101
|
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; 44:1361-1374. [PMID: 39988696 PMCID: PMC12052596 DOI: 10.1038/s41388-025-03302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
102
|
Bi J, Huang Y, Hu W, Liu Y. NOP2-Mediated m5C Methylation Modification of LMNB2 mRNA Facilitates Colorectal Cancer Progression. Cancer Med 2025; 14:e70970. [PMID: 40366008 PMCID: PMC12076344 DOI: 10.1002/cam4.70970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/19/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer-related mortality globally, yet current therapies exhibit suboptimal efficacy with limited prognostic improvement. RNA 5-methylcytosine (m5C), a posttranscriptional modification, has been implicated in tumorigenesis and progression across malignancies. In our previous study, the m5C methyltransferase NOP2 has been shown to promote proliferation, migration, and invasion of CRC cells, however, the underlying mechanism is still elusive. METHODS An integrated multi-omics strategy was employed, combining transcriptomic sequencing, RNA immunoprecipitation sequencing (RIP-seq), and methylated RNA immunoprecipitation sequencing (MeRIP-seq) to explore NOP2-regulated downstream genes mediating CRC progression via m5C methylation. Functional validation included in vitro and in vivo assays to assess tumor growth and metastasis. Rescue experiments were performed by overexpressing LMNB2 in NOP2-silenced CRC cells. RESULTS NOP2-dependent m5C modification of LMNB2 mRNA enhanced its stability, leading to elevated LMNB2 protein levels. This mechanism drove CRC tumor growth and metastasis both in vitro and in vivo. Overexpression of LMNB2 effectively rescued the suppressed malignant phenotypes induced by NOP2 knockdown, confirming LMNB2 as a critical downstream effector. CONCLUSION NOP2 catalyzes the m5C modification of LMNB2 mRNA to facilitate its stability, which contributes to the elevated LMNB2 protein level and CRC progression, suggesting the potential of NOP2 as a therapeutic target in the development of novel CRC treatment.
Collapse
Affiliation(s)
- Jinling Bi
- Department of OncologyThe Second People's Hospital of HefeiHefeiChina
- Department of OncologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yong Huang
- Department of OncologyThe Second People's Hospital of HefeiHefeiChina
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| | - Yulong Liu
- Department of OncologyThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| |
Collapse
|
103
|
Raunkilde L, Andersen RF, Thomsen CB, Hansen TF, Jensen LH. A prospective study of methylated ctDNA in patients undergoing treatment for liver metastases from colorectal cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109586. [PMID: 39847896 DOI: 10.1016/j.ejso.2025.109586] [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: 06/21/2024] [Revised: 12/12/2024] [Accepted: 01/06/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Decision regarding local treatment of colorectal liver metastases (CRLM) is a multidisciplinary assessment, and liver intervention should be performed when the metastases are deemed resectable. There is no standard biomarker to aid neither this decision nor the postoperative treatment decisions. The present prospective, observational study aimed to investigate the potential clinical utility of a combined tumor-specific and organ-specific methylated circulating DNA assay in the perioperative setting of CRLM. MATERIAL AND METHODS The study included 56 cases with CRLM. Blood samples were drawn preoperatively and postoperatively. Multiplex methylation analysis of the markers NPY, KANK1, and GAL3ST3 (meth-ctDNA) was performed using droplet digital PCR. RESULTS The assay detected preoperative and postoperative meth-ctDNA in 37 % and 46 % of patients, respectively. Patients with negative preoperative meth-ctDNA had a longer median PFS compared to those with positive preoperative meth-ctDNA (HR = 2.2, 95 % CI 1.2-3.9, p < 0.01). In a multivariate analysis, preoperative negative meth-ctDNA was identified as a strong independent predictor of PFS (HR = 3.3, 95 % CI 1.5-7.2, p < 0.01). Similarly, patients with negative postoperative meth-ctDNA had longer median PFS (HR = 3.0, 95 % CI = 1.6-5.6, p < 0.001) and OS (HR = 4.1, 95 % CI 1.9-9.1, p < 0.001) compared to those with positive postoperative meth-ctDNA. CONCLUSION Preoperative meth-ctDNA may serve as an important biomarker to inform the multidisciplinary assessment and treatment planning of CRLM. Negative meth-ctDNA may indicate the optimal timing for liver intervention, whereas positive meth-ctDNA may indicate initiation or re-orientation of chemotherapy, or immediate local intervention. Our results confirm postoperative negative meth-ctDNA as a strong prognostic marker of survival.
Collapse
Affiliation(s)
- Louise Raunkilde
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
| | - Rikke Fredslund Andersen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Clinical Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark
| | - Caroline Brenner Thomsen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Lars Henrik Jensen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100, Vejle, Denmark; Danish Colorectal Cancer Center South, Vejle Hospital, Beriderbakken 4, 7100, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| |
Collapse
|
104
|
McHugh KE, Pai RK, Grant RC, Gallinger S, Davison J, Ma C, Pai RK. Claudin 18.2 Expression in 1404 Digestive Tract Adenocarcinomas Including 1175 Colorectal Carcinomas: Distinct Colorectal Carcinoma Subtypes Are Claudin 18.2 Positive. Mod Pathol 2025; 38:100712. [PMID: 39826799 PMCID: PMC12103281 DOI: 10.1016/j.modpat.2025.100712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Claudin 18.2 (CLDN18.2) immunohistochemical (IHC) expression can be used to select patients with gastric/gastroesophageal junction adenocarcinomas for zolbetuximab (IMAB362) therapy, zolbetuximab (IMAB362) being a monoclonal antibody targeting CLDN18.2. The aim of this study was to correlate IHC expression of CLDN18.2 with clinicopathologic and molecular features in a large series of digestive tract cancers. IHC for CLDN18.2 was performed on tissue microarrays from 1404 adenocarcinomas including 155 gastric/gastroesophageal, 74 pancreatic ductal, and 1175 colorectal (576 in the initial test cohort; 599 in the subsequent validation cohort), and correlated with HER2 and mismatch repair (MMR) status. Cases were scored as CLDN18.2 positive or negative, with positivity defined as moderate-to-strong membranous staining in ≥75% of tumor cells. CLDN18.2 expression was correlated with clinicopathologic and molecular features for all colorectal adenocarcinomas. CLDN18.2 was positive in 39% (61/155) of gastric/gastroesophageal adenocarcinomas, 38% (28/74) of pancreatic ductal adenocarcinomas, and 3.4% (40/1175) of colorectal adenocarcinomas (P < .001). For gastric/gastroesophageal and pancreatic ductal adenocarcinoma, there was no correlation between CLDN18.2 expression and either HER2 or MMR status. In contrast, CLDN18.2-positive colorectal adenocarcinomas had distinct clinicopathologic and molecular features. CLDN18.2-positive colorectal adenocarcinomas were more frequently proximally located and were more often MMR deficient and BRAF V600E positive (all with P < .05). Quantitative pathologic analysis using the digital pathology biomarker QuantCRC (Aiforia) demonstrated marked differences in histologic features between CLDN18.2-positive and CLDN18.2-negative colorectal adenocarcinomas, with CLDN18.2-positive tumors having an increased tumor:stroma ratio and %mucin but decreased %immature stroma in both the test and validation cohorts (all with P < .05). In conclusion, CLDN18.2-positive colorectal adenocarcinomas are frequently MMR deficient and BRAF V600E mutated, and demonstrate distinct histologic features. Future studies addressing the efficacy of zolbetuximab therapy in this subset of colorectal cancers are needed.
Collapse
Affiliation(s)
- Kelsey E McHugh
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Robert C Grant
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Jon Davison
- Department of Pathology, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Changqing Ma
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Center and the University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
105
|
Guo Q, Wang K, Yu C, Yao L, Han Z. Retrospective Analysis of Therapeutic Efficacy and Prognosis of Neoadjuvant Chemotherapy in Patients With Mid to Low Locally Advanced Rectal Cancer. Am J Ther 2025; 32:e294-e297. [PMID: 40338691 DOI: 10.1097/mjt.0000000000001812] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Affiliation(s)
- Qinping Guo
- Department of General Surgery, Shanxi Bethune Hospital, Taiyuan, China
- General Surgery Department, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Kang Wang
- Department of General Surgery, Shanxi Bethune Hospital, Taiyuan, China
- General Surgery Department, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Chao Yu
- Department of General Surgery, Shanxi Bethune Hospital, Taiyuan, China
- General Surgery Department, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Li Yao
- Department of General Surgery, Beijing China-Japan Friendship Hospital, Beijing, China
| | - Zhenguo Han
- Department of General Surgery, Shanxi Bethune Hospital, Taiyuan, China
- General Surgery Department, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
106
|
Cohen D, Rogers C, Gabre J, Dionigi B. The Young: Early-Onset Colon Cancer. Clin Colon Rectal Surg 2025; 38:173-178. [PMID: 40292000 PMCID: PMC12020630 DOI: 10.1055/s-0044-1787883] [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: 04/30/2025]
Abstract
Early-onset colorectal cancer (EO-CRC), characterized by diagnosis before the age of 50 years, has emerged as a significant healthcare challenge with increasing global incidence. While traditional risk factors such as family history and inherited syndromes contribute to EO-CRC, a substantial proportion of cases remain sporadic, necessitating further investigation into additional etiological factors. Molecular studies reveal heterogeneity in EO-CRC, with distinct mutational profiles compared to late-onset CRC. Clinical management presents unique considerations, including challenges in early detection and treatment strategies tailored to younger patients. Addressing EO-CRC requires a multidisciplinary approach integrating epidemiological, molecular, and clinical insights to improve prevention, diagnosis, and therapeutic outcomes. Emerging research avenues explore novel biomarkers and therapeutic targets, offering promise for advancing understanding and management of this disease in contemporary oncology practice.
Collapse
Affiliation(s)
- David Cohen
- Department of Surgery, New York-Presbyterian/Columbia University Irving Medical Center, New York, New York
| | - Caitlin Rogers
- Herbert Irving Comprehensive Cancer Research Center, Columbia University, New York, New York
| | - Joel Gabre
- Herbert Irving Comprehensive Cancer Research Center, Columbia University, New York, New York
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Beatrice Dionigi
- Division of Colon and Rectal Surgery, New York-Presbyterian/Columbia University Medical Center, New York, New York
| |
Collapse
|
107
|
Paquier Z, Dhont J, Guiot T, Levillain H, Critchi G, Saude Conde R, Sclafani F, Flamen P, Reynaert N, Woff E, Hendlisz A. Development and Validation of an 18F-Fluorodeoxyglucose Positron Emission Tomography-Computed Tomography-Based Imaging Score to Predict 12-Week Life Expectancy in Advanced Chemorefractory Colorectal Cancer. JCO Clin Cancer Inform 2025; 9:e2400207. [PMID: 40305759 PMCID: PMC12058364 DOI: 10.1200/cci-24-00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/17/2024] [Accepted: 02/28/2025] [Indexed: 05/02/2025] Open
Abstract
PURPOSE Managing chemorefractory metastatic colorectal cancer (mCRC) requires a meticulous equilibrium between the efficacy and toxicity of interventions, a task compounded by the constrained life expectancy of the patient. While existing prognostic tools, such as the Colon Life nomogram, primarily focus on general patient conditions or a single diagnostic modality, they do not fully integrate the potential predictive value of multimodal data. This study aims to develop and validate an Imaging Score, integrating clinical and imaging features derived from whole-body 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography-computed tomography (PET-CT), predicting death probability within 12 weeks from treatment initiation for refractory disease. MATERIALS AND METHODS The development cohort comprises 254 patients from three clinical trials. Nine clinical variables and six imaging variables were assessed. After optimal subset selection through recursive Feature Elimination with cross-validation, a support vector classifier-trained machine learning model generated the Imaging Score. Validation was performed on a real-life patient cohort (n = 74). Model performance was assessed on discrimination (Harrell C-index) and calibration. RESULTS Final prognostic features included whole-body metabolically active tumor volume, Eastern Cooperative Oncology Group performance status, visceral fat density, number of metastatic sites, body mass index, maximum standardized distance, and months since diagnosis. The Imaging Score demonstrated robust discriminative ability in both the development (C-index, 0.797) and validation (C-index, 0.714) sets, outperforming the Colon Life nomogram that tended to overestimate 12-week mortality. CONCLUSION The Imaging Score, integrating 18F-FDG PET-CT imaging with clinical parameters, is an effective prognostic tool for patients with chemorefractory mCRC. This combination of imaging biomarkers with clinical factors improves discrimination, enhancing its potential for clinical decision making, patient stratification for chemorefractory treatments, and trial eligibility.
