Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Int J Colorectal Dis ; 39(1): 46, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38565736

ABSTRACT

PURPOSE: Lymph node metastasis (LNM) is a crucial factor that determines the prognosis of T1 colorectal cancer (CRC) patients. We aimed to develop a practical prediction model for LNM in T1 CRC. METHODS: We conducted a retrospective analysis of data from 825 patients with T1 CRC who underwent radical resection at a single center in China. All enrolled patients were randomly divided into a training set and a validation set at a ratio of 7:3 using R software. Risk factors for LNM were identified through multivariate logistic regression analyses. Subsequently, a prediction model was developed using the selected variables. RESULTS: The lymph node metastasis (LNM) rate was 10.1% in the training cohort and 9.3% in the validation cohort. In the training set, risk factors for LNM in T1 CRC were identified, including depressed endoscopic gross appearance, sex, submucosal invasion combined with tumor grade (DSI-TG), lymphovascular invasion (LVI), and tumor budding. LVI emerged as the most potent predictor for LNM. The prediction model based on these factors exhibited good discrimination ability in the validation sets (AUC: 79.3%). Compared to current guidelines, the model could potentially reduce over-surgery by 48.9%. Interestingly, we observed that sex had a differential impact on LNM between early-onset and late-onset CRC patients. CONCLUSIONS: We developed a clinical prediction model for LNM in T1 CRC using five factors that are easily accessible in clinical practice. The model has better predictive performance and practicality than the current guidelines and can assist clinicians in making treatment decisions for T1 CRC patients.


Subject(s)
Colorectal Neoplasms , Models, Statistical , Humans , Colorectal Neoplasms/surgery , Colorectal Neoplasms/pathology , Lymph Nodes/surgery , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Prognosis , Retrospective Studies , Risk Factors , Random Allocation , China
2.
Surg Endosc ; 38(7): 3783-3798, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38806955

ABSTRACT

BACKGROUND: Many studies reported the presence of adenomas with high-grade dysplasia (HGD) at index colonoscopy increased the incidence of advanced neoplasia (AN) and colorectal cancer (CRC) following. However, the conclusion remains obscure due to lack of studies on the specific population of adenomas with HGD. This study aimed to assess the long-term risk of AN and CRC after removal of adenomas with HGD. METHODS: A total of 814 patients who underwent adenomas with HGD removal between 2010 and 2019 were retrospectively analyzed. The outcomes were the incidences of AN and CRC during surveillance colonoscopy. Cox proportional hazards models were utilized to identify risk factors associated with AN and CRC. RESULTS: During more than 2000 person-years of follow-up, we found that AN and CRC incidence densities were 44.3 and 4.4 per 1000 person-years, respectively. The 10-year cumulative incidence of AN and CRC were 39.1% and 5.5%, respectively. In the multivariate model, synchronous low-risk polyps (HR 1.80, 95% CI 1.10-2.93) and synchronous high-risk polyps (HR 3.99, 95% CI 2.37-6.72) were risk factors for AN, whereas participation in surveillance colonoscopy visits (HR 0.56, 95% CI 0.36-0.88 for 1 visit; HR 0.10, 95% CI 0.06-0.19 for ≥ 2 visits) were associated with decreased AN incidence. Additionally, elevated baseline carcinoembryonic antigen (CEA) level (HR 10.19, 95% CI 1.77-58.59) was a risk factor for CRC, while participation in ≥ 2 surveillance colonoscopy visits (HR 0.11, 95% CI 0.02-0.56) were associated with decreased CRC incidence. Interestingly, for 11 patients who developed CRC after removal of adenomas with HGD, immunohistochemistry revealed that 8 cases (73%) were deficient mismatch repair CRCs. CONCLUSIONS: Patients who have undergone adenoma with HGD removal are at higher risk of developing AN and CRC, while surveillance colonoscopy can reduce the risk. Patients with synchronous polyps, or with elevated baseline CEA level are considered high-risk populations and require more frequent surveillance.


