, New York, New York
| | - Xiangying Chu
- School of Global Public Health, New York University, New York, New York
| | - Richard L. Bradshaw
- Department of Biomedical Informatics, University of Utah, Salt Lake City
- School of Medicine, University of Utah Health, Salt Lake City, Utah
| | | | | | - Brianne M. Daly
- Huntsman Cancer Institute, University of Utah, Salt Lake City
| | - Michael Flynn
- School of Medicine, University of Utah Health, Salt Lake City, Utah
| | - Amanda Gammon
- Huntsman Cancer Institute, University of Utah, Salt Lake City
| | - Rachel Hess
- Department of Population Health Sciences, University of Utah, Salt Lake City
- Department of Internal Medicine, University of Utah, Salt Lake City
| | - Cecelia Kessler
- Huntsman Cancer Institute, University of Utah, Salt Lake City
| | | | - Devin M. Mann
- Department of Population Health, New York University Grossman School of Medicine, New York University, New York, New York
| | - Rachel Monahan
- Perlmutter Cancer Center, NYU Langone Health, New York, New York
- Department of Population Health, New York University Grossman School of Medicine, New York University, New York, New York
| | - Sara Peel
- Huntsman Cancer Institute, University of Utah, Salt Lake City
| | - Kensaku Kawamoto
- Department of Biomedical Informatics, University of Utah, Salt Lake City
| | - Guilherme Del Fiol
- Department of Biomedical Informatics, University of Utah, Salt Lake City
| | | | - Saundra S. Buys
- Huntsman Cancer Institute, University of Utah, Salt Lake City
- Department of Internal Medicine, University of Utah, Salt Lake City
| | - Ophira Ginsburg
- Center for Global Health, National Cancer Institute, Rockville, Maryland
| | - Kimberly A. Kaphingst
- Huntsman Cancer Institute, University of Utah, Salt Lake City
- Department of Communication, University of Utah, Salt Lake City
| |
Collapse
|
5
|
Matboli M, Shafei AE, Ali MA, El-Din Ahmed TS, Naser M, Abdel-Rahman T, Anber N, Ali M. Role of extracellular LncRNA-SNHG14/miRNA-3940-5p/NAP12 mRNA in colorectal cancer. Arch Physiol Biochem 2021; 127:479-485. [PMID: 31397210 DOI: 10.1080/13813455.2019.1650070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND We aim to identify and analyze the expression of dyregulated RNAs in colorectal cancer (CRC). METHODS We selected a panel of RNAs specific to CRC composed of Nucleosome Assembly Protein 1 Like 2 (NAP1L2) mRNA, LNCRNA SNHG14 small nucleolar RNA host gene 14 (LNCRNA SNHG14) and homo sapiens microRNA-3940-5p(hsa-miRNA-3940-5p) from genetic and epigenetic databases. Validation of the chosen RNAs was achieved by real time quantitative PCR in sera of patients with CRC, versus controls groups (benign lesions and healthy individual). RESULTS We found that LLNCRNA SNHG14, hsa-miRNA-3940-5p and NAP1L2 mRNA had an excellent performance characteristics and more superior than CEA, and CA19.9 for differentiating CRC from controls. Combined expression of lncRNA SNHG14- hsa-miR-3940-5p and NAP1L2 mRNA had reached 100% sensitivity with accuracy 93%. Interestingly, serum hsa-miRNA-3940-5p could be an independent prognostic factor in CRC. CONCLUSION The extracellular lncRNA SNHG14- hsa-miR-3940-5p - NAP1L2 mRNA may aid in CRC management.KEY MESSAGESThe extracellular RNAs provide a potential class of noninvasive biomarkers with high specificity, accuracy and stability for detection of CRC.We used insilico data analysis followed by qPCR for detection of differential NAP1L2 gene expression with the selected epigenetic regulators.Our data presented interesting biomarker panel (NAP1L2 gene, lncRNA-SNHG14 and hsa-miR-3940-5p) that may be potential for CRC diagnosis and prognosis.