Collapse
Affiliation(s)
- Zelda Paquier
- Radiophysics and MRI Physics Laboratory, Université Libre De Bruxelles (ULB), Brussels, Belgium
- Department of Medical Physics, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Jennifer Dhont
- Radiophysics and MRI Physics Laboratory, Université Libre De Bruxelles (ULB), Brussels, Belgium
- Department of Medical Physics, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Thomas Guiot
- Radiophysics and MRI Physics Laboratory, Université Libre De Bruxelles (ULB), Brussels, Belgium
- Department of Medical Physics, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Hugo Levillain
- Radiophysics and MRI Physics Laboratory, Université Libre De Bruxelles (ULB), Brussels, Belgium
- Department of Medical Physics, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Gabriela Critchi
- Radiophysics and MRI Physics Laboratory, Université Libre De Bruxelles (ULB), Brussels, Belgium
| | - Rita Saude Conde
- Department of Gastrointestinal Oncology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Francesco Sclafani
- Department of Gastrointestinal Oncology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Patrick Flamen
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Nick Reynaert
- Radiophysics and MRI Physics Laboratory, Université Libre De Bruxelles (ULB), Brussels, Belgium
- Department of Medical Physics, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Erwin Woff
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Alain Hendlisz
- Department of Gastrointestinal Oncology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
108
|
van Stigt BJ, Lansdorp‐Vogelaar I, Spaander MCW, van Vuuren AJ, Dekker E, van Kemenade FJ, Nagtegaal ID, van Leerdam ME, Toes‐Zoutendijk E. Interval cancer risk after the upper age limit of screening has been reached: Informing risk stratification in FIT-based colorectal cancer screening. Int J Cancer 2025; 156:1783-1790. [PMID: 39697047 PMCID: PMC11887016 DOI: 10.1002/ijc.35294] [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: 07/11/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Upper age limits are currently fixed for all fecal immunochemical test (FIT)-based colorectal cancer (CRC) screening programs. A risk-stratified upper age limit may be beneficial. Therefore, we assessed differences in interval CRC risk among individuals who had reached the upper age limit of screening (75 years). Individuals with a negative FIT (<47 μg Hb/g feces) in the final round of the Dutch CRC screening program were selected from the national screening database and linked to the national cancer registry to identify CRCs diagnosed within 24 months (interval CRCs). Survival analyses assessed whether sex and last fecal hemoglobin (f-Hb) concentration were associated with interval CRC risk. A multivariable logistic regression assessed whether sex, last f-Hb concentration and screening round were associated with stage distribution (early vs. late). Last f-Hb concentrations were considered detectable when they were >0 μg Hb/g feces. Among 305,761 individuals with a complete follow-up (24 months), 661 were diagnosed with interval CRC (21.6 per 10,000 negative FITs). Individuals with detectable f-Hb (15%) were 5 times more likely to be diagnosed with interval CRC than those without (HR 4.87, 95%CI: 4.19-5.65). Moreover, their cancers were more often detected at a late stage compared to individuals without detectable f-Hb (OR 1.45, 95%CI: 1.06-2.01). Our results show that interval CRC risk among individuals aged ≥75 differs substantially by last f-Hb concentration, indicating a uniform age to stop screening is suboptimal. Future research, taking into account multiple screening rounds and FIT results, should determine the optimal risk-stratified screening strategy.
Collapse
Affiliation(s)
- Brenda J. van Stigt
- Department of Public HealthErasmus MC University Medical CentreRotterdamThe Netherlands
| | | | - Manon C. W. Spaander
- Department of Gastroenterology and HepatologyErasmus MC University Medical CentreRotterdamThe Netherlands
| | - Anneke J. van Vuuren
- Department of Gastroenterology and HepatologyErasmus MC University Medical CentreRotterdamThe Netherlands
| | - Evelien Dekker
- Department of Gastroenterology and HepatologyAmsterdam University Medical CenterAmsterdamThe Netherlands
| | | | - Iris D. Nagtegaal
- Department of PathologyRadboud University Medical CentreNijmegenThe Netherlands
| | - Monique E. van Leerdam
- Department of Gastrointestinal OncologyAntoni van Leeuwenhoek HospitalAmsterdamThe Netherlands
- Department of Gastroenterology and HepatologyLeiden University Medical CentreLeidenThe Netherlands
| | | |
Collapse
|
109
|
Wankhede D, Halama N, Kloor M, Edelmann D, Brenner H, Hoffmeister M. Prognostic Value of CD8+ T Cells at the Invasive Margin Is Comparable to the Immune Score in Nonmetastatic Colorectal Cancer: A Prospective Multicentric Cohort Study. Clin Cancer Res 2025; 31:1711-1718. [PMID: 40293274 DOI: 10.1158/1078-0432.ccr-24-3275] [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: 10/02/2024] [Revised: 11/23/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE The Immunoscore predicts colorectal cancer prognosis but faces adoption barriers because of complex software and reimbursement issues. This study used open-source methods to explore a simplified prognostic model in nonmetastatic colorectal cancer by focusing on single T-cell markers. EXPERIMENTAL DESIGN A multicentric prospective cohort study in patients with nonmetastatic colorectal cancer assessed CD3+ and CD8+ tumor-infiltrating lymphocytes (TIL) in the invasive margin (IM) and tumor core (TC) using QuPath. An immune cell score (ICS), based on TIL densities (CD3-IM, CD8-IM, CD3-TC, and CD8-TC), was calculated similarly to the Immunoscore. A split sample approach (70:30) estimated adjusted HRs for cancer-specific survival in training and validation sets. Classification and regression tree analysis identified the most prognostic TIL, and its model was compared with an ICS model for performance (Brier score) and discrimination (concordance probability estimate). RESULTS Over a median follow-up of 9.0 years, 203 colorectal cancer-specific deaths occurred among 1,260 patients. Classification and regression tree-selected CD8-IM was the most prognostic TIL at a cutoff of 231 cells/mm2. Patients with high CD8-IM had better cancer-specific survival than low CD8-IM in both training (HR 0.58, 95% confidence interval, 0.40-0.84) and validation sets (HR 0.35, 95% confidence interval, 0.21-0.60). Brier scores of CD8-IM and ICS survival models were comparable in both training and validation cohorts, whereas the survival discrimination of CD8-IM slightly outperformed the ICS in the validation set (concordance probability estimate: CD8-IM: 0.748; ICS: 0.730). CONCLUSIONS CD8-IM alone provided prognostic information comparable with the ICS. Simplified, cost-effective TIL assessments could improve clinical translation and guide adjuvant therapy in early-stage colorectal cancer.
Collapse
Affiliation(s)
- Durgesh Wankhede
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Department of Translational Immunotherapy (D240), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz Institute for Translational Oncology, Mainz, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Dominic Edelmann
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
110
|
Gao H, Zheng S, Liang J, Wang Y, Chen L, Li H, Chen Y, Zhang F, Shi H, Han A. m6A-induced DEAD-box RNA helicase 21 enhances lipid metabolism via 3‑hydroxy-3-methylglutaryl-CoA synthases 1 in colorectal cancer. Transl Oncol 2025; 55:102373. [PMID: 40127603 PMCID: PMC11979938 DOI: 10.1016/j.tranon.2025.102373] [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: 11/06/2024] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Altered lipid metabolism is a well-known hallmark of cancer. However, the underlying mechanisms of altered lipid metabolism in colorectal cancer (CRC) progression requires further investigation. Previously we have revealed that DEAD-box RNA helicase 21 (DDX21) promotes CRC metastasis via liquid-liquid phase separation. In this study, we identify DDX21 as a novel regulator of lipid metabolism in CRC. METHODS In vitro and in vivo assays illustrated the biological role of DDX21 and YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) in CRC lipid metabolism and progression. Bioinformatics analysis, ChIP, meRIP, RIP, RNA stability assay and dual-luciferase reporter assay were applied to explore the underlying molecular mechanisms. The expression levels and prognostic role of YTHDF1/DDX21/HMGCS1 axis in CRC patients were analyzed by immunohistochemical staining and Kaplan-Meier plotter. RESULTS DDX21 enhanced CRC progression via promoting lipid metabolism. Mechanistically, YTHDF1 enhanced DDX21 mRNA stability by recognizing its m6A-modified sites. DDX21 further binded to 3‑hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1) promoter region and directly activated HMGCS1 transcription. Moreover, our clinical data showed that a simultaneously high expression of YTHDF1, DDX21 and HMGCS1 predicted an unfavorable overall survival in CRC patients. CONCLUSIONS Our study demonstrates that the YTHDF1/DDX21/HMGCS1 axis promotes CRC progression via regulating lipid metabolism and DDX21 might be a promising target for CRC therapy.
Collapse
Affiliation(s)
- Huabin Gao
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuai Zheng
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiangtao Liang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuting Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Lin Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hui Li
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yongyu Chen
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Fenfen Zhang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Huijuan Shi
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Anjia Han
- Department of Pathology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
111
|
Thomas CE, Peters U. Genomic landscape of cancer in racially and ethnically diverse populations. Nat Rev Genet 2025; 26:336-349. [PMID: 39609636 DOI: 10.1038/s41576-024-00796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/30/2024]
Abstract
Cancer incidence and mortality rates can vary widely among different racial and ethnic groups, attributed to a complex interplay of genetic, environmental and social factors. Recently, substantial progress has been made in investigating hereditary genetic risk factors and in characterizing tumour genomes. However, most research has been conducted in individuals of European ancestries and, increasingly, in individuals of Asian ancestries. The study of germline and somatic genetics in cancer across racial and ethnic groups using omics technologies offers opportunities to identify similarities and differences in both heritable traits and the molecular features of cancer genomes. An improved understanding of population-specific cancer genomics, as well as translation of those findings across populations, will help reduce cancer disparities and ensure that personalized medicine and public health approaches are equitable across racial and ethnic groups.
Collapse
Affiliation(s)
- Claire E Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
112
|
Nguyen D, Palmquist S, Hwang K, Ma J, Salem U, Sun J, Wang X, Son JB, Ernst R, Wei P, Kaur H, Stanietzky N. T2-weighted imaging of rectal cancer using a 3D fast spin echo sequence with and without deep learning reconstruction: A reader study. J Appl Clin Med Phys 2025; 26:e70031. [PMID: 39976552 PMCID: PMC12059301 DOI: 10.1002/acm2.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/15/2024] [Accepted: 12/23/2024] [Indexed: 05/10/2025] Open
Abstract
PURPOSE To compare image quality and clinical utility of a T2-weighted (T2W) 3-dimensional (3D) fast spin echo (FSE) sequence using deep learning reconstruction (DLR) versus conventional reconstruction for rectal magnetic resonance imaging (MRI). METHODS The study included 50 patients with rectal cancer who underwent rectal MRI consecutively between July 7, 2020 and January 20, 2021 using a T2W 3D FSE sequence with DLR and conventional reconstruction. Three radiologists reviewed the two sets of images, scoring overall SNR, motion artifacts, and overall image quality on a 3-point scale and indicating clinical preference for DLR or conventional reconstruction based on those three criteria as well as image characterization of bowel wall layer definition, tumor invasion of muscularis propria, residual disease, fibrosis, nodal margin, and extramural venous invasion. RESULTS Image quality was rated as moderate or good for both DLR and conventional reconstruction for most cases. DLR was preferred over conventional reconstruction in all of the categories except for bowel wall layer definition. CONCLUSION Both conventional reconstruction and DLR provide acceptable image quality for T2W 3D FSE imaging of rectal cancer. DLR was clinically preferred over conventional reconstruction in almost all categories.
Collapse
Affiliation(s)
- Dan Nguyen
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Sarah Palmquist
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Ken‐Pin Hwang
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Jingfei Ma
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Usama Salem
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Jia Sun
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Xinzeng Wang
- GE HealthCareGlobal MR Applications and WorkflowHoustonTexasUSA
| | - Jong Bum Son
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Randy Ernst
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Peng Wei
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Harmeet Kaur
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Nir Stanietzky
- Department of RadiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
113
|
Zhong H, Jiang J, Hussain M, Zhang H, Chen L, Guan R. The Encapsulation Strategies for Targeted Delivery of Probiotics in Preventing and Treating Colorectal Cancer: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500304. [PMID: 40192333 PMCID: PMC12079478 DOI: 10.1002/advs.202500304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/01/2025] [Indexed: 05/16/2025]
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer worldwide. It is associated with imbalanced gut microbiota. Probiotics can help restore this balance, potentially reducing the risk of CRC. However, the hostile environment and constant changes in the gastrointestinal tract pose significant challenges to the efficient delivery of probiotics to the colon. Traditional delivery methods are often insufficient due to their low viability and lack of targeting. To address these challenges, researchers are increasingly focusing on innovative encapsulation technologies. One such approach is single-cell encapsulation, which involves applying nanocoatings to individual probiotic cells. This technique can improve their resistance to the harsh gastrointestinal environment, enhance mucosal adhesion, and facilitate targeted release, thereby increasing the effectiveness of probiotic delivery. This article reviews the latest developments in probiotic encapsulation methods for targeted CRC treatment, emphasizing the potential benefits of emerging single-cell encapsulation techniques. It also analyzes and compares the advantages and disadvantages of current encapsulation technologies. Furthermore, it elucidates the underlying mechanisms through which probiotics can prevent and treat CRC, evaluates the efficacy and safety of probiotics in CRC treatment and adjuvant therapy, and discusses future directions and potential challenges in the targeted delivery of probiotics for CRC treatment and prevention.
Collapse
Affiliation(s)
- Hao Zhong
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Jin Jiang
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Muhammad Hussain
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
- Moganshan Institute ZJUTKangqianDeqing313200China
| | - Haoxuan Zhang
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Ling Chen
- Sanya Branch of Hainan Academy of Inspection and TestingSan Ya572011China
| | - Rongfa Guan
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
- Moganshan Institute ZJUTKangqianDeqing313200China
| |
Collapse
|
114
|
Chen Y, Huang J, Fan Y, Huang L, Cai X. Understanding the cellular and molecular heterogeneity in colorectal cancer through the use of single-cell RNA sequencing. Transl Oncol 2025; 55:102374. [PMID: 40163910 PMCID: PMC11993189 DOI: 10.1016/j.tranon.2025.102374] [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: 02/15/2025] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
The very prevalent nature, genetic variability, and intricate tumor microenvironment (TUME) of colorectal cancer (COREC) are its defining features. In order to better understand the molecular and cellular make-up of COREC, this work used single-cell RNA sequencing (SRNAS) to isolate and characterize important cell types as well as their interactions within the TUME. Our analysis of 51,204 cells yielded six distinct types: epithelial, fibroblast, endothelial, T&NK, B, and myeloid. C3 B cells were shown to be the most active in immunological regulation, according to chemokine signaling study, which was one of seven clusters of B cells that were thoroughly subtyped. The examination of copy number variation (CONUV) revealed a great deal of genetic variability, especially in epithelial cells. We traced the activity of three key transcription factor clusters (M1, M2, and M3) across all B cell subtypes using transcription factor analysis. We created a predictive model that correctly sorts patients according to survival results by using marker genes from C3 B cells. In addition, the relationship between genetic changes and the immune system was better understood by tumor mutational burden (TUMUB) and immune infiltration studies. Our research sheds light on the genetic complexity and cellular variety of COREC, which in turn opens up new possibilities for targeted treatments and individualized approaches to patient care.
