151
|
Nieser KJ, Harris AHS, Binswanger IA, Clark SC, Finlay AK. Legal-involved veterans are less likely to receive guideline-concordant colorectal cancer screening. BMC Health Serv Res 2025; 25:333. [PMID: 40038662 DOI: 10.1186/s12913-025-12490-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Programs to improve health care for adults with criminal legal involvement, including those who have been released from incarceration in jails or prisons or who are under court or community supervison, understandably focus on treatment for mental illness, drug overdose, and suicide. However, criminal legal-involved adults also have higher risk of developing and dying from medical conditions, such as cancer, relative to the general population. Colorectal cancer (CRC) screening among legal-involved adults, particularly those who have been incarcerated, might be delayed or missed. METHODS We conducted an observational study of national Veterans Health Administration (VHA) electronic health record data to compare the CRC screening rate between legal-involved Veterans, identified through their contact with the Veterans Justice Programs, and non-legal-involved Veterans. We included patients ages 46 to 75 eligible for average-risk screening in fiscal year 2022. Our main outcome of guideline-concordant CRC screening included stool-based testing, CT colonography, flexible sigmoidoscopy, and colonoscopy. Comparisons were estimated using an unadjusted multilevel logistic regression model with a random intercept for facility. Secondary analyses included examining associations between patient-level factors and screening receipt using adjusted models as well as assessing the variation in screening rates across 129 VHA facilities. RESULTS There were 27,597 legal-involved and 3,467,396 non-legal-involved patients who met screening eligibility. Only 47% of legal-involved patients were up to date with screening, compared to 54% of non-legal-involved patients (OR = 0.77 [95% CI: 0.75 to 0.79]; risk difference = -6.5% [95% CI: -7.1% to -5.9%]). Adjusted odds of screening were higher for patients with an assigned primary care provider (OR = 2.49 [95% CI: 2.48 to 2.51]). Screening rates varied widely across facilities, ranging from 24 to 75% for legal-involved patients and from 30 to 68% for non-legal-involved patients. Legal-involved patients had significantly lower screening rates at 49 facilities and a higher rate at two facilities, compared to non-legal-involved patients. CONCLUSIONS Nearly half of VHA patients were behind on recommended CRC screening, and legal-involved VHA patients had even lower rates. Current VHA efforts to improve legal-involved patients' connection to primary care providers may result in improved screening rates.
Collapse
Affiliation(s)
- Kenneth J Nieser
- Center for Innovation to Implementation, VA Palo Alto Healthcare System, Palo Alto, CA, USA.
- Stanford-Surgery Policy Improvement Research and Education Center, Department of Surgery, Stanford University, Stanford, CA, USA.
| | - Alex H S Harris
- Center for Innovation to Implementation, VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Stanford-Surgery Policy Improvement Research and Education Center, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Ingrid A Binswanger
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO, USA
- Colorado Permanente Medical Group, Denver, CO, USA
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Health Systems Science, Bernard J. Tyson Kaiser Permanente School of Medicine, Pasadena, CA, USA
| | - Sean C Clark
- Department of Veterans Affairs, Veterans Justice Programs, Washington, DC, USA
| | - Andrea K Finlay
- Center for Innovation to Implementation, VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Veterans Affairs, National Center on Homelessness Among Veterans, Menlo Park, CA, USA
- Division of Health Systems Science, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
152
|
Xiang YC, Liu XY, Hai ZX, Lv Q, Zhang W, Liu XR, Peng D, Wen GX. Nomogram for predicting the development of pneumonia after colorectal cancer surgery. Sci Rep 2025; 15:7417. [PMID: 40033128 DOI: 10.1038/s41598-025-92106-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
The aim of this study was to analyze the factors contributing to the development of pneumonia after colorectal cancer (CRC) surgery and to develop a validated nomogram to predict the risk. We retrospectively collected information on patients who underwent radical CRC resection at a single clinical center from January 2011 to December 2021. The information was then randomly assigned to a training cohort and a validation cohort in a 7:3 ratio. Univariate and multivariate logistic regression analysis were performed on the training cohort to identify independent risk factors for the development of pneumonias, which were then included in the nomogram. Validation was performed in a validation cohort, area under the curve (AUC) and calibration curves were used to determine the predictive accuracy and discriminative power of the graphs, and decision curve analysis (DCA) was used to further substantiate the clinical efficacy of the nomogram. A total of 7130 patients were included in the study. Based on multivariate logistic regression analysis of the training cohort, age, sex, preoperative albumin, surgical methods, and surgical time were identified as independent risk factors for the development of pneumonia after CRC surgery, and a nomogram prediction model was established using the above five variables. The AUC was 0.745 in training cohort and 0.773 in validation cohort. This study established a nomogram that is a good predictor of the risk of developing pneumonia after CRC surgery and provided surgeons with a reference for personalized management of patients in the perioperative period.
Collapse
Affiliation(s)
- Ying-Chun Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiao-Yu Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhan-Xiang Hai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Quan Lv
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xu-Rui Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guang-Xu Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
153
|
Jacob J, Anami Y, High PC, Liang Z, Subramanian S, Ghosh SC, AghaAmiri S, Guernsey-Biddle C, Tran H, Rowe J, Azhdarinia A, Tsuchikama K, Carmon KS. Antibody-Drug Conjugates Targeting the EGFR Ligand Epiregulin Elicit Robust Antitumor Activity in Colorectal Cancer. Cancer Res 2025; 85:973-986. [PMID: 39693606 PMCID: PMC11875910 DOI: 10.1158/0008-5472.can-24-0798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 10/19/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
As colorectal cancer remains a leading cause of cancer-related death, identifying therapeutic targets and approaches is essential to improve patient outcomes. The EGFR ligand epiregulin (EREG) is highly expressed in RAS wild-type (WT) and mutant colorectal cancer, with minimal expression in normal tissues, making it an attractive target for antibody-drug conjugate (ADC) development. In this study, we produced and purified an EREG mAb, H231, which had high specificity and affinity for human and mouse EREG. H231 also internalized to lysosomes, which is important for ADC payload release. ImmunoPET and ex vivo biodistribution studies showed significant tumor uptake of zirconium-89-labeled H231, with minimal uptake in normal tissues. H231 was conjugated to either cleavable dipeptide or tripeptide chemical linkers attached to the DNA-alkylating payload duocarmycin DM, and the cytotoxicity of EREG ADCs was assessed in a panel of colorectal cancer cell lines. EREG ADCs incorporating tripeptide linkers demonstrated the highest potency in EREG-expressing colorectal cancer cells irrespective of RAS mutations. Preclinical safety and efficacy studies showed that EREG ADCs were well tolerated, neutralized EGFR pathway activity, caused significant tumor growth inhibition or regression, and increased survival in colorectal cancer cell line and patient-derived xenograft models. These data suggest that EREG is a promising target for the development of ADCs for treating colorectal cancer and other cancer types that express high levels of EREG. Although the efficacy of clinically approved anti-EGFR mAbs is largely limited by RAS mutational status, EREG ADCs may show promise for both RAS mutant and WT patients, thus improving existing treatment options. Significance: EREG-targeting antibody-drug conjugates demonstrate acceptable safety and robust therapeutic efficacy in RAS mutant and wild-type colorectal cancer, suggesting their potential as an alternative to EGFR-targeted therapy to benefit a broader patient population.
Collapse
Affiliation(s)
- Joan Jacob
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yasuaki Anami
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - Peyton C. High
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Zhengdong Liang
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - Shraddha Subramanian
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Sukhen C. Ghosh
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - Solmaz AghaAmiri
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - Cara Guernsey-Biddle
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Ha Tran
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Julie Rowe
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Ali Azhdarinia
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - Kendra S. Carmon
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
154
|
Liu Y, Zhang S, Liao W, Qian J, Lu C, Jin L. Ornithine decarboxylase antizyme 2 (OAZ2) in human colon adenocarcinoma: a potent prognostic factor associated with immunity. Sci Rep 2025; 15:7481. [PMID: 40032914 DOI: 10.1038/s41598-025-90066-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
Despite few studies focusing on the OAZ2 gene in colorectal cancer, its potential role in colon adenocarcinoma (COAD) prognosis and immune modulation remains underexplored. This study examines the expression and mechanistic involvement of OAZ2 in COAD using data from The Cancer Genome Atlas (TCGA) and additional laboratory experiments. We employed uni- and multivariate Cox hazard regression analyses to evaluate its prognostic significance and gene set enrichment analysis (GSEA) to identify related signaling pathways. Our findings demonstrate significantly lower OAZ2 expression in COAD tissues compared to normal counterparts (P < 0.05) and establish its value as an independent prognostic indicator (P < 0.05). Laboratory experiments further revealed that the protein and mRNA levels of OAZ2 are significantly diminished in COAD compared to adjacent normal tissues, while its antagonist AZIN2 shows elevated expression, suggesting a competitive interaction that may regulate tumor behavior. Overexpression of OAZ2 in RKO colorectal cancer cells significantly reduced their proliferation rate and impaired migration, confirming the functional impact of OAZ2 dysregulation in COAD. Gene Set Enrichment Analysis (GSEA) highlighted the involvement of OAZ2 in cardiac muscle contraction and oxidative phosphorylation pathways. Additionally, OAZ2's association with immune features such as tumor mutational burden (TMB), microsatellite instability (MSI), and immune infiltration underscores its integral role in the tumor microenvironment. These comprehensive findings position OAZ2 as a promising biomarker for COAD prognosis and a potential target for therapeutic intervention, with evidence supporting its regulatory effects on cell dynamics and tumor aggressiveness.
Collapse
Affiliation(s)
- Yiheng Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shengjie Zhang
- Department of Emergency, Nantong Third People'S Hospital, Affiliated Nantong Hospital 3 of Nantong University, No. 60, Qingnian Middle Road, Chongchuan District, Nantong, 226000, Jiangsu, People's Republic of China
| | - Wenjie Liao
- Department of Emergency, Lianyungang Second People'S Hospital, Affiliated to Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Jun Qian
- Department of Emergency, Nantong Third People'S Hospital, Affiliated Nantong Hospital 3 of Nantong University, No. 60, Qingnian Middle Road, Chongchuan District, Nantong, 226000, Jiangsu, People's Republic of China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, , Jiangsu, China
| | - Li Jin
- Department of Emergency, Nantong Third People'S Hospital, Affiliated Nantong Hospital 3 of Nantong University, No. 60, Qingnian Middle Road, Chongchuan District, Nantong, 226000, Jiangsu, People's Republic of China.
- Department of Emergency Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
155
|
Chen XQ, Cui SS, Chen YZ, Wang CY, Liu Q, Qi YK, Du SS. Efficient Delivery of Oncolytic Peptide LTX-315 by ZIF-8: pH-Responsive Release, Improved Stability, and Reduced Hemolysis. Mol Pharm 2025; 22:1449-1461. [PMID: 39913295 DOI: 10.1021/acs.molpharmaceut.4c01144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
The first-in-class oncolytic peptide LTX-315 has exhibited positive anticancer responses in multiple phase I/II clinical trials. Nevertheless, the linear peptide LTX-315 suffers from poor proteolytic stability and undesired toxicity, especially hemolysis, which may limit its widespread applications. Except for the direct structural modifications, drug delivery systems (DDSs) are expected to protect LTX-315 from degradation and shield its hemolytic properties. Therefore, the LTX-315 and zeolitic imidazolate framework (ZIF-8)-based nanoparticles (NPs) were constructed with a high LTX-315 encapsulation rate of 59.9%, utilizing the biomineralized "one-pot method" in an aqueous system. The release of LTX-315, in vitro anticancer potency, serum stability, anticancer durability, antimigration activity, hemolysis effect, subcellular localization, and the membrane disruption/permeation effects of LTX-315@ZIF-8 NPs were investigated. LTX-315@ZIF-8 NPs exhibited potent cytotoxicity against cancer cells. The serum stability experiment and time-inhibition curve assay indicated that ZIF-8 NPs could effectively improve the stability of LTX-315, prolong the duration of anticancer action, and enhance the cytostatic potency. Especially, the LTX-315@ZIF-8 NPs not only effectively attenuated the hemolytic toxicity of LTX-315 but also achieved the pH-responsive release of LTX-315. The mechanism investigation indicated that LTX-315@ZIF-8 NPs possessed potent membranolytic activity and reduced the mitochondrial membrane potential to trigger cell death. Collectively, this paper not only established a robust strategy to improve the stability and reduce the hemolytic properties of LTX-315 but also provided a reliable reference for the future delivery of oncolytic peptides.
Collapse
Affiliation(s)
- Xin-Qi Chen
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Su-Su Cui
- Suzhou Jinchang Street Bailian Community Health Service Center, Suzhou 215000, China
| | - Yu-Zhen Chen
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Cai-Yun Wang
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Qing Liu
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yun-Kun Qi
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Shan-Shan Du
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| |
Collapse
|
156
|
Fwelo P, Adekunle TE, Adekunle TB, Garza ER, Huang E, Lawrence WR, Ewing AP. Differential Colorectal Cancer Mortality Across Racial and Ethnic Groups: Impact of Socioeconomic Status, Clinicopathology, and Treatment-Related Factors. Cancer Med 2025; 14:e70612. [PMID: 40040375 PMCID: PMC11880620 DOI: 10.1002/cam4.70612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/06/2024] [Accepted: 01/04/2025] [Indexed: 03/06/2025] Open
Abstract
INTRODUCTION Non-Hispanic Black (Black) colorectal cancer (CRC) patients have a higher risk of mortality than most other racial/ethnic groups. Limited studies examine the contribution of socioeconomic (SES), clinicopathologic, or treatment variations to mortality disparities. This retrospective cohort investigation examined the extent to which SES, clinicopathologic, and treatment factors explain racial/ethnic differences in CRC mortality. METHODS We studied 146,515 individuals, 18+ years old, with a confirmed diagnosis of CRC within 2010-2017, identified from the Surveillance, Epidemiology, and End Results (SEER) database. We performed Cox regression analyses to examine the association of race and ethnicity, surgery type, and tumor site with all-cause mortality and CRC-specific mortality. We then performed mediation analysis to quantify the extent to which mortality differences were mediated by SES, clinicopathologic, and treatment factors. RESULTS Black patients had a significantly higher hazard of all-cause mortality than non-Hispanic White (White) patients. The White versus Black patients' comparison demonstrated that variations in SES and clinicopathologic factors significantly explained 46.63% (indirect effect HR: 0.92, 95% CI 0.91-0.93) and 10.87% (indirect effect HR: 0.98, 95% CI 0.97-0.99) of the excess all-cause mortality among Black patients, respectively. The Hispanic versus Black comparisons identified SES as the most influential mediator, explaining 19.68% of the excess all-cause mortality. The proportions mediating for CRC-specific mortality showed comparable outcomes to all-cause mortality. CONCLUSION Black patients had a greater risk for all-cause mortality and CRC-specific mortality attributed to SES and clinicopathologic variations compared to other racial/ethnic groups. Future studies should investigate equity in healthcare through interventions addressing SES-related disparities.