Subject(s)
Adenoma , Colonoscopy , Colorectal Neoplasms , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/surgery , Male , Female , Middle Aged , Retrospective Studies , Adenoma/pathology , Adenoma/surgery , Adenoma/epidemiology , Incidence , Risk Factors , Aged , Colonic Polyps/surgery , Colonic Polyps/pathology , Adult
3.
J Minim Access Surg ; 19(1): 130-137, 2023.
Article in English | MEDLINE | ID: mdl-36722537

ABSTRACT

Background: Inadequate bowel preparation leads to lower polyp detection rates, longer procedure times and lower cecal intubation rates. However, there is no consensus about high-quality bowel preparation, so our study evaluated graphical education and appropriate time before elective colonoscopy. Patients and Methods: We performed a secondary analysis of a national colorectal cancer screening programme of 738 patients. The patients were divided into a group given a graphical information manual (n = 242) or a word-only one (n = 496). They were also divided into groups according to the interval between bowel preparation and colonoscopy: 6-8 h (Group 1, n = 106), 9-12 h (Group 2, n = 228) and 13-17 h (Group 3, n = 402). All patients were scored according to the Boston Bowel Preparation Scale (BBPS) during the examination. Results: The bowel preparation of the graphical group was significantly better than the text group (P < 0.001). After adjustment, the bowel preparation score of Group 1 and Group 2 were both significantly higher than that of Group 3 (P = 0.012 and P = 0.032). Maximum BBPS was 6.31 when the interval time was 6.52 h (95% confidence interval: 5.95-6.66), and when the interval was <10 h, the BBPS was ≥6. Conclusion: High-quality bowel preparation was linked to graphical education and appropriate time before colonoscopy. We suggest that the interval between taking the first laxative and colonoscopy should be <10 h, preferably 6.5 h. Prospective multicentre research is needed to give more evidence of high-quality bowel preparation methods.

4.
BMC Cancer ; 22(1): 806, 2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35864459

ABSTRACT

BACKGROUND: For colorectal cancer, preoperative (neoadjuvant) chemotherapy is more effective than postoperative chemotherapy because it not only eradicates micrometastases more effectively but also reduces the risk of incomplete intraoperative resection and tumor cell shedding. For the treatment of acute left-sided malignant colorectal obstruction, colorectal stents as well as stoma are being used to relieve the obstructive colorectal cancer, and as a bridge to surgery, allowing easy mobilization and resection of the colon. Neoadjuvant chemotherapy combined with self-expandable metal stents (SEMS) or neoadjuvant chemotherapy combined with decompressing stoma (DS) can be used as a bridge to elective surgery (BTS) as an alternative to emergency surgery in patients with acute left-sided malignant colorectal obstruction, but its benefit is uncertain. The purpose of this study was to evaluate the safety and feasibility of neoadjuvant chemotherapy as a bridge to surgery in the treatment of acute left-sided malignant colorectal obstruction. METHODS: Data from patients who were admitted with acute left-sided malignant colorectal obstruction between January 2012 and December 2020 were retrospectively reviewed, and patients with gastrointestinal perforation or peritonitis were excluded. We performed one-to-two propensity score matching to compare the stoma requirement, postoperative complications, and other short-term oncological outcomes between the neoadjuvant chemotherapy group and surgery group. RESULTS: There were no differences in intraoperative blood loss, operative time, one-year postoperative mortality, and postoperative tumor markers between the two groups. The 1-year recurrence-free survival (RFS) rates of neoadjuvant chemotherapy group and surgery group were 96.8 and 91.3% (p = 0.562). The neoadjuvant chemotherapy group was able to reduce stoma rate 1 year after surgery (p = 0.047). Besides, the neoadjuvant group significantly reduced postoperative bowel function time (p < 0.001), postoperative hospital stay (p < 0.001), total hospital stay (p = 0.002), postoperative complications (p = 0.017), reduction in need to stay in the intensive care unit (ICU) (p = 0.042). CONCLUSIONS: Neoadjuvant chemotherapy as a bridge to elective surgery in patients with acute left-sided malignant colorectal obstruction is safe and has many advantages. Prospective multicenter studies with large samples are needed to further evaluate the feasibility of neoadjuvant chemotherapy.


Subject(s)
Colorectal Neoplasms , Intestinal Obstruction , Colorectal Neoplasms/complications , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/surgery , Feasibility Studies , Humans , Intestinal Obstruction/etiology , Intestinal Obstruction/pathology , Intestinal Obstruction/surgery , Neoadjuvant Therapy/adverse effects , Postoperative Complications , Prospective Studies , Retrospective Studies , Treatment Outcome
5.
Dis Colon Rectum ; 65(4): 590-598, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-34775404