Collapse
Affiliation(s)
| | - Jian Huang
- Wenzhou Central Hospital, Wenzhou, China
| | - Yufang Fan
- Wenzhou Central Hospital, Wenzhou, China
| | | | | |
Collapse
|
115
|
Li ZF, Zhang JN, Tian S, Sun C, Ma Y, Ye ZX. Dual-Time-Point Radiomics for Prognosis Prediction in Colorectal Liver Metastasis Treated with Neoadjuvant Therapy Before Radical Resection: A Two-Center Study. Ann Surg Oncol 2025; 32:3516-3525. [PMID: 39907877 DOI: 10.1245/s10434-025-16941-6] [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: 11/19/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Optimal prognostic stratification for colorectal liver metastases (CRLM) patients undergoing surgery with neoadjuvant therapy (NAT) remains elusive. This study aimed to develop and validate dual-time-point radiomic models for CRLM prognosis prediction using pre- and post-NAT imaging features. METHODS Radiomic features were extracted from four MRI sequences in 100 cases of CRLM patients who underwent NAT and radical resection. RAD scores were generated, and clinical/pathologic variables were incorporated into uni- and multivariate Cox regression analyses to construct prognosis models. Time-ROC, time-C index, decision curve analysis (DCA), and calibration curves assessed the predictive performance of Fong score and pre- and post-NAT models for overall survival (OS) and disease-free survival (DFS) in a testing set. RESULTS The final models included four variables for OS and three variables for DFS. The post-NAT models outperformed the pre-NAT models in time-ROC, time-C index, calibration, and DCA analysis, except for the 1-year DFS area under the curve (AUC). The Fong score models underperformed. The post-NAT OS RAD score effectively stratified patients into prognostic subgroups. CONCLUSIONS The radiomic models incorporating pre- and post-NAT MRI features and clinical/pathologic variables effectively stratified CRLM patients prognositically. The post-NAT models demonstrated superior performance.
Collapse
Affiliation(s)
- Zhuo-Fu Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | - Jia-Ning Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China
| | | | - Chao Sun
- Department of Radiology, Tianjin Union Medical Center, Tianjin, People's Republic of China
| | - Ying Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhao-Xiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, China; Tianjin Key Laboratory of Digestive Cancer; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, China.
| |
Collapse
|
116
|
Aoun RJN, Kalady MF. Hereditary Colorectal Cancer: From Diagnosis to Surgical Options. Clin Colon Rectal Surg 2025; 38:179-190. [PMID: 40292001 PMCID: PMC12020645 DOI: 10.1055/s-0044-1787884] [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: 04/30/2025]
Abstract
Hereditary colorectal cancer (CRC) syndromes account for up to 5% of CRC. Patients have an increased risk of CRC and extracolonic cancers, both of which develop at an early age. The main polyposis syndromes include familial adenomatous polyposis, MYH-associated polyposis, Peutz-Jeghers syndrome, juvenile polyposis syndrome, and PTEN hamartoma syndrome. The non-polyposis syndromes include Lynch syndrome and familial colorectal cancer type X. Each of the syndromes have distinct but sometimes overlapping phenotypes. Clinical evaluation and ultimately the underlying germline genetic pathogenic variants define the syndromes. Each syndrome has polyp, CRC, and extracolonic risks and management is based on early and timely surveillance with therapeutic and often extended prophylactic surgery. Surgical intervention strategies are individualized, considering not only the earlier onset of malignancies and heightened risks for metachronous cancers but also the patient's needs and quality of life. This article reviews the different diagnostic approaches to hereditary CRC and highlights subsequent disease-specific management and surgical decision-making strategies.
Collapse
Affiliation(s)
- Rami James N. Aoun
- Division of Colon and Rectal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Matthew F. Kalady
- Division of Colon and Rectal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
117
|
Zhang X, Di Y, Wang Y, Qin J, Ye L, Wen X, Ke Z, Wang Z, He W. SIRT5-mediated desuccinylation of PPA2 enhances HIF-1alpha-dependent adaptation to hypoxic stress and colorectal cancer metastasis. EMBO J 2025; 44:2514-2540. [PMID: 40164945 PMCID: PMC12048626 DOI: 10.1038/s44318-025-00416-1] [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: 10/08/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Metastasis is the primary cause of death in patients with colorectal cancer (CRC). Hypoxia is a hallmark of solid tumors that promotes cellular metabolic adaptation and dissemination. However, the mechanisms linking hypoxia-regulated metabolic adaptation to CRC metastasis remain unclear. Here, we found that inorganic pyrophosphatase 2 (PPA2) suppresses metastatic progression of CRC via its phosphatase function. PPA2 expression levels are reduced in CRC specimen and correlate with enhanced response to hypoxia by promoting hypoxia-inducible factor-1 (HIF-1) signaling to promote CRC cell glycolysis and dissemination. Mechanistically, PPA2 decreases HIF-1alpha stability through non-canonical ubiquitin-mediated proteasomal degradation via recruitment of E3 ligase NEDD4. Furthermore, PPA2 directly dephosphorylates NEDD4 at threonine 758 residue, resulting in its activation. Under hypoxic stress, NAD-dependent protein deacetylase sirtuin-5 promotes the dissociation of PPA2 and NEDD4 by inducing PPA2 desuccinylation at lysine 176, contributing to the improved stability of HIF-1alpha under hypoxic conditions. Our findings reveal a tumor-suppressive role of PPA2 in HIF-1alpha-dependent colorectal cancer, providing a potential therapeutic target and prognostic strategy.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yuqin Di
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Youpeng Wang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jiale Qin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lvlan Ye
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiangqiong Wen
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Medical College of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zunfu Ke
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Ziyang Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Weiling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China.
| |
Collapse
|
118
|
He D, Chen S, Wang X, Wen X, Gong C, Liu L, He G. Icaritin Represses Autophagy to Promote Colorectal Cancer Cell Apoptosis and Sensitized Low-Temperature Photothermal Therapy via Targeting HSP90-TXNDC9 Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412953. [PMID: 40184625 PMCID: PMC12120733 DOI: 10.1002/advs.202412953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/10/2025] [Indexed: 04/06/2025]
Abstract
Colorectal cancer (CRC) ranks among the leading causes of cancer-related dea ths worldwide, and the rising incidence and mortality of CRC underscores the urgent need for better understanding and management strategies. Icaritin (ICA) is the metabolites of icariin, a natural flavonoid glycoside compound derived from the stems and leaves of Epimedium. It has broad spectrum antitumor activity and inhibits the proliferation, migration, and invasion of CRC cells, and causes S phase cell cycle arrest. It exerts its antitumor effects against CRC through repressing autophagy to promote CRC cell apoptosis via interfering the HSP90-TXNDC9 interactions. The safety and efficacy of ICA are also affirmed in a mouse xenograft model. Additionally, to test whether ICA exerts synergistic effects with low-temperature photothermal therapy (LTPTT), a novel nanodrug delivery system, employing SiO2 nanocarriers, is designed aiming to load ICA with photothermal materials polydopamine (PDA), and folic acid (FA). This SiO2/Ica-PDA-FA multifunctional nanocomposite actively targets tumor tissues through the high affinity of FA for cancer cells. Once internalized, the acidic intracellular environment triggers the controlled release of ICA, inhibiting HSP90-TXNDC9 interactions. By LTPTT and ICA drug therapy under near-infrared illumination, a dual synergistic antitumor effect is achieved, holding promise for enhancing therapeutic outcomes in CRC treatment.
Collapse
Affiliation(s)
- Dan He
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Department of OncologyThe Second Affiliated Hospital of Chengdu Medical CollegeNuclear Industry 416 HospitalChengdu610051China
| | - Siliang Chen
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiaoyun Wang
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Xiang Wen
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Changyang Gong
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
| | - Lei Liu
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
| | - Gu He
- Division of Head & Neck Tumor Multimodality TreatmentCancer Center and Department of Dermatology & VenerologyWest China HospitalSichuan UniversityChengdu610041China
- Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Institute of Precision Drug Innovation and Cancer CenterThe Second Hospital of Dalian Medical UniversityDalian116023China
| |
Collapse
|
119
|
Zhan Z, Chen B, Lin W, Chen X, Huang R, Yang C, Guo Z. Rising Burden of Colon and Rectum Cancer in China: An Analysis of Trends, Gender Disparities, and Projections to 2030. Ann Surg Oncol 2025; 32:3361-3371. [PMID: 39836276 DOI: 10.1245/s10434-025-16905-w] [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: 08/19/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Colon and rectum cancer (CRC) is a major health burden in China, with notable gender disparities. This study was designed to analyze trends in CRC incidence, prevalence, and mortality from 1990 to 2021 and to project future trends. METHODS Using data from the Global Burden of Disease (GBD) Study 2021, we examined CRC burden in China, including incidence, prevalence, mortality, disability-adjusted life years (DALYs), years lived with disability (YLDs), and years of life lost (YLLs). Joinpoint regression, Bayesian age-period-cohort (BAPC) models, and age-period-cohort (APC) analysis identified trends and projected incidence up to 2030. RESULTS In 2021, CRC incidence was 658,321 cases, disproportionately affecting males, with an age-standardized incidence rate of 42.24 per 100,000 in males and 21.87 per 100,000 in females. The CRC-related deaths reached 275,129, with higher mortality in males (18.95 per 100,000) than females (9.34 per 100,000). The DALYs totaled 6,848,390 and largely impacted males. Joinpoint analysis showed a persistent increase in incidence and prevalence, especially in younger cohorts, whereas mortality declined slightly but began rising again after 2015. The BAPC analysis projected further incidence growth, particularly in males, through 2030. The APC analysis revealed higher CRC risk among younger cohorts, suggesting increasing early-onset CRC linked to lifestyle risk factors, such as smoking, high alcohol consumption, and diets low in fiber and calcium, with a stronger effect on males. CONCLUSIONS The increase of CRC incidence and prevalence in China, particularly among males, underscores the need for targeted prevention and early detection. Future research should address gender disparities and modifiable lifestyle risks through public health interventions.
Collapse
Affiliation(s)
- Zhouwei Zhan
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Bijuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Wei Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xiamei Chen
- Department of Operation, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Rui Huang
- Digestive Endoscopy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Chunkang Yang
- Department of Gastrointestinal Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Zengqing Guo
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
120
|
Borhani AA, Zhang P, Diergaarde B, Darwiche S, Chuperlovska K, Wang SC, Schoen RE, Su GL. Role of tumor-specific and whole-body imaging biomarkers for prediction of recurrence in patients with stage III colorectal cancer. Abdom Radiol (NY) 2025; 50:1907-1915. [PMID: 39487920 DOI: 10.1007/s00261-024-04656-3] [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: 09/06/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Imaging biomarkers are emerging as non-invasive predictors of cancer prognosis and clinical outcome. We assessed tumor-specific ("radiomics") and body composition imaging features ("morphomics") extracted from baseline pre-treatment CT for prediction of recurrence in patients with stage III colorectal cancer (CRC). METHODS Patients with newly diagnosed stage III CRC were enrolled in this prospective observational study. Patients with available preoperative scans were included (N = 101). The tumor, if visible, was manually segmented and first-order radiomics features were extracted with a commercially available software. The morphomics features (reflecting muscle, fat, and bone characteristics) were extracted in a standardized fashion using a proprietary software and the values were adjusted and normalized based on a reference standard. Time to recurrence was the final outcome. Correlation between demographics, clinical features, radiomics, and morphomics features and outcome were assessed using univariate and multivariate tests as well as Kaplan-Meier and log-rank tests. RESULTS Morphomic analysis was performed in all 101 patients. 60 patients had discrete tumors suitable for radiomics analysis. These patients had lower ECOG score (p < 0.05), more muscle mass (p > 0.05), and lower fat density (p > 0.05) compared to the patients in whom radiomics analysis could not be performed. Pathological stage (HR: 2.69; p = 0.03), CEA level after surgery (HR: 1.11 for 1 ng/mL; p < 0.005), bone mineral density (HR: 1.01 for 1 Hounsfield Unit; p < 0.01), and tumor skewness (HR: 0.33 for 1 unit; p < 0.05) had association with recurrence based on both univariate and multivariate analyses. A model using Cox's regression analyses was able to divide the patients into low-, medium-, and high-risk for recurrence. CONCLUSIONS Both radiomics and morphomics features were independently associated with the risk of CRC recurrence and, when combined, each contributed valuable information to explain risk of recurrence. TRIAL REGISTRATION Clinical trial.gov NCT02842203. Patient recruitment occurred between 22/07/2016 and 18/03/2020.
Collapse
Affiliation(s)
- Amir A Borhani
- Northwestern University, Evanston, USA.
- University of Pittsburgh, Pittsburgh, USA.
| | - Peng Zhang
- University of Michigan-Ann Arbor, Ann Arbor, USA
| | | | | | | | | | | | - Grace L Su
- University of Michigan-Ann Arbor, Ann Arbor, USA
| |
Collapse
|
121
|
Bai L, Liu X, Yuan Z, Xu G, Li X, Wan Z, Zhu M, Liang X, Li P, Lan Q, Yu H, Tang G, Huang M, Peng S, Lin J, Wang X, Luo Y, Wei G. Activation of IL-2/IL-2R pathway by Hedyotis diffusa polysaccharide improves immunotherapy in colorectal cancer. Int J Biol Macromol 2025; 306:141013. [PMID: 39954887 DOI: 10.1016/j.ijbiomac.2025.141013] [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: 09/02/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Colorectal cancer (CRC) is a prevalent and highly malignant tumor with a limited response to immune checkpoint inhibitor-based immunotherapy. There is an urgent need for novel immunomodulatory agents to enhance the immunotherapeutic response in CRC. Hedyotis diffusa, known for its immunomodulatory properties, has long been utilized as an adjunct in cancer treatment, positioning it as a potential source for discovering new tumor immunomodulators. In this study, we identified a polysaccharide derived from Hedyotis diffusa (HDP), comprising six monosaccharides: rhamnose, arabinose, galactose, glucose, xylose, and mannose. When combined with PD-1 and CTLA-4 inhibitors, HDP can boost systemic immunity in mice to enhance the effectiveness of immune checkpoint inhibitors in CRC therapy. HDP significantly increases the infiltration of CD4+ and CD8+ T cells into tumor microenvironment and upregulates the expression of key effector molecules derived from cytotoxic T cells. Mechanistic studies reveal that HDP activates the IL-2/IL-2R axis by upregulating IL-2 production and the expression of IL-2 receptor subunits, thereby promoting T cell proliferation. Collectively, this research introduces an innovative strategy to improve the efficacy of tumor immunotherapy by harnessing the immunomodulatory potential of polysaccharides. It also directs a roadmap for developing HDP as a promising immunomodulator for CRC treatment.