Collapse
Affiliation(s)
- Pierre Fwelo
- Department of Epidemiology, Human Genetics and Environmental SciencesUTHealth School of Public HealthHoustonTexasUSA
| | - Toluwani E. Adekunle
- Department of Psychology, Public Health ProgramCalvin University School of HealthGrand RapidsMichiganUSA
| | - Tiwaladeoluwa B. Adekunle
- Center for Education in Health Sciences, Institute for Public Health and MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Ella R. Garza
- Department of Epidemiology, Human Genetics and Environmental SciencesUTHealth School of Public HealthHoustonTexasUSA
| | - Emily Huang
- Department of Surgery, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Wayne R. Lawrence
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthRockvilleMarylandUSA
| | - Aldenise P. Ewing
- Division of Epidemiology, College of Public HealthThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
157
|
Huang S, Yu L, Xiong F, Zhang B, Ruan S. Alcohol consumption and risk of early onset colorectal cancer: A systematic review and meta-analysis. Colorectal Dis 2025; 27:e70046. [PMID: 40123461 DOI: 10.1111/codi.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/12/2024] [Accepted: 12/08/2024] [Indexed: 03/25/2025]
Abstract
AIM The existing evidence has shown a positive association between alcohol consumption and an increased risk of colorectal cancer (CRC). However, the evidence is primarily based on studies of CRC in all ages, and the role of alcohol in early onset colorectal cancer (EOCRC) remains to be determined. The aim of this study was to investigate an association between the increasing incidence of EOCRC and alcohol consumption. METHOD We systematically searched PubMed, EMBASE, Cochrane and Web of Science up to June 2024 for studies that evaluated the association of alcohol intake with EOCRC risk and report specific results (e.g. relative risk, OR or hazard ratio and corresponding 95% CI). Based on the varying designs of the included studies, the corresponding effect values were extracted and categorized into high alcohol consumption and low alcohol consumption groups; a random-effects model was adopted to estimate the pooled effect sizes for analysis. Furthermore, subgroup analyses and publication bias assessments were conducted. RESULTS Three cohort studies and eight case-control studies were eligible and included. The results were pooled in meta-analyses, which yielded a heightened risk of EOCRC for increased alcohol intake (OR = 1.56, 95% CI 1.28-1.89, I2 = 89.3%). In the subgroup analysis, no significant differences were found in the association between alcohol consumption and the risk of developing EOCRC across gender, location or tumour site. The results of sensitivity analysis and publication bias indicated that the conclusion was robust. CONCLUSIONS This meta-analysis provides possible evidence for an association between alcohol consumption and risk of EOCRC. More research is needed in the future to confirm these findings.
Collapse
Affiliation(s)
- Siyu Huang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - Lulin Yu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - Fengchun Xiong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| | - Bo Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Shanming Ruan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Zhejiang, Hangzhou, China
| |
Collapse
|
158
|
Charboneau AJ, Cragle C, Frankhouse J, Kanneganti S, Kaplan JA, Moonka R, Rashidi L, Simianu VV. "Impact of regional data reporting and feedback on rectal cancer surgery quality metrics in the Surgical Care Outcomes Assessment Program (SCOAP)". Surg Open Sci 2025; 24:74-79. [PMID: 40160674 PMCID: PMC11950755 DOI: 10.1016/j.sopen.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Background Management of rectal cancer is increasingly complex. Leading societies describe metrics of high-quality perioperative rectal cancer care with the goal of reducing variation in practice and improving outcomes. This study was designed to describe the impact of targeted feedback at the institutional level on improving achievement of rectal cancer quality metrics. Methods Adult elective rectal cancer resections performed at institutions that continuously participated in SCOAP between 2011 and 2022 were included for analysis. Quality metrics evaluated were preoperative MRI (MRI), determination of tumor location (TL), use of neoadjuvant chemoradiation (NAC), performance of a total mesorectal excision (TME), 12+ lymph nodes resected (LN), and composite negative margins (NM). In-depth feedback on these metrics was provided by SCOAP at the end of 2015 and 2019. Achievement of the metrics was evaluated before (2011-2016), between (2017-2019), and after (2020-2022) feedback events to determine effect on achievement. Results 1962 resections were performed at 19 institutions. There were statistically significant increases in MRI(2011-2016 = 32 %, 2017-2019 = 88 %, 2020-2022 = 92 %;p < 0.01), TME(47 %, 68 %, 80 %;p < 0.01), and LN(76 %, 86 %, 86 %;p < 0.01) after one or both feedback events. TL(67 %, 69 %, 70 %;p = 0.558), NAC(62 %, 63 %, 67 %;p = 0.124), and NM(98 %, 97 %, 96 %;p = 0.39) were not significantly different. Mean composite score for metrics increased after each feedback (2011-2016 = 3.8±1.4; 2017-2019 = 4.3±1.4; 2020-2022 = 4.5±1.5;p < 0.01). Conclusion Interval, in-depth feedback on rectal cancer quality process metrics was associated with increased achievement of several metrics and overall number of metrics achieved. Broader implementation of this feedback method could further advance the quality of rectal cancer surgical care.
Collapse
Affiliation(s)
- Alex J. Charboneau
- Colon and Rectal Surgery, Virginia Mason Medical Center, 1100 9 Ave, Seattle, WA 98101, USA
| | - Chad Cragle
- Colon and Rectal Surgery, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA
| | - Joseph Frankhouse
- Colon and Rectal Surgery, Legacy Medical Group, 2222 NW Lovejoy St, Suite 601, Building 1, Portland, OR 97210, USA
| | - Shalini Kanneganti
- Colon and Rectal Surgery, Virginia Mason Franciscan Health, 1708 S Yakima Ave, Suite 105, Tacoma, WA 98405, USA
| | - Jenny A. Kaplan
- Colon and Rectal Surgery, Virginia Mason Medical Center, 1100 9 Ave, Seattle, WA 98101, USA
| | - Ravi Moonka
- Colon and Rectal Surgery, Virginia Mason Medical Center, 1100 9 Ave, Seattle, WA 98101, USA
| | - Laila Rashidi
- Colon and Rectal Surgery, MultiCare, 3124 S 19 St, Suite 220, Tacoma, WA 98405, USA
| | - Vlad V. Simianu
- Colon and Rectal Surgery, Virginia Mason Medical Center, 1100 9 Ave, Seattle, WA 98101, USA
| |
Collapse
|
159
|
Wang H, Ji Y, Deng S, Qin XY, Ye XT, Sun YY, Che XY, Yang L, Huang CY, Chen Y, Liu YP. SQYC formula improves the efficacy of PD-1 monoclonal antibodies in MSS colorectal cancer by regulating dendritic cell mitophagy via the PINK1-Parkin pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156388. [PMID: 39826284 DOI: 10.1016/j.phymed.2025.156388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 01/02/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Microsatellite stable (MSS) colorectal carcinomas (CRCs) exhibit poor responsiveness to immunotherapy such as immune checkpoint inhibitors (ICIs). In the realm of clinical cancer treatment, traditional Chinese medicines (TCMs) are extensively utilized for their immunomodulatory properties. Shen Qi Yi Chang (SQYC), a clinical prescription for CRC treatment, improve the life quality of CRC patients and enhance their immune function. PURPOSE This study was to reveal the effect and mechanism of SQYC in improving the effect of PD-1 inhibitors in the treatment of MSS-type CRC. METHODS CT26-luc in situ CRC tumor model and human CRC organoid model was established to evaluate the anti-tumor efficacy of SQYC combined with PD-1 inhibitor. Flow cytometry analysis was utilized to investigate the effect of SQYC on the infiltration and immune function of TILs and DCs in the immune microenvironment. Following this, RNA sequencing analysis, seahorse, TEM and immunofluorescence were performed to regulation of SQYC on mitophagy in DCs cells. UPLC-Q-TOF/MS and molecular docking were used to reveal the key blood-entering components of SQYC-regulated PINK1-parkin pathway. RESULTS The SQYC-containing serum improved the efficacy of sintilimab in MSS CRC organoid model. After combined administration of 11.4 g/kg/day SQYC extract and 5 mg/kg α-PD-1, it was observed that SQYC enhanced the efficacy of PD-1 inhibitor against MSS CRC. Flow cytometry and immunofluorescence analysis revealed an augmented infiltration of tumor-infiltrating lymphocytes (TILs) and an improved antigen presentation function of dendritic cells (DCs). Notably, RNA sequencing analysis demonstrated an evident correlation with mitochondrial function related pathways following SQYC treatment. Mechanistically, SQYC promoted mitophagy in DCs via the PINK1-Parkin pathway, thereby improving mitochondrial quality, energy metabolism, and mitochondrial dynamics. Evaluation of the blood components of SQYC coupled with molecular docking, demonstrated good binding affinity with PINK1/PARKIN/LC3. CONCLUSION Our findings highlight SQYC as a promising candidate for improving immunotherapy in MSS CRC, suggesting that targeting PINK1-Parkin in DCs could represent a novel strategy for improving the efficacy of ICIs. Furthermore, it provides new theoretical and scientific underpinnings to enhance the clinical efficacy of immunosuppressants.
Collapse
Affiliation(s)
- Hong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Yi Ji
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Shan Deng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Xiao Ying Qin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Xie Tao Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Ye Yang Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Xiao Yu Che
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Lin Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Chu Yue Huang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China; Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing, 210028, China.
| | - Yu Ping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China; Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
160
|
Zhang YZ, Song M, Li S, Tie J, Zhu XG, Li YH, Wu AW, Cai Y, Wang WH. Comparison of prognosis for T4b rectal cancer with different pelvic compartment involvement treated using neoadjuvant chemoradiotherapy and implications for refinement of the current T staging system: A retrospective cohort study. Transl Oncol 2025; 53:102313. [PMID: 39908966 PMCID: PMC11847518 DOI: 10.1016/j.tranon.2025.102313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/25/2024] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
PURPOSE Although classified as one stage, T4b rectal cancer actually represents a group of heterogeneous diseases. Our study aimed to assess the prognosis difference of T4b rectal cancer involving inferior pelvic and other pelvic compartments. This information may be helpful in refinement of the current T staging system. METHODS We retrospectively analysed data from 195 patients with magnetic resonance imaging-identified locally advanced T4b rectal cancer who received neoadjuvant chemoradiotherapy between January 2010 and December 2019. 104 patients had only inferior pelvic compartment involvement (group A) while 91 patients had anterior, posterior or lateral pelvic compartment involvement (group B). Short-term and long-term outcomes were compared between the two groups. RESULTS After neoadjuvant therapy, 80.8 % patients (84/104) in group A and 92.3 % patients (84/91) in group B underwent surgery. The R0 resection rates were 97.6 % and 89.3 %, respectively. 8.7 % patients (9/104) in group A achieved clinical complete response and adopted watch-and-wait strategy. Patients in group A had significantly superior 5-year progression-free survival (PFS) (67.8 % vs. 55.5 %, P = 0.032) and overall survival (OS) (89.6 % vs. 71.8 %, P = 0.001) than group B. Multivariable Cox regression analysis also identified pelvic compartment involvement classification as an independent predictor of PFS (hazard ratio 1.776, P = 0.046) and OS (hazard ratio 3.477, P = 0.004). CONCLUSION T4b rectal cancers with involvement limited to the inferior pelvic compartment had superior prognosis compared to those involving other pelvic compartments. These differences should be investigated further and taken into consideration in refinement of the current T staging system.
Collapse
Affiliation(s)
- Yang-Zi Zhang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Maxiaowei Song
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shuai Li
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jian Tie
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiang-Gao Zhu
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yong-Heng Li
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ai-Wen Wu
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yong Cai
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Wei-Hu Wang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
161
|
Hu Q, Yang X, Sun Y. RE: Characteristics of a cost-effective blood test for colorectal cancer screening. J Natl Cancer Inst 2025; 117:570-571. [PMID: 39718768 DOI: 10.1093/jnci/djae340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 12/25/2024] Open
Affiliation(s)
- Qiang Hu
- School of Integrated Chinese and Western Medicine, Zhejiang Chinese Medicine University, Hangzhou 310053, China
- Department of General Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Xiyin Yang
- Department of Traditional Chinese Medicine, Community Health Service Center of Guali Town of Xiaoshan, Hangzhou 311200, China
| | - Yuanshui Sun
- Department of General Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| |
Collapse
|
162
|
Zhu X, Sun H, Wang Y, Hu G, Shao L, Zhang S, Liu F, Chi C, He K, Tang J, An Y, Tian J, Liu Z. Prediction of Lymph Node Metastasis in Colorectal Cancer Using Intraoperative Fluorescence Multi-Modal Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2025; 44:1568-1580. [PMID: 40030456 DOI: 10.1109/tmi.2024.3510836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The diagnosis of lymph node metastasis (LNM) is essential for colorectal cancer (CRC) treatment. The primary method of identifying LNM is to perform frozen sections and pathologic analysis, but this method is labor-intensive and time-consuming. Therefore, combining intraoperative fluorescence imaging with deep learning (DL) methods can improve efficiency. The majority of recent studies only analyze uni-modal fluorescence imaging, which provides less semantic information. In this work, we mainly established a multi-modal fluorescence imaging feature fusion prediction (MFI-FFP) model combining white light, fluorescence, and pseudo-color imaging of lymph nodes for LNM prediction. Firstly, based on the properties of various modal imaging, distinct feature extraction networks are chosen for feature extraction, which could significantly enhance the complementarity of various modal information. Secondly, the multi-modal feature fusion (MFF) module, which combines global and local information, is designed to fuse the extracted features. Furthermore, a novel loss function is formulated to tackle the issue of imbalanced samples, challenges in differentiating samples, and enhancing sample variety. Lastly, the experiments show that the model has a higher area under the receiver operating characteristic (ROC) curve (AUC), accuracy (ACC), and F1 score than the uni-modal and bi-modal models and has a better performance compared to other efficient image classification networks. Our study demonstrates that the MFI-FFP model has the potential to help doctors predict LNM and shows its promise in medical image analysis.
Collapse
|
163
|
Duan X, Yeerkenbieke G, Huang S, Feng Y. USP32 Promotes Colorectal Carcinoma Progression Through Activating NF-κB Signalling Pathway. J Cell Mol Med 2025; 29:e70457. [PMID: 40122703 PMCID: PMC11930632 DOI: 10.1111/jcmm.70457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Ubiquitin-specific protease 32 (USP32) plays a key role in cancer progression. However, its functions in colorectal carcinoma (CRC) are still unexplored. In our study, we explored the expression and clinical significance of USP32 in CRC as well as its relationship with the tumour microenvironment (TME). As a result, we found that USP32 is overexpressed in CRC and it is associated with poor outcomes in CRC patients. In addition, the expression of USP32 is significantly related to the activation of the NF-κB signalling pathway and the immune infiltrates of the TME. Wet experiments also confirmed that USP32 is critical for the proliferation, survival, and migration of CRC cells and tumour growth, which may be due to the activation of the NF-κB signalling pathway. In conclusion, targeting the USP32-NF-κB axis may be a novel treatment strategy for CRC patients.