ABSTRACT

BACKGROUND: Postoperative benign anastomotic stricture is associated with colorectal anastomosis following surgery for colorectal cancer. Endoscopic stricturotomy is a novel technique that has been demonstrated to be safe and effective for the treatment of colorectal anastomotic stricture in several case reports and series. OBJECTIVE: We designed this study to investigate the efficacy of endoscopic stricturotomy for postoperative benign anastomotic stricture in patients for colorectal cancer. The primary outcomes were stricture-recurrence-free survival and reoperation-free survival. DESIGN: This is a retrospective study. SETTING: This study presents a single-center experience. PATIENTS: This retrospective study included patients with colorectal cancer who underwent surgical resection and developed anastomotic stricture between January 2014 and June 2019 and were treated with endoscopic stricturotomy. MAIN OUTCOME MEASURES: Immediate technical success of endoscopic stricturotomy and the factors associated with success and recurrence were investigated. RESULTS: Endoscopic stricturotomy was performed in 57 patients, and immediate technical success was achieved in 84% of the patients. The mean follow-up was 31.3 (15.8) months (range, 9-74 months). Postoperative benign anastomotic stricture recurred in 11 patients after initial successful endoscopic stricturotomy; 10 of the 11 recurrent patients accepted reoperation. Univariate and multivariate analysis indicated that length of stricture ≥1 cm was an independent risk factor for failure of the initial endoscopic stricturotomy (OR, 9.423; 95% CI, 1.729-51.350; p = 0.010) and the recurrence of postoperative benign anastomotic stricture after the initial endoscopic stricturotomy (OR, 13.521; 95% CI, 2.305-79.306; p = 0.004). LIMITATIONS: The study was limited by its small sample size and retrospective design. CONCLUSIONS: Endoscopic stricturotomy is a safe and effective technique for postoperative benign anastomotic stricture. However, if the length of the stricture is ≥1 cm, endoscopic stricturotomy may not be effective, and recurrence of postoperative benign anastomotic stricture is also likely. See Video Abstract at http://links.lww.com/DCR/B739. ESTRICTUROTOMA ENDOSCPICA PARA PACIENTES CON ESTRICCIN ANASTOMTICA BENIGNA POSTOPERATORIA PARA EL CNCER COLORRECTAL: ANTECEDENTES:La estenosis anastomótica benigna postoperatoria se asocia con anastomosis colorrectal después de la cirugía para el cáncer colorrectal. La estricturotomia endoscópica es una técnica novedosa que se ha demostrado que es segura y efectiva para el tratamiento de la estenosis anastomótica colorrectal en varios informes de casos o series.OBJETIVO:Diseñamos este estudio para investigar la eficacia de la estricturotomia endoscópica para la estenosis anastomótica benigna postoperatoria en pacientes con cáncer colorrectal. El resultado primario fue la supervivencia libre de restricción estricta y la supervivencia libre de reoperación.DISEÑO:Este es un estudio retrospectivo.CONFIGURACIÓN:Este estudio presenta una experiencia de un solo centro.PACIENTES:Este estudio retrospectivo incluyó pacientes con cáncer colorrectal que se sometieron a resección quirúrgica y desarrollaron estenosis anastomótica entre enero de 2014 y junio de 2019 y tratados con estricturotomia endoscópica.MEDIDAS PRINCIPALES DE RESULTADO:Éxito técnico inmediato y estenosurotomía endoscópica, los factores asociados con el éxito y la recurrencia.RESULTADOS:Se realizó estricturotomia endoscópica en 57 pacientes, y se logró un éxito técnico inmediato en el 84% de los pacientes. El seguimiento medio fue de 31,3 (15,8) meses (rango, 9 a 74 meses), el POBAS se repitió en 11 pacientes después del éxito inicial de ESt. 10 de los 11 pacientes recurrentes aceptaron la reoperación. El análisis univariado y multivariado indicó que la longitud de la estenosis ≥1 cm era un factor de riesgo independiente para el fracaso de la estricturotomia endoscópica inicial (odds ratio = 9,423; IC del 95% = 1.729-51.350; p = 0.010) y la recurrencia de estenosis anastomótica benigna postoperatoria después de la estricturotomia endoscópica inicial (odds ratio = 13,521; IC del 95% = 2,305-79,306; p = 0.004).LIMITACIONES:El estudio estuvo limitado por su pequeño tamaño de muestra y diseño retrospectivo.CONCLUSIONES:La estricturotomia endoscópica es una técnica segura y efectiva para la estructura anastomótica benigna postoperatoria. Sin embargo, si la longitud de la estenosis es ≥1 cm, la estricturotomia endoscópica puede no ser efectiva y también es probable que se repita la estenosis anastomótica benigna postoperatoria. Consulte Video Resumen en http://links.lww.com/DCR/B739.