Collapse
Affiliation(s)
- Liangliang Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xiaoxia Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Ze Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Gaopo Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xuan Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zhongxian Wan
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei 445000, China
| | - Mingxuan Zhu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaoxia Liang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Peisi Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Qiqian Lan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Huichuan Yu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Guannan Tang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Mingzhe Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Shaoyong Peng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jinxing Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaolin Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Gang Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
122
|
Sakazaki A, Lui A, Wang M, Ngo K, Lugue MT, Aligireddy H, Nguyen M, Castro CL, Park K, Murakami S. Effects of Socioeconomic Status on Colorectal Cancer Incidence and Clinical Outcome Differences Among Asian American Populations: A Systematic Review. Cureus 2025; 17:e83806. [PMID: 40491647 PMCID: PMC12146441 DOI: 10.7759/cureus.83806] [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] [Accepted: 05/09/2025] [Indexed: 06/11/2025] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death among Asian Americans and Pacific Islanders (API) in the United States. Race, ethnicity, and socioeconomic status (SES) are known to impact outcomes of CRC, but the relationship is unclear in the context of the Asian American cohort and its diverse subgroups. This systematic review aims to gain insight into the relationship between CRC incidence and clinical outcomes in the Asian American community. A systematic literature search was conducted per the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) protocol using PubMed, Scopus, Excerpta Medica database (EMBASE), Cochrane, Cumulative Index to Nursing and Allied Health Literature (CINAHL), and Web of Science databases, accessed on August 13, 2023. Of the 2225 studies identified, a total of 14 studies were included in the analysis. Four studies concluded that there was no association or variable response subgroup-specific to SES measurements in CRC incidence in the Asian American population. However, there was evidence that the incidence of CRC varies among Asian American subgroups, using varying measures of SES. Seven of the eight studies that measured mortality or survival as the primary outcome found decreased mortality and increased survival in the API population despite changes in SES. Out of the six studies that measured incidence, four studies found no association with SES. A study found that Chinese Americans had a significant decrease in the CRC incidence and mortality across all SES categories. Japanese Americans experienced a significant decrease in the lowest SES category, while Koreans and Filipinos experienced a significant increase in both the lowest and highest SES categories. Therefore, grouping various Asian American ethnicities as a single monolithic "Asian" category is misleading. Although the incidence of CRC was thought to be low and decreasing, this review identified various subgroup-specific trends among 24 different Asian American subsets. For example, there was a decrease in CRC rates in two ethnic groups and an increase in the other two ethnic groups. The potential causes of these varying CRC incidence rates are likely multifactorial and may include inadequate screening rates, lack of CRC education, and cultural barriers. Further studies are needed to understand these mechanisms. This review recommends a more detailed classification of the API ethnic population but not as a single monolithic entity as Asian. It also emphasizes preventative CRC screening within the API communities due to lower rates of CRC screening among them.
Collapse
Affiliation(s)
- Amy Sakazaki
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| | - Austin Lui
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| | - Melody Wang
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| | - Katherine Ngo
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| | | | - Himani Aligireddy
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| | - Maria Nguyen
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| | | | - Kenneth Park
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| | - Shin Murakami
- College of Osteopathic Medicine, Touro University California, Vallejo, USA
| |
Collapse
|
123
|
Duffy L, Anderson M, Rowe E, Yan M, Abdelhak A, Garcia J, Muller C, Kumar A. Colorectal cancer care equity in underserved communities: Innovative solutions for screening, outreach & capacity in rural Washington. Am J Surg 2025; 243:116246. [PMID: 39986209 DOI: 10.1016/j.amjsurg.2025.116246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) mortality hotspots in rural Washington, notably Yakima and Richland, identified an 8-year earlier median age of death in non-white patients. Post-pandemic data from WA State Department of Health and MultiCare's electronic health records revealed a 40 % decrease in CRC screenings. METHODS During a 2023 CRC prevention summit, barriers and solutions were discussed focusing on rural Hispanic laborers as this population is often seen at partner locations, Yakima Valley Farm Workers Clinic (YVFWC). RESULTS Community outreach-Our team established a presence at Yakima's largest health fair, Fiesta de Salud. We also provided screening resources at Richland's farmers' markets. Planned events-- We will network with local community leaders and health providers to actualize screening (stool-based testing, hands-on endoscopy training). CONCLUSION Persistent CRC mortality disparities in Washington State underscores the need for targeted interventions. Partnering with organizations and engaging in community outreach aims to increase screening rates and enhance education.
Collapse
Affiliation(s)
- Lauren Duffy
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Marley Anderson
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Evelyn Rowe
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Michelle Yan
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Aliya Abdelhak
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Juliana Garcia
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Clemma Muller
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA
| | - Anjali Kumar
- Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99202, USA.
| |
Collapse
|
124
|
Babaei M, Abrishami A, Iranpour S, Saljooghi AS, Matin MM. Harnessing curcumin in a multifunctional biodegradable metal-organic framework (bio-MOF) for targeted colorectal cancer theranostics. Drug Deliv Transl Res 2025; 15:1719-1738. [PMID: 39302530 DOI: 10.1007/s13346-024-01707-6] [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] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
Despite significant advancements in managing colorectal cancer (CRC), the issues of efficient diagnosis and targeted therapy remain demanding. To address these challenges and improve treatment outcomes while reducing the cost and side effects, there is a need for more effective theranostic systems that combine diagnostic techniques with therapeutic modalities. This study introduces a pioneering approach for the synthesis of a porous bio-MOF (biodegradable metal-organic framework) using iron as the metal component and curcumin as the pharmaceutical ingredient. Subsequently, the developed drug delivery system was equipped with the anticancer drug doxorubicin (DOX), coated with biocompatible polyethylene glycol (PEG), and targeted with a CRC-specific aptamer (EpCAM). The physicochemical characterization confirmed the successful synthesis of the bio-MOF, demonstrating high encapsulation efficiency and pH-dependent release of DOX. In vitro studies for anticancer activity, cellular uptake, and mechanism of cell death demonstrated that in the case of positive EpCAM HT-29 cells, Apt-PEG-MOF@DOX had enhanced internalization that resulted in massive apoptosis. In vivo studies of the nanoparticles were then conducted in immunocompromised C57BL/6 mice bearing HT-29 tumors. These studies showed that the targeted platform could induce efficient tumor regression with reduced systemic toxicity. The targeted bio-MOF also exhibited MRI imaging properties useful for monitoring tumors. Significantly, the biocompatibility of the introduced bio-MOF was enhanced by pursuing the green synthesis method, which does not engage toxic solvents and strong acids. Overall, this multimodal system acts diversely as a tumor imaging agent and a therapeutic delivery platform suitable for CRC theranostics.
Collapse
Affiliation(s)
- Maryam Babaei
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Abrishami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sonia Iranpour
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Sh Saljooghi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
125
|
Yuan N, Ren R, Li D, Liang X, Pang X, Wang J, Feng X, Zhang Z. Advancing colorectal cancer diagnosis: The role of tumor-derived serum galanin and haptoglobin as novel biomarkers. J Cancer Res Ther 2025; 21:417-424. [PMID: 40317147 DOI: 10.4103/jcrt.jcrt_1742_24] [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] [Accepted: 01/24/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a widespread and serious global malignancy. This study aimed to investigate the clinical relevance of galanin (GAL) and haptoglobin (HP) as new diagnostic CRC biomarkers. METHODS An enzyme-linked immunosorbent assay was used to determine the GAL and HP levels in the serum of 88 patient with CRC and 88 healthy controls. The carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) contents were quantitatively evaluated via electrochemiluminescence assay. Receiver operating characteristic (ROC) curves were created to identify the diagnostic importance of single and combined CRC detection. The patients' relevant diagnostic and treatment data were collected from their medical records. Statistical analysis methods were employed to examine the relationship between these indicators and the clinical pathological parameters. RESULTS Patients with CRC exhibited significantly decreased and increased serum GAL and HP levels, respectively, compared with the healthy controls (P < 0.0001 for both). Furthermore, the HP level was positively correlated with tumor T stage (P = 0.0124). The area under the curve (AUC) of the ROC values for GAL and HP was 0.744 and 0.712, respectively, indicating their diagnostic efficiency. The combination of GAL and HP increased the AUC to 0.753, and when both were integrated with CEA and CA19-9, the AUC further increased to 0.893. CONCLUSION This study shows that serum GAL and HP can be used as potential noninvasive diagnostic biomarkers for CRC.
Collapse
Affiliation(s)
- Ning Yuan
- Department of Clinical Laboratory, Binhai New Area Hospital of TCM, Tianjin, China
| | - Ren Ren
- Intensive Care Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Dan Li
- Department of Clinical Laboratory, Binhai New Area Hospital of TCM, Tianjin, China
| | - Xinmei Liang
- Department of Clinical Laboratory, Binhai New Area Hospital of TCM, Tianjin, China
| | - Xiaochen Pang
- Chinese Medicine Clinical Research Center, Binhai New Area Hospital of TCM, Tianjin, China
| | - Jun Wang
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaodong Feng
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Zhijun Zhang
- Department of Clinical Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
- Shandong Provincial Key Medical and Health Laboratory of Anti-drug Resistant drug research, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| |
Collapse
|
126
|
Xu Y, Fu G, Chen X, He R, Fang Q, Tan X, Xu K, Li H, Tan J, Fang M, Fu K, Huang Q, Xiao S. Integrated single-nuclear RNA sequencing analysis reveals distinct characteristics of mucinous adenocarcinoma in right-sided colon cancer. Int J Biol Macromol 2025; 309:142744. [PMID: 40180102 DOI: 10.1016/j.ijbiomac.2025.142744] [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: 11/18/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Mucinous adenocarcinoma (MAC) is a unique histological subtype of colorectal cancer (CRC), which usually occurs in the right-sided colon with poor prognosis. Our previous studies reveled unique clinicopathological characteristic of MAC, but the distinct molecular features and tumor microenvironment (TME) characteristics remain clarify systematically. In this study, we conducted a single nuclear RNA sequencing (snRNA-seq) analysis for CRC tissues with different histological subtypes, including MAC, partial mucinous adenocarcinoma (pMAC) and non-specific adenocarcinoma (AC). Our results show that MAC has a unique transcriptome profile and a distinct single-cell characteristic. It exhibited a higher degree of tumor purity but a lower composition of TME. MAC had a distinct cell-cell communication microenvironment and a particular evolutionary trajectory. The EpithelialCells of MAC closely interacted with fibroblasts, endothelial cells, and mural cells clusters. Furthermore, molecular functional characteristics analysis revealed that MAC enriched EpithelialCells, Fibroblasts, and Endothelial cells clusters have a high active in glycolysis, inflammation, and angiogenesis respectively. This comprehensive study first demonstrated that MAC is a unique histological subtype of right-sided CRC in the single cell level, and elucidated its tumor microenvironment composition and biological function characteristics, which will help us better understand the intrinsic feature of MAC.
Collapse
Affiliation(s)
- Yunhua Xu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Institute of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Guang Fu
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiguang Chen
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Rongfang He
- The First Affiliated Hospital, Department of pathology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Qing Fang
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiangwen Tan
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The Second Affiliated Hospital, Department of hepatological Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kunming Xu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Hongming Li
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jie Tan
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Min Fang
- The First Affiliated Hospital, Department of Education and Training, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kai Fu
- Institute of Molecular Precision Medicine, Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| | - Qiulin Huang
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| | - Shuai Xiao
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Institute of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
127
|
Hanitrarimalala V, Prgomet Z, Hedhammar M, Tassidis H, Wingren AG. In vitro 3D modeling of colorectal cancer: the pivotal role of the extracellular matrix, stroma and immune modulation. Front Genet 2025; 16:1545017. [PMID: 40376304 PMCID: PMC12078225 DOI: 10.3389/fgene.2025.1545017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/23/2025] [Indexed: 05/18/2025] Open
Abstract
Colorectal cancer (CRC) is a leading global cancer with high mortality, especially in metastatic cases, with limited therapeutic options. The tumor microenvironment (TME), a network comprising various immune cells, stromal cells and extracellular (ECM) components plays a crucial role in influencing tumor progression and therapy outcome. The genetic heterogeneity of CRC and the complex TME complicates the development of effective, personalized treatment strategies. The prognosis has slowly improved during the past decades, but metastatic CRC (mCRC) is common among patients and is still associated with low survival. The therapeutic options for CRC differ from those for mCRC and include surgery (mostly for CRC), chemotherapy, growth factor receptor signaling pathway targeting, as well as immunotherapy. Malignant CRC cells are established in the TME, which varies depending on the primary or metastatic site. Herein, we review the role and interactions of several ECM components in 3D models of CRC and mCRC tumor cells, with an emphasis on how the TME affects tumor growth and treatment. This comprehensive summary provides support for the development of 3D models that mimic the interactions within the TME, which will be essential for the development of novel anticancer therapies.
Collapse
Affiliation(s)
- Veroniaina Hanitrarimalala
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Zdenka Prgomet
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - My Hedhammar
- KTH Royal Institute of Technology, Division of Protein Technology, Stockholm, Sweden
| | - Helena Tassidis
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| | - Anette Gjörloff Wingren
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| |
Collapse
|
128
|
Keiser E, Corbett AM, Chido-Amajuoyi O, Antoine A, Stehman C, Dorn I, Goines D, LoConte NK. Acceptability of Stool-Based DNA Colorectal Cancer Screening among Black/African-American Patients Served by Federally Qualified Health Centers. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2025:10.1007/s13187-025-02631-0. [PMID: 40307656 DOI: 10.1007/s13187-025-02631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/02/2025]
Abstract
Colorectal cancer (CRC) has an increased burden among Black/African-American populations. Following the COVID-19 pandemic, home-based CRC screening options are being used more frequently. We conducted focus groups to understand the acceptability of stool-based DNA testing for CRC screening in this population. Ten focus groups about the acceptability of various CRC screening modalities were held with Black/African-American participants at two federally qualified health centers (FQHCs) in Milwaukee, Wisconsin. Participants were separated into focus groups based on age and gender. Thematic analysis was carried out using NVivo. Across the groups, there were a total of 79 participants, of which 40.5% were aged 40-50 years ("younger participants"), 59.5% aged > 50 years ("older participants"), 53.2% male, and 46.8% female. Overall, knowledge was low regarding perceived risk of CRC. There was limited awareness of CRC screening options among younger patients and widespread lack of knowledge about stool-based DNA testing. Most respondents preferred colonoscopy as their first-choice screening test but were open to other screening tests. Stool-based DNA tests were more preferred among younger participants but was felt to be acceptable across all groups. Given the low awareness/knowledge of screening modalities identified in our study, educational interventions and shared decision making by primary care providers are needed.
Collapse
Affiliation(s)
- Evan Keiser
- School of Medicine and Public Health, Department of Medicine, University of Wisconsin, Madison, WI, USA.
| | - A Michelle Corbett
- Center for Urban Population Health, School of Medicine and Public Health, University of Wisconsin, Milwaukee, WI, USA
| | | | - Allison Antoine
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Carrie Stehman
- Center for Urban Population Health, School of Medicine and Public Health, University of Wisconsin, Milwaukee, WI, USA
| | - Isabella Dorn
- Outreach Community Health Center, Milwaukee, WI, USA
| | - David Goines
- Progressive Community Health Centers, Madison, WI, USA
| | | |
Collapse
|
129
|
Gao S, He Y, Jiang L, Yang J. Multimodal prehabilitation program for patients undergoing elective surgery for colorectal cancer: a scoping review. Front Oncol 2025; 15:1532624. [PMID: 40386553 PMCID: PMC12082037 DOI: 10.3389/fonc.2025.1532624] [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: 11/26/2024] [Accepted: 04/07/2025] [Indexed: 06/01/2025] Open
Abstract
Objectives Multimodal prehabilitation has been widely used in patients undergoing surgery for colorectal cancer and has improved clinical outcomes. The aim of this scoping review is to review the content and current state of clinical practice of multimodal prehabilitation programs. Methods A systematic literature review of multimodal prehabilitation studies in patients undergoing colorectal cancer surgery was conducted according to the PRISMA extension framework for scoping reviews. The literature was searched via the PubMed, Web of Science, SCOPUS, EMBASE and Cochrane Library databases. The results of the study included the components of multimodal prehabilitation (exercise, nutritional, and psychological interventions) and related evaluation indicators, duration, and compliance-related components. Results This review included 12 studies with 9 randomized controlled trials, 1 pilot intervention study, 1 cohort study, and 1 mock-target trial design. Specific protocols for multimodal rehabilitation training vary widely, ranging in duration from 2-8 weeks, and were implemented in healthcare facilities, the community, and at home. Adherence rates ranged from 50% to almost 100%. Common outcome indicators include the 6-minute walk test, comorbidities, length of hospitalization, health-related quality of life, and several anxiety assessment scales. Conclusion Current evidence suggests that multimodal preconditioning has a positive effect on the clinical prognosis of patients undergoing elective colorectal cancer surgery. However, owing to the heterogeneity of multimodal rehabilitation in terms of implementation protocols and evaluation metrics, many high-quality studies are still needed to explore the optimal model of multimodal rehabilitation and promote its standardization.
Collapse
Affiliation(s)
| | | | | | - Jie Yang
- Colorectal Cancer Center, West China Hospital, Sichuan University/West China School of
Nursing, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
130
|
Torshizi Esfahani A, Zafarjafarzadeh N, Vakili F, Bizhanpour A, Mashaollahi A, Karimi Kordestani B, Baratinamin M, Mohammadpour S. Gut microbiome in colorectal cancer: metagenomics from bench to bedside. JNCI Cancer Spectr 2025; 9:pkaf026. [PMID: 40045177 DOI: 10.1093/jncics/pkaf026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/27/2024] [Accepted: 02/27/2025] [Indexed: 05/27/2025] Open
Abstract
Colorectal cancer (CRC) is a major global health challenge. Emerging research highlights the pivotal role of the gut microbiota in influencing CRC risk, progression, and treatment response. Metagenomic approaches, especially high-throughput shotgun sequencing, have provided unprecedented insights into the intricate connections between the gut microbiome and CRC. By enabling comprehensive taxonomic and functional profiling, metagenomics has revealed microbial signatures, activities, and biomarkers associated with colorectal tumorigenesis. Furthermore, metagenomics has shown a potential to guide patient stratification, predict treatment outcomes, and inform microbiome-targeted interventions. Despite remaining challenges in multi-omics data integration, taxonomic gaps, and validation across diverse cohorts, metagenomics has propelled our comprehension of the intricate gut microbiome-CRC interplay. This review underscores the clinical relevance of microbial signatures as potential diagnostic and prognostic tools in CRC. Furthermore, it discusses personalized treatment strategies guided by this omics' approach.
Collapse
Affiliation(s)
- Amir Torshizi Esfahani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikta Zafarjafarzadeh
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Fatemeh Vakili
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Anahita Bizhanpour
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Amirhesam Mashaollahi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Bita Karimi Kordestani
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Mahdieh Baratinamin
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Somayeh Mohammadpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
131
|
Wang C, Chen M, Chen P, Han J, Hu H, Chen J, Wu Q, Zhao D, Wang T, Zhou J, Li Q, Zhou R, Wen Y, Yang J, Shi M, Wang Y. RBM15-mediated metabolic reprogramming boosts immune response in colorectal cancer. Front Immunol 2025; 16:1515568. [PMID: 40370450 PMCID: PMC12075365 DOI: 10.3389/fimmu.2025.1515568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Immune checkpoint blockade (ICB) therapy has shown promise in treating advanced colorectal cancer, particularly in patients with microsatellite instability-high (MSI-H) tumors. However, only a subset of these patients responds favorably, highlighting the need for strategies to improve immunotherapy efficacy. Methods To identify potential regulators of immunotherapy response, we conducted a comprehensive analysis of colorectal cancer datasets from The Cancer Genome Atlas (TCGA). We performed multi-omics analyses and functional assays in both human and murine colorectal cancer cell lines. Additionally, we evaluated tumor growth and immune cell infiltration using syngeneic mouse models. Results Our analysis revealed that RNA binding motif protein 15 (RBM15) is highly expressed in colorectal cancer and correlates with poor patient prognosis. Functional studies demonstrated that RBM15 loss led to increased expression of fumarate hydratase (FH). This led to decreased levels of fumarate, a metabolite known to suppress anti-tumor immune responses. In vivo, RBM15 depletion significantly delayed tumor progression and enhanced CD8⁺ T cell infiltration and activation in the tumor microenvironment. Discussion These findings identify RBM15 as a negative regulator of anti-tumor immunity in colorectal cancer. Targeting RBM15 may represent a novel strategy to boost immune responsiveness and improve outcomes for patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Chen Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyan Chen
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Panyu Chen
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jinlu Han
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Hu
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiong Chen
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiong Wu
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - De Zhao
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tongshuai Wang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Zhou
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Li
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runkai Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yugang Wen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yang
- Department of Pathology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
132
|
Huang Y, Min G, Wang H, Jiang L. A Prognostic Model for Senescence-Related LncRNA in a Novel Colon Adenocarcinoma Based on WGCNA and LASSO Regression. Biomedicines 2025; 13:1088. [PMID: 40426916 PMCID: PMC12109385 DOI: 10.3390/biomedicines13051088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/12/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Objective: This study aims to develop a prognostic model based on senescence-related long non-coding RNAs (lncRNAs) to predict the prognosis of patients with colon cancer and enhance their survival rates. Method: Differential expression analysis and Pearson correlation were employed to identify senescence-related lncRNAs in colon cancer. A risk prognosis model was constructed using univariate Cox regression analysis and Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis. The reliability of this model was validated through survival analysis, receiver operating characteristic (ROC) curves, bar charts, and calibration curves. Additionally, the relationship between the prognostic model, immune microenvironment, and drug sensitivity was explored. Results: A risk prognosis model comprising eight senescence-related lncRNAs (LINC02257, AL138921.1, ATP2B1-AS1, AC005332.7, AC007728.3, AC018755.4, AL390719.3, and THCAT158) was successfully established, demonstrating strong performance in predicting the overall survival rates of colon cancer patients (AUC = 0.733). A significant correlation was observed between the senescence-related lncRNA prognostic model and the tumor microenvironment, immune cell infiltration, and drug sensitivity (p < 0.05). Conclusions: The senescence-related lncRNA prognostic model developed in this work can accurately forecast the prognosis of colon cancer patients, offering new insights for personalized treatment approaches in colon cancer.
Collapse
Affiliation(s)
- Yichu Huang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Guangtao Min
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; (G.M.)
| | - Hongpeng Wang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; (G.M.)
| | - Lei Jiang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; (G.M.)
| |
Collapse
|
133
|
Zhang Y, Liu H. Aberrant DNMT1-mediated DACH1 methylation is associated with colorectal adenoma-to-carcinoma progression. Exp Biol Med (Maywood) 2025; 250:10469. [PMID: 40370966 PMCID: PMC12075005 DOI: 10.3389/ebm.2025.10469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
Colorectal cancer (CRC) remains a major contributor to cancer-related morbidity and mortality. While Dachshund homolog 1 (DACH1) was recognized as a critical regulator in cancer progression, its role in promoting or suppressing tumor development remains a subject of ongoing debate. This study aimed to elucidate the role of DACH1 in CRC progression and its underlying regulation mechanisms. The expression levels of Methyltransferase 1 (DNMT1) and DACH1, as well as its methylation status were assessed through a combination of TCGA data analysis and experimental validation using immunohistochemistry, PCR, methylation-specific PCR, and bisulfite sequencing RCR on 120 clinical samples, comprising normal mucosa, adenomas, and adenocarcinomas. The relationships among them were evaluated using Pearson or Spearman correlation analysis. The associations between the DACH1 and DNMT1 levels and clinicopathological parameters were examined to determine their clinical relevance. A progressive decrease in DACH1 expression and a concomitant increase in DACH1 promoter methylation and DNMT1 expression were observed from normal mucosa to adenoma and adenocarcinoma tissues. Higher DNMT1 expression and lower DACH1 expression were associated with poorer clinical outcomes, including worse tumor differentiation, lymphatic metastasis, and advanced tumor stages. Paired analysis of tissues from the same patient further validated their inverse expression patterns during CRC progression. DNMT1-mediated DACH1 epigenetic silencing plays a critical role in CRC progression, suggesting that the DNMT1-DACH1 regulatory axis may serve as a potential biomarker and therapeutic target in CRC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Pathology, Beijing Changping Traditional Chinese Medicine Hospital, Beijing, China
| | - Honggang Liu
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
134
|
Huang L, Zhao K, Lu H, He W, Miao D, Wang Y, Li B, Wang Q, Jiang S, Jia Y. Serum cholinesterase combined with platelet-to-hemoglobin ratio for predicting survival prognosis in stage I-III colorectal cancer patients undergoing radical surgery: a retrospective cohort study. BMC Gastroenterol 2025; 25:319. [PMID: 40301776 PMCID: PMC12042540 DOI: 10.1186/s12876-025-03932-w] [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: 12/20/2024] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Although surgery-based comprehensive therapy is highly effective for treating stage I-III colorectal cancer, heterogeneity in survival trajectories still exists, necessitating precise prognostic stratification. Serum cholinesterase (CHE) and the platelet-to-hemoglobin ratio (PHR) are emerging as potential prognostic markers reflecting inflammation, nutritional status, and tumor biology. This study aims to investigate their combined value in predicting survival outcomes for stage I-III CRC patients, potentially offering a cost-effective tool for personalized management. METHODS The study included 673 stage I-III CRC patients who underwent radical surgery at Harbin Medical University Cancer Hospital from January to August 2019 and January to March 2020. Comprehensive clinicopathological data were collected. The patients from 2019 were used for the primary research analysis. Kaplan-Meier analysis was used for survival comparisons, while Cox regression identified independent prognostic factors. Two nomograms were developed to predict the prognosis of DFS and OS and were validated in 2020 patient cohort. RESULTS A total of 475 patients from 2019 patient cohort were classified into three different risk groups: Group 1 (CHE ≥ 6213.3 U/L and PHR ≤ 3.03, n = 305), Group 2 (CHE < 6213.3 U/L or PHR > 3.03, n = 135), and Group 3 (CHE < 6213.3 U/L and PHR > 3.03, n = 35). Survival analysis indicated that CRC patients with low serum CHE levels and high PHR had a poorer prognosis (all p < 0.05), and the combined biomarker CHE-PHR effectively distinguished different prognostic risk groups (p < 0.001). Multivariate analysis identified Crea (p = 0.037), Eosi (p = 0.021), CA199 (p = 0.002), pTNM stage (p < 0.001), number of lymph nodes detected (p = 0.007), and CHE-PHR (p < 0.001) as independent prognostic factors for DFS, while CEA (p = 0.015), CA199 (p = 0.006), pTNM stage (p < 0.001), number of lymph nodes detected (p = 0.007), and CHE-PHR (p < 0.001) were independent prognostic factors for OS. In 2019 patient cohort t, the nomogram's AUC values for 1-, 3-, 5-year DFS are 0.825, 0.766, and 0.748, and for 1-, 3-, 5-year OS, they are 0.787, 0.743, and 0.756. In 2020 patient cohort, the AUC values are 0.776, 0.812, 0.736 for DFS, and 0.756, 0.818, 0.789 for OS. CONCLUSION Lower serum CHE and higher PHR levels are linked to poorer DFS and OS outcomes. The CHE-PHR indicator serves as an independent prognostic factor for stage I-III CRC patients post-surgery, aiding in predicting recurrence and metastasis.
Collapse
Affiliation(s)
- Luyu Huang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Kai Zhao
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Hongnan Lu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Wei He
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
- Zigong Third People's Hospital, Zigong, 643000, Sichuan, China
| | - Dazhuang Miao
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yan Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Bingcai Li
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Qi Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Shixiong Jiang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Yunhe Jia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
135
|
Song A, Liu B, Li W, Chen B, Gui P, Zhang H, Zhu C, Xu Y, Jiang T, Song J. Competitive binding between DDX21 and SIRT7 enhances NAT10-mediated ac 4C modification to promote colorectal cancer metastasis and angiogenesis- DDX21 promotes colorectal cancer metastasis. Cell Death Dis 2025; 16:353. [PMID: 40301349 PMCID: PMC12041575 DOI: 10.1038/s41419-025-07656-3] [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: 08/05/2024] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025]
Abstract
DExD- box helicase 21 (DDX21) is overexpressed in colorectal cancer (CRC) and is positively correlated with poor prognosis and the malignant phenotype of CRC. Functional characterization indicated that DDX21 promotes CRC metastasis and angiogenesis both in vitro and in vivo. N-acetyltransferase 10 (NAT10) is a key regulator of the N4-acetylcytidine (ac4C) modification of mRNA, regulating the stabilization of mRNA via ac4C modification. Here, we identified that DDX21 competitive binding with sirtuin 7 (SIRT7), inducing the overexpression of NAT10. Furthermore, DDX21 upregulates NAT10 expression to enhance ac4C modification and the stability of ATAD2, SOX4 and SNX5 mRNAs, which mediate CRC metastasis and angiogenesis. Overall, the present study revealed a mechanism of DDX21/NAT10-mediated mRNA stability in CRC, laying the foundation for the use of DDX21 as a therapeutic target to overcome metastasis and angiogenesis in CRC. DDX21 competitive binding with sirtuin 7 (SIRT7), inducing the overexpression of NAT10. Furthermore, DDX21 upregulates NAT10 expression to enhance ac4C modification and the stability of ATAD2, SOX4 and SNX5 mRNAs, which mediate CRC metastasis and angiogenesis.
Collapse
Affiliation(s)
- Angxi Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Bowen Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenjing Li
- Central Laboratory, Xuzhou NO.1 people's hospital, Xuzhou, China
| | - Bingyuan Chen
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
| | - Pengkun Gui
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hao Zhang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Can Zhu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China.
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
136
|
Abboud Y, Shah A, Fraser M, Montminy EM, Pan CW, Hajifathalian K, Gaglio PJ, Al-Khazraji A. Rising Incidence and Mortality of Early-Onset Colorectal Cancer in Young Cohorts Associated with Delayed Diagnosis. Cancers (Basel) 2025; 17:1500. [PMID: 40361427 PMCID: PMC12071177 DOI: 10.3390/cancers17091500] [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: 03/25/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Background and Aims: Prior data showed an increasing incidence of early-onset colorectal cancer (EO-CRC) in the US. However, there are limited comprehensive data on recent EO-CRC incidence and mortality per different age cohorts and tumor characteristics. We aimed to evaluate EO-CRC incidence and mortality in different populations. Methods: Age-adjusted EO-CRC incidence rates were calculated from the USCS database between 2001 and 2021. Age-adjusted EO-CRC mortality rates were calculated from the NCHS database between 2000 and 2022 and the SEER database between 2004 and 2021. The age groups were 20-44 years and 45-54 years. Tumors were categorized by anatomical location (right, transverse, left, and proximal) and stage at diagnosis (early and late). The annual and average annual percentage changes (AAPC) were estimated using joinpoint regression. Age-specific pairwise comparison was conducted. Results: A total of 474,601 patients were diagnosed with EO-CRC between 2001 and 2021. The EO-CRC incidence increased in patients aged 20-44 years faster than in those aged 45-54 years (AAPC = 1.51 vs. 0.73; AAPC difference = 0.78, p = 0.001). This was most notable in proximal colon tumors (AAPC difference = 0.88, p < 0.001). While the incidence of early-stage tumors decreased in recent years across all anatomical locations, late-stage tumors increased, mostly in the proximal colon (AAPC = 2.44). A total of 147,026 patients died from EO-CRC between 2000 and 2022. Mortality increased in patients aged 20-44 years faster than in patients aged 45-54 years, who had a stable trend (AAPC difference = 0.85, p < 0.001). The increase in mortality was noted in both early- and late-stage tumors. Conclusions: Our study provides epidemiologic evidence showing an alarming increase in EO-CRC incidence and mortality among patients aged 20-44 years compared to those aged 45-54 years, which is associated with delayed diagnosis, and mostly in proximal colon tumors. Expanding screening efforts to younger populations and addressing healthcare disparities are essential for improving early detection and outcomes for younger patients.
Collapse
Affiliation(s)
- Yazan Abboud
- Department of Internal Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (A.S.); (M.F.)
| | - Anand Shah
- Department of Internal Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (A.S.); (M.F.)
| | - Madison Fraser
- Department of Internal Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (A.S.); (M.F.)
| | - Eric M. Montminy
- Division of Gastroenterology and Hepatology, University of Chicago, Chicago, IL 60637, USA;
| | - Chun-Wei Pan
- Department of Internal Medicine, John H. Stroger Jr. Hospital of Cook County, Chicago, IL 60612, USA;
| | - Kaveh Hajifathalian
- Division of Gastroenterology and Hepatology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (K.H.); (P.J.G.); (A.A.-K.)
| | - Paul J. Gaglio
- Division of Gastroenterology and Hepatology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (K.H.); (P.J.G.); (A.A.-K.)
| | - Ahmed Al-Khazraji
- Division of Gastroenterology and Hepatology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (K.H.); (P.J.G.); (A.A.-K.)
| |
Collapse
|
137
|
McCready TM, Cohen EM, Laynor G, Chebli P, Liang PS, Renson A. Addressing the immigrant screening gap: A protocol for a systematic review on interventions to enhance colorectal cancer screening among immigrants in the United States. PLoS One 2025; 20:e0322038. [PMID: 40299980 PMCID: PMC12040244 DOI: 10.1371/journal.pone.0322038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/14/2025] [Indexed: 05/01/2025] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) screening rates are lower among immigrant populations in the United States (US) than the general population. Immigrant communities face structural barriers that disincentivize their engagement from CRC screening. A growing body of literature has evaluated the effects of interventions aimed at increasing CRC screening engagement among various immigrant groups, but there has not yet been a systematic synthesis of this literature. OBJECTIVE This review will systematically evaluate quantitative studies assessing the effects of interventions designed to increase CRC screening rates among immigrant populations residing in the US. METHODS We will conduct a comprehensive search of English language peer-reviewed and grey literature using specific keywords and database-specific structured vocabulary on interventions to improve CRC screening rates among immigrants published in 7 databases (PubMed, Cochrane Library (Wiley), CINAHL (EBSCO), ClinicalTrials.gov, Embase (Ovid), Scopus (Elsevier), and Web of Science) from January 1, 2000 to December 31, 2024. All studies will be imported into Covidence. Two reviewers will independently screen titles, abstracts, and full-texts for inclusion and exclusion criteria. Pilot screenings and consensus discussions will ensure accuracy and agreement in study selection and data extraction. Iterative data extraction of eligible studies will include critical appraisal using the Risk of Bias 2 (ROB2) for randomized controlled trials, while other study designs will be appraised with the risk of bias in nonrandomized studies of interventions (ROBINS-I) tool. Data synthesis will disaggregate pooled effect estimates by ethnicity, to the extent possible. The study protocol was pre-registered in International Prospective Register of Systematic reviews (PROSPERO): CRD42023488183. EXPECTED OUTPUTS This systematic review aims to generate an exhaustive summary of the evidence base, including a description of the intervention methods and settings, target populations, recruitment and retention strategies, partnerships and collaborations, and reported outcomes. The results will provide actionable recommendations for public health practitioners, healthcare providers, and policymakers developing tailored interventions and policies aimed at improving CRC screening uptake among diverse immigrant populations in the US.
Collapse
Affiliation(s)
- Taylor M. McCready
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Ethan M. Cohen
- Section of Internal Medicine, West Virginia University, West Virginia, West Virginia, United States of America
| | - Gregory Laynor
- Health Sciences Library, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Perla Chebli
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Peter S. Liang
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Medicine, Veterans Affairs New York Harbor Healthcare System, New York, New York, United States of America
| | - Audrey Renson
- Department of Population Health, New York University Grossman School of Medicine, New York, New York, United States of America
| |
Collapse
|
138
|
Bhowmick K, Yang X, Mohammad T, Xiang X, Molmenti CL, Mishra B, Dasarathy S, Krainer AR, Hassan MI, Crandall KA, Mishra L. Microbial metabolite ammonia disrupts TGF-β signaling to promote colon cancer. J Biol Chem 2025:108559. [PMID: 40311681 DOI: 10.1016/j.jbc.2025.108559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Colorectal cancer (CRC) is rising alarmingly in younger populations, potentially arising from factors such as obesity, pro-inflammatory gut microbiome and the accumulation of toxic metabolites. However, how metabolites such as ammonia impact key signaling pathways to promote CRC remains unclear. Our study investigates a critical link between gut microbiome alterations, ammonia, and their toxic effects on the TGF-β signaling pathway, driving CRC progression. We observe altered microbial populations in an obesity-induced mouse model of cancer, where ammonia promotes caspase-3-mediated cleavage of the SMAD3 adaptor βII-spectrin (SPTBN1). Cleaved SPTBN1 fragments form adducts with ammonia that induce pro-inflammatory cytokine expression and disrupt TGF-β signaling. Extending from AlphaFold docking simulations, we identified that ammonia interacts with N-terminal SPTBN1 potentially through residues D81, Y556, S663, Y666, N986, and D1177 to form hydrogen bonds that disrupt downstream SMAD3 signaling, altering TGF-β signaling to a protumorigenic phenotype. Blocking SPTBN1, through an SPTBN1-specific siRNA blocks ammonia toxicity and restores normal SMAD3/TGF-β signaling by reducing the abundance of SPTBN1 cleaved fragments in SW480 and Caco-2 (CRC) cell lines. In addition, our research establishes crosstalk between TGF-β signaling and a microbial sensor, carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), which is significantly overexpressed in CRC patients. We identified CEACAM1-SPTBN1 interactions at specific residues (E517 and Y520) within the immunoreceptor tyrosine-based inhibitory motif (ITIM) of CEACAM1 cytoplasmic domain, identifying distinguishing a potential axis that is harnessed by the altered microbiome. Our study identifies mechanistic insights into how microbial metabolites target TGF-β as a major signaling pathway to promote CRC.
Collapse
Affiliation(s)
- Krishanu Bhowmick
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA.
| | - Xiaochun Yang
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Xiyan Xiang
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA
| | - Christine L Molmenti
- Department of Occupational Medicine, Epidemiology and Prevention, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Feinstein Institutes for Medical Research, Institute of Cancer Research, Manhasset, NY, USA; Department of Surgery, Northwell Health, Manhasset, NY, USA
| | - Bibhuti Mishra
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Department of Neurology, Hempstead, NY, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | | | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Keith A Crandall
- Computational Biology Institute and Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington DC, USA
| | - Lopa Mishra
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA; Department of Surgery, George Washington University, Washington DC, USA.
| |
Collapse
|
139
|
Nascimento de Lima P, Maerzluft C, Ozik J, Collier N, Rutter CM. Stress-testing US colorectal cancer screening guidelines: Decennial colonoscopy from age 45 is robust to natural history uncertainty and colonoscopy sensitivity assumptions. Med Decis Making 2025:272989X251334373. [PMID: 40302197 DOI: 10.1177/0272989x251334373] [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: 05/01/2025]
Abstract
PurposeThe 2023 American College of Physicians (ACP) guidelines for colorectal cancer (CRC) screening are at odds with the United States Preventive Task Force (USPSTF) guidelines, with the former recommending screening starting at age 50 y and the latter at age 45 y. This article "stress tests" CRC colonoscopy screening strategies to investigate their robustness to uncertainties stemming from the natural history of disease and sensitivity of colonoscopy.MethodsThis study uses the CRC-SPIN microsimulation model to project the life-years gained (LYG) under several colonoscopy CRC screening strategies. The model was extended to include birth cohort effects on adenoma risk. We estimated natural history parameters under 2 different assumptions about the youngest age of adenoma initiation. For each, we generated 500 parameter sets to reflect uncertainty in the natural history parameters. We simulated 26 colonoscopy screening strategies and examined 4 different colonoscopy sensitivity assumptions, encompassing the range of sensitivities consistent with prior tandem colonoscopy studies. Across this set of scenarios, we identify efficient screening strategies and report posterior credible intervals for benefits of screening (LYG), burden (number of colonoscopies), and incremental burden-effectiveness ratios.ResultsProjected absolute screening benefits varied widely based on assumptions, but strategies starting at age 45 y were consistently in the efficiency frontier. Strategies in which screening starts at age 50 y with 10-y intervals were never efficient, saving fewer life-years than starting screening at age 45 y and performing colonoscopies every 15 y while requiring more colonoscopies per person.ConclusionsDecennial colonoscopy screening initiation at age 45 y remained a robust recommendation. Colonoscopy screening with a 10-y interval starting at age 50 y did not result in an efficient use of colonoscopies in any of the scenarios evaluated.HighlightsColorectal cancer colonoscopy screening strategies initiated at age 45 y were projected to yield more life-years gained while requiring the least number of colonoscopies across different model assumptions about disease natural history and colonoscopy sensitivity.Colonoscopy screening starting at age 50 y with a 10-y interval consistently underperformed strategies that started at age 45 y.
Collapse
Affiliation(s)
| | - Christopher Maerzluft
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jonathan Ozik
- Decision and Infrastructure Sciences, Argonne National Laborator, Argonne, IL, USA
| | - Nicholson Collier
- Decision and Infrastructure Sciences, Argonne National Laborator, Argonne, IL, USA
| | - Carolyn M Rutter
- Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
140
|
Cooke TM, Sofocleous CT, Petre EN, Alexander ES, Ziv E, Solomon SB, Sotirchos VS. Microwave Ablation of Colorectal Pulmonary Metastases Offers Excellent Local Tumor Control and Can Prolong Time Off Chemotherapy. Cardiovasc Intervent Radiol 2025:10.1007/s00270-025-04036-4. [PMID: 40295397 DOI: 10.1007/s00270-025-04036-4] [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: 11/27/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE To evaluate oncologic outcomes after microwave ablation (MWA) of colorectal pulmonary metastases, with focus on disease control without chemotherapy. MATERIALS AND METHODS This institutional review board-approved retrospective study examined patients with oligometastatic or oligoprogressive colorectal pulmonary metastases undergoing MWA between January 2011 and December 2021. Imaging response was assessed with CT at 4-8 weeks post-MWA, with subsequent cross-sectional follow-up imaging every 2-4 months. Local tumor progression-free survival (LTPFS), chemotherapy-free survival (CFS) and overall survival (OS) were calculated using Kaplan-Meier methodology. Variables were evaluated for predictive significance using the log-rank test and Cox regression. RESULTS Two hundred twenty-five patients (127 male, 98 female; median age: 55 years) with 720 pulmonary metastases underwent 400 MWA sessions (mean number of treated metastases per session: 1.8; range 1-9). Mean treated tumor size was 0.9 cm. LTPFS at 1, 2 and 3-years was 91.9%, 85.9% and 81.5%, respectively. Tumors ≥ 1 cm in size, pleural-based tumors and pre-MWA carcinoembryonic antigen (CEA) levels ≥ 10 ng/mL were associated with shorter LTPFS (all P < 0.001). 74.7% (168/225) of patients did not receive chemotherapy for at least two months after the initial MWA. Median CFS was 12 months (95% CI 7.8-16.2) and was significantly prolonged in patients with lung-only disease compared to those with concurrent extrapulmonary disease (34.4 vs. 4.0 months, P < 0.001). Median OS was 47 months (95% CI 36.7-57.3). CONCLUSION MWA of colorectal pulmonary metastases is associated with high local tumor control rates and can offer prolonged CFS, particularly for patients without concurrent extrapulmonary disease.
Collapse
Affiliation(s)
- Timothy M Cooke
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-112B, New York, NY, 10065, USA
| | - Constantinos T Sofocleous
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-112B, New York, NY, 10065, USA
| | - Elena N Petre
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-112B, New York, NY, 10065, USA
| | - Erica S Alexander
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-112B, New York, NY, 10065, USA
| | - Etay Ziv
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-112B, New York, NY, 10065, USA
| | - Stephen B Solomon
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-112B, New York, NY, 10065, USA
| | - Vlasios S Sotirchos
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, H-112B, New York, NY, 10065, USA.
| |
Collapse
|
141
|
Gong Y, Yuan X, Jiao Q, Yu Z. Unveiling fine-scale spatial structures and amplifying gene expression signals in ultra-large ST slices with HERGAST. Nat Commun 2025; 16:3977. [PMID: 40295488 PMCID: PMC12037780 DOI: 10.1038/s41467-025-59139-w] [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: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
We propose HERGAST, a system for spatial structure identification and signal amplification in ultra-large-scale and ultra-high-resolution spatial transcriptomics data. To handle ultra-large spatial transcriptomics (ST) data, we consider the divide and conquer strategy and devise a Divide-Iterate-Conquer framework especially for spatial transcriptomics data analysis, which can also be adopted by other computational methods for extending to ultra-large-scale ST data analysis. To tackle the potential over-smoothing problem arising from data splitting, we construct a heterogeneous graph network to incorporate both local and global spatial relationships. In simulations, HERGAST consistently outperforms other methods across all settings with more than a 10% increase in average adjusted rand index (ARI). In real-world datasets, HERGAST's high-precision spatial clustering identifies SPP1+ macrophages intermingled within colorectal tumors, while the enhanced gene expression signals reveal unique spatial expression patterns of key genes in breast cancer.
Collapse
Affiliation(s)
- Yuqiao Gong
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yuan
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhangsheng Yu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
142
|
Dong Q, Wang T, Bu X, Chen L, Zhong F, Liu C. Latent profile analysis of anxiety, depression, and resilience among elderly colorectal cancer patients in China. Sci Rep 2025; 15:14897. [PMID: 40295714 PMCID: PMC12037835 DOI: 10.1038/s41598-025-99493-9] [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: 10/30/2024] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
In China, most colorectal cancer patients aged 65 years and above often suffer from poor psychological states, including high levels of anxiety and depression, as well as low resilience, due to the disease and its related symptoms. Analyzing the heterogeneity of anxiety, depression, and resilience in these patients can help intervene timely to improve their psychological well-being and prognosis. It was a cross-sectional study, and participants were recruited at the Affiliated Hospital of Qingdao University. Latent profile analysis (LPA) was used to determine the optimal latent profile model, one-way analysis of variance (ANOVA) was conducted to compare the differences across each latent profile, multinomial logistic regression was employed to analyze the influencing factors. 221 older colorectal cancer patients were identified and classified as low negative emotions and high resilience (49.8%), medium negative emotions, low and unstable resilience (30.8%) and high negative emotions and medium resilience (19.4%). There were significant differences in the scores of anxiety, depression, resilience and social support among these patients. Multinomial logistic regression showed that age, gender, marital status, employment, tumor size, positive lymph nodes, degree of differentiation, and social support were influential in these three profiles. Three heterogeneous subgroups of anxiety, depression, and resilience were identified among older patients with colorectal cancer.
Collapse
Affiliation(s)
- Qian Dong
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Ting Wang
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Xiaolong Bu
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Liping Chen
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Feng Zhong
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong Province, China
| | - Cuiping Liu
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China.
| |
Collapse
|
143
|
Ma W, Hou C, Yang M, Wei Y, Mao J, Guan L, Zhao Z. Different MRI-based radiomics machine learning models to predict CD3+ tumor-infiltrating lymphocytes in rectal cancer. Front Oncol 2025; 15:1509207. [PMID: 40356764 PMCID: PMC12066337 DOI: 10.3389/fonc.2025.1509207] [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: 10/10/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Objectives This study aimed to develop and evaluate multiple machine learning models utilizing contrast-enhanced T1-weighted imaging (T1-CE) to differentiate between low-/high-infiltration of total T lymphocytes (CD3) in patients with rectal cancer. Methods We retrospectively selected 157 patients (103 men, 54 women) with pathologically confirmed rectal cancer diagnosed between March 2015 and October 2019. The cohort was randomly divided into a training dataset (n=109) and a test dataset (n=48) for subsequent analysis. Seven radiomic features were selected to generate three models: logistic regression (LR), random forest (RF), and support vector machine (SVM). The diagnostic performance of the three models was compared using the DeLong test. Additionally, Kaplan-Meier analysis was employed to assess disease-free survival (DFS) in patients with high and low CD3+ tumor-infiltrating lymphocyte (TIL) density. Results The three radiomics models performed well in predicting the infiltration of CD3+ TILS, with area under the curve (AUC) values of 0.871, 0.982, and 0.913, respectively, in the training set for the LR, RF, and SVM models. In the validation set, the corresponding AUC values were 0.869, 0.794, and 0.837, respectively. Among the radiomics models, the LR model exhibited superior diagnostic performance and robustness. The merged model, which integrated radiomics features from the SVM model and clinical features from the clinical model, outperformed the individual radiomics models, with AUCs of 0.8932 and 0.8829 in the training and test cohorts, respectively. Additionally, a lower expression level of CD3+ TILs in the cohort was independently correlated with DFS (P = 0.0041). Conclusion The combined model demonstrated a better discriminatory ability in assessing the abundance of CD3+ TILs in rectal cancer. Furthermore, the expression of CD3+ TILs was significantly correlated with DFS, highlighting its potential prognostic value. Advances in knowledge This study is the first attempt to compare the predictive TILs performance of three machine learning models, LR, RF, and SVM, based on the combination of radiomics and immunohistochemistry. The MRI-based combined model, composed of radiomics features from the SVM model and clinical features from the clinical model, exhibited better discriminatory capability for the expression of CD3+ TILs in rectal cancer.
Collapse
Affiliation(s)
- Weili Ma
- Department of Radiology, Shaoxing People’s Hospital, Key Laboratory of Functional Molecular Imaging of Tumor and Interventional Diagnosis and Treatment of Shaoxing City, Shaoxing, China
| | - Chuanling Hou
- Department of Pathology, Shaoxing People’s Hospital, Shaoxing, China
| | - Minxia Yang
- Department of Radiology, Shaoxing People’s Hospital, Key Laboratory of Functional Molecular Imaging of Tumor and Interventional Diagnosis and Treatment of Shaoxing City, Shaoxing, China
| | - Yuguo Wei
- Advanced Analytics, Global Medical Service, GE Healthcare, Hangzhou, China
| | - Jiwei Mao
- Department of Radiotherapy, Shaoxing People’s Hospital, Shaoxing, China
| | - Le Guan
- Department of Radiology, Shaoxing People’s Hospital, Key Laboratory of Functional Molecular Imaging of Tumor and Interventional Diagnosis and Treatment of Shaoxing City, Shaoxing, China
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People’s Hospital, Key Laboratory of Functional Molecular Imaging of Tumor and Interventional Diagnosis and Treatment of Shaoxing City, Shaoxing, China
| |
Collapse
|
144
|
Gosavi R, Nguyen TC, Teoh W, Warrier S, Heriot A, Narasimhan V. Neoadjuvant Therapy in Locally Advanced Colon Cancer: A Case for Selective Adoption in pMMR and Transformation in dMMR. ANZ J Surg 2025. [PMID: 40296274 DOI: 10.1111/ans.70151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/30/2025]
Affiliation(s)
- Rathin Gosavi
- Division of Cancer Surgery, Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Surgery (School of Clinical Sciences at Monash Health), Monash University, Melbourne, Victoria, Australia
| | - T C Nguyen
- Department of Surgery (School of Clinical Sciences at Monash Health), Monash University, Melbourne, Victoria, Australia
| | - William Teoh
- Department of Surgery (School of Clinical Sciences at Monash Health), Monash University, Melbourne, Victoria, Australia
| | - Satish Warrier
- Division of Cancer Surgery, Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alexander Heriot
- Division of Cancer Surgery, Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Vignesh Narasimhan
- Department of Surgery (School of Clinical Sciences at Monash Health), Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
145
|
Han Y, Han Y, Huang W, Liu Y, Wang Z, Zhao W, Zhang W. Effects of nurse-led interventions on enhancing patient-related outcomes in colorectal cancer management throughout the cancer care continuum: A systematic review and meta-analysis. Int J Nurs Stud 2025; 168:105100. [PMID: 40378810 DOI: 10.1016/j.ijnurstu.2025.105100] [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: 08/07/2024] [Revised: 04/13/2025] [Accepted: 04/25/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND Colorectal cancer ranks third in global incidence and second in cancer-related mortality worldwide. Despite persistent challenges across the care continuum, nurses play a pivotal role in optimizing patient outcomes through tailored interventions. However, robust evidence on the clinical efficacy and optimal delivery of nurse-led interventions remains limited. OBJECTIVE To describe the health problems of the participants and nurse-led interventions in accordance with the Omaha System and summarize evidence on the effects of these interventions throughout the continuum from colorectal cancer screening to survivorship. METHODS Eight databases, namely, Web of Science, the Cochrane Library, the Cumulative Index to Nursing and Allied Health Literature, EMBASE, PubMed, Scopus, China National Knowledge Infrastructure and Wanfang Data, were searched from their inception to May 2024. Additionally, the reference lists of the included studies were examined. Content analysis was applied to identify problems and interventions based on the Omaha System. Meta-analysis and descriptive analysis were adopted. Subgroup analyses were conducted on the basis of the study design, treatment history and mode of intervention delivery. RESULTS Twenty-eight studies identified problems and types of nurse-led interventions. There were 13 problems in the psychosocial, physiological and health-related behaviors domains. The nurse-led interventions included teaching, guidance and counseling; case management; and treatments and procedures. Nurse-led interventions improved screening rates for colonoscopy and fecal occult blood testing (odds ratio [OR] = 2.51; 95 % confidence interval [CI]: 2.16 to 2.92; p < 0.001; OR = 6.14; 95 % CI: 1.93 to 19.47; p = 0.002), the adequacy of bowel preparation (OR = 1.69; 95 % CI: 1.40 to 2.03; p < 0.001), stoma self-efficacy (standardized mean difference [SMD] = 2.48; 95 % CI: 0.71 to 4.25; p = 0.006), and quality of life (SMD = 0.72; 95 % CI: 0.21 to 1.22; p = 0.005), and reduced the incidence of stoma complications (OR = 0.28; 95 % CI: 0.18 to 0.42; p < 0.001), anxiety (SMD = -1.19; 95 % CI: -1.40 to -0.99; p < 0.001), and depression (SMD = -1.00; 95 % CI: -1.20 to -0.79; p < 0.001). However, nurse-led interventions were ineffective in reducing distress and addressing unmet supportive care needs. CONCLUSION Nurse-led interventions can reduce stoma complications, anxiety and depression and increase the screening rate for colorectal cancer, the adequacy of bowel preparation, stoma self-efficiency and quality of life. Nevertheless, future rigorous research is needed to validate their effects and inform wider implementation. REGISTRATION PROSPERO (CRD42024505730) registered on February 18, 2024.
Collapse
Affiliation(s)
- Yitong Han
- School of Nursing, Jilin University, Changchun, Jilin Province 130021, China
| | - Yujie Han
- School of Nursing, Jilin University, Changchun, Jilin Province 130021, China
| | - Wenshan Huang
- School of Nursing, Jilin University, Changchun, Jilin Province 130021, China
| | - Yantong Liu
- School of Nursing, Jilin University, Changchun, Jilin Province 130021, China
| | - Ziqi Wang
- School of Nursing, Jilin University, Changchun, Jilin Province 130021, China
| | - Wei Zhao
- School of Nursing, Jilin University, Changchun, Jilin Province 130021, China
| | - Wei Zhang
- School of Nursing, Jilin University, Changchun, Jilin Province 130021, China.
| |
Collapse
|
146
|
Spolverato G, Capelli G, Noel F, Steindler M, Gumbs AA. Pan-immune-inflammation in colon cancer: A prognostic biomarker and the role of tumor location in personalized care. World J Gastrointest Surg 2025; 17:101066. [PMID: 40291895 PMCID: PMC12019070 DOI: 10.4240/wjgs.v17.i4.101066] [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: 09/03/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Despite advances in surgery, chemotherapy, and radiotherapy, the treatment of colorectal cancer (CRC) requires more personalized approaches based on tumor biology and molecular profiling. While some relevant mutations have been associated with differential response to immunotherapy, such as RAS and BRAF mutations limiting response to anti-epithelial growth factor receptor drugs or microsatellite instability predisposing susceptibility to immune checkpoint inhibitors, the role of inflammation in dictating tumor progression and treatment response is still under investigation. Several inflammatory biomarkers have been identified to guide patient prognosis. These include the neutrophil-lymphocyte ratio, Glasgow prognostic score (GPS) and its modified version, lymphocyte-C-reactive protein ratio, and platelet-lymphocyte ratio. However, these markers are not yet included in the standard clinical management of patients with CRC, and further research is needed to evaluate their efficacy in different patient populations. A recent study by Wang et al, published in the World Journal of Gastroenterology, sheds light on the prognostic significance of pan-immune-inflammation value (PIV) in CRC, particularly concerning primary tumor location. Specifically, the authors found that a high PIV was strongly correlated with worse disease-free survival in patients with left-sided colon cancer, whereas no such association was observed in patients with right-sided colon cancer. Integrating tumor location into the prognostic assessment of CRC may improve our ability to more accurately identify high-risk patients and develop personalized treatment plans that are more likely to improve patient outcomes.
Collapse
Affiliation(s)
- Gaya Spolverato
- Department of Surgery, University of Padova, Padua 35122, Italy
| | - Giulia Capelli
- Department of Surgery, ASST Bergamo Est, Bergamo 24068, Lombardy, Italy
| | - Floriane Noel
- Department of Research, Sibylone, Paris 75002, France
| | | | - Andrew Alexander Gumbs
- Department of Surgery, University of Magdeburg, Magdeburg 39130, Saxony-Anhalt, Germany
- Department of Surgery, Service de Chirurgie Digestive Minimale Invasive, Hôpital Antoine Béclère, Assistance Publique-Hôpitaux de Paris, Clamart 92140, France
| |
Collapse
|
147
|
Tsai MH, Shahsavari D, Chen J, Moazzami B, Sridhar S. Racial/Ethnic Disparities in Early-Onset Colorectal Cancer Outcomes. J Racial Ethn Health Disparities 2025:10.1007/s40615-025-02450-5. [PMID: 40287583 DOI: 10.1007/s40615-025-02450-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND AND AIM Although some researchers have examined EO-CRC clinical presentations, much of this research has focused on non-US populations or single healthcare centers. Limited research has also explored outcomes across diverse racial/ethnic groups. Thus, we examined the relationship of five racial/ethnic groups (non-Hispanic White [NHW], non-Hispanic Black [NHB], American Indian/Alaskan Native [AI/AN], Asian/Pacific Islanders [PI], Hispanic) with EO-CRC tumor characteristics/histologic types and risk of CRC death. METHODS We conducted a retrospective cohort analysis using data from the 2006-2020 Surveillance, Epidemiology, and End Results Program. Multivariable Cox proportional hazards regression and logistical regression models were performed. RESULTS Among 46,956 patients, the lower 5-year survival rate was 64.8% among NHB patients (vs. 69.7% for AI/AN, 70.6% for Hispanic, 72.4% for Asian/PI, and 73.4% for NHW patients, p-value < 0.001). In multivariable analysis, NHB, Asian/PI, and Hispanic patients were 10-12% more likely to have late stage at diagnosis and had increased risk of CRC death by 9-37% than NHW patients (p-value < 0.05). Further, NHB patients were 52% more likely to have a right-sided CRC (OR, 1.52; 95% CI, 1.43-1.61), Asian/PI were 15% more likely to have high pathological grading (OR, 1.15; 95% CI, 1.06-1.25), and Hispanic patients were 25% more likely to have MAC/SC subtype (OR, 1.25; 95% CI, 1.16-1.35). CONCLUSION Effective patient-centered communication tailored to the specific needs of racial and ethnic minorities through primary care initiatives may have potential for improving early detection and outcomes, particularly for younger populations and racial minorities.
Collapse
Affiliation(s)
- Meng-Han Tsai
- Georgia Prevention Institute, Augusta University, 1120 15th Street, HS-1705, Augusta, GA, 30912, USA.
- Cancer Prevention, Control, & Population Health Program, Georgia Cancer Center, Augusta University, Augusta, GA, USA.
| | - Dariush Shahsavari
- Department of Medicine, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Jie Chen
- Department of Biostatistics, Data Science and Epidemiology, School of Public Health, Augusta University, Augusta, GA, USA
| | - Bobak Moazzami
- Internal Medicine, Graduate Medical Education, Northside Hospital Gwinnett, Lawrenceville, GA, USA
| | - Subbaramia Sridhar
- Department of Gastroenterology and Hepatology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
148
|
Li N, Zhang N, Wang G. Overexpression of MMP14 is associated with poor prognosis and immune cell infiltration in colon cancer. Front Oncol 2025; 15:1564375. [PMID: 40352589 PMCID: PMC12062125 DOI: 10.3389/fonc.2025.1564375] [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: 01/21/2025] [Accepted: 03/25/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Colorectal cancer (CRC) poses a significant risk of recurrence and distant metastases. This study investigated the regulatory role of Matrix metalloproteinase-14 (MMP14) in immune function and its impact on CRC prognosis. Methods we performed transcriptome sequencing on tumor and adjacent non-cancerous samples from four pairs of patients diagnosed with colorectal cancer. Single-cell transcriptome data were analyzed to explore MMP14 expression and immune microenvironment changes. mRNA expression profiles and clinical data were retrieved from public databases (TCGA and GEO). The association between MMP14 and pathways as well as immune regulators was analyzed. Co-expression genes of MMP14 relevant to prognosis were identified. A prognostic model was then constructed. MMP14 expression was examined using real-time fluorescence quantification PCR (qRT-PCR) and Western blotting (WB). Immunofluorescence was utilized to demonstrate MMP14 expression in colon cancer tissues, while Hematoxylin and eosin (HE) staining was employed to observe the histology of normal tissue and colon cancer tissue. Results Machine learning identified MMP14 as a candidate gene. MMP14 was overexpressed in CRC tissues and COLO205 cells. Single-cell transcriptome analysis revealed that MMP14 was highly expressed in fibrocyte cells within the liver metastasis group. Increased MMP14 levels correlated with poor overall survival (OS), progression-free survival (PFS), and advanced TNM stages. Functional assays indicated that silencing MMP14 in COLO205 cells enhanced apoptosis and upregulated the expression of the immune-related cytokine IL-1β. Furthermore, MMP14 exhibited significant correlations with immunomodulators, particularly immunostimulants and immunosuppressants, and was associated with immune cell infiltration within tumor tissues. Additionally, by utilizing co-expressed genes of MMP14 and conducting Cox regression analysis, we developed a risk prediction model comprising three genes (LIMK1, SPOCK3, SLC2A3). The risk scores derived from this model were found to correlate with OS and PFS. Discussion MMP14 plays a crucial role in CRC progression. Its overexpression is related to poor prognosis and immune cell infiltration. The prognostic model based on MMP14 co-expression genes may help predict CRC prognosis. However, further studies are needed to validate these findings, such as more in-vitro and in-vivo experiments. In conclusion, MMP14 can serve as a biomarker for evaluating CRC prognosis and immune cell infiltration.
Collapse
Affiliation(s)
- Na Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, Shaanxi, China
| | - Nan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, Shaanxi, China
| | - Guanghui Wang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, Shaanxi, China
| |
Collapse
|
149
|
El Muhtaseb MS, Ghanayem A, Almanaseer WN, Alshebelat H, Ghanayem R, Alsheikh GM, Al Karmi F, Al Aruri DO. Assessing awareness of colorectal cancer symptoms, risk factors and screening barriers among eligible adults in Jordan: a cross-sectional study. BMC Public Health 2025; 25:1544. [PMID: 40281527 PMCID: PMC12023689 DOI: 10.1186/s12889-025-22800-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the second most prevalent malignancy in Jordan. Because early detection can greatly improve treatment outcomes, it is crucial to increase awareness of signs and symptoms, risk factors, and the significance of routine CRC screenings. In this study, we aimed to assess awareness levels regarding CRC symptoms and risk factors among adults in Jordan and to identify barriers to CRC screening. METHODS This web-based cross-sectional study was conducted in Jordan from March 5, 2024 to July 9, 2024, and targeted people aged 50-75 years who had no history of CRC. The sample size was calculated via a convenience sampling method. Data were collected via a validated, culturally adapted survey. Descriptive analysis was used when appropriate. Analytic statistics were performed to predict participants' awareness of CRC symptoms and risk factors. RESULTS The study included 400 participants, with a mean age of 58.42 years (SD = 6.511). More than half of the respondents were females (56.5%). The mean awareness score of CRC symptoms among the study participants was 4.97/9 (SD = 1.18), whereas that of risk factors was 5.21/10 (SD = 1.53). The overall mean awareness score was 10.18/19 (SD = 2.65). The top three reported barriers to CRC screening were: not at risk due to absence of symptoms (61.8%), not at risk due to adopting a healthy lifestyle (56.8%), not at risk due to absence of family history (51.8%). CONCLUSION Colorectal cancer awareness among the population was relatively low, with significant symptoms and risk factors being overlooked by the participants. In addition to that, notable barriers to screening, especially fear and embarrassment of the screening test, have surfaced. This prompts the need for more cancer education and healthcare provider involvement to overcome screening barriers and promote participation in screening programs to enable early detection.
Collapse
Affiliation(s)
- M S El Muhtaseb
- Department of General Surgery, School of Medicine, University of Jordan, Amman, Jordan
| | | | | | | | | | - Ghadeer M Alsheikh
- Department of General Surgery, Jordan University Hospital, University of Jordan, Amman, Jordan
| | - Fahed Al Karmi
- Department of General Surgery, Jordan University Hospital, University of Jordan, Amman, Jordan
| | - Daoud O Al Aruri
- Department of General Surgery, Jordan University Hospital, University of Jordan, Amman, Jordan.
| |
Collapse
|
150
|
Zhou Y, Gao Y, Peng Y, Cai C, Han Y, Chen Y, Deng G, Ouyang Y, Shen H, Zeng S, Du Y, Xiao Z. QKI-induced circ_0001766 inhibits colorectal cancer progression and rapamycin resistance by miR-1203/PPP1R3C/mTOR/Myc axis. Cell Death Discov 2025; 11:192. [PMID: 40263288 PMCID: PMC12015279 DOI: 10.1038/s41420-025-02478-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and remains a significant challenge due to high rates of drug resistance and limited therapeutic options. Circular RNAs (circRNAs) are increasingly recognized for their roles in CRC initiation, progression, and drug resistance. However, no circRNA-based therapies have yet entered clinical development, underscoring the need for comprehensive detection and mechanistic studies of circRNAs in CRC. Here, we identified and characterized a circular RNA, circ_0001766 (hsa_circ_0001766), through microarray analysis of CRC tissues. Our results showed that circ_0001766 is downregulated in CRC tissues and closely associated with patient survival and metastasis. Functional experiments demonstrated that circ_0001766 inhibits CRC cell proliferation, migration and invasion both in-vitro and in-vivo. Mechanistically, hypoxia downregulates Quaking (QKI), an RNA-binding protein essential for the biogenesis of circ_0001766 by binding to introns 1 and 3 of PDIA4 pre-mRNA. Reduced QKI expression under hypoxic conditions leads to decreased circ_0001766 levels in CRC. Circ_0001766 acts as a competitive endogenous RNA, sponging miR-1203 to prevent the degradation of PPP1R3C mRNA. Loss of circ_0001766 results in decreased PPP1R3C expression, leading to the activation of mTOR signaling and increased phosphorylation of Myc, which promotes CRC progression and rapamycin resistance. Our study reveals that overexpression of circ_0001766 or PPP1R3C in CRC cells inhibits the mTOR and Myc pathway, thereby resensitizing cells to rapamycin. The combination of circ_0001766 or PPP1R3C with rapamycin markedly inhibits CRC cell proliferation and induces apoptosis by reducing rapamycin-induced Myc phosphorylation. In summary, our study elucidates a critical circ_0001766/miR-1203/PPP1R3C axis that modulates CRC progression and rapamycin resistance. Our findings highlight circ_0001766 as a promising therapeutic target in CRC, providing a new avenue for enhancing the efficacy of existing treatments and overcoming drug resistance.
Collapse
Grants
- This study was supported by grants from the National Natural Science Foundation of China (No. 82373275, 81974384, 82173342 & 82203015), the China Postdoctoral Science Foundation (No.2023JJ40942), three projects from the Nature Science Foundation of Hunan Province (No.2021JJ3109, 2021JJ31048, 2023JJ40942), Nature Science Foundation of Changsha (No.73201), CSCO Cancer Research Foundation (No.Y-HR2019-0182 & Y-2019Genecast-043), the Key Research and Development Program of Hainan Province (No.ZDYF2020228 & ZDYF2020125), Natural Science Foundation (Youth Funding) of Hunan Province of China (2022JJ40458), Hunan Provincial Natural Science Foundation of China (2024JJ6662), The Youth Science Foundation of Xiangya Hospital (2023Q01) and Scientific Research Program of Hunan Provincial Health Commission (202203105261). The graphical abstract was created using BioRender (BioRender.com).This study was supported by grants from the National Natural Science Foundation of China (No. 82373275, 81974384, 82173342 & 82203015), the China Postdoctoral Science Foundation (No.2023JJ40942), three projects from the Nature Science Foundation of Hunan Province (No.2021JJ3109, 2021JJ31048, 2023JJ40942), Nature Science Foundation of Changsha (No.73201), CSCO Cancer Research Foundation (No.Y-HR2019-0182 & Y-2019Genecast-043), the Key Research and Development Program of Hainan Province (No.ZDYF2020228 & ZDYF2020125), Natural Science Foundation (Youth Funding) of Hunan Province of China (2022JJ40458), Hunan Provincial Natural Science Foundation of China (2024JJ6662), The Youth Science Foundation of Xiangya Hospital (2023Q01) and Scientific Research Program of Hunan Provincial Health Commission (202203105261). The graphical abstract was created using BioRender (BioRender.com).
Collapse
Affiliation(s)
- Yulai Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX, USA
| | - Yan Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gongping Deng
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Yanhong Ouyang
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yangfeng Du
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China.
| | - Zemin Xiao
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China.
| |
Collapse
|