Collapse
Affiliation(s)
- Xiaofan Duan
- School of MedicineTongji UniversityShanghaiChina
- Department of OncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
| | - Gaoshaer Yeerkenbieke
- School of MedicineTongji UniversityShanghaiChina
- Department of OncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
| | - Siping Huang
- Department of surgical oncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
| | - Yanjun Feng
- Department of OncologyShanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghaiChina
- Shanghai East Hospital, Nanjing Medical UniversityShanghaiChina
- Department of OncologyShanghai Artemed HospitalShanghaiChina
| |
Collapse
|
164
|
Martínez-Castedo B, Camblor DG, Martín-Arana J, Carbonell-Asins JA, García-Micó B, Gambardella V, Huerta M, Roselló S, Roda D, Gimeno-Valiente F, Cervantes A, Tarazona N. Minimal residual disease in colorectal cancer. Tumor-informed versus tumor-agnostic approaches: unraveling the optimal strategy. Ann Oncol 2025; 36:263-276. [PMID: 39675560 DOI: 10.1016/j.annonc.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) analysis has emerged as a minimally invasive tool for detecting minimal residual disease (MRD) in colorectal cancer (CRC) patients. This enables dynamic risk stratification, earlier recurrence detection and optimized post-surgical treatment. Two primary methodologies have been developed for ctDNA-based MRD detection: tumor-informed strategies, which identify tumor-specific mutations through initial tissue sequencing to guide ctDNA monitoring, and tumor-agnostic approaches, which utilize predefined panels to detect common cancer-associated genomic or epigenomic alterations directly from plasma without prior tissue analysis. The debate over which is superior in terms of sensitivity, specificity, cost-effectiveness and clinical feasibility remains unsolved. DESIGN This review summarizes studies published up to November 2024, exploring the utility and performance of tumor-informed and tumor-agnostic approaches for ctDNA analysis in CRC. We evaluate the strengths and limitations of each methodology, focusing on sensitivity, specificity and clinical outcomes. RESULTS Both strategies demonstrate clinical utility in post-operative risk stratification and guiding adjuvant chemotherapy decisions in CRC patients. Tumor-informed approaches generally exhibit superior sensitivity and specificity for recurrence prediction, attributed to their personalized tumor profile designs. However, these methods are limited by the need for prior tissue sequencing and higher associated costs. In contrast, tumor-agnostic approaches offer broader applicability due to their reliance on plasma-only analysis, although with relatively lower sensitivity. Technological advancements, including fragmentomics and multi-omic integrations, are expanding the capabilities of ctDNA-based MRD detection, enhancing the performance of both approaches. CONCLUSIONS While tumor-informed strategies currently offer higher precision in MRD detection, tumor-agnostic approaches are gaining traction due to their convenience and improving performance metrics. The integration of novel technologies in ongoing clinical trials may redefine the optimal approach for MRD detection in CRC, paving the way for more personalized and adaptive patient management strategies.
Collapse
Affiliation(s)
- B Martínez-Castedo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - D G Camblor
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - J Martín-Arana
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - J A Carbonell-Asins
- Biostatistics Unit, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - B García-Micó
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - V Gambardella
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - M Huerta
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - S Roselló
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - D Roda
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain
| | - F Gimeno-Valiente
- Cancer Evolution and Genome Instability Laboratory, University College London Cancer Institute, London, UK
| | - A Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain.
| | - N Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
165
|
Nadukkandy AS, Blaize B, Kumar CD, Mori G, Cordani M, Kumar LD. Non-coding RNAs as mediators of epithelial to mesenchymal transition in metastatic colorectal cancers. Cell Signal 2025; 127:111605. [PMID: 39842529 DOI: 10.1016/j.cellsig.2025.111605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/06/2025] [Accepted: 01/12/2025] [Indexed: 01/24/2025]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, necessitating the development of innovative treatment strategies. Recent research has underscored the significant role of non-coding RNAs (ncRNAs) in CRC pathogenesis, offering new avenues for diagnosis and therapy. In this review, we delve into the intricate roles of various ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in CRC progression, epithelial-mesenchymal transition (EMT), metastasis, and drug resistance. We highlight the interaction of these ncRNAs with and regulation of key signaling pathways, such as Wnt/β-catenin, Notch, JAK-STAT, EGFR, and TGF-β, and the functional relevance of these interactions in CRC progression. Additionally, the review highlights the emerging applications of nanotechnology in enhancing the delivery and efficacy of ncRNA-based therapeutics, which could address existing challenges related to specificity and side effects. Future research directions, including advanced diagnostic tools, targeted therapeutics, strategies to overcome drug resistance, and the integration of personalized medicine approaches are discussed. Integrating nanotechnology with a deeper understanding of CRC biology offers the potential for more effective, targeted, and personalized strategies, though further research is essential to validate these approaches.
Collapse
Affiliation(s)
- Aisha Shigna Nadukkandy
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Britny Blaize
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India
| | - Chethana D Kumar
- Department of Surgical ICU, Christian Medical College, IDA Scudder Road, Vellore 632004, Tamil Nadu, India
| | - Giulia Mori
- Department Of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Lekha Dinesh Kumar
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad 500007, India.
| |
Collapse
|
166
|
Lehtonen TM, Koskenvuo LE, Lepistö AH. Early-onset rectal cancer: Experience of a single-center, high-volume unit. Scand J Surg 2025; 114:22-34. [PMID: 39396124 DOI: 10.1177/14574969241282543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
BACKGROUND AND OBJECTIVE The incidence of early-onset colorectal cancer among the young (<50 years) has been reported to have risen in last decades. This retrospective study aimed to investigate the characteristics of early-onset rectal cancers (EO-RCs) and potential changes in proportion of EO-RCs, and further to report the mortality and recurrence rates of EO-RCs. METHODS In the years 2007-2021, 2557 rectal cancer (RC) patients were operated in Helsinki University Hospital and of them 147 were 18-49 years old. Cumulative overall survival (OS), disease-specific survival, and disease-free survival were calculated using the Kaplan-Meier analysis. RESULTS The percentual amount of the EO-RCs varied between 2.5% and 11.3% annually and there was no perceivable trend. Majority were adenocarcinomas (98.7%), of which 8.8% were mucinous. Predisposing factors such as Lynch syndrome, polyposis, or ulcerative colitis were seen in 26 patients (17.7%) and in 10 of 22 patients (45.5%) under 35 years. The cumulative 5-year OS was 91.9% in stage I, 93.3% in stage II, 86.7% in stage III, and 50.0% in stage IV disease. Metastatic recurrence was found in 22 cases (18.4%) and local recurrence in 8 patients (5.4%) and 6 patients had both. CONCLUSIONS In our cohort of 147 EO-RC patients, OS was good and the clinical course did not seem to differ much from the course of RC in general population.
Collapse
Affiliation(s)
- Taru M Lehtonen
- Department of Surgery HUS Helsinki University Hospital and University of Helsinki, Jorvi Hospital,Espoo 20540, Finland
| | - Laura E Koskenvuo
- Department of Surgery, Helsinki University Hospital and University of Helsinki, Jorvi, Finland
| | - Anna H Lepistö
- Department of Surgery, Helsinki University Hospital and University of Helsinki, Jorvi, Finland
- Applied Tumor Genomics, Research Programs Unit, University of Helsinki, Helsinki, Finland
| |
Collapse
|
167
|
Saka AH, Giaquinto AN, McCullough LE, Tossas KY, Star J, Jemal A, Siegel RL. Cancer statistics for African American and Black people, 2025. CA Cancer J Clin 2025; 75:111-140. [PMID: 39976243 PMCID: PMC11929131 DOI: 10.3322/caac.21874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 02/21/2025] Open
Abstract
African American and other Black individuals (referred to as Black people in this article) have a disproportionate cancer burden, including the lowest survival of any racial or ethnic group for most cancers. Every 3 years, the American Cancer Society estimates the number of new cancer cases and deaths for Black people in the United States and compiles the most recent data on cancer incidence (herein through 2021), mortality (through 2022), survival, screening, and risk factors using population-based data from the National Cancer Institute and the Centers for Disease Control and Prevention. In 2025, there will be approximately 248,470 new cancer cases and 73,240 cancer deaths among Black people in the United States. Black men have experienced the largest relative decline in cancer mortality from 1991 to 2022 overall (49%) and in almost every 10-year age group, by as much as 65%-67% in the group aged 40-59 years. This progress largely reflects historical reductions in smoking initiation among Black teens, advances in treatment, and earlier detections for some cancers. Nevertheless, during the most recent 5 years, Black men had 16% higher mortality than White men despite just 4% higher incidence, and Black women had 10% higher mortality than White women despite 9% lower incidence. Larger inequalities for mortality than for incidence reflect two-fold higher death rates for prostate, uterine corpus, and stomach cancers and for myeloma, and 40%-50% higher rates for colorectal, breast, cervical, and liver cancers. The causes of ongoing disparities are multifactorial, but largely stem from inequalities in the social determinants of health that trace back to structural racism. Increasing diversity in clinical trials, enhancing provider education, and implementing financial incentives to ensure equitable care across the cancer care continuum would help close these gaps.
Collapse
Affiliation(s)
- Anatu H Saka
- Cancer Surveillance Research, American Cancer Society, Atlanta, Georgia, USA
| | - Angela N Giaquinto
- Cancer Surveillance Research, American Cancer Society, Atlanta, Georgia, USA
| | | | - Katherine Y Tossas
- Department of Social and Behavioral Sciences, School of Public Health, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jessica Star
- Risk Factors and Screening Research, American Cancer Society, Atlanta, Georgia, USA
| | - Ahmedin Jemal
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| | - Rebecca L Siegel
- Cancer Surveillance Research, American Cancer Society, Atlanta, Georgia, USA
| |
Collapse
|
168
|
Ren Q, Vermeulen L. Shaping the microenvironment in peritoneal metastases. NATURE CANCER 2025; 6:412-414. [PMID: 39966609 DOI: 10.1038/s43018-024-00878-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Affiliation(s)
- Qihao Ren
- Discovery Oncology, Genentech Inc., South San Francisco, CA, USA.
| | - Louis Vermeulen
- Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
169
|
Sayed IM, Chakraborty A, Inouye K, Dugan L, Tocci S, Advani I, Park K, Gaboyan S, Kasaraneni N, Ma B, Hazra TK, Das S, Crotty Alexander LE. E-cigarettes increase the risk of adenoma formation in murine colorectal cancer model. Arch Toxicol 2025; 99:1223-1236. [PMID: 39786590 DOI: 10.1007/s00204-024-03932-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
E-cigarettes (E.cigs) cause inflammation and damage to human organs, including the lungs and heart. In the gut, E.cig vaping promotes inflammation and gut leakiness. Further, E.cig vaping increases tumorigenesis in oral and lung epithelial cells by inducing mutations and suppressing host DNA repair enzymes. It is well known that cigarette (cig) smoking increases the risk of colorectal cancer (CRC). To date, it is unknown whether E.cig vaping impacts CRC development. A mouse model of human familial adenomatous polyposis (CPC-APC) was utilized wherein a mutation in the adenomatous polyposis coli (APC) gene, CDX2-Cre-APCMin/+, leads to the development of colon adenomas within 11-16 weeks. Mice were exposed to air (controls), E.cig vaping, cig, or both (dual exposure). After 4 weeks of 2 h exposures per day (1 h of each for dual exposures), the colon was collected and assessed for polyp number and pathology scores by microscopy. Expression of inflammatory cytokines and cancer stem cell markers were quantified. DNA damage such as double-strand DNA breaks was evaluated by immunofluorescence, western blot, and gene-specific long amplicon qPCR. DNA repair enzyme levels (NEIL-2, NEIL-1, NTH1, and OGG1) were quantified by western blot. Proliferation markers were assessed by RT-qPCR and ELISA. CPC-APC mice exposed to E.cig, cig, and dual exposure developed a higher number of polyps compared to controls. Inflammatory proteins, DNA damage, and cancer stemness markers were higher in E-cig, cig, and dual-exposed mice as well. DNA damage was found to be associated with the suppression of DNA glycosylases, particularly with NEIL-2 and NTH1. E.cig and dual exposure both stimulated cancer cell stem markers (CD44, Lgr-5, DCLK1, and Ki67). The effect of E.cigs on polyp formation and CRC development was less than that of cigs, while dual exposure was more tumorigenic than either of the inhalants alone. E.cig vaping promotes CRC by stimulating inflammatory pathways, mediating DNA damage, and upregulating transcription of cancer stem cell markers. Critically, combining E.cig vaping with cig smoking leads to higher levels of tumorigenesis. Thus, while the chemical composition of these two inhalants, E.cigs and cigs, is highly disparate, they both drive the development of cancer and when combined, a highly common pattern of use, they can have additive or synergistic effects.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, CA, 92093, USA.
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA.
| | - Anirban Chakraborty
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kaili Inouye
- Department of Pathology, University of California, San Diego, CA, 92093, USA
| | - Leanne Dugan
- Department of Pathology, University of California, San Diego, CA, 92093, USA
| | - Stefania Tocci
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Ira Advani
- Department of Medicine, University of California, San Diego, CA, 92093, USA
| | - Kenneth Park
- Department of Medicine, University of California, San Diego, CA, 92093, USA
| | - Samvel Gaboyan
- Department of Medicine, University of California, San Diego, CA, 92093, USA
| | - Nikita Kasaraneni
- Department of Medicine, University of California, San Diego, CA, 92093, USA
| | - Benjamin Ma
- Department of Medicine, University of California, San Diego, CA, 92093, USA
| | - Tapas K Hazra
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA, 92093, USA.
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA, 01854, USA.
| | - Laura E Crotty Alexander
- Department of Medicine, University of California, San Diego, CA, 92093, USA.
- Medicine Service, VA San Diego Healthcare System, San Diego, CA, 92161, USA.
| |
Collapse
|
170
|
Li L, He J, Hu H, Wang Y, Li W, Huang S, Rownoak J, Xu S, Xie F, Wang J, Mi W, Cai J, Ye Y, Liu S, Wang J, Li Y. Linear-Array Endoscopic Ultrasound and Narrow-Band Imaging Measure the Invasion Depth of Nonpedunculated Rectal Lesions With Comparable Accuracy Based on a Randomized Controlled Trial. Am J Gastroenterol 2025; 120:562-569. [PMID: 39051647 DOI: 10.14309/ajg.0000000000002975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Linear-array endoscopic ultrasound (EUS) and narrow-band imaging (NBI) are both used to estimate the invasion depth of nonpedunculated rectal lesions (NPRLs). However, it is unclear which procedure is more accurate. This randomized controlled trial aimed to compare the diagnostic accuracy of linear EUS and NBI for estimating the invasion depth of NPRLs. METHODS This study is a single-center, randomized, tandem trial. Eligible patients with NPRLs were randomly assigned to A group (assessment with EUS followed by NBI) or B group (assessment with NBI followed by EUS). The invasion depth of each lesion was independently measured by each procedure and categorized as mucosal to slight submucosal (M-SM s , invasion depth <1,000 μm) or deep submucosal (SM d , invasion depth ≥1,000 μm) invasion, with postoperative pathology as the standard of measurement. The primary outcome was diagnostic accuracy, and secondary outcomes included sensitivity, specificity, and procedure time. RESULTS Eighty-six patients with NPRLs were enrolled, and 79 patients were finally analyzed, including 39 cases in the A group and 40 cases in the B group. Comparable diagnostic accuracies were observed between EUS and NBI (96.2% vs 93.7%, P = 0.625). EUS identified lesions with deep submucosal invasion with 81.8% sensitivity while that of NBI was 63.6% ( P = 0.500). The specificity of both EUS and NBI was 98.5%. The procedure time was also similar between EUS and NBI (5.90 ± 3.44 vs 6.4 ± 3.94 minutes, P = 0.450). Furthermore, the combined use of EUS and NBI did not improve diagnostic accuracy compared with EUS or NBI alone (94.9% vs 96.2% vs 93.7%, P = 0.333). DISCUSSION Linear EUS and NBI measure the invasion depth of NPRLs with comparable accuracy. The combination of the 2 methods does not improve the diagnostic accuracy. Single NBI should be preferred, considering its simplicity and convenience in clinical practice.