Subject(s)
Anastomotic Leak , Rectal Neoplasms , Anastomosis, Surgical/adverse effects , Anastomotic Leak/etiology , Constriction, Pathologic/etiology , Constriction, Pathologic/surgery , Humans , Postoperative Complications/surgery , Rectal Neoplasms/surgery , Retrospective Studies
6.
J Surg Oncol ; 126(4): 757-771, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35661159

ABSTRACT

BACKGROUND: Patients with nonmetastatic pT3-4 colon cancers are prone to develop metachronous peritoneal carcinomatosis (mPC). Risk factors for mPC and the influence of mutant kirsten rat sarcoma viral oncogene (KRAS)/neuroblastoma rat sarcoma (NRAS)/v-raf murine sarcoma viral oncogene homolog B1 (BRAF) and DNA mismatch repair (MMR) status on mPC remain to be described in these patients. METHOD: All enrolled patients were identified from the prospectively collected colorectal cancer database of a tertiary referral hospital between 2013 and 2018. Multivariate analysis was used to identify risk factors associated with mPC. RESULTS: Of the 1689 patients with nonmetastatic pT3-4 colon carcinoma, 8.4% (142/1689) progressed to mPC. Endoscopic obstruction (HR = 3.044, p < 0.001), elevated CA125 (HR = 1.795, p = 0.009), pT (T4a vs. T3, HR = 2.745, p < 0.001; T4b vs. T3, HR = 3.167, p = 0.001), pN (N1 vs. N0, HR = 2.592, p < 0.001; N2 vs. N0, HR = 4.049, p < 0.001), less than 12 lymph nodes harvested (HR = 2.588, p < 0.001), mucinous or signet ring cell carcinoma (HR = 1.648, p = 0.038), perineural invasion (HR = 1.984, p < 0.001), and adjuvant chemotherapy (HR = 1.522, p = 0.039) were strongly related to mPC but that mutant KRAS/NRAS/BRAF and MMR status was not associated with mPC. CONCLUSION: This study identified the high-risk factors for mPC in patients with nonmetastatic pT3-4 colon carcinoma, and these factors should be considered in selective preventive therapy and close follow-up for patients subsequently deemed to have high risk for mPC.


Subject(s)
Carcinoma, Signet Ring Cell , Colonic Neoplasms , Colorectal Neoplasms , Peritoneal Neoplasms , Animals , Carcinoma, Signet Ring Cell/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/surgery , Colorectal Neoplasms/pathology , Mice , Mutation , Neoplasm Staging , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Risk Factors
7.
Int J Colorectal Dis ; 37(7): 1621-1634, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35704090

ABSTRACT

PURPOSE: Watch and wait strategy is a safe and effective alternative to surgery in patients with locally advanced rectal cancer (LARC) who have achieved pathological complete response (pCR) after neoadjuvant therapy (NAT); present restaging methods do not meet clinical needs. This study aimed to construct a machine learning (ML) model to predict pCR preoperatively. METHODS: LARC patients who received NAT were included to generate an extreme gradient boosting-based ML model to predict pCR. The group was divided into a training set and a tuning set at a 7:3 ratio. The SHapley Additive exPlanations value was used to quantify feature importance. The ML model was compared with a nomogram model developed using independent risk factors identified by conventional multivariate logistic regression analysis. RESULTS: Compared with the nomogram model, our ML model improved the area under the receiver operating characteristics from 0.72 to 0.95, sensitivity from 43 to 82.2%, and specificity from 87.1 to 91.6% in the training set, the same trend applied to the tuning set. Neoadjuvant radiotherapy, preoperative carbohydrate antigen 125 (CA125), CA199, carcinoembryonic antigen level, and depth of tumor invasion were significant in predicting pCR in both models. CONCLUSION: Our ML model is a potential alternative to the existing assessment tools to conduct triage treatment for patients and provides reference for clinicians in tailoring individual treatment: the watch and wait strategy is used to avoid surgical trauma in pCR patients, and non-pCR patients receive surgical treatment to avoid missing the optimal operation time window.


Subject(s)
Neoadjuvant Therapy , Rectal Neoplasms , Chemoradiotherapy/methods , Humans , Machine Learning , Neoadjuvant Therapy/methods , Rectal Neoplasms/pathology , Retrospective Studies , Treatment Outcome
8.
Int J Clin Oncol ; 27(4): 749-755, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35079898