Collapse
Affiliation(s)
- Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Ain Shams Faculty of Medicine, Cairo, Egypt
| | - Ayman E Shafei
- Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Mahmoud A Ali
- Biomedical Research Department, Armed Forces College of Medicine, Cairo, Egypt
| | | | - Mahmoud Naser
- Biomedical Research Department, Armed Forces College of Medicine, Cairo, Egypt
| | - Tamer Abdel-Rahman
- Tropical Medicine Department, Ain Shams Faculty of Medicine, Cairo, Egypt
| | - Nahla Anber
- Fellow of Biochemistry, Mansoura University, Cairo, Egypt
| | - Marwa Ali
- Medical Biochemistry and Molecular Biology Department, Ain Shams Faculty of Medicine, Cairo, Egypt
| |
Collapse
|
6
|
Chavez-Yenter D, Kimball KE, Kohlmann W, Lorenz Chambers R, Bradshaw RL, Espinel WF, Flynn M, Gammon A, Goldberg E, Hagerty KJ, Hess R, Kessler C, Monahan R, Temares D, Tobik K, Mann DM, Kawamoto K, Del Fiol G, Buys SS, Ginsburg O, Kaphingst KA. Patient Interactions With an Automated Conversational Agent Delivering Pretest Genetics Education: Descriptive Study. J Med Internet Res 2021; 23:e29447. [PMID: 34792472 PMCID: PMC8663668 DOI: 10.2196/29447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/22/2021] [Accepted: 09/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background Cancer genetic testing to assess an individual’s cancer risk and to enable genomics-informed cancer treatment has grown exponentially in the past decade. Because of this continued growth and a shortage of health care workers, there is a need for automated strategies that provide high-quality genetics services to patients to reduce the clinical demand for genetics providers. Conversational agents have shown promise in managing mental health, pain, and other chronic conditions and are increasingly being used in cancer genetic services. However, research on how patients interact with these agents to satisfy their information needs is limited. Objective Our primary aim is to assess user interactions with a conversational agent for pretest genetics education. Methods We conducted a feasibility study of user interactions with a conversational agent who delivers pretest genetics education to primary care patients without cancer who are eligible for cancer genetic evaluation. The conversational agent provided scripted content similar to that delivered in a pretest genetic counseling visit for cancer genetic testing. Outside of a core set of information delivered to all patients, users were able to navigate within the chat to request additional content in their areas of interest. An artificial intelligence–based preprogrammed library was also established to allow users to ask open-ended questions to the conversational agent. Transcripts of the interactions were recorded. Here, we describe the information selected, time spent to complete the chat, and use of the open-ended question feature. Descriptive statistics were used for quantitative measures, and thematic analyses were used for qualitative responses. Results We invited 103 patients to participate, of which 88.3% (91/103) were offered access to the conversational agent, 39% (36/91) started the chat, and 32% (30/91) completed the chat. Most users who completed the chat indicated that they wanted to continue with genetic testing (21/30, 70%), few were unsure (9/30, 30%), and no patient declined to move forward with testing. Those who decided to test spent an average of 10 (SD 2.57) minutes on the chat, selected an average of 1.87 (SD 1.2) additional pieces of information, and generally did not ask open-ended questions. Those who were unsure spent 4 more minutes on average (mean 14.1, SD 7.41; P=.03) on the chat, selected an average of 3.67 (SD 2.9) additional pieces of information, and asked at least one open-ended question. Conclusions The pretest chat provided enough information for most patients to decide on cancer genetic testing, as indicated by the small number of open-ended questions. A subset of participants were still unsure about receiving genetic testing and may require additional education or interpersonal support before making a testing decision. Conversational agents have the potential to become a scalable alternative for pretest genetics education, reducing the clinical demand on genetics providers.
Collapse
Affiliation(s)
- Daniel Chavez-Yenter
- Department of Communication, University of Utah, Salt Lake City, UT, United States.,Cancer Control and Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Kadyn E Kimball
- Cancer Control and Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Wendy Kohlmann
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | | | - Richard L Bradshaw
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, United States
| | - Whitney F Espinel
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Michael Flynn
- University of Utah Health, Salt Lake City, UT, United States
| | - Amanda Gammon
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Eric Goldberg
- Department of Medicine, New York University Grossman School of Medicine, New York University, New York, NY, United States
| | - Kelsi J Hagerty
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Rachel Hess
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, United States
| | - Cecilia Kessler
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Rachel Monahan
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, United States.,Department of Population Health, New York University Grossman School of Medicine, New York University, New York, NY, United States
| | - Danielle Temares
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, United States
| | - Katie Tobik
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Devin M Mann
- Department of Population Health, New York University Grossman School of Medicine, New York University, New York, NY, United States
| | - Kensaku Kawamoto
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, United States
| | - Guilherme Del Fiol
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, United States
| | - Saundra S Buys
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Ophira Ginsburg
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, United States.,Department of Population Health, New York University Grossman School of Medicine, New York University, New York, NY, United States
| | - Kimberly A Kaphingst
- Department of Communication, University of Utah, Salt Lake City, UT, United States.,Cancer Control and Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, United States
| |
Collapse
|
7
|
Bai Y, Cho Lee W, Li G, So WKW. Development and feasibility of an evidence-based and theory-driven tailored mHealth communication intervention to increase colonoscopy screening rate in first-degree relatives of people with colorectal cancer. Eur J Oncol Nurs 2021; 56:102063. [PMID: 34847402 DOI: 10.1016/j.ejon.2021.102063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 08/27/2021] [Accepted: 11/08/2021] [Indexed: 01/22/2023]
Abstract
PURPOSE To develop and investigate the feasibility of an evidence-based and theory-driven tailored communication to increase colonoscopy screening rates amongst first-degree relatives of colorectal cancer patients. METHOD Based on the tailoring process and identified evidence from the systematic review conducted by the research team, the tailored communication was developed from four aspects: (1) tailoring variables (e.g., demographic, behavioural, and psychosocial characteristics), (2) decision rules, (3) tailored messages and (4) delivery plan. Expert (n = 5) and layman review (n = 5) were conducted to ensure the content validity of decision rules and tailored messages. A single-blinded, family-based cluster randomised controlled trial (n = 21) tested the feasibility and acceptability of the intervention. RESULTS A three-session mobile-based tailored intervention with clear decision rules and 27 tailored messages were developed. In the feasibility study, the recruitment rate was 34.4%. The response rate at 1-month post-intervention was 61.9%. After a voice call was added for nonresponders, the response rate at the 3-month post-intervention increased to 81%. All participants were satisfied with the intervention and agreed that the intervention helped them understand the risks and appropriate screening recommendations. CONCLUSION The development of a three-session mobile-based tailored intervention with an integrated tailoring decision and message system was reported in this study. Given its remote nature, the mobile-based tailored intervention may encounter challenges in family recruitment and online assessment. Suggestions on (1) study design to avoid contamination, (2) recruitment approaches and (3) strategies to promote response to online questionnaires were made for a future definitive trial.
Collapse
Affiliation(s)
- Yang Bai
- School of Nursing, Sun Yat-Sen University, Guangzhou, China; The Nethersole School of Nursing, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Wong Cho Lee
- The Nethersole School of Nursing, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gairui Li
- Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Winnie K W So
- The Nethersole School of Nursing, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
8
|
Loosen SH, Roderburg C, Alizai PH, Roeth AA, Schmitz SM, Vucur M, Luedde M, Schöler D, Paffenholz P, Tacke F, Trautwein C, Luedde T, Neumann UP, Ulmer TF. Comparative Analysis of Circulating Biomarkers for Patients Undergoing Resection of Colorectal Liver Metastases. Diagnostics (Basel) 2021; 11:diagnostics11111999. [PMID: 34829346 PMCID: PMC8622404 DOI: 10.3390/diagnostics11111999] [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: 09/06/2021] [Revised: 10/07/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Surgical tumor resection has evolved as a potentially curative therapy for patients with resectable colorectal liver metastases (CRLM). However, disease recurrence is common and the available preoperative stratification strategies are often imprecise to identify the ideal candidates for surgical treatment, resulting in a postoperative 5-year survival rate below 50%. Data on the prognostic value of CEA, CA19-9 and other common laboratory parameters after CRLM resection are scarce and partly inconclusive. Here, we analyzed the prognostic potential of circulating CEA and CA19-9 in comparison to other standard laboratory markers in resectable CRLM patients. Serum levels of tumor markers and other laboratory parameters were analyzed in 125 patients with CRLM undergoing tumor resection at a tertiary referral center. Results were correlated with clinical data and outcome. Both tumor markers were significantly elevated in CRLM patients compared to healthy controls. Interestingly, elevated levels of CEA, CA19-9 and C-reactive protein (CRP) were associated with an unfavorable prognosis after CRLM resection in Kaplan-Meier curve analysis. However, only CEA and not CA19-9 or CRP serum levels were an independent prognostic marker in multivariate Cox regression analysis. Our data demonstrate that circulating levels of CEA rather than CA19-9 might be a valuable addition to the existing preoperative stratification algorithms to identify patients with a poor prognosis after CRLM resection.
Collapse
Affiliation(s)
- Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (C.R.); (M.V.); (D.S.); (T.L.)
- Correspondence:
| | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (C.R.); (M.V.); (D.S.); (T.L.)
| | - Patrick H. Alizai
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; (P.H.A.); (A.A.R.); (S.M.S.); (U.P.N.); (T.F.U.)
| | - Anjali A. Roeth
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; (P.H.A.); (A.A.R.); (S.M.S.); (U.P.N.); (T.F.U.)
| | - Sophia M. Schmitz
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; (P.H.A.); (A.A.R.); (S.M.S.); (U.P.N.); (T.F.U.)
| | - Mihael Vucur
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (C.R.); (M.V.); (D.S.); (T.L.)
| | - Mark Luedde
- KGP Bremerhaven, Postbrookstraße 105, 27574 Bremerhaven, Germany;
| | - David Schöler
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (C.R.); (M.V.); (D.S.); (T.L.)
| | - Pia Paffenholz
- Department of Urology, University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany;
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany;
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (C.R.); (M.V.); (D.S.); (T.L.)
| | - Ulf P. Neumann
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; (P.H.A.); (A.A.R.); (S.M.S.); (U.P.N.); (T.F.U.)
- Department of Surgery, Maastricht University Medical Centre (MUMC), 5800 Maastricht, The Netherlands
| | - Tom F. Ulmer
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; (P.H.A.); (A.A.R.); (S.M.S.); (U.P.N.); (T.F.U.)
| |
Collapse
|
9
|
Gu GL, Zhang Z, Zhang YH, Yu PF, Dong ZW, Yang HR, Yuan Y. Detection and analysis of common pathogenic germline mutations in Peutz-Jeghers syndrome. World J Gastroenterol 2021; 27:6631-6646. [PMID: 34754157 PMCID: PMC8554407 DOI: 10.3748/wjg.v27.i39.6631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/31/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Different types of pathogenic mutations may produce different clinical phenotypes, but a correlation between Peutz-Jeghers syndrome (PJS) genotype and clinical phenotype has not been found. Not all patients with PJS have detectable mutations of the STK11/LKB1 gene, what is the genetic basis of clinical phenotypic heterogeneity of PJS? Do PJS cases without STK11/LKB1 mutations have other pathogenic genes? Those are clinical problems that perplex doctors. AIM The aim was to investigate the specific gene mutation of PJS, and the correlation between the genotype and clinical phenotype of PJS. METHODS A total of 24 patients with PJS admitted to the Air Force Medical Center, PLA (formerly the Air Force General Hospital, PLA) from November 1994 to January 2020 were randomly selected for inclusion in the study. One hundred thirty-nine common hereditary tumor-related genes including STK11/LKB1 were screened and analyzed for pathogenic germline mutations by high-throughput next-generation sequencing (NGS). The mutation status of the genes and their relationship with clinical phenotypes of PJS were explored. RESULTS Twenty of the 24 PJS patients in this group (83.3%) had STK11/LKB1 gene mutations, 90% of which were pathogenic mutations, and ten had new mutation sites. Pathogenic mutations in exon 7 of STK11/LKB1 gene were significantly lower than in other exons. Truncation mutations are more common in exons 1 and 4 of STK11/LKB1, and their pathogenicity was significantly higher than that of missense mutations. We also found SLX4 gene mutations in PJS patients. CONCLUSION PJS has a relatively complicated genetic background. Changes in the sites responsible for coding functional proteins in exon 1 and exon 4 of STK11/LKB1 may be one of the main causes of PJS. Mutation of the SLX4 gene may be a cause of genetic heterogeneity in PJS.
Collapse
Affiliation(s)
- Guo-Li Gu
- Department of General Surgery, Air Force Medical Center, Chinese People's Liberation Army, Beijing 100142, China
| | - Zhi Zhang
- Department of General Surgery, Air Force Medical Center, Chinese People's Liberation Army, Beijing 100142, China
| | - Yu-Hui Zhang
- Department of General Surgery, Air Force Medical Center, Chinese People's Liberation Army, Beijing 100142, China
- Graduate School, Hebei North University, Zhangjiakou 075000, Hebei Province, China
| | - Peng-Fei Yu
- Department of General Surgery, Air Force Medical Center, Chinese People's Liberation Army, Beijing 100142, China
| | - Zhi-Wei Dong
- Department of General Surgery, Air Force Medical Center, Chinese People's Liberation Army, Beijing 100142, China
| | - Hai-Rui Yang
- Department of General Surgery, Air Force Medical Center, Chinese People's Liberation Army, Beijing 100142, China
| | - Ying Yuan
- Department of Medical Oncology, Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| |
Collapse
|
10
|
LINC01287 facilitates proliferation, migration, invasion and EMT of colon cancer cells via miR-4500/MAP3K13 pathway. BMC Cancer 2021; 21:782. [PMID: 34229645 PMCID: PMC8259379 DOI: 10.1186/s12885-021-08528-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/21/2021] [Indexed: 02/08/2023] Open
Abstract
Background Accumulated studies indicate that aberrant expression of long noncoding RNAs (lncRNAs) is associated with tumorigenesis and progression of colon cancer. In the present study, long intergenic non-protein coding RNA 1287 (LINC01287) was identified to up-regulate in colon cancer by transcriptome RNA-sequencing, but the exact function remained unclear. Methods Transcriptome RNA-sequencing was conducted to identify dysregulated lncRNAs. Expression of LINC01287 was evaluated by real-time quantitative PCR. The downstream targets of LINC01287 and miR-4500 were verified by luciferase reporter assay, pull down assay and western blot. The potential functions of LINC01287 were evaluated by cell viability assay, colony formation assay, soft agar assay, flow cytometry, transwell migration and invasion assay, and tumor xenograft growth in colon cancer cells. Results Our results indicated that LINC01287 was up-regulated in colon cancer patients. High LINC01287 expression was associated with advanced TNM stage, lymph node metastasis, distant metastasis and shorter overall survival. Knockdown of LINC01287 inhibited cell growth, colony formation in plates and soft agar, transwell cell migration and invasion, and epithelial-mesenchymal transition (EMT) of colon cancer cells, while LINC01287 overexpression had contrary effects. In addition, LINC01287 mediated MAP3K13 expression by sponging miR-4500, thus promoted NF-κB p65 phosphorylation. Restored MAP3K13 expression or miR-4500 knockdown partially abrogated the effects of silencing LINC01287 in colon cancer cells. Conclusion Our findings demonstrated that the LINC01287/miR-4500/MAP3K13 axis promoted progression of colon cancer. Therefore, LINC01287 might be a potential therapeutic target and prognostic marker for colon cancer patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08528-7.
Collapse
|
11
|
Huang CC, Liu FR, Feng Q, Pan XY, Song SL, Yang JL. RGD4C peptide mediates anti-p21Ras scFv entry into tumor cells and produces an inhibitory effect on the human colon cancer cell line SW480. BMC Cancer 2021; 21:321. [PMID: 33765976 PMCID: PMC7993510 DOI: 10.1186/s12885-021-08056-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/17/2021] [Indexed: 01/12/2023] Open
Abstract
Background We prepared an anti-p21Ras scFv which could specifically bind with mutant and wild-type p21Ras. However, it cannot penetrate the cell membrane, which prevents it from binding to p21Ras in the cytoplasm. Here, the RGD4C peptide was used to mediate the scFv penetration into tumor cells and produce antitumor effects. Methods RGD4C-EGFP and RGD4C-p21Ras-scFv recombinant expression plasmids were constructed to express fusion proteins in E. coli, then the fusion proteins were purified with HisPur Ni-NTA. RGD4C-EGFP was used as reporter to test the factors affecting RGD4C penetration into tumor cell. The immunoreactivity of RGD4C-p21Ras-scFv toward p21Ras was identified by ELISA and western blotting. The ability of RGD4C-p21Ras-scFv to penetrate SW480 cells and colocalization with Ras protein was detected by immunocytochemistry and immunofluorescence. The antitumor activity of the RGD4C-p21Ras-scFv was assessed with the MTT, TUNEL, colony formation and cell migration assays. Chloroquine (CQ) was used an endosomal escape enhancing agent to enhance endosomal escape of RGD4C-scFv. Results RGD4C-p21Ras-scFv fusion protein were successfully expressed and purified. We found that the RGD4C fusion protein could penetrate into tumor cells, but the tumor cell entry of was time and concentration dependent. Endocytosis inhibitors and a low temperature inhibited RGD4C fusion protein endocytosis into cells. The change of the cell membrane potential did not affect penetrability. RGD4C-p21Ras-scFv could penetrate SW480 cells, effectively inhibit the growth, proliferation and migration of SW480 cells and promote this cells apoptosis. In addition, chloroquine (CQ) could increase endosomal escape and improve antitumor activity of RGD4C-scFv in SW480 cells. Conclusion The RGD4C peptide can mediate anti-p21Ras scFv entry into SW480 cells and produce an inhibitory effect, which indicates that RGD4C-p21Ras-scFv may be a potential therapeutic antibody for the treatment of ras-driven cancers.
Collapse
Affiliation(s)
- Chen-Chen Huang
- School of Medicine, Kunming University of Science and Technology, 727 South Jing Ming Road, Chenggong County, Kunming, 650500, Yunnan Province, China.,Department of Pathology, 920th Hospital of Joint Logistics Support Force of PLA, 212Daguan Rd, Xishan District, Kunming, 650032, Yunnan Province, China
| | - Fang-Rui Liu
- School of Medicine, Kunming University of Science and Technology, 727 South Jing Ming Road, Chenggong County, Kunming, 650500, Yunnan Province, China
| | - Qiang Feng
- Department of Pathology, 920th Hospital of Joint Logistics Support Force of PLA, 212Daguan Rd, Xishan District, Kunming, 650032, Yunnan Province, China
| | - Xin-Yan Pan
- Department of Pathology, 920th Hospital of Joint Logistics Support Force of PLA, 212Daguan Rd, Xishan District, Kunming, 650032, Yunnan Province, China
| | - Shu-Ling Song
- Department of Pathology, 920th Hospital of Joint Logistics Support Force of PLA, 212Daguan Rd, Xishan District, Kunming, 650032, Yunnan Province, China
| | - Ju-Lun Yang
- School of Medicine, Kunming University of Science and Technology, 727 South Jing Ming Road, Chenggong County, Kunming, 650500, Yunnan Province, China. .,Department of Pathology, 920th Hospital of Joint Logistics Support Force of PLA, 212Daguan Rd, Xishan District, Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
12
|
Jing Z, Xi Y, Yin J, Shuwen H. Biological roles of piRNAs in colorectal cancer. Gene 2020; 769:145063. [PMID: 32827685 DOI: 10.1016/j.gene.2020.145063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/17/2020] [Indexed: 12/25/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide and a major cause of cancer-related deaths. Numerous studies have suggested that piwi-interacting RNAs (piRNAs), a new type of non-coding RNA (ncRNA), are closely related to the occurrence and development of cancer. piRNAs have been shown to regulate the occurrence of CRC by modulating multiple molecular signaling pathways. Here, the roles of piRNAs in CRC were reviewed to provide evidence for their potential as molecular targets for CRC.
Collapse
Affiliation(s)
- Zhuang Jing
- Graduate School of Nursing, Huzhou University, Zhejiang, No. 1 Bachelor Road, Huzhou, Zhejiang Province 313000, PR China
| | - Yang Xi
- Department of Oncology, Huzhou Cent Hospital, Affiliated Cent Hospital HuZhou University, 198 Hongqi Rd, Huzhou, Zhejiang 313000, PR China
| | - Jin Yin
- Department of Laboratory Medicine, Huzhou Cent Hospital, Affiliated Cent Hospital HuZhou University, 198 Hongqi Rd, Huzhou, Zhejiang 313000, PR China
| | - Han Shuwen
- Department of Oncology, Huzhou Cent Hospital, Affiliated Cent Hospital HuZhou University, 198 Hongqi Rd, Huzhou, Zhejiang 313000, PR China.
| |
Collapse
|
13
|
Hohl SD, Shankaran V, Bell-Brown A, Issaka RB. Text Message Preferences for Surveillance Colonoscopy Reminders Among Colorectal Cancer Survivors. HEALTH EDUCATION & BEHAVIOR 2020; 47:581-591. [PMID: 32449386 PMCID: PMC7398620 DOI: 10.1177/1090198120925413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background. Surveillance colonoscopy 1-year after colorectal cancer (CRC) surgery effectively reduces CRC mortality, yet less than half of survivors undergo this procedure. Text message reminders can improve CRC screening and other health behaviors, but use of this strategy to address barriers to CRC surveillance has not been reported. Objectives. The goal of this qualitative study was to assess CRC survivor perspectives on barriers to colonoscopy to inform the design of a theory-based, short message service (SMS) intervention to increase surveillance colonoscopy utilization. Method. CRC survivors in Western Washington participated in one of two focus groups to explore perceived barriers to completing surveillance colonoscopy and preferences for SMS communication. Content analysis using codes representative of the health belief model and prospect theory constructs were applied to qualitative data. Results. Thirteen CRC survivors reported individual-, interpersonal-, and system-level barriers to surveillance colonoscopy completion. Participants were receptive to receiving SMS reminders to mitigate these barriers. They suggested that reminders offer supportive, loss-framed messaging; include educational content; and be personalized to communication preferences. Finally, they recommended that reminders begin no earlier than 9 months following CRC surgery and not include response prompts. Conclusions. Our study demonstrates that CRC survivors perceive SMS reminders as an acceptable, valuable tool for CRC surveillance. Furthermore, there may be value in integrating theoretical frameworks to design, implement, and evaluate SMS interventions to address barriers to CRC surveillance. As physicians play a key role in CRC surveillance, provider- and system-level interventions that could additively improve the impact of SMS interventions are also worth exploring.
Collapse
Affiliation(s)
- Sarah D. Hohl
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Veena Shankaran
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| | - Ari Bell-Brown
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rachel B. Issaka
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Zhang Z, Duan FX, Gu GL, Yu PF. Mutation analysis of related genes in hamartoma polyp tissue of Peutz-Jeghers syndrome. World J Gastroenterol 2020; 26:1926-1937. [PMID: 32390703 PMCID: PMC7201153 DOI: 10.3748/wjg.v26.i16.1926] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/29/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Peutz-Jeghers syndrome (PJS) is a rare disease with clinical manifestations of pigmented spots on the lips, mucous membranes and extremities, scattered gastrointestinal polyps, and susceptibility to tumors. The clinical heterogeneity of PJS is obvious, and the relationship between clinical phenotype and genotype is still unclear. AIM To investigate the mutation status of hereditary colorectal tumor-associated genes in hamartoma polyp tissue of PJS patients and discuss its relationship with the clinicopathological data of PJS. METHODS Twenty patients with PJS were randomly selected for this study and were treated in the Air Force Medical Center (former Air Force General Hospital) PLA between 2008 and 2017. Their hamartoma polyp tissues were used for APC, AXIN2, BMPR1A, EPCAM, MLH1, MLH3, MSH2, MSH6, MUTYH, PMS1, PMS2, PTEN, SMAD4, and LKB1/STK11 gene sequencing using next-generation sequencing technology. The correlations between the sequencing results and clinical pathological data of PJS were analyzed. RESULTS Fourteen types of LKB1/STK11 mutations were detected in 16 cases (80.0%), of which 8 new mutations were found (3 types of frameshift deletion mutations: c.243delG, c.363_364delGA, and c.722delC; 2 types of frameshift insertions: c. 144_145insGCAAG, and c.454_455insC; 3 types of splice site mutations: c.464+1G>T, c.464+1G>A, and c.598-1G>A); 9 cases (45.0%) were found to have 18 types of heterozygous mutations in the remaining 13 genes except LKB1/STK11. Of these, MSH2: c.792+1G>A, MSH6: c.3689C>G, c.4001+13C>CTTAC, PMS1: c.46C>t, and c.922G>A were new mutations. CONCLUSION The genetic mutations in hamartoma polyp tissue of PJS are complex and diverse. Moreover, other gene mutations in PJS hamartoma polyp tissue were observed, with the exception of LKB1/STK11 gene, especially the DNA mismatch repair gene (MMR). Colorectal hamartoma polyps with LKB1/STK11 mutations were larger in diameter than those with other gene mutations.
Collapse
Affiliation(s)
- Zhi Zhang
- Air Force Clinical College (Air Force Medical Center) of Anhui Medical University, Beijing 100142, China
| | - Fu-Xiao Duan
- Department of General Surgery, the General Hospital of Northern Theater Command PLA, Shenyang 110016, Liaoning Province, China
| | - Guo-Li Gu
- Department of General Surgery, Air Force Medical Center, PLA, Beijing 100142, China
| | - Peng-Fei Yu
- Department of General Surgery, Air Force Medical Center, PLA, Beijing 100142, China
| |
Collapse
|
15
|
Wang Z, Zang A, Wei Y, An L, Hong D, Shi Y, Zhang J, Su S, Fang G. Hyaluronic Acid Capped, Irinotecan and Gene Co-Loaded Lipid-Polymer Hybrid Nanocarrier-Based Combination Therapy Platform for Colorectal Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1095-1105. [PMID: 32210538 PMCID: PMC7076892 DOI: 10.2147/dddt.s230306] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 02/05/2020] [Indexed: 12/15/2022]
Abstract
Background The current approach for treating colorectal cancer favors the use of drug and gene combination therapy, and targeted nano-systems are gaining considerable attention for minimizing toxicity and improving the efficacy of anticancer treatment. The aim of this study was to develop ligand-modified, irinotecan and gene co-loaded lipid-polymer hybrid nanocarriers for targeted colorectal cancer combination therapy. Methods Hyaluronic acid modified, irinotecan and gene co-loaded LPNs (HA-I/D-LPNs) were prepared using a solvent-evaporation method. Their average size, zeta potential, drug and gene loading capacity were characterized. The in vitro and in vivo gene transfection and anti-tumor ability of this nano-system were evaluated on colorectal cancer cells and mice bearing colorectal cancer model. Results HA-I/D-LPNs had a size of 182.3 ± 5.1, over 80% drug encapsulation efficiency and over 90% of gene loading capacity. The peak plasma concentration (Cmax) and half-life (T1/2) achieved from HA-I/D-LPNs were 41.31 ± 1.58 μg/mL and 12.56 ± 0.67 h. HA-I/D-LPNs achieved the highest tumor growth inhibition efficacy and the most prominent transfection efficiency in vivo. Conclusion HA-I/D-LPNs exhibited the most remarkable tumor inhibition efficacy and best gene transfection efficiency in the tumor, which could prove the effects of the drug and gene combination therapy.
Collapse
Affiliation(s)
- Zhiyu Wang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Aimin Zang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Yaning Wei
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Lin An
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Dan Hong
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Yan Shi
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Jingnan Zhang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Shenyong Su
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| | - Guotao Fang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, People's Republic of China
| |
Collapse
|
16
|
Challenges with colorectal cancer staging: results of an international study. Mod Pathol 2020; 33:153-163. [PMID: 31383959 DOI: 10.1038/s41379-019-0344-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/18/2022]
Abstract
Challenges exist with standardized colorectal cancer reporting despite adoption of the American Joint Committee on Cancer-Staging Manual 8th edition. We performed this study to gauge current practice patterns among a diverse group of surgical pathologists. A web-based questionnaire depicting problematic issues and images related to colorectal carcinoma staging was circulated among 118 surgical pathologists and their responses were correlated with their geographic location (North America vs. Europe vs. others), nature of practice (academic vs. community), the sign-out model (gastrointestinal subspecialty vs. general surgical pathology), and years of professional experience. We found that a substantial number of practicing pathologists ignore recommended-staging criteria in specific settings, particularly with respect to assessment of advanced T stage. Tumors that communicated with the serosa through inflammatory foci were staged as pT3 (49%) or pT4a (51%) by nearly equal numbers of pathologists regardless of level of experience, the sign-out model, or geographic location. Only 65% assigned T stage and margin status based on extent of viable tumor in the neoadjuvant setting. One-third of pathologists, particularly those in Europe (p = 0.015), classified acellular mucin deposits as N1 disease when detected in treatment-naive cases. Nearly 50% of pathologists classified isolated tumor cells (i.e., deposits <0.2 mm) in lymph nodes as metastatic disease (i.e., pN1, p = 0.02). Our results suggest that pathologists ignore recommendations that are based on insufficient data and apply individualized criteria when faced with situations that are not addressed in the American Joint Committee on Cancer Staging Manual 8th edition. These variations in practice limit the ability to compare outcome data across different institutions and draw attention to areas that require further study.
Collapse
|
17
|
Wu WM, Wang Y, Jiang HR, Yang C, Li XQ, Yan B, Zhou Y, Xu WH, Lin T. Colorectal Cancer Screening Modalities in Chinese Population: Practice and Lessons in Pudong New Area of Shanghai, China. Front Oncol 2019; 9:399. [PMID: 31214488 PMCID: PMC6558000 DOI: 10.3389/fonc.2019.00399] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/29/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Parallel test of risk stratification and two-sample qualitative fecal immunochemical tests (FITs) are used to screen colorectal cancer (CRC) in Shanghai, China. This study was designed to identify an optimal initial screening modality based on available data. Methods: A total of 538,278 eligible residents participated in the program during the period of January 2013 to June 2017. Incident CRC was collected through program reporting system and by record linkage with the Shanghai Cancer Registry up to December 2017. Logistic regression model was applied to identify significant factors to calculate risk score for CRC. Cutoff points of risk score were determined based on Youden index and defined specificity. Sensitivity, specificity, and positive predictive values (PPVs) were computed to evaluate validity of assumed screening modalities. Results: A total of 446 CRC were screen-detected, and 777 interval or missed cases were identified through record linkage. The risk score system had an optimal cutoff point of 19 and performed better in detecting CRC and predicting long-term CRC risk than did the risk stratification. When using a cutoff point of 24, parallel test of risk score, and FIT were expected to avoid 56 interval CRCs with minimal decrease in PPV and increase in colonoscopy. However, the observed detection rates were much lower than those expected due to low compliance to colonoscopy. Conclusions: Risk score is superior to risk stratification used in the program, particularly when combined with FIT. Compliance to colonoscopy should be improved to guarantee the effectiveness of CRC screening in the population.
Collapse
Affiliation(s)
- Wei-miao Wu
- Fudan University School of Public Health, Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), Shanghai, China
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
| | - Yingying Wang
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
- Center of Disease Prevention and Control in Pudong New Area of Shanghai, Shanghai, China
| | - Hui-ru Jiang
- Fudan University School of Public Health, Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), Shanghai, China
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
| | - Chen Yang
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
- Center of Disease Prevention and Control in Pudong New Area of Shanghai, Shanghai, China
| | - Xiao-qiang Li
- Fudan University School of Public Health, Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), Shanghai, China
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
| | - Bei Yan
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
- Center of Disease Prevention and Control in Pudong New Area of Shanghai, Shanghai, China
| | - Yi Zhou
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
- Center of Disease Prevention and Control in Pudong New Area of Shanghai, Shanghai, China
| | - Wang-hong Xu
- Fudan University School of Public Health, Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China (Fudan University), Shanghai, China
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
| | - Tao Lin
- Fudan University Pudong Institute of Preventive Medicine, Pudong New Area, Shanghai, China
- Center of Disease Prevention and Control in Pudong New Area of Shanghai, Shanghai, China
| |
Collapse
|
18
|
Abstract
Tumor stage, as determined by the Tumor, Node, Metastasis (TNM) staging system, is the single most influential factor determining treatment decisions and outcome among patients with colorectal cancer. Several stage-related elements in pathology reports consistently pose diagnostic challenges: recognition of serosal penetration by tumor (ie, pT3 vs pT4a), evaluation of regional lymph nodes, distinction between tumor deposits and effaced lymph nodes, and assessment of tumor stage in the neoadjuvant setting. This article discusses each of these issues in detail and provides practical tips regarding colorectal cancer staging.
Collapse
Affiliation(s)
- Rhonda K Yantiss
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 525 East 68th Street, New York, NY 10065, USA.
| |
Collapse
|
19
|
Shuwen H, Qing Z, Yan Z, Xi Y. Competitive endogenous RNA in colorectal cancer: A systematic review. Gene 2017; 645:157-162. [PMID: 29273554 DOI: 10.1016/j.gene.2017.12.036] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/30/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Abstract
Colorectal cancer is one of the most common malignant tumours. Competitive endogenous RNA (ceRNA) networks have been hypothesized, in which various RNAs regulate each other's expression using microRNA response elements (MREs). Recent evidence has highlighted the crucial regulatory roles of ceRNA networks in colorectal cancer. In this review, we summarize the present research methods as well as the currently known ceRNA competitors and targets in colorectal cancer. In addition, we discuss the significance of ceRNA and shortcomings of current studies of colorectal cancer.
Collapse
Affiliation(s)
- Han Shuwen
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, Zhejiang Province, China
| | - Zhou Qing
- Department of Critical Care Medicine, Huzhou Central Hospital, Huzhou, Zhejiang Province, China
| | - Zheng Yan
- Department of Pathology, Huzhou Central Hospital, Huzhou, Zhejiang Province, China
| | - Yang Xi
- Department of Intervention and Radiotherapy, Huzhou Central Hospital, Huzhou, Zhejiang Province, China.
| |
Collapse
|
20
|
Ji B, Cheng X, Cai X, Kong C, Yang Q, Fu T, Wang Y, Song Y. CK20 mRNA Expression in Serum as a Biomarker for Colorectal Cancer Diagnosis: A Meta-analysis. Open Med (Wars) 2017; 12:347-353. [PMID: 29043299 PMCID: PMC5639391 DOI: 10.1515/med-2017-0050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
The aim of this study was to evaluate the diagnostic value of serumCK20 mRNA as a biomarker for colorectal cancer diagnosis by meta-analysis. Clinical studies related to serum CK20 mRNA expression for colorectal cancer diagnosis were searched in the databases of Pubmed, Cochrane Library, Embase, ISI Web of Knowledge, CNKI and Wanfang. The number of true positive (tp), false positive (fp), false negative (fn) and true negative (tn) of the original included publications were extracted by two reviewers independently. The diagnostic sensitivity, specificity, positive likely hood ratio (+LR), negative likelyhood ratio (-LR), diagnostic odds ratio (DOR) and area under the symmetric ROC curve (AUC) were pooled by random or fixed effect method according to the statistical heterogeneity among the studies. After screening the databases, nineteen publications met the inclusion criteria and were finally included in this meta-analysis. The diagnostic sensitivity and specificity were pooled by random effect model(I2>50%). The pooled diagnostic sensitivity and specificity of CK20 mRNA in serum as biomarker for colorectal cancer were 0.49 (95% CI:0.46 to 0.51) and 0.94 (95%CI:0.92-0.96) respectively. The pooled +LR and -LR were 10.90 (95%CI:5.78 to 20.55) and 0.51 (95%CI:0.45 to 0.57) respectively by random-effect method. The pooled DOR was 22.31 with the 95% CI of 11.65 to 42.71. The pooled area under the ROC curve (AUC) was 0.72for CK20 mRNA in serum as a biomarker for colorectal cancer diagnosis. Conclusion Serum CK20 mRNA expression was significantly elevated in colorectal cancer patients which could be a promising serum biomarker for colorectal cancer diagnosis with high specificity.
Collapse
Affiliation(s)
- Baoyan Ji
- Department of Oncology, Qinghai Province People's Hospital, Xining Qinghai81000, China
| | - Xiongfei Cheng
- Department of Oncology Center, The People's Hospital of Shiyan City HubeiProvince442000China
| | - Xiaojun Cai
- Department of Oncology Center, The People's Hospital of Shiyan City HubeiProvince442000China
| | - Chuiyan Kong
- Department of Oncology, The People's Hospital of Wuzhou GuangxiProvince543000, China
| | - Qingyan Yang
- Department of Oncology, The 188th Hospital of PLA, Chaozhou GuangdongProvince521000, China
| | - Ting Fu
- Department of Oncology Center, The People's Hospital of Shiyan City HubeiProvince442000China
| | - Yahang Wang
- Department of General surgery, People's Hospital Fenghua District Ningbo City ZhejiangProvince, 315500, PR China
| | - Ying Song
- Department of Oncology Center, The People's Hospital of Shiyan City HubeiProvince442000China
| |
Collapse
|
21
|
Zeng JH, Liang L, He RQ, Tang RX, Cai XY, Chen JQ, Luo DZ, Chen G. Comprehensive investigation of a novel differentially expressed lncRNA expression profile signature to assess the survival of patients with colorectal adenocarcinoma. Oncotarget 2017; 8:16811-16828. [PMID: 28187432 PMCID: PMC5370003 DOI: 10.18632/oncotarget.15161] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023] Open
Abstract
Growing evidence has shown that long non-coding RNAs (lncRNAs) can serve as prospective markers for survival in patients with colorectal adenocarcinoma. However, most studies have explored a limited number of lncRNAs in a small number of cases. The objective of this study is to identify a panel of lncRNA signature that could evaluate the prognosis in colorectal adenocarcinoma based on the data from The Cancer Genome Atlas (TCGA). Altogether, 371 colon adenocarcinoma (COAD) patients with complete clinical data were included in our study as the test cohort. A total of 578 differentially expressed lncRNAs (DELs) were observed, among which 20 lncRNAs closely related to overall survival (OS) in COAD patients were identified using a Cox proportional regression model. A risk score formula was developed to assess the prognostic value of the lncRNA signature in COAD with four lncRNAs (LINC01555, RP11-610P16.1, RP11-108K3.1 and LINC01207), which were identified to possess the most remarkable correlation with OS in COAD patients. COAD patients with a high-risk score had poorer OS than those with a low-risk score. The multivariate Cox regression analyses confirmed that the four-lncRNA signature could function as an independent prognostic indicator for COAD patients, which was largely mirrored in the validating cohort with rectal adenocarcinoma (READ) containing 158 cases. In addition, the correlative genes of LINC01555 and LINC01207 were enriched in the cAMP signaling and mucin type O-Glycan biosynthesis pathways. With further validation in the future, our study indicates that the four-lncRNA signature could serve as an independent biomarker for survival of colorectal adenocarcinoma.
Collapse
Affiliation(s)
- Jiang-Hui Zeng
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Liang Liang
- Department of General Surgery, First Affiliated Hospital of Guangxi Medical University (West Branch), Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rui-Xue Tang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Xiao-Yong Cai
- Department of General Surgery, First Affiliated Hospital of Guangxi Medical University (West Branch), Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Jun-Qiang Chen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Dian-Zhong Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| |
Collapse
|