Collapse
Affiliation(s)
- Lingzhi Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaming He
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyan Hu
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingying Wang
- Economics of Global Health and Infectious Diseases Unit, Melbourne Health Economics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Weixin Li
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shaohui Huang
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jahan Rownoak
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shenglin Xu
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Xie
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junfen Wang
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenting Mi
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianqun Cai
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaping Ye
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Side Liu
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Wang
- Department of Pathology, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yue Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Zhongshan Torch Development Hospital, Zhongshan, China
| |
Collapse
|
171
|
Leary JB, Hu J, Leal A, Davis SL, Kim S, Lentz R, Friedrich T, Herter W, Messersmith WA, Lieu CH. Risk Without Reward: Differing Patterns of Chemotherapy Use Do Not Improve Outcomes in Stage II Early-Onset Colon Cancer. JCO Oncol Pract 2025; 21:333-340. [PMID: 39047212 PMCID: PMC11925348 DOI: 10.1200/op.24.00159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/11/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024] Open
Abstract
PURPOSE Rising rates of early-onset colon cancer (EOCC) present challenges in deciding how to optimally treat patients. Although standard of care for stage II CC is surgical resection, adding chemotherapy for high-risk disease, evidence suggests treatment selection may differ by age. We investigated whether adjuvant chemotherapy (AC) administration rates differ between patients with early- and later-onset stage II CC. METHODS Data originated from the nationwide Flatiron Health electronic health record (EHR)-derived deidentified database spanning January 1, 2003, to August 1, 2021. Adults with stage II CC were grouped as age 18-49 years (EOCC) and those age 50 years or older (later-onset colon cancer [LOCC]). Demographics, Eastern Cooperative Oncology Group score, tumor stage and site, and chemotherapy were included. Primary outcomes included rates of AC administration by age and ethnicity; secondary outcomes included overall survival (OS) and time to metastatic disease (TTMD). Univariate and multivariable logistic regression models evaluated relationships between chemotherapy administration, age, and ethnicity, adjusting for significant covariates. RESULTS One thousand sixty-five patients were included. Median age of patients with EOCC was 45.0 years versus 69.0 years for patients with LOCC. Adjusted multivariate analysis showed patients with EOCC received AC significantly more often than patients with LOCC. Non-Hispanic patients received AC at significantly lower rates than Hispanic patients in both cohorts. Subanalysis of stage IIA patients showed that patients with EOCC were more likely to receive AC than patients with LOCC. No significant differences in OS or TTMD were observed by age regardless of AC administration in stage II overall; however, patients with stage IIA EOCC receiving AC had significantly longer TTMD than those not receiving AC. CONCLUSION AC was given preferentially in stage II EOCC, even in stage IIA, despite deviation from guidelines. This may expose low-risk patients to unnecessary toxicities and suggests bias toward treating younger patients more aggressively, despite unclear evidence for better outcomes.
Collapse
Affiliation(s)
- Jacob B. Leary
- Department of Medicine, University of Washington, Seattle, WA
| | - Junxiao Hu
- Biostatistics Shared Resource, University of Colorado Cancer Center, Aurora, CO
| | - Alexis Leal
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - S. Lindsey Davis
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Sunnie Kim
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Robert Lentz
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tyler Friedrich
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Whitney Herter
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Wells A. Messersmith
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Christopher H. Lieu
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
172
|
Kang Q, Hu X, Chen Z, Liang X, Xiang S, Wang Z. The METTL3/TRAP1 axis as a key regulator of 5-fluorouracil chemosensitivity in colorectal cancer. Mol Cell Biochem 2025; 480:1865-1889. [PMID: 39287889 PMCID: PMC11842504 DOI: 10.1007/s11010-024-05116-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
Colorectal cancer (CRC) remains a significant clinical challenge, with 5-Fluorouracil (5-FU) being the frontline chemotherapy. However, chemoresistance remains a major obstacle to effective treatment. METTL3, a key methyltransferase involved in RNA methylation processes, has been implicated in CRC carcinogenesis. However, its role in modulating CRC sensitivity to 5-FU remains elusive. In this study, we aimed to investigate the role and mechanisms of METTL3 in regulating 5-FU chemosensitivity in CRC cells. Initially, we observed that 5-FU treatment inhibited cell viability and induced apoptosis, accompanied by a reduction in METTL3 expression in HCT-116 and HCT-8 cells. Subsequent assays including drug sensitivity, EdU, colony formation, TUNEL staining, and flow cytometry revealed that METTL3 depletion enhanced 5-FU sensitivity and increased apoptosis induction both in vitro and in vivo. Conversely, METTL3 overexpression conferred resistance to 5-FU in both cell lines. Moreover, knockdown of METTL3 in 5-FU-resistant CRC cell lines HCT-116/FU and HCT-15/FU significantly decreased 5-FU tolerance and induced apoptosis upon 5-FU treatment. Mechanistically, we found that METTL3 regulated 5-FU sensitivity and apoptosis induction by modulating TRAP1 expression. Further investigations using m6A colorimetric ELISA, dot blot, MeRIP-qPCR and RNA stability assays demonstrated that METTL3 regulated TRAP1 mRNA stability in an m6A-dependent manner. Additionally, overexpression of TRAP1 mitigated the cytotoxic effects of 5-FU on CRC cells. In summary, our study uncovers the pivotal role of the METTL3/TRAP1 axis in modulating 5-FU chemosensitivity in CRC. These findings provide new insights into the mechanisms underlying CRC resistance to 5-FU and may offer potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Qingjie Kang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaoyu Hu
- Chongqing Medical University, Chongqing, 400016, China
| | - Zhenzhou Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaolong Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Song Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
173
|
Greene M, Gohil S, Camardo M, Ozbay AB, Limburg P, Lovelace J. Adherence to mt-sDNA testing for colorectal cancer screening among new users in a US Black population. Curr Med Res Opin 2025; 41:513-520. [PMID: 40029239 DOI: 10.1080/03007995.2025.2475074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
OBJECTIVE Colorectal cancer (CRC) poses significant mortality risks, particularly among Black individuals, who experience the highest CRC incidence and mortality rates in the United States. This study examined adherence to multi-target stool DNA (mt-sDNA) testing in this population. METHODS This retrospective cohort analysis used Exact Sciences Laboratories (ESL)-linked claims data from January 2017 to December 2023 on Black patients in the United States aged 45 and older. High-risk individuals, those with payers other than commercial plans, managed care organizations, Medicare Advantage, Medicaid, or Medicare, and individuals with mt-sDNA prescriptions outside the study period were excluded. Adherence was defined as the percentage of patients returning the test kit with valid results within 365 days of shipment. Logistic regression analysis was used to identify factors associated with adherence. RESULTS Among 434,951 patients included in the study, the overall adherence to mt-sDNA testing was 62.0% (N = 266,981), with a mean time to adherence of 27.6 days (SD = 44.17). Females, older adults (76+ years), and non-metropolitan residents had higher adherence than males, younger adults, and metropolitan patients (all p < 0.001), respectively. Patients with orders from GI specialists had higher adherence than other prescribing clinicians (NP/PA: OR = 0.39, OB/GYN: OR = 0.54, Other: OR = 0.38, PCP: OR = 0.50; all p < 0.001). Digital outreach, especially SMS and email combination, was also associated with higher adherence (OR = 1.25, p < 0.001). CONCLUSIONS This large, national study found a 62.0% adherence rate to mt-sDNA testing among Black individuals. Higher adherence was associated with being female, older age, non-metropolitan residence, and digital outreach. While the findings highlight the promise of mt-sDNA, further research is needed to explore its full potential in improving CRC screening adherence across different demographic groups.
Collapse
Affiliation(s)
| | - Shrey Gohil
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | - Mark Camardo
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | | | - Paul Limburg
- Exact Sciences Corporation, Madison, Wisconsin, USA
| | - Jerry Lovelace
- Nebraska Department of Correctional Services, Lincoln, Nebraska, USA
| |
Collapse
|
174
|
Ao W, Wang N, Chen X, Wu S, Mao G, Hu J, Han X, Deng S. Multiparametric MRI-Based Deep Learning Models for Preoperative Prediction of Tumor Deposits in Rectal Cancer and Prognostic Outcome. Acad Radiol 2025; 32:1451-1464. [PMID: 39438175 DOI: 10.1016/j.acra.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/28/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate the predictive value of a deep learning model based on multiparametric MRI (mpMRI) for tumor deposit (TD) in rectal cancer (RC) patients and to analyze their prognosis. MATERIALS AND METHODS Data from 529 RC patients who underwent radical surgery at two centers were retrospectively collected. 379 patients from center one were randomly divided into a training set (n = 265) and an internal validation (invad) set (n = 114) in a 7:3 ratio. 150 patients from center two were included in the external validation (exvad) set. Univariate and multivariate analyses were performed to identify independent clinical predictors and to construct a clinical model. Preoperative mpMRI images were utilized to extract deep features through the ResNet-101 model. Following feature selection, a deep learning model was developed. A nomogram was created by combining the clinical model with the deep learning model. The clinical applicability of each model was assessed using ROC curves, decision curve analysis (DCA), clinical impact curves (CIC), and deLong test. Kaplan-Meier survival analysis was conducted to evaluate prognostic outcome among patients. RESULTS Among the 529 patients, 142 (26.8%) were TD positive. In the training set, clinical model was constructed based on clinical independent predictors (cT and cN). 30 deep features were selected to calculate the deep learning radscore (DLRS) and develop the deep learning (DL) model. The AUC values for the clinical model were 0.724, 0.836, and 0.763 in the training set, invad set, and exvad set, respectively. The AUC values for the DL model were 0.903, 0.853, and 0.874, respectively. The nomogram achieved higher AUC values of 0.925, 0.919, and 0.9, respectively. The DeLong test indicated that the predictive performance of the nomogram was superior to both the DL model and the clinical model in training and invad sets. Kaplan-Meier survival analysis showed that both the DL model and the nomogram effectively stratified patients into high-risk and low-risk groups for 3-year DFS (p < 0.05). CONCLUSION The nomogram, which integrates mpMRI-based deep radiomic features and clinical characteristics, effectively predicts preoperative TD status in RC. Both the DL model and the nomogram can effectively stratify patients' 3-year DFS risk.
Collapse
Affiliation(s)
- Weiqun Ao
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (W.A., G.M., S.D.)
| | - Neng Wang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China (N.W., S.W.)
| | - Xu Chen
- Hangzhou Dianzi University Zhuoyue Honors College, Hangzhou, Zhejiang Province, China (X.C.)
| | - Sikai Wu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China (N.W., S.W.)
| | - Guoqun Mao
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (W.A., G.M., S.D.)
| | - Jinwen Hu
- Department of Radiology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China (J.H.)
| | - Xiaoyu Han
- Department of Pathology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (X.H.)
| | - Shuitang Deng
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (W.A., G.M., S.D.).
| |
Collapse
|
175
|
Zhang L, Qian J, Zhang X, Lv Y, Zhao J, Wang S, Xu H. Dual‐Responsive PPy‐AIPH@LA Nanoplatform for Synergistic Photothermal and Thermodynamic Therapy of Colorectal Cancer. ChemistrySelect 2025; 10. [DOI: 10.1002/slct.202500263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
AbstractIn this study, we introduce a novel nanoplatform, polypyrrole (PPy)‐2,2′‐Azobis[2‐(2‐imidazolin‐2‐yl)propane] dihydrochloride (AIPH)@lauric acid (LA) (PPy‐AIPH@LA) nanoparticles (NPs), designed to overcome these limitations through synergistic photothermal therapy (PTT) and photodynamic therapy (PDT). This dual‐responsive system incorporates PPy for efficient photothermal conversion, AIPH for thermos‐responsive and oxygen‐independent free radical generation, and LA as a thermally responsive encapsulation layer. The LA coating melts upon 808 nm near‐infrared laser irradiation, releasing AIPH and free radicals to enable precise spatiotemporal activation of therapeutic effects. PPy‐AIPH@LA demonstrates exceptional photothermal conversion efficiency (55.74%) and generates sufficient radicals to enhance PDT efficacy, even in hypoxic tumor microenvironments. In vitro studies revealed concentration‐dependent tumor cell ablation and inhibition of migration, while in vivo experiments showed that the combined PTT‐PDT treatment achieved an impressive 90.7% tumor growth inhibition rate in a mouse colon cancer cells CT‐26 murine model, with no significant systemic toxicity. Molecular analyses further revealed modulations in pathways associated with tumor metabolism, apoptosis, and immune escape, highlighting the comprehensive therapeutic potential of this nanoplatform. These findings underscore the potential of PPy‐AIPH@LA as a safe, effective, and minimally invasive nanotherapeutic platform for combating CRC and other solid tumors.
Collapse
Affiliation(s)
- Liang Zhang
- School of Materials and Chemistry University of Shanghai for Science and Technology No. 516 Jungong Road Shanghai 200093 P. R. China
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Jiahao Qian
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Xinyuan Zhang
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Yanwei Lv
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Jiulong Zhao
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| | - Shige Wang
- School of Materials and Chemistry University of Shanghai for Science and Technology No. 516 Jungong Road Shanghai 200093 P. R. China
| | - Hao Xu
- Department of Gastroenterology Changhai Hospital Naval Medical University No. 168 Changhai Road Shanghai 200433 P. R. China
| |
Collapse
|
176
|
Relander P, Rauhaniemi E, Löyttyniemi E, Salminen K, Carpelan A, Koffert J. First local results of the Finnish FIT-based colorectal cancer screening program - high yield, low complications. Scand J Gastroenterol 2025; 60:219-224. [PMID: 39893517 DOI: 10.1080/00365521.2025.2458062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The aim of screening for colorectal cancer (CRC) is to find the cancer in its early stages, thereby improving the prognosis of cancer patients by preventing cancer-related deaths. In Finland, the national CRC screening program was initiated in 2022, with fecal immunochemical test (FIT) being the primary screening test. The FIT-threshold used was 25 µg hemoglobin/g feces. The aim of this retrospective study was to evaluate the results of the first screening round that was implemented by the wellbeing services county of Southwest Finland. MATERIALS AND METHODS Participants were screened for CRC between March 1st, 2022 and April 14th, 2023. Participants aged 60-70 years had their health records scrutinized retrospectively. RESULTS Out of 36 397 FIT-invitees 23 388 (64%) returned a FIT-sample. 1407 (6%) subjects gave a FIT-positive stool sample of which 1118 (79%) attended the recommended screening colonoscopy. A total of 63 (6%) CRCs were found. 31 (49%) CRCs were classified as early stage I tumors, 12 (19%) of which were solely suitable for endoscopic treatment. Endoscopically removable adenomas were detected in 709 (63%) of the colonoscopies, which resulted in a recommendation of a 3-year follow-up colonoscopy for 427 (38%) cases. There were 3 (0.27%) acute polypectomy related complications and 5 (0.45%) late post-colonoscopy complications. CONCLUSIONS This is the first study to show the prevalence of CRC amongst participants of the newly implemented Finnish national CRC-screening program. Nearly half of the patients with CRC were diagnosed in the early stage. The adenoma detection rate was high.