ABSTRACT

BACKGROUND: The incidence of early-onset colorectal cancer (EOCRC) is rapidly increasing worldwide in decade when screening of colorectal cancer (CRC) is more prevalent. The clinicopathological and molecular characteristics of EOCRC have not yet been clarified. This study aims to evaluate clinicopathological and molecular features among EOCRC and late-onset colorectal cancer (LOCRC) patients according to different tumor locations. METHODS: We identified CRC patients from a prospectively maintained CRC database between January 2015 and December 2018. The clinicopathological and molecular characteristics including dMMR, mutation of PIK3CA, BRAF and KRAS were compared between EOCRC and LOCRC. The relationships according to different tumor locations were assessed. RESULTS: Totally 4468 patients were analyzed in this study. Compared to LOCRC patients, EOCRC patients were more likely to have status of dMMR (OR, 2.52; P < 0.001), regardless of tumor location. EOCRC patients were more likely to be detected with mutation of PIK3CA (OR, 1.24; P = 0.041), which only tended to exist in the left-side colon (OR, 1.51; P = 0.06), but not in the right-side colon or rectum. No significant difference was found for BRAF or KRAS mutation, but mutation of KRAS was more frequently found in left-side colon (OR, 1.34; P = 0.04) among EOCRC patients. CONCLUSION: Status of dMMR, mutation of PIK3CA, BRAF and KRAS was different between EOCRC and LOCRC patients according to different tumor locations, which implied that EOCRC might be a unique subgroup of CRC patients. Further investigations of molecular and genetic differences should be performed to help define new diagnosing and therapeutical strategies for EOCRC patients.


Subject(s)
Colorectal Neoplasms , Colorectal Neoplasms/pathology , Humans , Incidence , Mutation , Proto-Oncogene Proteins B-raf/genetics , Rectum/pathology
9.
J Clin Lab Anal ; 36(10): e24678, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36036768

ABSTRACT

BACKGROUND: The inhibitor of ß-catenin and T-cell factor (ICAT) is a direct negative regulator of the canonical Wnt signaling pathway, which is an attractive therapeutic target for colorectal cancer (CRC). Accumulating evidence suggests that ICAT interacts with other proteins to exert additional functions, which are not yet fully elucidated. METHODS: The overexpression of ICAT of CRC cells was conducted by lentivirus infection and plasmids transfection and verified by quantitative real-time reverse transcription-polymerase chain reaction (real-time RT-PCR) and Western blotting. The effect of ICAT on the mobility of CRC cells was assessed by wound healing assay and transwell assay in vitro and lung metastasis in vivo. New candidate ICAT-interacting proteins were explored and verified using the STRING database, silver staining, co-immunoprecipitation mass spectrometry analysis (Co-IP/MS), and immunofluorescence (IF) staining analysis. RESULT: Inhibitor of ß-catenin and T-cell factor overexpression promoted in vitro cell migration and invasion and tumor metastasis in vivo. Co-IP/MS analysis and STRING database analyses revealed that junction plakoglobin (JUP), a homolog of ß-catenin, was involved in a novel protein interaction with ICAT. Furthermore, JUP downregulation impaired ICAT-induced migration and invasion of CRC cells. In addition, ICAT overexpression activated the NF-κB signaling pathway, which led to enhanced CRC cell migration and invasion. CONCLUSION: Inhibitor of ß-catenin and T-cell factor promoted CRC cell migration and invasion by interacting with JUP and the NF-κB signaling pathway. Thus, ICAT could be considered a protein diagnostic biomarker for predicting the metastatic ability of CRC.


Subject(s)
Colorectal Neoplasms , beta Catenin , Adaptor Proteins, Signal Transducing , Biomarkers , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/metabolism , Neoplasm Metastasis , TCF Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism , gamma Catenin/metabolism
10.
Proc Natl Acad Sci U S A ; 116(13): 6286-6291, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30862736

ABSTRACT

Disruption of mucosal immunity plays a critical role in the pathogenesis of inflammatory bowel disease, yet its mechanism remains not fully elucidated. Here, we found that activating transcription factor 3 (ATF3) protects against colitis by regulating follicular helper T (TFH) cells in the gut. The expression of ATF3 in CD4+ T cells was negatively correlated with the severity of ulcerative colitis in clinical patients. Mice with ATF3 deficiency in CD4+ T cells (CD4creAtf3fl/fl ) were much more susceptible to dextran sulfate sodium-induced colitis. The frequencies of TFH cells, not other T cell subsets, were dramatically decreased in Peyer's patches from CD4creAtf3fl/fl mice compared with Atf3fl/fl littermate controls. The defective TFH cells significantly diminished germinal center formation and IgA production in the gut. Importantly, adoptive transfer of TFH or IgA+ B cells caused significant remission of colitis in CD4creAtf3fl/fl mice, indicating the TFH-IgA axis mediated the effect of ATF3 on gut homeostasis. Mechanistically, B cell lymphoma 6 was identified as a direct transcriptional target of ATF3 in CD4+ T cells. In summary, we demonstrated ATF3 as a regulator of TFH cells in the gut, which may represent a potential immunotherapeutic target in colitis.


Subject(s)
Activating Transcription Factor 3/immunology , Activating Transcription Factor 3/pharmacology , Colitis/drug therapy , Colitis/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Adoptive Transfer , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Colitis/pathology , Colitis, Ulcerative , Colon/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Gene Expression Profiling , Homeostasis , Immunity, Mucosal/immunology , Immunoglobulin A , Immunotherapy , Mice , Peyer's Patches/immunology , T-Lymphocyte Subsets
11.
J Transl Med ; 19(1): 256, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112180

ABSTRACT

BACKGROUND: We aimed to develop a radiomic model based on pre-treatment computed tomography (CT) to predict the pathological complete response (pCR) in patients with rectal cancer after neoadjuvant treatment and tried to integrate our model with magnetic resonance imaging (MRI)-based radiomic signature. METHODS: This was a secondary analysis of the FOWARC randomized controlled trial. Radiomic features were extracted from pre-treatment portal venous-phase contrast-enhanced CT images of 177 patients with rectal cancer. Patients were randomly allocated to the primary and validation cohort. The least absolute shrinkage and selection operator regression was applied to select predictive features to build a radiomic signature for pCR prediction (rad-score). This CT-based rad-score was integrated with clinicopathological variables using gradient boosting machine (GBM) or MRI-based rad-score to construct comprehensive models for pCR prediction. The performance of CT-based model was evaluated and compared by receiver operator characteristic (ROC) curve analysis. The LR (likelihood ratio) test and AIC (Akaike information criterion) were applied to compare CT-based rad-score, MRI-based rad-score and the combined rad-score. RESULTS: We developed a CT-based rad-score for pCR prediction and a gradient boosting machine (GBM) model was built after clinicopathological variables were incorporated, with improved AUCs of 0.997 [95% CI 0.990-1.000] and 0.822 [95% CI 0.649-0.995] in the primary and validation cohort, respectively. Moreover, we constructed a combined model of CT- and MRI-based radiomic signatures that achieve better AIC (75.49 vs. 81.34 vs.82.39) than CT-based rad-score (P = 0.005) and MRI-based rad-score (P = 0.003) alone did. CONCLUSIONS: The CT-based radiomic models we constructed may provide a useful and reliable tool to predict pCR after neoadjuvant treatment, identify patients that are appropriate for a 'watch and wait' approach, and thus avoid overtreatment. Moreover, the CT-based radiomic signature may add predictive value to the MRI-based models for clinical decision making.


Subject(s)
Neoadjuvant Therapy , Rectal Neoplasms , Area Under Curve , Humans , Magnetic Resonance Imaging , Rectal Neoplasms/diagnostic imaging , Rectal Neoplasms/therapy , Retrospective Studies , Tomography, X-Ray Computed
12.
Int J Colorectal Dis ; 36(9): 1965-1979, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34297196

ABSTRACT

PURPOSE: The role of villous architecture in the prognosis of colon adenocarcinoma remains unclear. This study aimed to investigate the prognostic factors of colon adenocarcinoma with different types of villous architecture and to establish nomograms for predicting cancer-specific mortality. METHODS: This retrospective study included 10,427 patients with colon adenocarcinoma arising in adenomas with villous architectures. The patients were stratified into the tubulovillous adenocarcinoma cohort and villous adenocarcinoma cohort. The prognostic risk factors, which were incorporated into nomograms for survival prediction, were determined by the log-rank test and Cox hazard models. The Harrell's Concordance Index (C-index) and calibration curve were utilized to evaluate the prediction accuracy. RESULTS: The pathological type of villous architecture was independently associated with the mortality of the entire population. Age, race, tumor size, T/N/M stage, and chemotherapy were independent risk factors of mortality in both cohorts. Interestingly, tumor differentiation was a prognostic factor for tubulovillous adenocarcinoma rather than villous adenocarcinoma, while the retrieved lymph node number was a prognostic factor for villous adenocarcinoma rather than tubulovillous adenocarcinoma. Survival analysis showed that the mortality rate of villous adenocarcinoma was higher than that of tubulovillous adenocarcinoma (HR 1.361, P < 0.001). We then established nomograms to predict the mortality of both cohorts and found excellent discrimination and predictive accuracy (C-index 0.842 and 0.821). CONCLUSION: Villous architecture is a determinant of colon adenocarcinoma outcomes, which might prompt reports of villous architecture in colon adenocarcinoma specimens by pathologists. Our population-based nomograms could be useful for predicting the survival of patients with colon adenocarcinoma and guiding individualized treatments.