Collapse
Affiliation(s)
- Pyry Relander
- University of Turku and Turku University Hospital, Abdominal Center-Gastroenterology Turku, Turku, Finland
| | - Elli Rauhaniemi
- University of Turku and Turku University Hospital, Abdominal Center-Gastroenterology Turku, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Kimmo Salminen
- University of Turku and Turku University Hospital, Abdominal Center-Gastroenterology Turku, Turku, Finland
| | - Anu Carpelan
- Abdominal Center-Gastrointestinal surgery Turku, University of Turku and Turku University Hospital, Turku, Finland
| | - Jukka Koffert
- University of Turku and Turku University Hospital, Abdominal Center-Gastroenterology Turku, Turku, Finland
| |
Collapse
|
177
|
Alptekin HM, Küçükkaya B. Level of Information and Awareness About Colorectal Cancer and Its Screening Program in Türkiye: A Community-Based Cross-Sectional Study. Public Health Nurs 2025; 42:869-879. [PMID: 39715079 DOI: 10.1111/phn.13519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
OBJECTIVE Colorectal cancer (CRC) has high mortality if it is detected in advanced stages, persons can lower their risk of CRC by adopting a healthy lifestyle that may lead to prevention. Screening is the most powerful public health tool to reduce mortality, considering the challenges of making substantial lifestyle changes or implementing widespread primary prevention strategies to lower the risk of CRC. This study aims to find out the levels of information and awareness about CRC and its screening program in individuals living in Türkiye. DESIGN This study was a community-based cross-sectional. SAMPLE This study was performed from November 2019 to November 2022 with a web-based survey circulated through social media and communication platforms. The research was conducted with 4146 participants. MEASUREMENTS The "Survey Form" was used in the data collection. Descriptive statistics and logistic regression were used in the data analysis. RESULTS The study found that 43.3% of participants had information about CRC and its screening program. Among all participants, 94.7% had never undergone CRC screening, and 55.6% were aware of a CRC awareness month. The study also revealed that participants who were knowledgeable about CRC screening were more likely to have a family history of cancer, a personal history of cancer, prior awareness of CRC, and previous information about the disease. Additionally, they were more likely to have undergone CRC screening before and to believe that CRC screening is necessary. CONCLUSION It was found that the majority of participants had no information on CRC and its screening program and never underwent CRC screening before. More studies should be performed to find out the factors negatively affecting the participation of individuals in the CRC screening program.
Collapse
Affiliation(s)
- Hatice Merve Alptekin
- Nursing Department, Division of Surgical Nursing, Faculty of Health Sciences, Kocaeli University, Kocaeli, Turkey
| | - Burcu Küçükkaya
- Nursing Department, Division of Gynecology and Obstetrics Nursing, Faculty of Health Sciences, Bartın University, Bartın, Turkey
| |
Collapse
|
178
|
Lee SO, Joo SH, Cho SS, Yoon G, Choi YH, Park JW, Weon KY, Shim JH. Licochalcone D Exerts Antitumor Activity in Human Colorectal Cancer Cells by Inducing ROS Generation and Phosphorylating JNK and p38 MAPK. Biomol Ther (Seoul) 2025; 33:344-354. [PMID: 39933827 PMCID: PMC11893492 DOI: 10.4062/biomolther.2024.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/12/2024] [Accepted: 10/04/2024] [Indexed: 02/13/2025] Open
Abstract
Anticancer activities of Licochalcone D (LCD) in human colorectal cancer (CRC) cells HCT116 and oxaliplatin-resistant HCT116 (HCT116-OxR) were determined. Cell viability assay and soft agar assay were used to analyze antiproliferative activity of LCD. Flow cytometry was performed to determine effects of LCD on apoptosis, cell cycle distribution, reactive oxygen species (ROS), mitochondrial membrane potential (MMP) dysfunction, and multi-caspase activity in CRC cells. Western blot analysis was used to monitor levels of proteins involved in cell cycle and apoptosis signaling pathways. LCD suppressed the growth and anchorageindependent colony formation of both HCT116 and HCT116-OxR cells. Cell cycle analysis by flow cytometry indicated that LCD induced cell cycle arrest and increased cells in sub-G1 phase. In parallel with the antiproliferative effect of LCD, LCD up-regulated levels of p21 and p27 while downregulating cyclin B1 and cdc2. In addition, phosphorylation levels of JNK and p38 mitogen-activated protein kinase (MAPK) were increased by LCD. Inhibition of these kinases somehow prevented the antiproliferative effect of LCD. Moreover, LCD increased ROS and deregulated mitochondrial membrane potential, leading to the activation of multiple caspases. An ROS scavenger N-acetyl-cysteine (NAC) or pan-caspase inhibitor Z-VAD-FMK prevented the antiproliferative effect of LCD, supporting that ROS generation and caspase activation mediated LCD-induced apoptosis in CRC cells. In conclusion, LCD exerted antitumor activity in CRC cells by inducing ROS generation and phosphorylation of JNK and p38 MAPK. These results support that LCD could be further developed as a chemotherapeutic agent for treating CRC.
Collapse
Affiliation(s)
- Seung-On Lee
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Sang Hoon Joo
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Kwon-Yeon Weon
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| |
Collapse
|
179
|
Shi Y, Zhao J, Xu S, Zhu H, Wang Y, Zhao B, Sun Z, He S, Hou X. A sodium alginate-based injectable hydrogel system for locoregional treatment of colorectal cancer by eliciting pyroptosis and apoptosis. Int J Biol Macromol 2025; 294:139345. [PMID: 39746425 DOI: 10.1016/j.ijbiomac.2024.139345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/18/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Effective delivery of sufficient doxorubicin (DOX) molecules in tumors is hindered by the complex biological barriers. Herein, a DOX-loaded sodium alginate-based injectable hydrogel (DOX@MHB-conj-SAgel) was designed by the Michael addition reactions between the sulfydryl in cross-linkers and the double bonds in a derivative of sodium alginate. The DOX@MHB-conj-SAgel was administrated to CT26 tumor-bearing mice via peritumoral injection for locoregional treatment of colorectal cancer by inducing apoptosis and pyroptosis. The released DOX molecules were localized within tumors, resulting in pronounced anti-tumor effects as evidenced by reduced tumor volumes, less tumor weight , higher inhibition rate, and diminished Ki67 staining . Meanwhile, DOX triggered pyroptosis in CT26 cells through the activation of caspase-3 to cleave gasdermin E, promoting potent anti-tumor immunity. The pyroptosis in CT26 cells was verified by characteristic cellular changes, including cell swelling, membrane blebbing, and cell rupture. Additionally, the immunogenicity of CT26 cells triggered by pyroptosis was demonstrated by the immunofluorescence imaging of GSDME, CD3, CD4, CD8 T cells, and CD31 in tumor sections. The hydrogel-based locoregional therapy represents a novel platform that combines the pyroptosis and apoptosis of DOX, thereby enhancing the therapeutic efficacy against colorectal cancer.
Collapse
Affiliation(s)
- Yongli Shi
- College of pharmacy, Xinxiang Medical University, 453003, Xinxiang, PR China.
| | - Jingya Zhao
- College of pharmacy, Xinxiang Medical University, 453003, Xinxiang, PR China
| | - Suyue Xu
- College of pharmacy, Xinxiang Medical University, 453003, Xinxiang, PR China
| | - Huiqing Zhu
- College of pharmacy, Xinxiang Medical University, 453003, Xinxiang, PR China
| | - Yuxin Wang
- College of pharmacy, Xinxiang Medical University, 453003, Xinxiang, PR China
| | - Bingqian Zhao
- Basic medicine college, Xinxiang Medical University, 453003, Xinxiang, PR China
| | - Zeyu Sun
- First clinical college, Xinxiang Medical University, 453003, Xinxiang, PR China
| | - Sisi He
- Department of Oncology, the Second Affiliated Hospital of Zunyi Medical University, 563000, Guizhou, PR China.
| | - Xueyan Hou
- College of pharmacy, Xinxiang Medical University, 453003, Xinxiang, PR China; Pingyuan Laboratory, 453007, Xinxiang, Henan, PR China.
| |
Collapse
|
180
|
Tan S, Li S, Xia L, Jiang X, Ren Z, Peng Q, Peng M, Yang W, Xu X, Oyang L, Shen M, Wang J, Li H, Wu N, Tang Y, Liao Q, Lin J, Zhou Y. Long non‑coding RNA ABHD11‑AS1 inhibits colorectal cancer progression through interacting with EGFR to suppress the EGFR/ERK signaling pathway. Int J Oncol 2025; 66:20. [PMID: 39950321 PMCID: PMC11844336 DOI: 10.3892/ijo.2025.5726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/15/2025] [Indexed: 02/23/2025] Open
Abstract
Long non‑coding (lnc)RNAs participate in colorectal cancer (CRC) occurrence and progression. The present study aimed to investigate whether lncRNA ABHD11‑AS1 regulates malignant biological behavior of CRC cells. Bioinformatic analysis, reverse transcription‑quantitative PCR and in situ hybridization revealed that ABHD11‑AS1 expression was decreased in CRC samples and associated with an unfavorable prognosis. ABHD11‑AS1 overexpression significantly decreased proliferation, migration and invasion of CRC cells, whereas ABHD11‑AS1 inhibition had the opposite effects. ABHD11‑AS1 interacted with EGFR to inhibit EGFR phosphorylation and attenuate EGFR/ERK signaling, which in turn suppressed the malignant biological behavior of CRC cells. The tumor suppressor function of ABHD11‑AS1 was attenuated by the EGFR agonist NSC228155. Finally, resveratrol (RSV) inhibited CRC cell proliferation, migration and invasion, which may be associated with RSV‑induced decrease in SPT6 homolog, histone chaperone and transcription elongation factor protein expression and increase in ABHD11‑AS1 transcript levels. ABHD11‑AS1 inhibited the phosphorylation of EGFR and decreased EGFR/ERK signaling by interacting with EGFR, thereby delaying the progression of CRC. The ABHD11‑AS1/EGFR/ERK axis may be a novel therapeutic target for preventing CRC progression.
Collapse
Affiliation(s)
- Shiming Tan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Shizhen Li
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Longzheng Xia
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Xianjie Jiang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Zongyao Ren
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Qiu Peng
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Mingjing Peng
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Wenjuan Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
| | - Xuemeng Xu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Linda Oyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Mengzhou Shen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Jiewen Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Haofan Li
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
| | - Nayiyuan Wu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Yanyan Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Qianjin Liao
- Department of Oncology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China
| | - Jinguan Lin
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Yujuan Zhou
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
181
|
Hosseini-Carroll P, Dominitz JA, Jacobson BC, May FP, Issaka RB, Schmitt CM, Adams TL, Boston IJ, Bucobo JC, Day L, Khaykis IB, Marino MJ, Rex DK, Sun E, Wynter JA, Christie JA. American Society for Gastrointestinal Endoscopy Colorectal Cancer Screening Project National Summit. Gastrointest Endosc 2025; 101:511-519. [PMID: 40024634 DOI: 10.1016/j.gie.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 03/04/2025]
Affiliation(s)
| | - Jason A Dominitz
- Veterans Health Administration and University of Washington, Seattle, Washington, USA
| | | | - Folasade P May
- David Geffen School of Medicine/UCLA and VA Greater LA Healthcare System, Los Angeles, California, USA
| | - Rachel B Issaka
- Fred Hutchison Cancer Center and University of Washington, Seattle, Washington, USA
| | | | | | - Iman J Boston
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Lukejohn Day
- University of California San Francisco, San Francisco, California, USA
| | | | - Mark J Marino
- University of Mississippi Medical Center and Baptist GI, Jackson, Mississippi, USA; Metropolitan Gastroenterology Associates, Metairie, Louisiana, USA
| | - Douglas K Rex
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Edward Sun
- Peconic Bay Medical Center, Northwell Health, East Setauket, New York, USA
| | | | | |
Collapse
|
182
|
Planey AM, Wong S, Planey DA, Winata F, Ko MJ. Longer travel times to acute hospitals are associated with lower likelihood of cancer screening receipt among rural-dwelling adults in the U.S. South. Cancer Causes Control 2025; 36:297-308. [PMID: 39576391 DOI: 10.1007/s10552-024-01940-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/09/2024] [Indexed: 03/22/2025]
Abstract
PURPOSE Given rural hospitals' role in providing outpatient services, we examined the association between travel burdens and receipt of cancer screening among rural-dwelling adults in the U.S. South region. METHODS First, we estimated network travel times and distances to access the nearest and second nearest acute care hospital from each rural census tract in the U.S. South. After appending the Centers for Disease Control's PLACES dataset, we fitted generalized linear mixed models. RESULTS Longer distances to the second nearest hospital are negatively associated with breast, colorectal, and cervical cancer screening receipt among eligible rural-dwelling adults. Rural-dwelling women in counties with 1 closure had reduced likelihood of breast cancer screening. Residence in a partial- or whole-county Health Professional Shortage Area (HPSA) was negatively associated with cancer screening receipt. Specialist (OB/GYN and gastroenterologist) supply was positively associated with receipt of cancer screening. Uninsurance was positively associated with cervical and breast cancer screening receipt. Medicaid expansion was associated with increased breast and cervical cancer screening. CONCLUSIONS Rural residents in partial-county primary care HPSAs had the lowest rates of breast, cervical, and colorectal cancer screening, compared with whole-county HPSAs and non-shortage areas. These residents also faced the greatest distances to their nearest and second nearest hospital. This is notable because rural residents in the South face greater travel burdens for cancer care compared with residents in other regions. Finally, the positive association between uninsurance and breast and cervical cancer screening may reflect the CDC's National Breast and Cervical Cancer Early Detection Program's effectiveness.
Collapse
Affiliation(s)
- Arrianna Marie Planey
- Department of Health Policy and Management, Gillings School of Global Public Health, University of North Carolina, McGavran-Greenberg, CB #1105C, Chapel Hill, NC, 27599-7411, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Sandy Wong
- Department of Geography, The Ohio State University, Columbus, OH, USA
| | - Donald A Planey
- Department of City and Regional Planning, University of North Carolina, Chapel Hill, NC, USA
| | - Fikriyah Winata
- Department of Geography, Texas A&M University, College Station, TX, USA
| | - Michelle J Ko
- Department of Public Health Sciences, School of Medicine, University of California, Davis, CA, USA
| |
Collapse
|
183
|
Zhang C, Tao X, Pan J, Huang L, Dong Z, Lin J, Su H, Zhu Y, Du H, Xiao B, Chen M, Wu L, Yu H. The Effect of Computer-Aided Device on Adenoma Detection Rate in Different Implement Scenarios: A Real-World Study. J Gastroenterol Hepatol 2025; 40:692-705. [PMID: 39663912 DOI: 10.1111/jgh.16847] [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: 10/05/2024] [Revised: 11/11/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Several recent studies have found that the efficacy of computer-aided polyp detection (CADe) on the adenoma detection rate (ADR) diminished in real-world settings. The role of unmeasured factors in AI-human interaction, such as monitor approaches, remains unknown. This study aimed to validate the effectiveness of CADe in the real world and assess the impact of monitor approaches. METHODS A retrospective propensity score-matched cohort study was conducted using routine data from a tertiary endoscopy center in China before and after the implementation of CADe. Four propensity score-matched cohorts were established: Cohort 1: pre-CADe matched with dual-monitor CADe-assisted group; Cohort 2: dual-monitor CADe-assisted with single-monitor CADe-assisted group; Cohort 3: pre-CADe with single-monitor CADe-assisted group; and Cohort 4: pre-CADe with CADe period. ADR was set as the primary outcome. RESULTS There were 5390, 6083, and 6131 eligible patients in the pre-CADe group, dual-monitor group, and single-monitor group, respectively. In the matched analysis, results indicated that regardless of the monitor setup, CADe-assisted groups showed a trend of increased ADR compared with the pre-CADe period (CADe period: OR 1.141, 95% CI 1.047-1.243; p = 0.003; dual-monitor: OR 1.178, 95% CI 1.069-1.299, p = 0.001; single-monitor: OR 1.094, 95% CI 0.998-1.200, p = 0.056). Moreover, no significant difference between different monitor approaches was observed, although dual-monitor setup showed an increasing tendency on ADR compared with single-monitor setup (OR 1.069, 95% CI 0.985-1.161, p = 0.109). CONCLUSION CADe shows great potential to improve ADR during colonoscopy in the real world. Meanwhile, changes in monitor setup do not significantly impact the assistance capability of CADe. Further research dedicated to evaluating the unmeasured elements in the AI-clinician hybrid for better implementation of CADe would be beneficial.
Collapse
Affiliation(s)
- Chenxia Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiao Tao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, Zhejiang Province, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang Province, China
| | - Li Huang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zehua Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiejun Lin
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, Zhejiang Province, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang Province, China
| | - Huang Su
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, Zhejiang Province, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang Province, China
| | - Yijie Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongliu Du
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bing Xiao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingkai Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lianlian Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
184
|
Biswas S, Lee JE, Xie G, Masclef L, Ren Z, Côté J, Affar EB, Ge K, Kutateladze TG. Colorectal cancer hot spot mutations attenuate the ASXL-MLL4 interaction. J Biol Chem 2025; 301:108333. [PMID: 39984049 PMCID: PMC11957774 DOI: 10.1016/j.jbc.2025.108333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025] Open
Abstract
Human additional sex combs like (ASXL) proteins are involved in the maintenance of both transcriptional activation and repression through their ability to bind multiple chromatin regulators, including two tumor suppressors: deubiquitinase BAP1 and methyltransferase MLL4 (KMT2D). The ASXL genes are often altered in colorectal cancer (CRC), and ASXL1 is one of the four hub genes related to the pathogenesis of CRC. Here, we show that MLL4 and BAP1 interdependently target specific genomic regions and positively or negatively regulate expression of a subset of genes in the human colon carcinoma HCT116 cells. MLL4 and BAP1 colocalize on a subset of enhancers and promoters in an interdependent manner. Genomic distribution of BAP1 in CRC cells differs from that in ESCs, with substantially more BAP1 binding sites identified on enhancers and promoters in HCT116 cells. MLL4 occupancy on MLL4+ BAP1+ genomic regions depends on functional ASXLs that interact with both MLL4 and BAP1, and CRC-relevant mutations attenuate the formation of the MLL4-ASXL complex. Mutational analysis and binding assays identified CRC hot spot mutations in ASXLs. Our findings suggest that alterations in the genomic distribution of the MLL4-ASXL-BAP1 axis and CRC hot spot mutations in ASXLs perturb normal transcriptional programs and may trigger pathogenic events in colon cancer.
Collapse
Affiliation(s)
- Soumi Biswas
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ji-Eun Lee
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Guojia Xie
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Louis Masclef
- Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada
| | - Zhizhong Ren
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Jacques Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Quebec City, Québec, Canada
| | - El Bachir Affar
- Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada; Department of Medicine, University of Montréal, Montréal, Québec, Canada
| | - Kai Ge
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA.
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado, USA.
| |
Collapse
|
185
|
Huang J, Chan VCW, Chen M, Liew JJM, Liu X, Zhong C, Lin J, Hang J, Zhong CC, Yuan J, Xu W, Withers M, Chan AT, Wong MCS. Revisiting the starting age of colorectal cancer screening for the average-risk Asian population: a cost-effectiveness analysis. Gastrointest Endosc 2025:S0016-5107(25)00141-5. [PMID: 40024296 DOI: 10.1016/j.gie.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND AND AIMS One of the most prevalent and fatal tumors, colorectal cancer (CRC), has a significant impact on the use of healthcare services. Although Hong Kong's CRC screening program has been successful, it does not prioritize preventing early-onset CRC in people under age 50 years. This study aimed to assess the cost-effectiveness of different starting ages for CRC screening among an Asian population. METHODS We conducted a simulation study involving 100,000 individuals in Hong Kong who were screened using either the fecal immunochemical test (FIT) or colonoscopy as primary screening methods at ages 40, 45, and 50 until age 75. The performance of different strategies was evaluated based on life-years gained, and cost-effectiveness was measured using the incremental cost-effectiveness ratio (ICER). RESULTS The ICERs for initiating FIT screening at age 50, screening starting at age 45, and screening starting at age 40 were U.S. dollars (USD) 53,262, USD 67,892, and USD 86,554, respectively. For colonoscopy, the ICERs for initiating screening at ages 50, 45, and 40 were USD 267,669, USD 312,848, and USD 372,090, respectively. Overall, the FIT strategy was found to be less costly. At 70%, 80%, and 90% compliance rates, the FIT at age 45 gained 2135, 2296, and 2438 life-years, respectively, whereas colonoscopy at age 45 gained 2725, 2798, and 2855 life-years, respectively. With increased compliance rates, the FIT could save a similar number of life-years as colonoscopy with lower cost. CONCLUSIONS Initiating CRC screening at age 45 using the FIT in Hong Kong was determined to be a well-balanced and cost-effective strategy. This approach demonstrated a cost advantage over starting screening at age 40 and resulted in more lives saved compared with screening at age 50.
Collapse
Affiliation(s)
- Junjie Huang
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR
| | - Victor C W Chan
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR
| | - Mingtao Chen
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR
| | - Jamie Jie Mei Liew
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR
| | - Xianjing Liu
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Chaoying Zhong
- Department of Electrical Engineering and Automation, Guangdong Ocean University, Guangdong, China
| | - Jianli Lin
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Junjie Hang
- Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Guangdong, China
| | - Claire Chenwen Zhong
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR.
| | - Jinqiu Yuan
- Clinical Research Center, Big Data Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Guangdong, China
| | - Wanghong Xu
- The School of Public Health, Fudan University, Shanghai, China
| | - Mellissa Withers
- Department of Population and Health Sciences, Institute for Global Health, University of Southern California, Los Angeles, California, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - Martin C S Wong
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Centre for Health Education and Health Promotion, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR; The School of Public Health, Fudan University, Shanghai, China; The School of Public Health, Peking University, Beijing, China; The School of Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Guangdong, China.
| |
Collapse
|
186
|
Wang L, Lin X, Qi GY, Chen GP. Relationship between expression of lncRNA HOTAIR and miR-302b-3p and circulating tumor cells in colon cancer and their value for prognostic prediction. Shijie Huaren Xiaohua Zazhi 2025; 33:140-147. [DOI: 10.11569/wcjd.v33.i2.140] [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: 12/03/2024] [Revised: 01/24/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND In recent years, studies have reported that the abnormal expression of the long non-coding RNA (lncRNA) HOTAIR and microRNA (miRNA)-302b-3p is associated with the occurrence and development of malignant tumors. However, there is limited research on their relationship with circulating tumor cells and their prognostic value, which warrants further investigation.
AIM To investigate the relationship between the expression of HOTAIR and miR-302b-3p and circulating tumour cells in colon cancer, and to explore their value for prognostic prediction.
METHODS A total of 160 patients with colon cancer who underwent radical resection at the Traditional Chinese Medicine Hospital of Linhai City from May 2019 to May 2023 were selected as an observation group, and another 160 healthy volunteers who underwent physical examination at the same hospital's physical examination center during the same period were selected as a control group. The expression of HOTAIR and miR-302b-3p in the two groups was compared. The relationship between the expression of HOTAIR and miR-302b-3p and pathological characteristics was analyzed. The recurrence and metastasis of colon cancer patients were recorded, and the predictive value of HOTAIR and miR-302b-3p expression for the recurrence and metastasis of colon cancer was evaluated.
RESULTS The expression of serum HOTAIR in the observation group was higher than that of the control group, while the expression of miR-302b-3p was lower than that of the control group (P < 0.05). Multiple linear regression analysis showed that clinical stage, lymph node metastasis, and circulating tumor cells were all factors affecting the expression of serum HOTAIR and miR-302b-3p in colon cancer patients (P < 0.05). Among the 160 patients with colon cancer, 2 were lost to follow-up, and 28 experienced recurrence or metastasis. The expression of serum HOTAIR in patients with recurrence or metastasis was higher than that of patients without recurrence or metastasis, while the expression of miR-302b-3p was lower than that of patients without recurrence or metastasis (P < 0.05). The area under the curve (AUC) of serum HOTAIR and miR-302b-3p for predicting the risk of postoperative recurrence and metastasis in patients with colon cancer was 0.747 (0.672-0.813) and 0.785 (0.713-0.846), respectively. The optimal cutoff values were 3.14 and 0.48, respectively. The AUC of the combination of the two for predicting the risk of postoperative recurrence and metastasis was 0.880 (0.919-0.926), with a sensitivity and specificity of 85.71% and 75.38%, respectively. The predictive efficacy was superior to that of each indicator alone.
CONCLUSION The expression of serum HOTAIR is up-regulated and the expression of miR-302b-3p is down-regulated in patients with colon cancer, which is related to clinical stage, lymph node metastasis, and positive circulating tumor cells. Detecting the levels of both can help predict patient prognosis.
Collapse
Affiliation(s)
- Ling Wang
- Department of Gastroenterology, Linhai Hospital of Traditional Chinese Medicine, Taizhou 317000, Zhejiang Province, China
| | - Xia Lin
- Department of Pathology, Linhai First People's Hospital, Taizhou 317000, Zhejiang Province, China
| | - Guo-Yong Qi
- Department of Gastroenterology, Linhai Hospital of Traditional Chinese Medicine, Taizhou 317000, Zhejiang Province, China
| | - Gui-Ping Chen
- Department of Gastrointestinal Surgery, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou 310001, Zhejiang Province, China
| |
Collapse
|
187
|
Cheloff AZ, Kim L, Pochapin MB, Shaukat A, Popov V. Accuracy of Visual Estimation for Measuring Colonic Polyp Size: A Systematic Review and Meta-Analysis. Am J Gastroenterol 2025:00000434-990000000-01609. [PMID: 40019167 DOI: 10.14309/ajg.0000000000003391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Measurement of colorectal polyps is typically performed through visual estimation, which is prone to bias. Studies have evaluated the accuracy of visual estimation and utility of assistive tools, but results have been mixed. The aim of this study was to clarify the accuracy of visual estimation as a measurement tool and the benefits of artificial intelligence. METHODS MEDLINE and Embase were searched through October 2024. Extraction and quality assessment were performed independently by 2 authors. The primary outcome was the pooled absolute mean difference in size between visual estimation and control. Secondary outcomes included subgroup analysis of expert vs trainee status, accuracy of artificial intelligence, study origin (East vs West), comparator type, definition of accuracy, polyp size, direction of estimation, and image type. RESULTS Thirty-five studies with 42,964 polyp measurements were included in our analysis. All studies were of high quality, and there was no evidence of publication bias. The pooled absolute mean difference from comparator was 1.68 mm (confidence interval 1.21-2.15) with high variability explained by differences in the comparator, direction of estimation, image type, and size of the polyp. Overall accuracy was 60% with high variability as well, with increased accuracy with video displayed over photographs. Artificial intelligence improved accuracy with an odds ratio of 7.46. DISCUSSION Visual estimation is an inaccurate and imprecise way to measure colorectal polyps. Further research is needed to determine the impact on clinical outcomes related to colorectal cancer. Investment in new technology to aid in polyp measurement is an important next step.
Collapse
Affiliation(s)
- Abraham Z Cheloff
- Division of Gastroenterology and Hepatology, NYU Langone Health, New York, New York, USA
| | | | | | | | | |
Collapse
|
188
|
Sahoo K, Sundararajan V. IL-1β and associated molecules as prognostic biomarkers linked with immune cell infiltration in colorectal cancer: an integrated statistical and machine learning approach. Discov Oncol 2025; 16:252. [PMID: 40019680 PMCID: PMC11871282 DOI: 10.1007/s12672-025-01989-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
PURPOSE Colorectal cancer (CRC) is the third most common cancer globally, necessitating novel biomarkers for early diagnosis and treatment. This study proposes an efficient pipeline leveraging an integrated bioinformatics and machine learning framework to enhance the identification of diagnostic and prognostic biomarkers for CRC. METHODS A selection of methylated differentially expressed genes (MeDEGs) and features (genes) was made using both statistical and Machine learning (ML) approaches from publically available datasets. These genes were subjected to STRING network construction and hub genes estimation, separately. Also, essential miRNAs (micro-RNAs) and TFs (Transcription factors) as regulatory elements were revealed and findings were validated through scRNA-seq analysis, promoter methylation, gene expression levels correlated with pathological stage, and interaction with tumor-infiltrating immune cells. RESULTS Through an integrated analysis pipeline, we identified 27 hub genes, among which CTNNB1, GSK3B, IL-1β, MYC, PXDN, TP53, EGFR, SRC, COL1A1, and TGBF1 showed better diagnostic behaviour. Machine learning approach includes the development of K-Nearest Neighbors (KNN), Artificial Neural Networks (ANN), and Random Forest (RF) models using TCGA datasets, achieving an accuracy range between 99 and 100%. The Area Under the Curve (AUC) value for each model is 1.00, signifying good classification performance. The high expression of some diagnostic genes was associated with poor prognosis, concluding IL-1β as both a prognostic and diagnostic biomarker. Additionally, the NF-κB and microRNAs (miR-548d-3p, miR-548-ac) and TFs (NFκB and STAT5A) play a major role in the comprehensive regulatory network for CRC. Furthermore, hub genes such as IL-1β, TGFB1, and COL1A1 were significantly correlated with immune infiltrates, suggesting their potential role in CRC progression. CONCLUSION Overall, the elevated expression of IL-1β coupled with abnormal DNA methylation, and its consequent effect on the PI3K/Akt signaling pathway are relevant prognostic and therapeutic marker in CRC. Additional molecular candidates reveal insights into the epigenetic regulatory targets of CRC and their association with immune cell infiltration.
Collapse
Affiliation(s)
- Karishma Sahoo
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Vino Sundararajan
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
189
|
Hu M, Xu Y, Wang Y, Huang Z, Wang L, Zeng F, Qiu B, Liu Z, Yuan P, Wan Y, Ge S, Zhong D, Xiao S, Luo R, He J, Sun M, Zhuang X, Guo N, Cui C, Gao J, Zhou H, He X. Gut microbial-derived N-acetylmuramic acid alleviates colorectal cancer via the AKT1 pathway. Gut 2025:gutjnl-2024-332891. [PMID: 40015949 DOI: 10.1136/gutjnl-2024-332891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/15/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Gut microbial metabolites are recognised as critical effector molecules that influence the development of colorectal cancer (CRC). Peptidoglycan fragments (PGFs) produced by microbiota play a crucial role in maintaining intestinal homeostasis, but their role in CRC remains unclear. OBJECTIVE Here, we aimed to explore the potential contribution of PGFs in intestinal tumourigenesis. DESIGN The relative abundance of peptidoglycan synthase and hydrolase genes was assessed by metagenomic analysis. Specific PGFs in the faeces and serum of CRC patients were quantified using targeted mass spectrometry. The effects of PGF on intestinal tumourigenesis were systematically evaluated using various murine models of CRC and organoids derived from CRC patients. Downstream molecular targets were screened and evaluated using proteome microarray, transcriptome sequencing and rescue assays. RESULTS Metagenomic analysis across seven independent cohorts (n=1121) revealed a comprehensive reduction in peptidoglycan synthase gene relative abundance in CRC patients. Targeted mass spectrometry identified significant depletion of a specific PGF, N-acetylmuramic acid (NAM) in CRC patients, which decreased as tumours progressed (p<0.001). NAM significantly inhibits intestinal tumourigenesis in various models, including Apc Min/+, AOM/DSS-treated and MC38 tumour-bearing mice. Additionally, NAM inhibits the growth of patient-derived CRC organoids in a concentration-dependent manner. Mechanistically, NAM inhibits the activation of AKT1 by directly binding to it and blocking its phosphorylation, which is a partial mediator of NAM's anticancer effects. CONCLUSION The PGF NAM protects against intestinal tumourigenesis by targeting the AKT1 signalling pathway. NAM may serve as a novel potential preventive and therapeutic biomarker against CRC.
Collapse
Affiliation(s)
- Mengyao Hu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Xu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuqing Wang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhenhe Huang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Wang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Fanan Zeng
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bowen Qiu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zefeng Liu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peibo Yuan
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Wan
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuang Ge
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dian Zhong
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Siyu Xiao
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Rongrong Luo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaqi He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Meiling Sun
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoduan Zhuang
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Nannan Guo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunhui Cui
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Gao
- Department of Gastroenterology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaolong He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Southern Medical University, Guangzhou, Guangdong, China
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
190
|
Yi LL, Lao XJ, Lu LY, Zhu CL, Zhou DY, Li ST, Fan ML, Peng QL. Value of brief hematological characteristics in differentiating carcinoembryonic-antigen-negative colorectal cancer from benign colorectal diseases. World J Gastrointest Surg 2025; 17:101403. [DOI: 10.4240/wjgs.v17.i2.101403] [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: 09/13/2024] [Revised: 11/07/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most common malignancies worldwide, with a significant subset of patients exhibiting absence of carcinoembryonic-antigen (CEA) expression. The lack of effective diagnostic method for CEA-negative CRC prevents its early treatment.
AIM To identify potentially valuable biomarkers for identifying CEA-negative CRC, the hematological characteristics of patients with CEA-negative CRC was investigated.
METHODS In this retrospective analysis, 74 patients were included who had been pathologically confirmed to have CEA-negative CRC, along with 79 individuals diagnosed with benign colorectal conditions. The utility of various biomarkers was evaluated using analysis of the receiver operating characteristic (ROC) curve.
RESULTS Compared with patients with benign colorectal diseases, those with CEA-negative CRC had lower hemoglobin-to-red blood cell distribution width ratio (HRR) and lymphocyte-to-red blood cell distribution width ratio (LRR), and higher platelet-to-lymphocyte ratio (PLR) (P < 0.05). Correlation analysis showed that HRR was negatively correlated with T stage (r = -0.237), LRR was negatively correlated with T stage (r = -0.265) and distant metastasis (r = -0.321), and PLR was positively correlated with T stage (r = 0.251) (all P < 0.05). ROC analysis indicated that HRR outperformed LRR and PLR in identifying CEA-negative CRC. Combining HRR and PLR provided the highest area under the curve (area under the curve = 0.808; sensitivity = 82.43%; specificity = 68.35%) for distinguishing CEA-negative CRC from benign colorectal diseases.
CONCLUSION HRR, LRR, and PLR alone or in combination could be used to distinguish CEA-negative CRC from benign colorectal diseases.
Collapse
Affiliation(s)
- Li-Ling Yi
- Department of Clinical Laboratory, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, Guangxi Zhuang Autonomous Region, China
| | - Xian-Jun Lao
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Liu-Yi Lu
- Department of Clinical Laboratory, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, Guangxi Zhuang Autonomous Region, China
| | - Chun-Ling Zhu
- Department of Clinical Laboratory, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, Guangxi Zhuang Autonomous Region, China
| | - Dong-Yi Zhou
- Department of Clinical Laboratory, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, Guangxi Zhuang Autonomous Region, China
| | - Si-Ting Li
- Department of Clinical Laboratory, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, Guangxi Zhuang Autonomous Region, China
| | - Meng-Li Fan
- Department of Clinical Laboratory, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, Guangxi Zhuang Autonomous Region, China
| | - Qi-Liu Peng
- Department of Clinical Laboratory, Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530201, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
191
|
Xu L, Wang J, Peng Y, Wu C, Wang S, Zhang X, Liang C, Wan S, Yang C, Fu Q, Xia Y, Huang X, Xu L. Clinical comparative study of the modified superior mesenteric artery approach in total laparoscopic radical resection for right colon cancer - a single-center retrospective study. World J Surg Oncol 2025; 23:67. [PMID: 40016739 PMCID: PMC11869699 DOI: 10.1186/s12957-025-03725-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/21/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND To explore the safety and feasibility of the modified approach for accessing the superior mesenteric artery (SMA) in total laparoscopic radical resection for right colon cancer. METHODS This single-center retrospective study included 107 patients who underwent total laparoscopic radical resection of right colon cancer at The First Affiliated Hospital of Wannan Medical College between August 2022 and December 2023. 53 patients were in the modified SMA approach (modified group) and 54 patients were in the traditional SMA approach (control group). The control group and modified group underwent total laparoscopic radical resection of right colon cancer, and the following baseline and pathological characteristics of the two groups were compared: intraoperative condition, postoperative recovery, and postoperative complications. Our modified surgical method was to isolate the mesocolon using a cranial(the ligament of Treitz) -to- caudal(the pedicle of ileocolic) pathway and the orderly ligation of blood vessels in the SMA. RESULTS There was no statistically significant difference in the baseline characteristics or pathological data between the two groups. Compared with the traditional SMA approach, the modified SMA approach had a shorter surgical time(P < 0.001) and vascular dissection time (P < 0.001) and less intraoperative blood loss (P = 0.000). There was no statistically significant difference in the number of total harvested lymph nodes or positive harvested lymph nodes between the two groups of patients (P > 0.05); There was no statistically significant difference in postoperative hospital stay, time to first flatus, time to pull out drainage tube and drainage between the two groups of patients (P > 0.05), and there was no statistically significant difference in the incidence of complications between the two groups of patients (P > 0.05). CONCLUSION The modified SMA approach in totally laparoscopic radical resection for right colon cancer can shorten the surgical and vascular dissection time, reduce intraoperative bleeding and reduce the surgical difficulty and intraoperative risk of the SMA approach for right colon cancer. In clinical practice, its safety and feasibility are relatively high, and it is worth promoting. TRIAL REGISTRATION The study was approved by the Ethics Committee of The First Affiliated Hospital of Wannan Medical College and registered with the China Clinical Trials Registry (ChiCTR2300075919, Date of Registration:2023-09-19- retrospective registration) http://www.chictr.org.cn/index.aspx .
Collapse
Affiliation(s)
- Lishuai Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Jiawei Wang
- Department of General Surgery, Maanshan Maternal and Child Health Care Hospital, No.446 Jiashan Road, Ma'anshan, Anhui, 243000, China
| | - Yue Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Chengwei Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Song Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Xu Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Changming Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Senlin Wan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Cheng Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Qingsheng Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Yabin Xia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China
| | - Xiaoxu Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China.
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China.
| | - Li Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, No.2, Zheshan West Road, Wuhu, Anhui, Anhui, 241001, China.
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, Anhui, China.
| |
Collapse
|
192
|
Machlab S, Lorenzo-Zúñiga V, Pantaleon MA, Sábado F, Arieira C, Pérez Arellano E, Cotter J, Carral D, Turbí Disla C, Gorjão R, Esteban JM, Rodriguez S. Real-world effectiveness and safety of 1L polyethylene glycol and ascorbic acid for bowel preparation in patients aged 80 years or older. Endosc Int Open 2025; 13:a25259938. [PMID: 40018074 PMCID: PMC11866040 DOI: 10.1055/a-2525-9938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/15/2025] [Indexed: 03/01/2025] Open
Abstract
Background and study aims Clinical trials and real-world studies show a 1L polyethene glycol and ascorbic acid solution (1L PEG-ASC) to be an effective and safe bowel preparation for colonoscopy in the general population. Here, the effectiveness and safety of 1L PEG-ASC were evaluated in patients aged 80 years or older in a real-world setting. Patients and methods A post-hoc analysis of an observational, multicenter, retrospective study assessed the effectiveness and safety of 1L PEG-ASC on outpatients aged ≥ 80 years old undergoing colonoscopy at eight centers in Spain and Portugal. Cleansing quality was assessed using the Boston Bowel Preparation Scale, with overall scores ≥ 6 and all segmental scores ≥ 2 considered adequate colon cleansing, and overall scores ≥ 8 or 3 in the right colon considered high-quality cleansing. Cecal intubation rate, withdrawal time, polyp and adenoma detection rates (ADR), and adverse events (AEs) were also monitored. Results Data were analyzed from 423 patients aged ≥ 80 years; mean age 83.5 years (±3.2) and 49.2% males. The adequate colon cleansing success rate was 88.9%, with high-quality cleansing of the overall and right colon achieved in 54.1% and 46.1% of patients, respectively. Colonoscopy was complete in 94.1% of cases and the ADR was 51.3%. At least one AE was experienced by 4.5% of participants, the most frequent being mild dehydration (2.8%) and nausea (1.2%). Conclusions This post-hoc analysis confirms 1L PEG-ASC to be an effective and safe bowel cleansing preparation for patients aged 80 years or older in a real-world setting.
Collapse
Affiliation(s)
- Salvador Machlab
- Gastroenterology, Parc Taulí Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí I3PT, Sabadell, Spain
| | | | | | - Fernando Sábado
- Gastroenterology, Consorcio Hospitalario Provincial de Castelló, Castellón, Spain
| | - Cátia Arieira
- Gastroenterology, Hospital da Senhora da Oliveira, Guimarães, Portugal
| | | | - José Cotter
- Gastroenterology, Hospital da Senhora da Oliveira, Guimarães, Portugal
- School of Medicine, Universidade do Minho, Life and Health Sciences Research Institute (ICVS), Braga/Guimarães, Portugal
- Gastroenterology, ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - David Carral
- Gastroenterology, Hospital San Rafael, A Coruña, Spain
| | - Carmen Turbí Disla
- Medical Affairs, Norgine, Harefield, United Kingdom of Great Britain and Northern Ireland
| | - Ricardo Gorjão
- Gastroenterology, Hospital CUF Descobertas, Lisboa, Portugal
| | | | | |
Collapse
|
193
|
Du L, Min H, Meng Y, Zhang K, Wang L, Zhang Y, Sun P, Zhang W, Qi Y, Wu G. LRG1-Targeted Camptothecin Nanomicelles with Simultaneous Delivery of Olaparib for Enhanced Colorectal Cancer Chemotherapy. NANO LETTERS 2025; 25:3130-3140. [PMID: 39947227 DOI: 10.1021/acs.nanolett.4c05354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Camptothecin (CPT), an effective topoisomerase I inhibitor used in colorectal cancer chemotherapy, often faces limitations due to severe toxicities. Addressing this, we developed OCENM, a nanomedicine featuring the CPT-ET conjugate─comprising a cathepsin B-sensitive linker and CPT linked to the ET peptide targeting leucine-rich alpha-2-glycoprotein 1 (LRG1) and encapsulating Olaparib, a potent poly ADP-ribose polymerase (PARP) inhibitor. OCENM aims for precise CPT delivery to colorectal cancer sites overexpressing LRG1, while Olaparib disrupts compromised DNA repair pathways, enhancing DNA damage and promoting increased tumor cell apoptosis. Our results show OCENM accumulates preferentially in colorectal cancer models through LRG1 targeting and triggers synergistic tumor cell apoptosis through the dual action of enhanced DNA damage and inhibited repair mechanisms. This study not only highlights the potential of LRG1 as a strategic target for nanomedicine delivery but also underscores the therapeutic promise of combining CPT with Olaparib for colorectal cancer treatment.
Collapse
Affiliation(s)
- Lin Du
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Huan Min
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yongkang Meng
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Kejuan Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Longdi Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Yana Zhang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450003, China
| | - Peichun Sun
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Wei Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| | - Yingqiu Qi
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Gang Wu
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial Research Center for Precision Control Engineering of Digestive Tract Tumors, Zhengzhou 450003, China
| |
Collapse
|
194
|
Dai Z, Li T, Lai K, Wang X, Zhou P, Hu K, Zhou Y. Serum metabolic characteristics associated with the deterioration of colorectal adenomas. Sci Rep 2025; 15:6845. [PMID: 40000732 PMCID: PMC11861597 DOI: 10.1038/s41598-025-91444-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) can evolve from colorectal adenomas, which can be further classified into non-advanced adenomas (NAAs) and advanced adenomas (AAs) based on their clinical characteristics. Their prognoses are vastly different, with patients with NAAs having significantly lower recurrence and CRC-related mortality rates than those with AA or CRC. Although serum metabolomics has shown promise for the early diagnosis of CRC, the differences in serum metabolite composition between NAA and AA still need to be further elucidated. This study aimed to explore the mechanism of CRC occurrence and development based on the unique serum metabolic fingerprints of different stages of CRC and to discover a new non-invasive diagnostic method based on serum metabolomics. A clinical CRC progression cohort containing healthy control (NC; n = 40), NAA (n = 40), AA (n = 40), and CRC (n = 22) groups was constructed, and untargeted metabolomic analysis based on liquid chromatography/mass spectrometry was performed to analyze the serum metabolite characteristics of each group. A semi-quantitative analysis of intergroup metabolite differences was conducted, focusing on specific metabolites that differed in the NAA and AA groups. Finally, variable metabolites were selected based on least absolute shrinkage and selection operator (LASSO) regression analysis, and receiver operating characteristic curves were plotted to evaluate the efficacy of the serum metabolite-based diagnostic model in distinguishing NC/NAA populations from AA/CRC populations. Metabolomic analysis revealed significant differences in the composition of metabolites in the NC and NAA groups compared to the CRC group, whereas the serum metabolites of the AA group were similar to those of the CRC group. The levels of 33 metabolites were significantly different in the serum of AA/CRC patients compared to that of NAA patients, and their functions included glycerophospholipid, sphingolipid, and caffeine metabolism. LASSO regression analysis identified 57 differential metabolite variables between the NC/NAA and AA/CRC groups. The diagnostic model constructed using the random forest algorithm had the best discrimination effect, with areas under the curve of 1.000 (95% confidence interval [CI] 1.000-1.000) and 0.685 (95% CI 0.540-0.830) for the training and testing sets, respectively. The composition of serum metabolites is specific to the different stages of CRC development. The serum metabolite composition of patients with AAs was similar to that of patients with CRC. Auxiliary diagnostic measures based on serum metabolites have the potential to guide the follow-up and treatment of patients with adenoma.
Collapse
Affiliation(s)
- Ze Dai
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo, 315020, Zhejiang, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo Key Laboratory, Ningbo, 315020, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Tong Li
- Department of Colorectal-Anal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
| | - Kecong Lai
- Digestive Department, The Second People's Hospital of Beilun District, Ningbo, 315020, Zhejiang, China
| | - Xiaomei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo, 315020, Zhejiang, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo Key Laboratory, Ningbo, 315020, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Peng Zhou
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Kefeng Hu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo, 315020, Zhejiang, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo Key Laboratory, Ningbo, 315020, Zhejiang, China
| | - Yuping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China.
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo, 315020, Zhejiang, China.
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo Key Laboratory, Ningbo, 315020, Zhejiang, China.
| |
Collapse
|
195
|
Zhang X, Chen W, Wan S, Qu B, Liao F, Cheng D, Zhang Y, Ding Z, Yang Y, Yuan Q. Spatially Selective MicroRNA Imaging in Human Colorectal Cancer Tissues Using a Multivariate-Gated Signal Amplification Nanosensor. J Am Chem Soc 2025; 147:6679-6687. [PMID: 39933117 DOI: 10.1021/jacs.4c16001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
MicroRNA (miRNA) is involved in the genesis in viand development of colorectal cancer. The in vivo imaging of miRNA at the tumor sites is essential for understanding its role in colorectal cancer pathology and therapeutic target identification. However, achieving accurate imaging of miRNA at the tumor sites is hindered by the low abundance of miRNAs in tumor cells and nonspecific signal leakage in normal tissues. Here, we report a multivariate-gated catalytic hairpin assembly (CHA) nanosensor for the specific amplified imaging of microRNA-21 (miR-21) in human colorectal cancer tissues to reveal the underlying miR-21-associated molecular mechanism. The endogenous glutathione and exogenous near-infrared multivariate-gated design in combination with CHA probes improves the signal strength of target miR-21 and reduces the background interference. The nanosensor enables specific amplified imaging of miR-21 in vivo, and the signal-to-background ratios are 1.6-fold compared with traditional CHA methods. With the assistance of the designed nanosensor, we achieve the preliminary identification of tumor tissues and normal tissues from human clinical surgical resection samples. The overexpressed miR-21 is found to suppress the core mismatch repair recognition protein human mutS homologue 2 involved in DNA damage recognition and repair to inhibit the therapeutic efficacy of colorectal cancer. The strategy of probe design, which combines multivariate-gated activation methods with a signal amplification system, is applicable for accurate miRNA imaging and disease-relevant molecular mechanism research.
Collapse
Affiliation(s)
- Xiaoming Zhang
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Wenhui Chen
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Songlin Wan
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Bing Qu
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Fei Liao
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Di Cheng
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Yun Zhang
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Zhao Ding
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Yanbing Yang
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
| | - Quan Yuan
- College of Chemistry and Molecular Sciences, Department of Colorectal and Anal Surgery of Zhongnan Hospital of Wuhan University, Clinical Center of Intestinal and Colorectal Diseases of Hubei Province, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, School of Microelectronics, Wuhan University, Wuhan 430072, P. R. China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
196
|
Deng Y, Wang Y, Yang J, Luo X, Qiu J, Long R, Zhang C, Li J, Tang G, Chen L, Zuo J. Global, Regional, and National Disease Burden and Prediction Analysis of Colorectal Cancer Attributable to Tobacco, Alcohol, and Obesity From 1990 to 2030. Front Oncol 2025; 15:1524308. [PMID: 40078186 PMCID: PMC11896864 DOI: 10.3389/fonc.2025.1524308] [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: 11/07/2024] [Accepted: 01/15/2025] [Indexed: 03/14/2025] Open
Abstract
Background Colorectal cancer (CRC) ranks among the highest in incidence and mortality rates globally. A significant portion of Colorectal cancer cases and deaths can be attributed to modifiable risk factors, with smoking, alcohol use, and high body mass index (BMI) being the three most prominent. However, the impact of these risk factors on Colorectal cancer across regions, genders, and age groups remains insufficiently characterized. Methods Utilizing data from the Global Burden of Disease (GBD) study 2019, restrictive cubic splines (RCS) and quantile regression analyses are applied to explore the relationship between the Socio-Demographic Index (SDI) and ASMR or ASDR. Additionally, gender differences, changes across different SDI levels, and age group trends in smoking, alcohol use, and high BMI over the 30-year period are analyzed. The Bayesian age-period-cohort (BAPC) model is employed to predict mortality trends from 2020 to 2030, aiming to explore the epidemiological and sociodemographic transitions in the Colorectal cancer disease burden attributed to smoking, alcohol use, and high BMI. Results In 2019, the number of colorectal cancer deaths globally attributable to risk factors as smoking, alcohol consumption, and obesity increased to 142,931, 52,495, and 85,882 cases respectively, collectively accounting for approximately one-third of all Colorectal cancer-related deaths. Notably, there is an upward trend in early-onset Colorectal cancer mortality associated with these factors. Discussion To reduce the burden of Colorectal cancer, it is recommended to enhance health education, promote smoking cessation and alcohol moderation, and increase the coverage and participation in Colorectal cancer screening, which are crucial for lowering Colorectal cancer mortality rates. These findings are vital for the development of public health policies and intervention measures to reduce the global disease burden. They provide guidance for Colorectal cancer prevention across different regions, genders, and age groups worldwide.
Collapse
Affiliation(s)
- Yuqi Deng
- Department of Health, Inspection and Quarantine, School of Public Health, University of South China, Hengyang, China
| | - Yajie Wang
- Department of Health, Inspection and Quarantine, School of Public Health, University of South China, Hengyang, China
| | - Jinsai Yang
- Computer Institute, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Xinyu Luo
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jieya Qiu
- Transformation Research Lab, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Rou Long
- Transformation Research Lab, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Chaohui Zhang
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiale Li
- Computer Institute, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Guiyang Tang
- Transformation Research Lab, Hengyang Medical School, University of South China, Hunan, Hengyang, China
| | - Lili Chen
- Department of Health, Inspection and Quarantine, School of Public Health, University of South China, Hengyang, China
| | - Jianhong Zuo
- Department of Health, Inspection and Quarantine, School of Public Health, University of South China, Hengyang, China
- Computer Institute, Hengyang Medical School, University of South China, Hunan, Hengyang, China
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Transformation Research Lab, Hengyang Medical School, University of South China, Hunan, Hengyang, China
- The Third Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
197
|
Tang WW, Battistone B, Bauer KM, Weis AM, Barba C, Fadlullah MZH, Ghazaryan A, Tran VB, Lee SH, Agir ZB, Nelson MC, Victor ES, Thibeaux A, Hernandez C, Tantalla J, Tan AC, Rao D, Williams M, Drummond MJ, Beswick EJ, Round JL, Ekiz HA, Voth WP, O'Connell RM. A microRNA-regulated transcriptional state defines intratumoral CD8 + T cells that respond to immunotherapy. Cell Rep 2025; 44:115301. [PMID: 39951377 PMCID: PMC11924119 DOI: 10.1016/j.celrep.2025.115301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/24/2024] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
The rising incidence of advanced-stage colorectal cancer (CRC) and poor survival outcomes necessitate new and effective therapies. Immune checkpoint inhibitors (ICIs), specifically anti-PD-1 therapy, show promise, yet clinical determinants of a positive response are suboptimal. Here, we identify microRNA-155 (miR-155) as necessary for CD8+ T cell-infiltrated tumors through an unbiased in vivo CRISPR-Cas9 screen identifying functional tumor antigen-specific CD8+ T cell-expressed microRNAs. T cell miR-155 is required for anti-PD-1 responses and for a vital intratumor CD8+ T cell differentiation cascade by repressing Ship-1, inhibiting Tcf-1 and stemness, and subsequently enhancing Cxcr6 expression, anti-tumor immunity, and effector functions. Based on an underlying miR-155-dependent CD8+ T cell transcriptional profile, we identify a gene signature that predicts ICI responses across 12 diverse cancers. Together, our findings support a model whereby miR-155 serves as a central regulator of CD8+ T cell-dependent cancer immunity and ICI responses that may be leveraged for future therapeutics.
Collapse
Affiliation(s)
- William W Tang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ben Battistone
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Kaylyn M Bauer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Allison M Weis
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Cindy Barba
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Muhammad Zaki Hidayatullah Fadlullah
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Arevik Ghazaryan
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Van B Tran
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Soh-Hyun Lee
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Z Busra Agir
- Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey
| | - Morgan C Nelson
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Emmanuel Stephen Victor
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Amber Thibeaux
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Colton Hernandez
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Jacob Tantalla
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Aik C Tan
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Dinesh Rao
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew Williams
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Micah J Drummond
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT 84108, USA
| | - Ellen J Beswick
- Division of Digestive Disease and Nutrition, Department of Internal Medicine, University of Kentucky, Lexington, KY 40508, USA
| | - June L Round
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - H Atakan Ekiz
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Biology and Genetics, İzmir Institute of Technology, İzmir, Turkey; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Warren P Voth
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Ryan M O'Connell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
198
|
Haynes J, Manogaran P. Mechanisms and Strategies to Overcome Drug Resistance in Colorectal Cancer. Int J Mol Sci 2025; 26:1988. [PMID: 40076613 PMCID: PMC11901061 DOI: 10.3390/ijms26051988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related mortality worldwide, with a significant impact on public health. Current treatment options include surgery, chemotherapy, radiotherapy, molecular-targeted therapy, and immunotherapy. Despite advancements in these therapeutic modalities, resistance remains a significant challenge, often leading to treatment failure, poor progression-free survival, and cancer recurrence. Mechanisms of resistance in CRC are multifaceted, involving genetic mutations, epigenetic alterations, tumor heterogeneity, and the tumor microenvironment. Understanding these mechanisms at the molecular level is crucial for identifying novel therapeutic targets and developing strategies to overcome resistance. This review provides an overview of the diverse mechanisms driving drug resistance in sporadic CRC and discusses strategies currently under investigation to counteract this resistance. Several promising strategies are being explored, including targeting drug transport, key signaling pathways, DNA damage response, cell death pathways, epigenetic modifications, cancer stem cells, and the tumor microenvironment. The integration of emerging therapeutic approaches that target resistance mechanisms aims to enhance the efficacy of current CRC treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Jennifer Haynes
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive, Huntington, WV 25701, USA;
| | | |
Collapse
|
199
|
Pang NQ, Lau J, Tan KK. Health-Related Quality of Life After Colorectal Cancer Surgery in Older Patients Compared to the Young: A Prospective Multicentre Observational Study. J Appl Gerontol 2025:7334648251321560. [PMID: 39993183 DOI: 10.1177/07334648251321560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025] Open
Abstract
Introduction: The impact of colorectal surgery on older adults' (≥65 years) health-related quality of life (HRQoL) is rarely reported. Methods: This prospective multi-institutional cohort study involving 217 participants investigated factors affecting older colorectal cancer (CRC) patients' HRQoL following surgery with the EORTC QLQ-C30 and the HADS. Results: The trend of the global QoL and individual subscales show an overall dip in QoL from baseline to pre-discharge, before slowly rising until the 6-month mark. Baseline score of individual outcome measures was the only constant predictor of the outcome measure over time. Conclusion: Age was not found to be a significant predictor of HRQoL in older adults undergoing colorectal surgery, while baseline scores of individual outcome measures consistently predicted post-operative HRQoL scores. Interventions should be targeted at specific subgroups of older adults scheduled for colorectal surgery, such as those with lower baseline scores.
Collapse
Affiliation(s)
- Ning Qi Pang
- Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore
- National University Centre for Organ Transplantation (NUCOT), National University Hospital, National University Health System, Singapore
| | - Jerrald Lau
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ker-Kan Tan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Colorectal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore
| |
Collapse
|
200
|
Sheng Z, Luo S, Huang L, Deng YN, Zhang N, Luo Y, Zhao X, Chen Y, Li Q, Dai R, Liang S. SENP1-mediated deSUMOylation of YBX1 promotes colorectal cancer development through the SENP1-YBX1-AKT signaling axis. Oncogene 2025:10.1038/s41388-025-03302-6. [PMID: 39988696 DOI: 10.1038/s41388-025-03302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/18/2025] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
Aberrant SUMOylation is associated with the progression of colorectal cancer (CRC). The SUMO-specific protease 1 (SENP1)-induced deSUMOylation of different target substrates plays specific roles in CRC. In this study, we dissected the SENP1-interacting protein complex by employing protein co-immunoprecipitation enrichment in combination with His6-SUMO1T95K-tagging mass spectrometry (MS) identification, and identified YBX1 as a novel substrate of SENP1. Further studies revealed that SENP1 interacted with YBX1 and consequently catalyzed YBX1 deSUMOylation at K26 residue preferentially. SENP1-mediated deSUMOylation enhanced the pro-tumor activity of YBX1 protein by maintaining the interaction of YBX1 with DDX5, thereby activating AKT phosphorylation signaling and promoting CRC tumor growth. Indeed, SENP1 knockdown elevated YBX1 SUMOylation and disrupted the interaction between YBX1 with DDX5, which significantly inhibited CRC cell proliferation and migration. And overexpression of K26 mutant YBX1 (YBX1-K26R) protein rescued the anti-tumor effect of SENP1 depletion compared with the wild-type YBX1 (YBX1-WT). Moreover, the expression levels of SENP1 and YBX1 were both increased in CRC specimens and associated with poor outcomes in CRC patients. In general, our studies have revealed SENP1-mediated YBX1 protein deSUMOylation promotes CRC progression through the activation of AKT phosphorylation signaling, suggesting that targeting the SENP1-YBX1-AKT signaling axis is a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Zenghua Sheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Shu Luo
- Department of Medical Oncology, Suining First People's Hospital, Suining, Sichuan, PR China
| | - Lan Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Ya-Nan Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Nan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yinheng Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Xinyu Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Ying Chen
- Department of Medical Oncology, Suining First People's Hospital, Suining, Sichuan, PR China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Ruiwu Dai
- Department of General Surgery, General Hospital of Western Theater Command, Chengdu, PR China
| | - Shufang Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|