Subject(s)
Adenocarcinoma , Nomograms , Adenocarcinoma/pathology , Colon/pathology , Humans , Neoplasm Staging , Prognosis , Retrospective Studies , SEER Program
13.
J Cell Mol Med ; 24(16): 9349-9361, 2020 08.
Article in English | MEDLINE | ID: mdl-32628809

ABSTRACT

Gut microbiota and short-chain fatty acids (SCFAs) are associated with the development of various human diseases. In this study, we examined the role of astragaloside IV in modulating mouse gut microbiota structure and the generation of SCFAs, as well as in slow transit constipation (STC). An STC model was established by treating mice with loperamide, in which the therapeutic effects of astragaloside IV were evaluated. The microbiota community structure and SCFA content were analysed by 16S rRNA gene sequencing and gas chromatography-mass spectrometry, respectively. The influence of butyrate on STC was assessed using a mouse model and Cajal cells (ICC). Astragaloside IV promoted defecation, improved intestinal mobility, suppressed ICC loss and alleviated colonic lesions in STC mice. Alterations in gut microbiota community structure in STC mice, such as decreased Lactobacillus reuteri diversity, were improved following astragaloside IV treatment. Moreover, astragaloside IV up-regulated butyric acid and valeric acid, but decreased isovaleric acid, in STC mouse stools. Butyrate promoted defecation, improved intestinal mobility, and enhanced ICC proliferation by regulating the AKT-NF-κB signalling pathway. Astragaloside IV promoted intestinal transit in STC mice and inhibited ICC loss by regulating the gut microbiota community structure and generating butyric acid.


Subject(s)
Butyric Acid/metabolism , Constipation/drug therapy , Feces/microbiology , Gastrointestinal Microbiome , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Antidiarrheals/pharmacology , Constipation/chemically induced , Constipation/metabolism , Constipation/pathology , Female , Loperamide/toxicity , Male , Mice
14.
Int J Colorectal Dis ; 35(9): 1703-1709, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32458392

ABSTRACT

BACKGROUND: Postoperative anastomotic bleeding is considered a rare but life-threatening complication. There is no standard treatment strategy for this emergency condition. The aim of this study was to report our experiences in the management of postoperative anastomotic bleeding in patients with colorectal cancer. METHODS: We analyzed the general characteristics, treatments, and outcomes of patients with anastomotic bleeding after surgery for colorectal carcinoma at the Sixth Affiliated Hospital of Sun Yat-Sen University between July 2013 and September 2019 retrospectively. A univariate and multivariate analysis was performed to find protective factors for endoscopic hemostasis. Risk factors for anastomotic leakage after colonoscopy were also analyzed. RESULTS: A total of 9870 patients underwent surgeries for colorectal carcinoma between July 2013 and September 2019. Colonoscopies were performed in 78 cases with postoperative anastomotic bleeding. The effective rate of initial endoscopic hemostasis was 81% (63/78). In univariate and multivariate analysis, hemoclip therapy (odds ratio = 4.572; 95%CI 1.305-16.017; P = 0.017) and postoperative anastomotic bleeding within 5 days (odds ratio = 3.639; 95%CI 1.045-12.675; P = 0.042) are protective factors for endoscopic hemostasis. Comorbidity was associated with an increased risk for anastomotic leakage. CONCLUSIONS: Colonoscopy seems to be an effective way to achieve hemostasis for patients with anastomotic bleeding after surgery for colorectal carcinoma. It may be more effective in the early postoperative period, and hemoclip appears to be the first choice to control postoperative anastomotic bleeding.


Subject(s)
Colorectal Neoplasms , Postoperative Hemorrhage , Anastomosis, Surgical/adverse effects , Anastomotic Leak/etiology , Anastomotic Leak/surgery , Colorectal Neoplasms/complications , Colorectal Neoplasms/surgery , Humans , Postoperative Hemorrhage/etiology , Postoperative Hemorrhage/therapy , Postoperative Period , Retrospective Studies , Risk Factors
15.
Rev Esp Enferm Dig ; 112(1): 34-40, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31823640

ABSTRACT

INTRODUCTION: the evidence with regard to the benefit of laparoscopic surgery for pancreatoduodenectomy is conflicting. The aim of this meta-analysis was to compare the short-term outcomes in patients undergoing laparoscopic or open pancreatoduodenectomy via randomized controlled trial studies. METHODS: PubMed, Embase and Cochrane Library databases were searched for studies addressing laparoscopic versus open pancreatoduodenectomy up to February 2019. Only randomized controlled trial studies were included. RESULTS: three randomized controlled trial studies were identified, which included a total of 224 patients. Statistically significant differences were found with regard to estimated blood loss in favor of laparoscopic pancreatoduodenectomy (WMD, -150.9 ml; 95% CI, -167.61 to -134.18; p < 0.001) but with longer operative time (WMD, 97.66 min; 95% CI, 21.28 to 174.05; p = 0.01). No significant differences were found for severe postoperative complications (defined as Clavien-Dindo grade ≥ III complications), complication-related mortality within 90 days, blood transfusion requirements, length of hospital stay, postoperative pancreatic fistula, postpancreatectomy hemorrhage, bile leakage, delayed gastric emptying, surgical site infection, readmission rate, reoperation rate, harvested lymph nodes and R0 resection rate. CONCLUSIONS: the perioperative safety of laparoscopic pancreatoduodenectomy, which may have an advantage of lower estimated blood loss, is comparable to that of open pancreatoduodenectomy. Currently, a small volume of cases may be an important reason that affects the evaluation between laparoscopic and open pancreatoduodenectomy. Further evaluation of laparoscopic pancreatoduodenectomy will require large randomized control trials.


Subject(s)
Blood Loss, Surgical/statistics & numerical data , Laparoscopy/adverse effects , Pancreaticoduodenectomy/adverse effects , Postoperative Complications/mortality , Blood Transfusion/statistics & numerical data , Female , Gastric Emptying , Humans , Length of Stay , Lymph Node Excision , Male , Operative Time , Pancreatic Fistula/etiology , Postoperative Hemorrhage , Randomized Controlled Trials as Topic , Surgical Wound Infection , Treatment Outcome
17.
Int J Colorectal Dis ; 34(12): 2201-2204, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31735986

ABSTRACT

BACKGROUND: The incidence of colorectal cancer, especially located in distal colorectum, is rising markedly in young patients. Conventional adenomas and serrated polyps have been widely recognized as precursors of colorectal cancer. AIM: To investigate the correlation of polyp feature with polyp location in patients under age 50. METHOD: Patients under age 50 who had received colonoscopy were included from 2010 to 2018. Clinical data including number, location, size, and histopathology of polyps were collected. Odd ratios and 95% confidence interval of adenomas with their location were calculated. RESULT: In total, 25,636 patients aged 18-49 were enrolled, among which 4485 patients had polyps, with polyp detection rate of 17.5%. A total of 2484 and 2387 patients had conventional adenomas and serrated polyps, respectively. 76.0% advanced adenomas and 69.5% ≥ 10-mm serrated polyps were located in the distal colorectum. The detection rate of advanced adenomas was higher in patients aged 45-49. Patients with adenomas especially advanced adenomas in the distal colorectum were more likely to have advanced adenoma in the proximal colon. CONCLUSION: Among patients under age 50, advanced adenomas and ≥ 10-mm serrated polyps were predominantly in the distal colorectum. Advanced adenomas tended to be found in patients aged 45-49. Our results might explain the rising trend of distal colorectal cancer and emphasize the necessity for colonoscopy screening among these populations.


Subject(s)
Adenomatous Polyps/pathology , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Adenomatous Polyps/epidemiology , Adolescent , Adult , Age Distribution , China/epidemiology , Colonic Polyps/epidemiology , Colonoscopy , Colorectal Neoplasms/epidemiology , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Retrospective Studies , Tumor Burden , Young Adult
18.
Cancer Sci ; 109(2): 354-362, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29160937

ABSTRACT

Invasion and metastasis are crucially important factors in the survival of malignant tumors. Epithelial-mesenchymal transition (EMT) is an early step in metastatic progression and the presence of cancer stem cells is closely related to tumor survival, proliferation, metastasis, and recurrence. Herein we report that ectopic overexpression of microRNA 26b (miR-26b) in colorectal cancer (CRC) cell lines promoted EMT and stem cell-like phenotypes in vitro. Furthermore, miR-26b directly targeted and suppressed multiple tumor suppressors, including phosphatase and tensin homolog (PTEN) and wingless-type MMTV integration site family member 5A (WNT5A). Notably, miR-26b is markedly upregulated in tumor samples from patients with lymphatic metastases. These results indicate that miR-26b promotes CRC metastasis by downregulating PTEN and WNT5A, and may represent a therapeutic target for metastatic CRC.


Subject(s)
Colorectal Neoplasms/genetics , Down-Regulation , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Wnt-5a Protein/genetics , Caco-2 Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL