1
|
Li S, Liu T, Li C, Zhang Z, Zhang J, Sun D. Overcoming immunotherapy resistance in colorectal cancer through nano-selenium probiotic complexes and IL-32 modulation. Biomaterials 2025; 320:123233. [PMID: 40081224 DOI: 10.1016/j.biomaterials.2025.123233] [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: 10/14/2024] [Revised: 02/11/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND AND OBJECTIVE Colorectal cancer (CRC) is a major global health burden, with immunotherapy often limited by immune tolerance and resistance. This study introduces an innovative approach using Selenium Nanoparticles-Loaded Extracellular Vesicles combined with Interleukin-32 and Engineered Probiotic Escherichia coli Nissle 1917 (SeNVs@NE-IL32-EcN) to enhance CD8+ T cell-mediated immune responses and overcome immunotherapy resistance. METHODS Single-cell RNA sequencing (scRNA-seq) and transcriptomic analyses were performed to identify key immune cells and regulators involved in CRC immunotherapy resistance, focusing on CD8+ T cells and the regulatory factor IL32. A humanized xenograft mouse model was used to evaluate the impact of IL32 and SeNVs@NE-IL32-EcN on tumor growth and immune responses. The SeNVs@NE-IL32-EcN complex was synthesized through a reverse micelle method and functionalized using extracellular vesicles. Its morphology, size, antioxidant activity, and safety were characterized using electron microscopy, dynamic light scattering (DLS), and in vitro co-culture assays. RESULTS Single-cell analyses revealed a significant reduction in CD8+ T cell infiltration in immunotherapy-resistant CRC patients. IL32 was identified as a key regulator enhancing CD8+ T cell cytotoxic activity through granzyme B and IFN-γ secretion. Treatment with SeNVs@NE-IL32-EcN significantly improved the proliferation and activity of CD8+ T cells and reduced tumor progression in humanized mouse models. In vitro and in vivo results demonstrated the complex's biocompatibility, antioxidant properties, and ability to enhance CRC immunotherapy while mitigating immune tolerance. CONCLUSION SeNVs@NE-IL32-EcN offers a novel nano-biomaterial strategy that integrates nanotechnology and probiotic therapy to enhance CD8+ T cell-mediated immunity and overcome CRC immunotherapy resistance. This study lays the foundation for future therapeutic applications in cancer treatment by advancing immune-modulating biomaterials.
Collapse
Affiliation(s)
- Shiquan Li
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Tao Liu
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Chenyao Li
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhiyuan Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Jiantao Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China
| | - Di Sun
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Mehanna MG, El-Halawany AM, Al-Abd AM, Alqurashi MM, Bukhari HA, Kazmi I, Al-Qahtani SD, Bawadood AS, Anwar F, Al-Abbasi FA. 6-Paradol enhances sorafenib's effects against colorectal cancer and promotes active metabolite formation. Pathol Res Pract 2025; 269:155876. [PMID: 40086339 DOI: 10.1016/j.prp.2025.155876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/03/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
AIM This research aimed to investigate the effect of 6-Paradol on sorafenib cytotoxicity in colorectal cancer (CRC) cell lines (HT-29, HCT-116, LS-174T, CaCo-2) through its effects on cellular processes. BACKGROUND CRC, which ranks as the third most prevalent cancer, arises in the colon or rectum due to a multifaceted interplay of dietary, lifestyle, genetic, and age-related factors. Sorafenib (multikinase inhibitor), a drug targeting multiple cancer signals, shows promise against liver, thyroid, and kidney tumors but faces resistance in CRC. 6-Paradol (Zingiberaceae), a natural compound, offers the potential for overcoming this resistance. OBJECTIVE The current study investigates whether 6-paradol potentiates sorafenib's anti-cancer activity in CRC cells by promoting increased drug entrapment, uptake, and metabolism. METHOD Sulpharodamine B assay, caspase-3 activity, PARP cleavage, cell cycle distribution analysis, and P-gp efflux activity were assessed for Cytotoxicity using CRC cell lines (HT-29, HCT-116, LS-174T, CaCo-2). RESULT Sorafenib showed potent cytotoxicity across CRC cell lines (IC50: 2.0-11.1 µM). Notably, 6-Paradol synergistically enhanced its effect in HT-29 and HCT-116 cells, reducing IC50 from 338 ± 55.7 to 107 ± 5.3 µM, respectively. 6-Paradol synergized with sorafenib, reducing HCT-116 cell proliferation from from 14.25 ± 0.62 to 20.7 ± 0 fg/cell and Paradol didn't show any significant change in c-PARP concentration which equaled 289.82 ± 27.32 pg/cell, suggesting enhanced cytotoxicity. 6-Paradol exposure resulted in a concentration-dependent (1-100 µM) increase in intracellular p-gp accumulation in CaCo-2, HCT-116, and HT-29. Co-incubation with 6-paradol significantly enhanced sorafenib cellular internalization within CaCo-2 cells, resulting in a concomitant decrease in its extracellular concentration from 1491 ± 132 ng/ml to 216 ± 39 ng/ml. 6-Paradol increased sorafenib intracellular accumulation ranges from 239 ± 16 to 327 ± 34 pg/cell, respectively in HCT-116 cells. 6-Paradol significantly increased the intracellular conversion of sorafenib to its active N-oxide metabolite and facilitated its retention within the CaCo-2 cell compartment. CONCLUSION In conclusion, 6-Paradol marked elevation in sorafenib's cytotoxic profile by influencing its cellular uptake and metabolism. Future research should expand in vitro studies to diverse cell lines, conducting in vivo elucidating underlying mechanisms and rigorously evaluating safety and toxicity.
Collapse
Affiliation(s)
- Mohamed G Mehanna
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ali M El-Halawany
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr-El-Ainy Street, Cairo 11562, Egypt.
| | - Ahmed M Al-Abd
- Pharmacology Department, Medical Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt.
| | - May M Alqurashi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Hussam A Bukhari
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Salwa D Al-Qahtani
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia.
| | - Azizah Salim Bawadood
- Basic Medical Sciences Department, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Wang W, Li X, Wang H, Huang C, Zhu L, Wang H, Zhang W. CircDIAPH1 Promotes Liver Metastasis and Development of Colorectal Cancer by Initiation of CEACAM6 Expression. Mol Carcinog 2025; 64:897-910. [PMID: 39987565 DOI: 10.1002/mc.23896] [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: 11/08/2024] [Revised: 01/25/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025]
Abstract
Liver metastasis is a critical factor influencing the 5-year survival rate in colorectal cancer (CRC). However, the biological function of most circRNAs in liver metastasis of CRC is still unknown. In this study, we identified differentially expressed circRNAs associated with liver metastasis (LM-DE-circRNAs). A total of 247 LM-DE-circRNAs were identified, and crucial signaling pathways, including the regulation of actin cytoskeleton, were significantly enriched, featuring six LM-DE-circRNAs. Notably, circDIAPH1 (hsa_circ_0074323), with the highest AUC value, emerged as a potential biomarker for CRC liver metastasis (CRLM). Functional assays following circDIAPH1 knockdown demonstrated induced apoptosis, suppressed proliferation, reduced metastasis, and invasion in CRC cell lines in vitro. The circDIAPH1 knockdown attenuated tumor growth in a cell-derived xenograft model. Furthermore, circDIAPH1 knockdown lessened the liver metastasis. Transcriptome profiling revealed that CEACAM6 was the most downregulated gene while circDIAPH1 was knocked down, and possesses high expression value in CRC. Most importantly, we found that circDIAPH1 recruited transcription factor FOXA1 to bind in the promoter region of CEACAM6 and initiated CEACAM6 expression. Additionally, the study identified the transcription factor BRD4 as a regulator of circDIAPH1 expression in CRC. In conclusion, this study reveals that circDIAPH1 recruits FOXA1 to initiate CEACAM6 expression, promoting liver metastasis and development of CRC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xu Li
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hantao Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Cheng Huang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Laicheng Zhu
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Hao Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei Zhang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
6
|
Ding YW, Chen G, Hua HJ, Lu C. Correlation of Epstein-Barr virus infection with pathological characteristics of colorectal cancer and its short-term prognosisr after chemotherapy. Shijie Huaren Xiaohua Zazhi 2025; 33:316-323. [DOI: 10.11569/wcjd.v33.i4.316] [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: 03/09/2025] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 04/28/2025] Open
Abstract
BACKGROUND In recent years, studies have demonstrated that Epstein-Barr virus (EBV) may play a role in the occurrence of colorectal cancer. Currently, clinical reports on EBV involved in tumor cell proliferation, differentiation, and migration are mainly concentrated in gastric cancer, nasopharyngeal carcinoma, B-cell lymphoma, etc., which have the functions of promoting tumor development and regulating the body's immune microenvironment. The relationship between EBV and colorectal cancer has been less explored.
AIM To analyze the correlation between EBV infection and the pathological characteristics of colorectal cancer as well as its short-term prognosis after chemotherapy to provide reference for its clinical diagnosis, treatment, and prognosis evaluation.
METHODS A total of 45 EBV-positive and 180 EBV-negative colorectal cancer patients admitted to Jinhua Central Hospital from May 2021 to March 2024 were selected. The relationship between EBV infection and the clinicopathological characteristics of colorectal cancer was analyzed, and the EBV-DNA load was compared in patients with different pathological characteristics in the EBV-positive group. The correlation between EBV-DNA load and pathological characteristics of colorectal cancer was analyzed. The short-term prognosis of colorectal cancer patients after chemotherapy was recorded. The pathological characteristics and EBV infection status was compared in patients with different short-term prognoses. The factors that affect the patients' short-term prognosis were analyzed. The predictive value of EBV infection and pathological characteristics for the short-term prognosis of colorectal cancer patients was analyzed.
RESULTS The proportions of patients with stage Ⅲ + Ⅳ, low differentiation, deep invasion (T3 + T4), and lymph node metastasis in the EBV-positive group were higher than those of the EBV-negative group (P < 0.05). EBV-DNA load differed significantly in patients with different degrees of differentiation, infiltration depth, tumor stages, and lymph node metastasis in the EBV-positive group (P < 0.05). EBV-DNA load in patients with EBV-infected colorectal cancer was negatively correlated with the degree of differentiation, and positively correlated with tumor stage, lymph node metastasis, and infiltration depth (P < 0.05). Lymph node metastasis, degree of differentiation, tumor stage, infiltration depth, and EBV infection were all independent factors affecting the short-term prognosis of colorectal cancer patients (P < 0.05). The area under the curve of the combination of tumor stage, infiltration depth, lymph node metastasis, degree of differentiation, and EBV infection in predicting the short-term prognosis of colorectal cancer patients was 0.882 (95% confidence interval: 0.832-0.921), with a sensitivity of 84.93% and specificity of 91.45% (P < 0.05).
CONCLUSION EBV infection is significantly correlated with tumor stage, differentiation degree, depth of invasion, lymph node metastasis, and short-term prognosis after chemotherapy in colorectal cancer patients. The combination of pathological characteristics with EBV detection has high predictive value for the short-term prognosis of colorectal cancer patients.
Collapse
Affiliation(s)
- Ying-Wei Ding
- Department of Emergency Medicine, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang Province, China
| | - Gang Chen
- Department of Emergency Medicine, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang Province, China
| | - Hong-Jun Hua
- Department of Emergency Medicine, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang Province, China
| | - Chong Lu
- Department of Emergency Medicine, Jinhua Municipal Central Hospital, Jinhua 321000, Zhejiang Province, China
| |
Collapse
|
7
|
Zheng ZP, Zhang YG, Long MB, Ji KQ, Peng JY, He K. Construction of a risk prediction model for postoperative cognitive dysfunction in colorectal cancer patients. World J Gastrointest Surg 2025; 17:104459. [PMID: 40291874 PMCID: PMC12019063 DOI: 10.4240/wjgs.v17.i4.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most prevalent and lethal malignant tumors worldwide. Currently, surgical intervention was the primary treatment modality for CRC. However, increasing studies have revealed that CRC patients may experience postoperative cognitive dysfunction (POCD). AIM To establish a risk prediction model for POCD in CRC patients and investigate the preventive value of dexmedetomidine (DEX). METHODS A retrospective analysis was conducted on clinical data from 140 CRC patients who underwent surgery at the People's Hospital of Qian Nan from February 2020 to May 2024. Patients were allocated into a modeling group (n = 98) and a validation group (n = 42) in a 7:3 ratio. General clinical data were collected. Additionally, in the modeling group, patients who received DEX preoperatively were incorporated into the observation group (n = 54), while those who did not were placed in the control group (n = 44). The incidence of POCD was recorded for both cohorts. Data analysis was performed using statistical product and service solutions 20.0, with t-tests or χ 2 tests employed for group comparisons based on the data type. Least absolute shrinkage and selection operator regression was applied to identify influencing factors and reduce the impact of multicollinear predictors among variables. Multivariate analysis was carried out using Logistic regression. Based on the identified risk factors, a risk prediction model for POCD in CRC patients was developed, and the predictive value of these risk factors was evaluated. RESULTS Significant differences were observed between the cognitive dysfunction group and the non-cognitive dysfunction group in diabetes status, alcohol consumption, years of education, anesthesia duration, intraoperative blood loss, intraoperative hypoxemia, use of DEX during surgery, intraoperative use of vasoactive drugs, surgical time, systemic inflammatory response syndrome (SIRS) score (P < 0.05). Multivariate Logistic regression analysis identified that diabetes [odds ratio (OR) = 4.679, 95% confidence interval (CI) = 1.382-15.833], alcohol consumption (OR = 5.058, 95%CI: 1.255-20.380), intraoperative hypoxemia (OR = 4.697, 95%CI: 1.380-15.991), no use of DEX during surgery (OR = 3.931, 95%CI: 1.383-11.175), surgery duration ≥ 90 minutes (OR = 4.894, 95%CI: 1.377-17.394), and a SIRS score ≥ 3 (OR = 4.133, 95%CI: 1.323-12.907) were independent risk factors for POCD in CRC patients (P < 0.05). A risk prediction model for POCD was constructed using diabetes, alcohol consumption, intraoperative hypoxemia, non-use of DEX during surgery, surgery duration, and SIRS score as factors. A receiver operator characteristic curve analysis of these factors revealed the model's predictive sensitivity (88.56%), specificity (70.64%), and area under the curve (AUC) (AUC = 0.852, 95%CI: 0.773-0.919). The model was validated using 42 CRC patients who met the inclusion criteria, demonstrating sensitivity (80.77%), specificity (81.25%), and accuracy (80.95%), and AUC (0.805) in diagnosing cognitive impairment, with a 95%CI: 0.635-0.896. CONCLUSION Logistic regression analysis identified that diabetes, alcohol consumption, intraoperative hypoxemia, non-use of DEX during surgery, surgery duration, and SIRS score vigorously influenced the occurrence of POCD. The risk prediction model based on these factors demonstrated good predictive performance for POCD in CRC individuals. This study offers valuable insights for clinical practice and contributes to the prevention and management of POCD under CRC circumstances.
Collapse
Affiliation(s)
- Zhen-Ping Zheng
- Department of Anesthesiology, The People’s Hospital of Qian Nan, Duyun 558000, Guizhou Province, China
| | - Yong-Guo Zhang
- Department of Anesthesiology, The People’s Hospital of Qian Nan, Duyun 558000, Guizhou Province, China
| | - Ming-Bo Long
- Department of Anesthesiology, The People’s Hospital of Qian Nan, Duyun 558000, Guizhou Province, China
| | - Kui-Quan Ji
- Department of Anesthesiology, The People’s Hospital of Qian Nan, Duyun 558000, Guizhou Province, China
| | - Jin-Yan Peng
- Department of Anesthesiology, The People’s Hospital of Qian Nan, Duyun 558000, Guizhou Province, China
| | - Kai He
- Department of Anesthesiology, The People’s Hospital of Qian Nan, Duyun 558000, Guizhou Province, China
| |
Collapse
|
8
|
Li WB, Li J, Yu W, Gao JH. Short-term efficacy of laparoscopic radical resection for colorectal cancer and risk of unplanned reoperation after surgery. World J Gastrointest Surg 2025; 17:102442. [PMID: 40291873 PMCID: PMC12019040 DOI: 10.4240/wjgs.v17.i4.102442] [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: 01/08/2025] [Revised: 02/11/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Surgery is the first choice of treatment for patients with colorectal cancer. Traditional open surgery imparts great damage to the body of the patient and can easily cause adverse stress reactions. With the continuous development of medical technology, laparoscopic minimally invasive surgery has shown great advantages for the treatment of patients with celiac disease. AIM To investigate the short-term efficacy of laparoscopic radical surgery and traditional laparotomy for the treatment of colorectal cancer, and the differences in the risk analysis of unplanned reoperation after operation. METHODS As the research subjects, this study selected 100 patients with colorectal cancer who received surgical treatment at the Yulin First Hospital from January 2018 to January 2022. Among them, 50 patients who underwent laparoscopic radical resection were selected as the research group and 50 patients who underwent traditional laparotomy were selected as the control group. Data pertaining to clinical indexes, gastrointestinal hormones, nutrition indexes, the levels of inflammatory factors, quality of life, Visual Analog Scale score, and the postoperative complications of the two groups of patients before and after treatment were collected, and the therapeutic effects in the two groups were analyzed and compared. RESULTS Compared with the control group, perioperative bleeding, peristalsis recovery time, and hospital stays were significantly shorter in the research group. After surgery, the levels of gastrin (GAS) and motilin (MTL) were decreased in both groups, and the fluctuation range of GAS and MTL observed in the research group was significantly lower than that recorded in the control group. The hemoglobin (Hb) levels increased after surgery, and the level of Hb in the research group was significantly higher compared with the control group. After the operation, the expression levels of tumor necrosis factor-α, interleukin-6, and C-reactive protein and the total incidence of complications were significantly lower in the research group compared with the control group. One year after the operation, the quality of life of the two groups was greatly improved, with the quality of life in the research group being significantly better. CONCLUSION Laparoscopy was effective for colorectal surgery by reducing the occurrence of complications and inflammatory stress reaction; moreover, the quality of life of patients was significantly improved, which warrants further promotion.
Collapse
Affiliation(s)
- Wen-Bin Li
- Department of General Surgery, Yulin First Hospital, Yulin 719000, Shaanxi Province, China
| | - Jiang Li
- Department of General Surgery, Yulin First Hospital, Yulin 719000, Shaanxi Province, China
| | - Wei Yu
- Department of General Surgery, Yulin First Hospital, Yulin 719000, Shaanxi Province, China
| | - Jian-Hua Gao
- Department of General Surgery, Yulin First Hospital, Yulin 719000, Shaanxi Province, China
| |
Collapse
|
9
|
Tang QY, Yu W. Logistic regression analysis of pathological features of bone metastasis in colorectal cancer and related influencing factors after surgery. World J Gastrointest Surg 2025; 17:100851. [PMID: 40291899 PMCID: PMC12019058 DOI: 10.4240/wjgs.v17.i4.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignant tumor in the digestive system, whose main treatment comprises surgical resection, radiotherapy and chemotherapy, and targeted drug therapy. At present, the radical resection of CRC is the main way of achieving an early cure. AIM To investigate the logistic regression analysis of bone metastasis after CRC surgery and related influencing factors. METHODS We selected 100 patients who underwent surgery for CRC and were admitted from February 2018 to February 2024, collected the general data of bone metastasis, and collected the pathological characteristics of patients with bone metastasis. Next, we divided them into groups with and without bone metastasis (Bone metastases group, n = 44; no bone metastases group, n = 56), compared the clinical data of the two groups, and analyzed the risk factors of bone metastasis using logistic regression analysis. RESULTS Among the 100 patients, the mean age was 54.33 ± 8.45 years, and most were male (54.55%). The proportion of patients with lytic bone changes was 43.18%. The most common location of combined bone metastasis was the pelvis, whereas only 5 patients had limb transfer. There was a higher incidence of lung than of pancreatic or liver metastases. Regression analysis showed that the primary location of the cancer was rectal cancer. Lymph node involvement, lung metastasis, and no postoperative chemotherapy were the risk factors for postoperative bone metastasis in patients who underwent surgery for CRC (P < 0.05). CONCLUSION Rectal cancer, lymph node involvement, complicated pulmonary metastasis, and no postoperative chemotherapy treatment can help predict high risk of bone metastasis in CRC.
Collapse
Affiliation(s)
- Qiu-Yan Tang
- The First Department of Oncology, Jiujiang First People’s Hospital, Jiujiang 332000, Jiangxi Province, China
| | - Wei Yu
- Department of Orthopedics, Jiujiang First People’s Hospital, Jiujiang 332000, Jiangxi Province, China
| |
Collapse
|
10
|
Zhou K, Yu Y, Li W, Zhu M. Clostridium butyricum Regulates the Inflammatory and Immunoregulatory Pathway Through NFKB1 in Colorectal Cancer Treatment. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10547-w. [PMID: 40279041 DOI: 10.1007/s12602-025-10547-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Colorectal cancer (CRC) ranks among the top three most prevalent malignancies globally and is a leading cause of cancer-related mortality. Traditional therapeutic approaches usually cause significant adverse effects, highlighting the urgent demand for alternative, more effective treatments. Probiotics have gained attentions as potential cancer therapy due to their beneficial impacts on host health. Clostridium butyricum (Cl. butyricum) has shown anticancer properties in recent studies, though the underlying mechanisms remain inadequately understood. This study presents an integrative analysis of network pharmacology and proteomics to elucidate the key targets of Cl. butyricum in CRC treatment. The network pharmacology analysis identified 72 overlapping genes, and functional analysis of these genes indicated that most pathways were related to pathways in cancer and inflammation, and butyrate emerging as the pivotal product of Cl. butyricum due to its strong associations with the identified hub genes. In parallel, proteomics analysis revealed 168 differential expressed proteins (DEPs) in Cl. butyricum-treated HCT-116 cells, comprising 78 upregulated and 90 downregulated proteins. These DEPs were primarily enriched in apoptosis and inflammatory pathways. PPI analysis further highlighted NFKB1 as key contributors to the anticancer effects of Cl. butyricum. The integrative analysis revealed a significant convergence of pathways enrichment patterns, particularly in inflammatory and immune-related pathways. Computational and experimental validation identified NFKB1 as a pivotal molecular target in CRC intervention. These collective findings elucidate the mechanistic basis of the antitumor properties of Cl. butyricum, highlighting its regulatory effects on NFKB1 through both inflammatory and, to a lesser extent, immunoregulatory pathways.
Collapse
Affiliation(s)
- Kun Zhou
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China
| | - Yue Yu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China
| | - Wei Li
- College of Medicine, Translational Medicine Research Institute, Yangzhou University, Yangzhou, 225001, China
| | - Mingchang Zhu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China.
- College of Environmental and Safety Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China.
| |
Collapse
|
11
|
Lu Y, Zhao X, Liu H, Zhang Y, Guo L, Wang L. The prognostic significance of tumor-specific growth factor in colorectal cancer: an observation study. BMC Gastroenterol 2025; 25:280. [PMID: 40259216 PMCID: PMC12013182 DOI: 10.1186/s12876-025-03902-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 04/16/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND The correlation between tumor-specific growth factor (TSGF) and the prognosis of colorectal cancer (CRC)has not been extensively studied. Research was designed to evaluate the potential of TSGF as a predictive marker for the prognosis of patients with CRC. METHODS The serum TSGF was quantified and stratified into elevated/normal groups based on the established threshold detected using the rate method. The log-rank test was employed for survival analysis to evaluate the 3-year overall survival (OS). Prognostic independence was verified through Cox proportional hazards model. The receiver operating characteristic (ROC) curves were plotted to assess the clinical application value of TSGF. RESULTS A total of 2223 patients were incorporated in our study. The survival analysis revealed that elevated TSGF levels were associated with poorer survival in CRC. The multivariate Cox analysis demonstrated TSGF as an independent prognostic factor. The signature covering TSGF with T staging, N staging, CEA, CA199 demonstrated superior predictive capability based on the Area Under the Curve (AUC) (Delong's test p = 0.025). CONCLUSION TSGF is an independent factor correlating with the 3-year OS of stage I-III CRC patients. Moreover, our new signature including TSGF can effectively improve the prognostic evaluation of CRC, thereby contributing to precision medication.
Collapse
Affiliation(s)
- Yanjun Lu
- Department of Colorectal Surgery, Cancer Hospital, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Xinxuan Zhao
- Department of Colorectal Surgery, Cancer Hospital, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Haiyi Liu
- Department of Colorectal Surgery, Cancer Hospital, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yixun Zhang
- Department of Colorectal Surgery, Cancer Hospital, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Linghong Guo
- Department of Colorectal Surgery, Cancer Hospital, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Lichun Wang
- Department of Colorectal Surgery, Cancer Hospital, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
12
|
Yuan T, Liu Y, Wu R, Qian M, Wang W, Li Y, Zhu H, Wang J, Ge F, Zeng C, Dai X, Hu R, Zhou T, He Q, Zhu H, Yang B. Josephin Domain Containing 2 (JOSD2) inhibition as Pan-KRAS-mutation-targeting strategy for colorectal cancer. Nat Commun 2025; 16:3623. [PMID: 40240366 PMCID: PMC12003847 DOI: 10.1038/s41467-025-58923-y] [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: 07/30/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
KRAS is the most common mutated oncogenes in colorectal cancer (CRC), yet effective therapeutic strategies for targeting multiple KRAS mutations remained challenging. The prolonged protein stability of KRAS mutants contribute to their robust tumor-promoting effects, but the underlying mechanism is elusive. Herein by screening deubiquitinases (DUBs) siRNA library, we identify Josephin domain containing 2 (JOSD2) functions as a potent DUB that regulates the protein stability of KRAS mutants. Mechanistically, JOSD2 directly interacts with and stabilizes KRAS variants across different mutants, by reverting their proteolytic ubiquitination; while KRAS mutants reciprocally inhibit the catalytic activity of CHIP, a bona fide E3 ubiquitin ligase for JOSD2, thus forming a JOSD2/KRAS positive feedback circuit that significantly accelerates KRAS-mutant CRC growth. Inhibition of JOSD2 by RNA interference or its pharmacological inhibitor promotes the polyubiquitination and proteasomal degradation of KRAS mutants, and preferentially impede the growth of KRAS-mutant CRC including patient-derived cells/xenografts/organoids (PDCs/PDXs/PDOs) over that harboring wild-type KRAS. Collectively, this study not only reveals the crucial roles of JOSD2/KRAS positive feedback circuit in KRAS-mutant CRC, but also provides a rationale to target JOSD2 as the promising pan-KRAS-mutation-targeting strategy for the treatment of a broad population of CRC patients with KRAS variant across different mutant types.
Collapse
Affiliation(s)
- Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yue Liu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ruilin Wu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meijia Qian
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Weihua Wang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yonghao Li
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hongdao Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jia'er Wang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fujing Ge
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chenming Zeng
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyang Dai
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
| | - Ronggui Hu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- School of Medicine, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
13
|
Tian HP, Xiao ZX, Su BW, Li YX, Peng H, Meng CY. Impact of SLC16A8 on tumor microenvironment and angiogenesis in colorectal cancer: New therapeutic target insights. World J Gastrointest Oncol 2025; 17:99188. [PMID: 40235880 PMCID: PMC11995316 DOI: 10.4251/wjgo.v17.i4.99188] [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: 07/16/2024] [Revised: 11/08/2024] [Accepted: 01/15/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND SLC16A8, a lactate efflux transporter, is upregulated in various cancers, but its effects on tumor microenvironments remain understudied. This research explores its role in colorectal cancer (CRC) and the impact on the associated microenvironment consisting of vascular endothelial cells. AIM To explore the role in CRC and the impact on the associated microenvironment consisting of vascular endothelial cells. METHODS Hypoxic conditions prompted examination of SLC16A8 expression, glycolysis, lactate efflux, and Warburg effect correlations in CRC cell lines. Co-culture with HUVEC allowed for endothelial-mesenchymal transition (EndMT) characterization, revealing lactate efflux's influence. Knockdown of SLC16A8 in CRC cells enabled relevant phenotype tests and tumorigenesis experiments, investigating tumor growth, blood vessel distribution, and signaling pathway alterations. RESULTS SLC16A8 expression was significantly upregulated in CRC tissues compared to adjacent normal tissues and correlated with disease progression (P < 0.05). Under hypoxic conditions, HIF-1α induced SLC16A8 expression, leading to enhanced metabolic reprogramming and increased lactate production. siRNA-mediated SLC16A8 knockdown effectively reversed hypoxia-induced changes, including reduced glucose consumption and lactate production. Co-culture experiments revealed that SLC16A8 knockdown significantly inhibited hypoxia-induced EndMT in HUVEC cells. In vivo studies demonstrated that SLC16A8 knockdown suppressed tumor growth, reduced Ki67 expression, and decreased HIF-1α levels. Furthermore, SLC16A8 silencing led to decreased expression of key metabolic enzymes PKM2 and LDHA, indicating its role in glycolytic regulation. CONCLUSION Our findings reveal that SLC16A8 functions as a critical mediator of hypoxia-induced metabolic reprogramming in CRC progression.
Collapse
Affiliation(s)
- Hong-Peng Tian
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Zhong-Xiang Xiao
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Bo-Wen Su
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Yi-Xuan Li
- Department of Premarital and Prenatal Examination, Nanchong Shunqing District Maternal and Child Health Hospital, Nanchong 637000, Sichuan Province, China
| | - Hong Peng
- Department of Anorectal Surgery, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Chang-Yuan Meng
- Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| |
Collapse
|
14
|
Zhang W, Yang C, Lu Y, Tang C, Zhao M, Wang Z, Gao J, Hu S, Chen Z. Cancer-associated fibroblasts gene signature: a novel approach to survival prediction and immunotherapy guidance in colon cancer. Front Immunol 2025; 16:1532306. [PMID: 40264753 PMCID: PMC12011795 DOI: 10.3389/fimmu.2025.1532306] [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: 11/21/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Background Fibroblasts can regulate tumour development by secreting various factors. For COAD survival prediction and CAFs-based treatment recommendations, it is critical to comprehend the heterogeneity of CAFs and find biomarkers. Methods We identified fibroblast-associated specific marker genes in colon adenocarcinoma by single-cell sequencing analysis. A fibroblasts-related gene signature was developed, and colon adenocarcinoma patients were classified into high-risk and low-risk cohorts based on the median risk score. Additionally, the impact of these risk categories on the tumor microenvironment was evaluated. The ability of CAFGs signature to assess prognosis and guide treatment was validated using external cohorts. Ultimately, we verified MAN1B1 expression and function through in vitro assays. Results Relying on the bulk RNA-seq and scRNA-seq data study, we created a predictive profile with 11 CAFGs. The profile effectively differentiated survival differences among cohorts of colon adenocarcinoma patients. The nomogram further effectively predicted the prognosis of COAD patients, with low-risk patients having a better prognosis. A higher immune infiltration rate and lower IC50 values of anticancer drugs were significant in the high-risk group. In cellular experiments, Following MAN1B1 knockdown, in cell assays, the colony formation, migration, and invasion ability of HCT116 and HT29 cell lines decreased. Conclusion Our CAFG signature provides important insights into the role of CAF cells in influencing COAD prognosis. It may also serve as a guide for selecting immunotherapy options and predicting chemotherapy responses in COAD patients.
Collapse
Affiliation(s)
- Wenbing Zhang
- Department of General Surgery, Anqing First People’s Hospital of Anhui Medical University, Anqing, China
| | - Chi Yang
- Department of General Surgery, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, QingPu District Central Hospital Shanghai, No. 1158, Gong Yuan Dong Road, Shanghai, China
| | - Ye Lu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Chenling Tang
- The First People’s Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mengyu Zhao
- Department of General Surgery, Anqing First People’s Hospital of Anhui Medical University, Anqing, China
| | - Zhaohui Wang
- Department of General Surgery, Anqing First People’s Hospital of Anhui Medical University, Anqing, China
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Shuangjiu Hu
- Department of General Surgery, Anqing First People’s Hospital of Anhui Medical University, Anqing, China
| | - Zhihua Chen
- The First People’s Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
15
|
Wang Z, Li T, Sun M, Liu N, Zhang H, Feng Z, Lei N. Metabolomics- and Proteomics-Based Disease Diagnostic Classifier Model for the Prediction and Diagnosis of Colorectal Carcinoma. J Proteome Res 2025; 24:2096-2111. [PMID: 40108892 PMCID: PMC11977543 DOI: 10.1021/acs.jproteome.5c00010] [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: 01/12/2025] [Revised: 02/28/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Colorectal carcinoma (CRC) is a leading cause of cancer-related deaths globally. Diagnostic biomarkers are essential for risk stratification and early detection, potentially enhancing patient survival. Our study aimed to explore the potential biomarkers of CRC at the protein and metabolic levels. METHODS Blood serum from CRC patients and healthy controls was analyzed using metabolomic and proteomic techniques. A conjoint analysis was conducted, and samples were split into training and validation sets (7:3 ratio) to develop and evaluate a disease diagnosis classifier model. Immunohistochemistry (IHC) analyses were conducted to validate the results. RESULTS We identified 631 differential metabolites and 61 differentially expressed proteins (DEPs) in CRC, involved in pathways such as arginine and proline metabolism, central carbon metabolism in cancer, and signaling pathways including TGF-β, mTOR, PI3K-Akt, and others. Key proteins (CILP2, SLC3A2, EXTL2, hydroxypyruvate isomerase (HYI), ENPEP, LRG1, CTSS, thyrotropin-releasing hormone-degrading ectoenzyme (TRHDE), SELE, and HSPA1A) showed significant expression differences between CRC patients and controls. IHC results showed that compared with the paracancerous tissues, the expression of CILP2, EXTL2, and HYI was significantly downregulated in the CRC tissues (P < 0.05). The classifier model, comprising l-arginine, Harden-Young ester, l-aspartic acid, oxoglutaric acid, l-proline, octopine, l-valine, and progesterone, achieved AUC values of 0.998 and 0.914 in training and validation data sets, respectively. CONCLUSIONS The identified metabolites and DEPs are promising CRC biomarkers. The developed classifier model based on eight metabolites demonstrates high accuracy for CRC assessment and diagnosis.
Collapse
Affiliation(s)
- Zhaorui Wang
- Translational
Medicine Research Center, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Tianyuan Li
- Department
of Gastroenterology, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Mengyao Sun
- Department
of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Na Liu
- Translational
Medicine Research Center, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Haozhe Zhang
- Translational
Medicine Research Center, The Fifth Clinical
Medical College of Henan University of Chinese Medicine (Zhengzhou
People’s Hospital), Zhengzhou, Henan 450000, China
| | - Zhikun Feng
- Department
of Pathology, The Fifth Clinical Medical
College of Henan University of Chinese Medicine (Zhengzhou People’s
Hospital), Zhengzhou, Henan 450000, China
| | - Ningjing Lei
- Department
of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
16
|
Xu X, Zhang Y, Huang G, Perekatt A, Wang Y, Chen L. Advances and applications of gut organoids: modeling intestinal diseases and therapeutic development. LIFE MEDICINE 2025; 4:lnaf012. [PMID: 40276096 PMCID: PMC12018802 DOI: 10.1093/lifemedi/lnaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Gut organoids are 3D cellular structures derived from adult or pluripotent stem cells, capable of closely replicating the physiological properties of the gut. These organoids serve as powerful tools for studying gut development and modeling the pathogenesis of intestinal diseases. This review provides an in-depth exploration of technological advancements and applications of gut organoids, with a focus on their construction methods. Additionally, the potential applications of gut organoids in disease modeling, microenvironmental simulation, and personalized medicine are summarized. This review aims to offer perspectives and directions for understanding the mechanisms of intestinal health and disease as well as for developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoting Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Yuping Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Guoxin Huang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou 515041, China
| | - Ansu Perekatt
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Yan Wang
- Center for Translation Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
- Institute of Microphysiological Systems, Southeast University, Nanjing 211189, China
| |
Collapse
|
17
|
Yue Y, Su L, Wang Y, Li X, Xiao X, Xie J, Yan S. Banxia Xiexin Decoction inhibits colitis-associated colorectal cancer development by modulating STAT3 signaling and gut microbiota. CHINESE HERBAL MEDICINES 2025; 17:380-391. [PMID: 40256716 PMCID: PMC12009064 DOI: 10.1016/j.chmed.2024.02.004] [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: 05/10/2023] [Revised: 01/15/2024] [Accepted: 02/02/2024] [Indexed: 01/06/2025] Open
Abstract
Objective To investigate the therapeutic effects of Banxia Xiexin Decoction (BXD), a herbal medicine formula, on inflammation and the imbalance of the gut microbiota in a rat model of colorectal cancer (CRC) induced by azoxymethane (AOM) /dextran sulfate sodium (DSS). Methods A total of 75 male C57BL/6 mice were randomly divided into five groups: normal control group (NC), model group (MODEL), low-dose BXD treatment group (L-BXD), high-dose BXD treatment (H-BXD) group and MS treatment group (MS). BXD and MS were used in CRC mice at the doses of 3.915 g/kg, 15.66 g/kg, 0.6 g/kg for 3 weeks consecutively. Histopathological changes in the colon were observed using hematoxylin-eosin (HE) staining. The content of inflammatory factors in serum was detected by an enzyme-linked immunosorbent assay (ELISA), and the expression of mRNA and protein of genes related to immunity, apoptosis, inflammation, and inflammatory factors was evaluated. Changes in the intestinal flora of mouse fecal were determined based on high-throughput sequencing of the 16S rRNA microbial gene. Results Compared to the model group, the low-dose BXD and high-dose BXD groups decreased the number of colon tumors, reversed weight loss, and shortened colon length of mice. The pathological examination showed that BXD alleviated the malignancy of intestinal tumors. It also suppressed signal transducer and activator of transcription 3 (STAT3), matrix metalloproteinase-9 (MMP-9), and transforming growth factor beta 1 (TGF-β1) expression, while increasing the expression of the tight junction protein ZO-1 in colon tissues. Additionally, the levels of key pathway proteins involved in inflammation (phosphorylated-STAT3, Bcl-2, COX-2) and cell cycle regulatory molecules (c-Myc and PCNA) were reduced. According to 16S rRNA sequence analysis, BXD enhanced the relative abundance of potentially beneficial bacteria, while that of cancer-related bacteria decreased. Conclusion BXD plays a preventive role in developing colorectal cancer; its mechanisms are related to the inhibition of inflammation and tumor proliferation, as well as maintenance of intestinal homeostasis.
Collapse
Affiliation(s)
- Yinzi Yue
- Department of General Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Lianlin Su
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yahui Wang
- Department of Anorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Xiaoman Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoyan Xiao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jin Xie
- Department of General Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215009, China
| | - Shuai Yan
- Department of Anorectal Surgery, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215009, China
| |
Collapse
|
18
|
Chang Y, Long M, Shan H, Liu L, Zhong S, Luo JL. Combining gut microbiota modulation and immunotherapy: A promising approach for treating microsatellite stable colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104629. [PMID: 39864533 DOI: 10.1016/j.critrevonc.2025.104629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide, ranking third in incidence and second in mortality. While immunotherapy has shown promise in patients with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), its effectiveness in proficient mismatch repair (pMMR) or microsatellite stable (MSS) CRC remains limited. Recent advances highlight the gut microbiota as a potential modulator of anti-tumor immunity. The gut microbiome can significantly influence the efficacy of immune checkpoint inhibitors (ICIs), especially in pMMR/MSS CRC, by modulating immune responses and systemic inflammation. This review explores the role of the gut microbiota in pMMR/MSS CRC, the mechanisms by which it may enhance immunotherapy, and current strategies for microbiota modulation. We discuss the potential benefits of combining microbiota-targeting interventions with immunotherapy to improve treatment outcomes for pMMR/MSS CRC patients.
Collapse
Affiliation(s)
- Yujie Chang
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Min Long
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Hanguo Shan
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hunan 410008, China.
| |
Collapse
|
19
|
Fang ZH, Hao AH, Qi YG. Imaging features and correlation with short-term prognosis in laparoscopic radical resection of colorectal cancer. World J Gastrointest Surg 2025; 17:99782. [PMID: 40162386 PMCID: PMC11948101 DOI: 10.4240/wjgs.v17.i3.99782] [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: 10/31/2024] [Revised: 12/09/2024] [Accepted: 01/11/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant tumor with high morbidity and mortality rates worldwide. With the development of medical imaging technology, imaging features are playing an increasingly important role in the prognostic evaluation of CRC. Laparoscopic radical resection is a common surgical approach for treating CRC. However, research on the link between preoperative imaging and short-term prognosis in this context is limited. We hypothesized that specific preoperative imaging features can predict the short-term prognosis in patients undergoing laparoscopic CRC resection. AIM To investigate the imaging features of CRC and analyze their correlation with the short-term prognosis of laparoscopic radical resection. METHODS This retrospective study conducted at the Affiliated Cancer Hospital of Shandong First Medical University included 122 patients diagnosed with CRC who underwent laparoscopic radical resection between January 2021 and February 2024. All patients underwent magnetic resonance imaging (MRI) and were diagnosed with CRC through pathological examination. MRI data and prognostic indicators were collected 30 days post-surgery. Logistic regression analysis identified imaging features linked to short-term prognosis, and a receiver operating characteristic (ROC) curve was used to evaluate the predictive value. RESULTS Among 122 patients, 22 had irregular, low-intensity tumors with adjacent high signals. In 55, tumors were surrounded by alternating signals in the muscle layer. In 32, tumors extended through the muscular layer and blurred boundaries with perienteric adipose tissue. Tumor signals appeared in the adjacent tissues in 13 patients with blurred gaps. Logistic regression revealed differences in longitudinal tumor length, axial tumor length, volume transfer constant, plasma volume fraction, and apparent diffusion coefficient among patients with varying prognostic results. ROC analysis indicated that the areas under the curve for these parameters were 0.648, 0.927, 0.821, 0.809, and 0.831, respectively. Sensitivity values were 0.643, 0.893, 0.607, 0.714, and 0.714, and specificity 0.702, 0.904, 0.883, 0.968, and 0.894 (P < 0.05). CONCLUSION The imaging features of CRC correlate with the short-term prognosis following laparoscopic radical resection. These findings provide valuable insights for clinical decision-making.
Collapse
Affiliation(s)
- Ze-Hui Fang
- Department of Imaging, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Ai-Hua Hao
- Department of Imaging, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Yuan-Gang Qi
- Department of Imaging, The Affiliated Cancer Hospital of Shandong First Medical University (Shandong Cancer Hospital), Jinan 250117, Shandong Province, China
| |
Collapse
|
20
|
Huang C, Tang B, Chen W, Chen J, Zhang H, Bai M. Multiomic traits reveal that critical irinotecan-related core regulator FSTL3 promotes CRC progression and affects ferroptosis. Cancer Cell Int 2025; 25:115. [PMID: 40140870 PMCID: PMC11938592 DOI: 10.1186/s12935-025-03753-7] [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: 12/09/2024] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Irinotecan is a widely used chemotherapy drug in colorectal cancer (CRC). The evolution and prognosis of CRC involve complex mechanisms and depend on the drug administered, especially for irinotecan. However, the specific mechanism and prognostic role of irinotecan-related regulators remain to be elucidated. METHODS Data from public databases were used to explore the multiomic traits of irinotecan-related regulators through bioinformatics analysis. RT‒qPCR, western blotting, transmission electron microscopy and flow cytometry were used as experimental validations. RESULTS Iriscore (irinotecan-related score) was constructed based on irinotecan-related regulators, and a high iriscore predicted a poor prognosis, poor therapeutic response and the MSS/MSI-L status. Single-cell analysis revealed that FSTL3 and TMEM98 were mainly expressed in CRC stem cells. Potential transcription factors (E2F1, STAT1, and TTF2) and therapeutic drugs (telatinib) that target irinotecan-related regulators were identified. FSTL3 was the core risk irinotecan-related regulator. Some ferroptosis regulators (GPX4, HSPB1 and RGS4) and related metabolic pathways (lipid oxidation and ROS metabolism) were correlated significantly with FSTL3. In vitro, irinotecan inhibited the expression of FSTL3 and ferroptotic defence proteins (GPX4 and SLC7A11), and induced lipid peroxidation and intracellular Fe (2+) ions concentration increased. CONCLUSIONS We confirmed that irinotecan-related regulators, especially FSTL3, have effective prognostic value in CRC and speculated that FSTL3 may promote CRC progression and affect ferroptosis, which is beneficial for identifying candidate targeted irinotecan-related regulators and accurate individualized treatment strategies for CRC.
Collapse
Affiliation(s)
- Chengyi Huang
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenjuan Chen
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China
| | - Jinggang Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Huojun Zhang
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Minghua Bai
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
21
|
Neira M, Mena C, Torres K, Simón L. The Potential Benefits of Curcumin-Enriched Diets for Adults with Colorectal Cancer: A Systematic Review. Antioxidants (Basel) 2025; 14:388. [PMID: 40298630 PMCID: PMC12024020 DOI: 10.3390/antiox14040388] [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: 02/16/2025] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Conventional treatments such as chemotherapy and radiotherapy are often associated with severe side effects and limited effectiveness. Curcumin, a polyphenol derived from Curcuma longa, has demonstrated anti-inflammatory and anticancer properties. A systematic review of the recent scientific literature followed PRISMA guidelines to evaluate the benefits of a curcumin-enriched diet for adults with colorectal cancer. Articles published between 2018 and 2024 were retrieved from PubMed, SciELO, Google Scholar, and Scopus. Studies meeting the inclusion criteria focused on curcumin, adults, and colorectal cancer outcomes. The administration of curcumin-containing products was associated with improved survival rates, enhanced quality of life, tumor reduction, and anti-inflammatory effects. A curcumin-enriched diet shows potential as an effective adjunct therapy for CRC patients, though its limited bioavailability and potential side effects, such as gastrointestinal discomfort, pose challenges. Addressing these limitations through larger cohorts, extended study durations, and improved formulations to enhance bioavailability is essential. Such efforts could enable the development of personalized dietary recommendations for CRC management.
Collapse
Affiliation(s)
| | | | - Keila Torres
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501015, Chile; (M.N.); (C.M.)
| | - Layla Simón
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501015, Chile; (M.N.); (C.M.)
| |
Collapse
|
22
|
Shi YH, Liu JL, Cheng CC, Li WL, Sun H, Zhou XL, Wei H, Fei SJ. Construction and validation of machine learning-based predictive model for colorectal polyp recurrence one year after endoscopic mucosal resection. World J Gastroenterol 2025; 31:102387. [PMID: 40124266 PMCID: PMC11924002 DOI: 10.3748/wjg.v31.i11.102387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/25/2025] [Accepted: 02/14/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND Colorectal polyps are precancerous diseases of colorectal cancer. Early detection and resection of colorectal polyps can effectively reduce the mortality of colorectal cancer. Endoscopic mucosal resection (EMR) is a common polypectomy procedure in clinical practice, but it has a high postoperative recurrence rate. Currently, there is no predictive model for the recurrence of colorectal polyps after EMR. AIM To construct and validate a machine learning (ML) model for predicting the risk of colorectal polyp recurrence one year after EMR. METHODS This study retrospectively collected data from 1694 patients at three medical centers in Xuzhou. Additionally, a total of 166 patients were collected to form a prospective validation set. Feature variable screening was conducted using univariate and multivariate logistic regression analyses, and five ML algorithms were used to construct the predictive models. The optimal models were evaluated based on different performance metrics. Decision curve analysis (DCA) and SHapley Additive exPlanation (SHAP) analysis were performed to assess clinical applicability and predictor importance. RESULTS Multivariate logistic regression analysis identified 8 independent risk factors for colorectal polyp recurrence one year after EMR (P < 0.05). Among the models, eXtreme Gradient Boosting (XGBoost) demonstrated the highest area under the curve (AUC) in the training set, internal validation set, and prospective validation set, with AUCs of 0.909 (95%CI: 0.89-0.92), 0.921 (95%CI: 0.90-0.94), and 0.963 (95%CI: 0.94-0.99), respectively. DCA indicated favorable clinical utility for the XGBoost model. SHAP analysis identified smoking history, family history, and age as the top three most important predictors in the model. CONCLUSION The XGBoost model has the best predictive performance and can assist clinicians in providing individualized colonoscopy follow-up recommendations.
Collapse
Affiliation(s)
- Yi-Heng Shi
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
- The First Clinical Medical College of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Jun-Liang Liu
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Cong-Cong Cheng
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
- The First Clinical Medical College of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Wen-Ling Li
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
- The First Clinical Medical College of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Han Sun
- Department of Gastroenterology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou 221009, Jiangsu Province, China
| | - Xi-Liang Zhou
- Department of Gastroenterology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou 221009, Jiangsu Province, China
| | - Hong Wei
- Department of Gastroenterology, Xuzhou New Health Hospital, North Hospital of Xuzhou Cancer Hospital, Xuzhou 221007, Jiangsu Province, China
| | - Su-Juan Fei
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| |
Collapse
|
23
|
Shi L, Wang H, Sun Y, Xu N, Pei A, Zhang N. Development and validation of a disulfidptosis-related prognostic model for colorectal cancer using multi-omics analysis. Discov Oncol 2025; 16:338. [PMID: 40095116 PMCID: PMC11914417 DOI: 10.1007/s12672-025-02055-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
This study aims to integrate multi-omic and clinical data concerning disulfidptosis-related genes (DRGs) to facilitate molecular typing and prognosis in colorectal cancer (CRC). Public databases provided CRC transcriptome and clinical data, enabling differential expression, genomic analyses, pathway enrichment, survival analysis, and subtyping based on the expression levels of 15 DRGs identified in published studies. Differentially expressed genes (DEGs) between subtypes were identified to create a disulfidptosis prognostic model using LASSO and Cox regression analyses. This model was evaluated by comparing risk scores, survival curves, cellular infiltration, and drug sensitivity between high- and low-risk groups. Analyses revealed differential expression, mutations, and copy number variations (CNV) in DRGs in CRC. Survival analysis demonstrated significant prognostic differences among DRG expression subtypes. GSVA and ssGSEA highlighted DRGs' regulatory roles in CRC. DEGs identified between DRG expression subtypes led to the classification into subtypes A and B. A disulfidptosis prognostic model, including genes VSIG4, SCG2, INHBB, DDC, CXCL13, KLK10, CXCL10, and CCL11A, was developed to stratify patients into high- and low-risk groups. This model displayed strong predictive capability (AUC = 0.700) and calibration. The risk score was also strongly associated with immune cell infiltration, stromal cell score, and stem cell index in the CRC tumor microenvironment. Drug sensitivity analysis indicated that high-risk samples were more responsive to most medications. We established a robust disulfidptosis prognostic model for CRC through comprehensive multi-omics analysis. Our findings provide valuable insights into the role of DRGs in CRC progression and disease management, presenting an important resource for further research.
Collapse
Affiliation(s)
- Lei Shi
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Huimei Wang
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Yongxiao Sun
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Na Xu
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Aiyue Pei
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China.
| | - Nan Zhang
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China.
| |
Collapse
|
24
|
Li F, Dai Y, Tang C, Peng L, Huang H, Chen Y, Xu Y, Chen X, Wang Q, Lin Y. Elevated UBC9 expression and its oncogenic role in colorectal cancer progression and chemoresistance. Sci Rep 2025; 15:9123. [PMID: 40097547 PMCID: PMC11914596 DOI: 10.1038/s41598-025-93868-8] [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: 07/09/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Colorectal cancer (CRC) is a highly prevalent and fatal malignancy, with incidence and mortality rates rising globally. While elevated UBC9 expression has been implicated in various cancers, its specific role in CRC remains poorly understood. This study aims to investigate the expression levels, prognostic significance, and functional roles of UBC9 in CRC. We assessed the expression and prognostic value of UBC9 mRNA and protein in colorectal cancer separately using multiple databases and immunohistochemical techniques. Additionally, in vitro functional assays and in vivo zebrafish tumor models were employed to elucidate the role of UBC9 in CRC cell proliferation, migration, invasion, and chemoresistance. UBC9 expression was significantly upregulated in CRC tissues. Elevated UBC9 levels were associated with poor prognosis in chemotherapy-treated CRC patients. Gene Set Enrichment Analysis revealed that pathways related to MYC targets, DNA repair, and oxidative stress response were enriched in groups with high UBC9 expression. Immune profiling indicated reduced infiltration of CD4+ memory-activated T cells and NK cells in tumors with elevated UBC9 levels. Functional assays demonstrated that UBC9 knockdown inhibited CRC cell proliferation, migration, and invasion, and sensitized cells to oxaliplatin, which was further validated using zebrafish xenograft models. UBC9 is crucial for CRC progression, genomic instability, and chemoresistance. It represents a potential prognostic biomarker and therapeutic target, particularly for enhancing chemotherapy efficacy in CRC patients.
Collapse
Affiliation(s)
- Feng Li
- Department of Pathology, Shengli Clinical Medical College of Fujian Medical University and Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| | - Yongmei Dai
- Departments of Oncology, Shengli Clinical Medical College of Fujian Medical University and Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Chenchen Tang
- Longyan First Hospital Affiliated to Fujian Medical University, Longyan, 361000, China
| | - Lu Peng
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350001, China
| | - Haijian Huang
- Department of Pathology, Shengli Clinical Medical College of Fujian Medical University and Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Yuluo Chen
- Fujian Normal University, Fuzhou, 350001, China
| | - Yining Xu
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China
| | - Xuequn Chen
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China.
- Rehabilitation Technology Innovation Center by Joint Collaboration of Ministry of Education and Fujian Province, Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China.
| | - Qingshui Wang
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China.
| | - Yao Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350001, China.
| |
Collapse
|
25
|
Wei L, Wang L, Liu YG, Gao LF. Expression significance of biomarker MORC4 in colorectal cancer patients and its relationship with pathological features and prognosis. World J Gastrointest Oncol 2025; 17:102434. [PMID: 40092938 PMCID: PMC11866243 DOI: 10.4251/wjgo.v17.i3.102434] [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: 11/07/2024] [Revised: 12/08/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignant gastrointestinal tumors worldwide, with high incidence and mortality rates. AIM To investigate the expression significance of the chromatin-remodeling protein MORC family CW-type zinc finger 4 (MORC4) as a biomarker in CRC patients, and to explore its relationship with pathological features and prognosis. METHODS A total of 143 CRC specimens and 57 adjacent tissue specimens, surgically removed from our hospital between January 2020 and January 2021, were collected. MORC4 protein expression was assessed using immunohistochemistry after paraffin embedding. The relationship between MORC4 protein expression and clinicopathological characteristics of patients was analyzed. Kaplan-Meier survival curves were plotted to analyze the relationship between MORC4 protein expression and prognosis in CRC patients. RESULTS Compared with adjacent tissues, the expression rate of MORC4 protein in CRC tissues was significantly higher (P < 0.05). No significant difference was observed in the high expression rate of MORC4 protein in CRC tissues among patients of different gender, age, tumor location, tumor diameter, and primary tumor status (P > 0.05). However, significant differences were found in the high expression rate of MORC4 protein in patients with different degrees of differentiation, lymph node metastasis, distant metastasis, tumor-lymph node-metastasis stage, and serum carcinoembryonic antigen levels (P < 0.05). Compared with patients with low MORC4 expression, patients with high MORC4 expression had a worse prognosis (P < 0.05). CONCLUSION The upregulation of MORC4 expression in CRC patients is closely related to disease severity and prognosis, suggesting its potential as an evaluation biomarker, which warrants further investigation.
Collapse
Affiliation(s)
- Li Wei
- Department of General Surgery, Cangzhou Central Hospital, Cangzhou 061000, Hebei Province, China
| | - Liang Wang
- Department of General Surgery, Cangzhou Central Hospital, Cangzhou 061000, Hebei Province, China
| | - Ya-Gang Liu
- Department of General Surgery, Cangzhou Central Hospital, Cangzhou 061000, Hebei Province, China
| | - Li-Fei Gao
- Department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou 061000, Hebei Province, China
| |
Collapse
|
26
|
Sun J, Chen Y, Xu Z, Wang W, Li P. Notch signaling in the tumor immune microenvironment of colorectal cancer: mechanisms and therapeutic opportunities. J Transl Med 2025; 23:315. [PMID: 40075484 PMCID: PMC11900264 DOI: 10.1186/s12967-025-06282-z] [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: 11/29/2024] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related morbidity and mortality worldwide, driven by a complex interplay of genetic, environmental, and immune-related factors. Among the pivotal pathways implicated in CRC tumorigenesis, the Notch signaling pathway is instrumental in governing cell fate decisions, tissue renewal, homeostasis, and immune cell development. As a highly conserved mechanism, Notch signaling not only modulates tumor cell behavior but also shapes the immune landscape within the tumor microenvironment (TME). Aberrant Notch signaling in CRC fosters immune evasion and tumor progression through its effects on the balance and functionality of immune cells, including myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs). Elevated Notch pathway activation correlates with advanced clinicopathological features and poorer clinical outcomes, highlighting its relevance as both a prognostic biomarker and a therapeutic target. Therapeutic approaches aimed at inhibiting the Notch pathway, such as γ-secretase inhibitors (GSIs) or monoclonal antibodies (mAbs) in combination with other therapies, have demonstrated promising efficacy in preclinical and clinical settings. This review examines the impact of Notch signaling on CRC immunity, elucidating its regulatory mechanisms within immune cells and its role in promoting tumor progression. Additionally, this review discusses therapeutic strategies targeting Notch signaling, including GSIs, mAbs, and potential combination therapies designed to overcome resistance and improve patient outcomes. By elucidating the multifaceted role of Notch within the CRC TME, this review underscores its potential as a target for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Jiachun Sun
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, 471003, China
| | - Yi Chen
- Department of Children's gastroenterology, Anhui Children's Hospital, Hefei, Anhui, 230000, China
| | - Ziyi Xu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, 471003, China
| | - Weizheng Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, 471003, China
| | - Penghui Li
- Department of Gastrointestinal surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, No. 24 Jinghua Road, Jianxi District, Luoyang, Henan, 471000, China.
| |
Collapse
|
27
|
Zhang X, Fang H, Wu W, Jiang C, Wang H, Shi Y. LPIN3 promotes colorectal cancer growth by dampening intratumoral CD8 + T cell effector function. Cancer Immunol Immunother 2025; 74:135. [PMID: 40042548 PMCID: PMC11883066 DOI: 10.1007/s00262-025-03989-2] [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: 12/16/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
LPIN3 has emerged as a key factor in a variety of malignancies, although its precise role in colorectal cancer (CRC) remains unclear. By analyzing the data from The Cancer Genome Atlas, we discovered that the expression pattern of LPIN3 and the relevant makeup of the immune microenvironment were immensely diverse among tumors. LPIN3 is abundantly expressed in CRC and may enhance tumor growth by activating the β-catenin signaling pathway. In addition, we discovered that LPIN3 might reduce tumor antigen presentation signals, hence suppressing CD8+ T cell-mediated cytotoxicity. Furthermore, high expression of LPIN3 predicts decreased CD8+ T cell infiltration and effector function via bioinformatics analysis. Indeed, CD8+ T cell-mediated cytotoxicity as well as CD8+ T cell infiltration and activation in vivo were strengthened by LPIN3 knockdown. To sum up, our results highlight the part that LPIN3 plays in driving the progression of CRC by regulating β-catenin signaling and CD8+ T cell activity.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hao Fang
- Department of Gastroenterology, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Wenliang Wu
- Division of Gastrointestinal Surgery, Department of Surgery, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Congqing Jiang
- Department of Colorectal and Anal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Yifei Shi
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
28
|
Shahzad M, Hameed H, Amjad A, Khan MA, Qureshi IS, Hameed A, Saeed A, Munir R. An updated landscape on nanopharmaceutical delivery for mitigation of colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2107-2125. [PMID: 39361171 DOI: 10.1007/s00210-024-03482-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 03/19/2025]
Abstract
Globally, colorectal cancer (CRC) continues to rank among the leading causes of cancer-related death. Systemic toxicity, multidrug resistance, and nonspecific targeting often pose challenges to conventional therapy for CRC. Because it is a complex disease with a complex genetic and environmental pathophysiology, advanced therapeutic strategies are needed. Nanotechnology presents a potential solution that may maximize therapeutic efficacy while minimizing negative effects by enabling personalized delivery of anticancer drugs. This review focuses on recent developments in colorectal drug delivery systems based on nanotechnology. Numerous nanomaterials, including liposomes, dendrimers, micelles, exosomes, and gold nanoparticles, are developed and used. Distinctive characteristics of mentioned nanocarriers are discussed along with strategies that can be employed for enhancing the delivery of drugs to colorectal cancer cells. The review also quotes the most relevant preclinical and clinical studies that show how these nanomaterials improve drug solubility, stability, and targeted delivery while overcoming the shortcomings of conventional therapies. Nanotechnology has made CRC treatment very efficient and advanced, which has opened up new possibilities for targeted drug delivery. Preclinical and clinical studies have also proved that the use of nano-formulations in colon-specific delivery systems have significant results, indicating potential for better patient outcomes. Future research can be done in order to overcome the hurdles regarding biocompatibility, expansion, and regulatory challenges. Large-scale clinical trials and nanomaterial formulation optimization should be the main goals of future research to confirm the efficacy and safety of these novel treatments.
Collapse
Affiliation(s)
- Maria Shahzad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Ayesha Amjad
- Faculty of Food Technology and Nutrition Sciences, Lahore University of Biological and Applied Sciences, Lahore, 54000, Pakistan
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Inaba Shujaat Qureshi
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Asad Saeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| |
Collapse
|
29
|
Sun X, Gong Y, Xie T, Fu Z, Lu D, Wei B, Cai Y, Yao W, Shen J. Nanoscale Liposomes Co-Loaded with Irinotecan Hydrochloride and Thalidomide for Colorectal Cancer Synergistic Therapy. Macromol Biosci 2025; 25:e2400478. [PMID: 39704649 DOI: 10.1002/mabi.202400478] [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: 09/29/2024] [Revised: 12/03/2024] [Indexed: 12/21/2024]
Abstract
Irinotecan hydrochloride (CPT-11) is one of the first-line drugs used in the clinical treatment of colorectal cancer (CRC). However, the concomitant adverse effect of delayed diarrhea has hindered its clinical use. CPT-11 combined with Thalidomide (THA) therapy is considered a palliative strategy. To optimize the synergistic treatment of CPT-11 and THA, co-loaded liposomes are constructed using cholesterol, lecithin, and 1, 2-Distearoyl-sn-glycero-3-phosphoethanolamine-Poly(ethylene glycol) (DSPE-PEG) as the "immune and gut microbiota regulator." The co-loaded liposomes, which possess good stability, are prepared by the solvent injection method. After the treatment with the co-loaded liposomes, tumor growth in CRC-bearing mice is significantly inhibited. In particular, the co-loaded liposomes demonstrate favorable diarrhea-relieving effects through the modulation of inflammatory cytokines and gut microbiota. These findings suggest that the co-loaded liposomes have great potential as a combined drug-delivery platform for CRC therapy.
Collapse
Affiliation(s)
- Xuanrong Sun
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yubei Gong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Ting Xie
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zixi Fu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Dongze Lu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Bin Wei
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yue Cai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Wenlong Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Xue Yuan Road No. 38, Beijing, 100191, China
| | - Jie Shen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
30
|
Kuang J, Li J, Zhou S, Li Y, Lin J, Huang W, Yuan X. Genomic and micro-environmental insights into drug resistance in colorectal cancer liver metastases. Discov Oncol 2025; 16:241. [PMID: 40009285 DOI: 10.1007/s12672-025-01976-8] [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: 12/01/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is known for its high heterogeneity, with liver metastases significantly impairing survival outcomes. Understanding the tumor microenvironment (TME) and genomic alterations in metastatic sites is crucial for developing personalized therapies that overcome drug resistance and improve prognosis. METHODS We profiled 54 CRC liver metastases, comparing them with 198 other metastatic lesions and normal liver tissues. By analyzing immune cell infiltration, stromal interactions, and key genomic alterations, we constructed an 11-gene prognostic model to predict survival and immunotherapy outcomes. RESULTS CRC liver metastases with high-risk profiles demonstrated enriched follicular helper T cells, activated dendritic cells, and M2 macrophages in the TME. Frequent mutations in APC, TP53, KRAS, and PIK3CA were identified, alongside altered EGFR signaling. The 11-gene model effectively stratified patients by prognosis and predicted immunotherapy responses, emphasizing the therapeutic potential of targeting resistance mechanisms. CONCLUSIONS This study reveals how genomic and TME-driven factors contribute to drug resistance in CRC liver metastases. Integrating these insights with clinical data could advance precision therapies, addressing the evolving challenge of tumor drug resistance in CRC.
Collapse
Affiliation(s)
- Junjie Kuang
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Jun Li
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Siwei Zhou
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Yi Li
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Jinbo Lin
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Weizhen Huang
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China.
| | - Xia Yuan
- The Second Department of Oncology, Cancer Center, Huizhou First Hospital, Huizhou, Guangdong, China.
| |
Collapse
|
31
|
Chen J, Wang J, Zheng Q. Disitamab vedotin combined with pyrotinib as salvage treatment in Her-2-amplified treatment refractory metastatic colorectal cancer: a case report. Front Pharmacol 2025; 16:1431422. [PMID: 40051563 PMCID: PMC11883190 DOI: 10.3389/fphar.2025.1431422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Background Human epidermal growth factor receptor 2 (HER-2) amplification has been identified in approximately 3% of patients with metastatic colorectal cancer (mCRC). Owing to the lack of established anti-ERBB2 therapeutic approaches, mCRC patients with Her-2 amplification rarely receive targeted treatments. Moreover, conventional chemotherapy regimens are not ideal for these patients, leaving options in the advanced stage limited to best supportive care or participation in clinical drug trials. Case presentation This report presents a case of a patient with Her-2-amplified refractory mCRC treated with a salvage regimen combining Disitamab Vedotin and Pyrotinib, resulting in a partial response and progression-free survival for 6 months, which is still ongoing. Conclusion This case study suggests that the anti-Her-2 regimen involving Disitamab Vedotin and Pyrotinib may offer a potential salvage treatment option for patients with Her-2-amplified mCRC patients. However, further validation in larger cohorts is necessary in future studies.
Collapse
Affiliation(s)
- Jianxin Chen
- Department of Medical Oncology, Quzhou People′s Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, Zhejiang, China
| | - Jian Wang
- Department of Medical Oncology, Quzhou People′s Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, Zhejiang, China
- The Third Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qinhong Zheng
- Department of Medical Oncology, Quzhou People′s Hospital, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, Zhejiang, China
| |
Collapse
|
32
|
Kumar A, Kaw P. Clinicopathological and radiological characteristics and prediction of survival in colon cancer. World J Gastrointest Oncol 2025; 17:101516. [PMID: 39958557 PMCID: PMC11756002 DOI: 10.4251/wjgo.v17.i2.101516] [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: 09/17/2024] [Revised: 10/14/2024] [Accepted: 11/04/2024] [Indexed: 01/18/2025] Open
Abstract
There are various histological characteristics which have been proposed to predict the survival rate in colon cancer. However, there is no definitive model to accurately predict the survival. Therefore, it is important to find out one model for the prediction of survival in colon cancer which may also include the preoperative, and operative factors in addition to histopathology.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Payal Kaw
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| |
Collapse
|
33
|
Nishiki H, Ura H, Togi S, Hatanaka H, Fujita H, Takamura H, Niida Y. Integrated Analysis of Somatic DNA Variants and DNA Methylation of Tumor Suppressor Genes in Colorectal Cancer. Int J Mol Sci 2025; 26:1642. [PMID: 40004106 PMCID: PMC11855003 DOI: 10.3390/ijms26041642] [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: 01/06/2025] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
DNA methylation of tumor suppressor genes in cancer is known to be a mechanism for silencing gene expression, but much remains unknown about its extent and relationship to somatic variants at the DNA sequence level. In this study, we comprehensively analyzed DNA methylation and somatic variants of all gene regions across the genome of the major tumor suppressor genes, APC, TP53, SMAD4, and mismatch repair genes in colorectal cancer using a novel next-generation sequencing-based analysis method. The Targeted Methyl Landscape (TML) shows that DNA hypermethylation patterns of these tumor suppressor genes in colorectal cancer are more complex and widespread than previously thought. Extremely high levels of DNA methylation were observed in relatively long regions around exon 1A of APC and exon 1 and surrounding region of MLH1. DNA hypermethylation occurred whether or not somatic DNA variants were present in the tumor. Even in tumors where the loss of heterozygosity has been demonstrated by somatic variants alone, additional methylation of the same gene can occur. Our data demonstrate that somatic variants and hypermethylation of these tumor suppressor genes were considered independent, parallel events, not exclusive of each other or having one event affecting the other.
Collapse
Affiliation(s)
- Hisashi Nishiki
- General and Digestive Surgery, Kanazawa Medical University, Uchinada 920-0293, Japan; (H.N.); (H.F.); (H.T.)
| | - Hiroki Ura
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada 920-0293, Japan (S.T.)
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Sumihito Togi
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada 920-0293, Japan (S.T.)
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Hisayo Hatanaka
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada 920-0293, Japan (S.T.)
| | - Hideto Fujita
- General and Digestive Surgery, Kanazawa Medical University, Uchinada 920-0293, Japan; (H.N.); (H.F.); (H.T.)
| | - Hiroyuki Takamura
- General and Digestive Surgery, Kanazawa Medical University, Uchinada 920-0293, Japan; (H.N.); (H.F.); (H.T.)
| | - Yo Niida
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada 920-0293, Japan (S.T.)
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| |
Collapse
|
34
|
Pretzsch E, Peschel CA, Rokavec M, Torlot L, Li P, Hermeking H, Werner J, Klauschen F, Neumann J, Jung A, Kumbrink J. Five-Gene Expression Signature Associated With Acquired FOLFIRI Resistance and Survival in Metastatic Colorectal Cancer. J Transl Med 2025; 105:104107. [PMID: 39954853 DOI: 10.1016/j.labinv.2025.104107] [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: 08/15/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
FOLFIRI, a combination of folinic acid, 5-fluorouracil, and irinotecan, is one of the recommended first-line chemotherapeutic treatments for metastatic colorectal cancer. Unfortunately, acquired FOLFIRI resistance represents a common obstacle in the treatment of metastatic colorectal cancer patients. Thus, we aimed to identify mechanisms, gene alterations, and gene expression signatures contributing to acquired FOLFIRI resistance by mimicking this problem in a cell culture model and subsequent translation in clinical data sets. Three FOLFIRI-resistant colorectal cancer (CRC) cell lines were established by continuous FOLFIRI treatment. Comparative mutation screening (161 genes) and transcriptomics (pathway and differential expression analyses) were performed in parental and resistant cells. Data reconciliation was performed in GSE62322, a clinical FOLFIRI responder data set (intrinsic resistance). Relapse-free survival (RFS) associations of identified differentially expressed genes and potential gene signatures were investigated in 8 clinical CRC data sets. No mutual genetic alterations were found in FOLFIRI-resistant derivatives. Resistant cell lines displayed activation of mitogen-activated protein kinase, immune response, and epithelial-mesenchymal transition pathways. Twelve differentially expressed genes, significantly differentially expressed in at least 2 of the 3 resistant cell lines, were identified. Comparison with GSE62322 and subsequent survival analyses revealed a 5-gene FOLFIRI signature comprised of CAV2, TNC, TACSTD2, SERPINE2, and PERP that was associated with RFS in multiple data sets including the cancer genome atlas CRC (hazard ratio [HR] =2.634, P = 4.53 × 10-6), in pooled samples of all data sets (all stages [N = 1981]: HR = 1.852, P = 6.44 × 10-13; stage IV [N = 260]: HR = 2.462, P = 5.22 × 10-9). A multivariate Cox regression analysis identified the 5-gene signature as an independent prognostic factor in the cancer genome atlas data set (HR = 1.89, P = .0202). Our analyses revealed a 5-gene FOLFIRI resistance signature associated with RFS that may help predict FOLFIRI resistance and thus avoid unnecessary ineffective treatment. Signature members might also represent targets to fight FOLFIRI resistance.
Collapse
Affiliation(s)
- Elise Pretzsch
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Christiane A Peschel
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Matjaz Rokavec
- Experimental and Molecular Pathology, Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lucien Torlot
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Germany
| | - Pan Li
- Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heiko Hermeking
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Experimental and Molecular Pathology, Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Frederick Klauschen
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Neumann
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Jung
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jörg Kumbrink
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany; Institute of Pathology, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
35
|
Lyu H, Chen X, Cheng Y, Zhang T, Wang P, Wong JHY, Wang J, Stasiak L, Sun L, Yang G, Wang L, Yue F. Pioneer factor GATA6 promotes colorectal cancer through 3D genome regulation. SCIENCE ADVANCES 2025; 11:eads4985. [PMID: 39919174 PMCID: PMC11804904 DOI: 10.1126/sciadv.ads4985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025]
Abstract
Colorectal cancer (CRC) is one of the most lethal and prevalent malignancies. While the overexpression of pioneer factor GATA6 in CRC has been linked with metastasis, its role in genome-wide gene expression dysregulation remains unclear. Through studies of primary human CRC tissues and analysis of the TCGA data, we found that GATA6 preferentially binds at CRC-specific active enhancers, with enrichment at enhancer-promoter loop anchors. GATA6 protein also physically interacts with CTCF, suggesting its critical role in 3D genome organization. The ablation of GATA6 through AID and CRISPR systems severely impaired cancer cell clonogenicity and proliferation. Mechanistically, GATA6 knockout induced global loss of CRC-specific open chromatins and extensive alterations of critical enhancer-promoter interactions for CRC oncogenes. Last, we showed that GATA6 knockout greatly reduced tumor growth and improved survival in mice. Together, we revealed a previously unidentified mechanism by which GATA6 contributes to the pathogenesis of colorectal cancer.
Collapse
Affiliation(s)
- Huijue Lyu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xintong Chen
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yang Cheng
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Te Zhang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ping Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Josiah Hiu-yuen Wong
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Juan Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lena Stasiak
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leyu Sun
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Guangyu Yang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Lu Wang
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
36
|
Zhang Y, Liu YJ, Mei J, Yang ZX, Qian XP, Huang W. An Analysis Regarding the Association Between DAZ Interacting Zinc Finger Protein 1 (DZIP1) and Colorectal Cancer (CRC). Mol Biotechnol 2025; 67:527-547. [PMID: 38334905 DOI: 10.1007/s12033-024-01065-1] [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: 11/09/2023] [Accepted: 12/21/2023] [Indexed: 02/10/2024]
Abstract
Colorectal cancer (CRC) is the third most common malignant disease worldwide, and its incidence is increasing, but the molecular mechanisms of this disease are highly heterogeneous and still far from being fully understood. Increasing evidence suggests that fibrosis mediated by abnormal activation of fibroblasts based in the microenvironment is associated with a poor prognosis. However, the function and pathogenic mechanisms of fibroblasts in CRC remain unclear. Here, combining scrna-seq and clinical specimen data, DAZ Interacting Protein 1 (DZIP1) was found to be expressed on fibroblasts and cancer cells and positively correlated with stromal deposition. Importantly, pseudotime-series analysis showed that DZIP1 levels were up-regulated in malignant transformation of fibroblasts and experimentally confirmed that DZIP1 modulates activation of fibroblasts and promotes epithelial-mesenchymal transition (EMT) in tumor cells. Further studies showed that DZIP1 expressed by tumor cells also has a driving effect on EMT and contributes to the recruitment of more fibroblasts. A similar phenomenon was observed in xenografted nude mice. And it was confirmed in xenograft mice that downregulation of DZIP1 expression significantly delayed tumor formation and reduced tumor size in CRC cells. Taken together, our findings suggested that DZIP1 was a regulator of the CRC mesenchymal phenotype. The revelation of targeting DZIP1 provides a new avenue for CRC therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
- Department of Oncology, Nanjing Tianyinshan Hospital, Nanjing, 211199, Jiangsu, China
| | - Yuan-Jie Liu
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jia Mei
- Department of Pathology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Zhao-Xu Yang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Xiao-Ping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Clinical Cancer Institute of Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Wei Huang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Hanzhong Road No.155, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
37
|
Gila F, Khoddam S, Jamali Z, Ghasemian M, Shakeri S, Dehghan Z, Fallahi J. Personalized medicine in colorectal cancer: a comprehensive study of precision diagnosis and treatment. Per Med 2025; 22:59-81. [PMID: 39924822 DOI: 10.1080/17410541.2025.2459050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025]
Abstract
Colorectal cancer is a common and fatal disease that affects many people globally. CRC is classified as the third most prevalent cancer among males and the second most frequent cancer among females worldwide. The purpose of this article is to examine how personalized medicine might be used to treat colorectal cancer. The classification of colorectal cancer based on molecular profiling, including the detection of significant gene mutations, genomic instability, and gene dysregulation, is the main topic of this discussion. Advanced technologies and biomarkers are among the detection methods that are explored, demonstrating their potential for early diagnosis and precise prognosis. In addition, the essay explores the world of treatment possibilities by providing light on FDA-approved personalized medicine solutions that provide individualized and precise interventions based on patient characteristics. This article assesses targeted treatments like cetuximab and nivolumab, looks at the therapeutic usefulness of biomarkers like microsatellite instability (MSI) and circulating tumor DNA (ctDNA), and investigates new approaches to combat resistance. Through this, our review provides a thorough overview of personalized medicine in the context of colorectal cancer, ultimately highlighting its potential to revolutionize the field and improve patient care.
Collapse
Affiliation(s)
- Fatemeh Gila
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Khoddam
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Jamali
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohmmad Ghasemian
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayan Shakeri
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Xia J, Shen Y, Jiang Q, Li X, Yan Y, Xu Z, Zhou L. Poly (ADP-Ribose) Polymerase 1 Induces Cyclic GMP-AMP Synthase-stimulator of Interferon Genes Pathway Dysregulation to Promote Immune Escape of Colorectal Cancer Cells. J Immunother 2025; 48:35-45. [PMID: 39787528 DOI: 10.1097/cji.0000000000000543] [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: 04/26/2024] [Accepted: 09/10/2024] [Indexed: 01/12/2025]
Abstract
Colorectal cancer (CRC) ranks third globally in cancer incidence and mortality, posing a significant human concern. Recent advancements in immunotherapy are noteworthy. This study explores immune modulation for CRC treatment. Initially targeting poly (ADP-ribose) polymerase 1 (PARP-1), a gene overexpressed in CRC tissues per The Cancer Genome Atlas, we examined its correlation with immune cell infiltration using the Tumor Immune Estimation Resource tool. Quantitative reverse transcription polymerase chain reaction assessed PARP-1 mRNA and inflammation-related gene expression in tumor tissues and cells. Assessing CD8 + T-cell proliferation and cytotoxicity towards HCT116 cells involved carboxyfluorescein diacetate succinimidyl ester and lactate dehydrogenase kits. Chemotaxis was gauged using a Transwell system in a CD8 + T-cell coculture setup, with immunofluorescence revealing cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) levels in HCT116 cells. Enzyme-linked immunosorbent assay kits measured CD8 + T-cell cytokine secretion. The findings suggested that PARP-1 was overexpressed in CRC tissues and cells and this overexpression was positively correlated with Treg cell infiltration. Overexpression of PARP-1 could significantly reduce the proportion of cGAS and STING-positive cells in HCT116 cells, dampen the proliferation, tumor-killing capacity, and chemotaxis of CD8 + T cells, and inhibit the secretion of related cytokines. The introduction of STING agonists could reverse the effects caused by overexpressed PARP-1. In vivo experiments affirmed the independent anti-tumor effects of PARP-1 inhibitors and STING agonists, synergistically inhibiting tumor growth. Silencing PARP-1 in HCT116 cells potentially boosts CD8 + T-cell activity against these cells through the cGAS-STING pathway.
Collapse
Affiliation(s)
- Jianhong Xia
- Department of Radiation Oncology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| | - Yue Shen
- Department of Radiation Oncology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| | - Qian Jiang
- Department of Radiation Oncology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| | - Xin Li
- Department of Radiation Oncology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| | - Yan Yan
- Department of Radiation Oncology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| | - Zhi Xu
- Medical Affairs, ICON Public Limited Company (ICON Plc), Beijing, China
| | - Liqing Zhou
- Department of Radiation Oncology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an, China
| |
Collapse
|
39
|
Jia J, Huang Y, Chen Q, Hou J, Liu Y, Xie L, Li X, Yang C. DR6 Augments Colorectal Cancer Cell Growth, Invasion, and Stemness by Activating AKT/NF-κB Pathway. Biochem Genet 2025; 63:606-622. [PMID: 38478147 PMCID: PMC11832796 DOI: 10.1007/s10528-024-10673-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/02/2024] [Indexed: 02/19/2025]
Abstract
This study aims to elucidate the role and mechanisms of Death Receptor 6 (DR6), a member of the tumor necrosis factor receptor superfamily, in the malignant progression of colorectal cancer (CRC). The association of DR6 expression levels and CRC patient survival was examined using the CRC cohort data from GEPIA database. The functional role of DR6 in CRC cells was investigated by performing loss-of-function and gain-of-function experiments based on CCK-8 proliferation assay, transwell migration and invasion assay, and sphere-forming assays. Xenograft model of CRC cells in nude mouse was established to evaluate the impact of DR6 knockdown on CRC tumorigenesis. Elevated expression of DR6 was correlated with an unfavorable prognosis in CRC patients. In vitro functional assays demonstrated that silencing DR6 considerably suppressed the proliferation, migration, invasion, and stemness of CRC cells, whereas its overexpression showed an opposite effect. DR6 knockdown also attenuated tumor formation of CRC cells in the nude mice. Mechanistically, silencing DR6 reduced the phosphorylation of AKT and NF-κB in CRC cells, and the treatment with an AKT activator (SC79) abrogated the inhibitory effects of DR6 knockdown on the malignant features of CRC cells. Our data suggest that DR6 contributes to the malignant progression of CRC by activating AKT/NF-κB pathway, indicating its clinical potential as a prognostic marker and therapeutic target for CRC.
Collapse
Affiliation(s)
- Jing Jia
- Department of Gastrointestinal Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, 250 Dongjie, Licheng District, Quanzhou, 362002, Fujian, China
| | - Yisen Huang
- Department of Gastroenterology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362002, Fujian, China
| | - Qiwei Chen
- Department of Gastrointestinal Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, 250 Dongjie, Licheng District, Quanzhou, 362002, Fujian, China
| | - Jianbin Hou
- Department of Gastrointestinal Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, 250 Dongjie, Licheng District, Quanzhou, 362002, Fujian, China
| | - Yan Liu
- Department of Gastrointestinal Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, 250 Dongjie, Licheng District, Quanzhou, 362002, Fujian, China
| | - Lifeng Xie
- Department of Gastrointestinal Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, 250 Dongjie, Licheng District, Quanzhou, 362002, Fujian, China
| | - Xinyu Li
- Department of Gastrointestinal Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, 250 Dongjie, Licheng District, Quanzhou, 362002, Fujian, China.
| | - Chunkang Yang
- Department of Gastrointestinal Surgery, Fujian Cancer Hospital, Clinical Oncology School of Fujian Medical University, Fuma Road Fengban, Jin'an District, Fuzhou, 362002, Fujian, China.
| |
Collapse
|
40
|
Fei F, Lu P, Ni J. Peripheral blood CD8 + CD28+ T cells as predictive biomarkers for treatment response in metastatic colorectal cancer. Biomarkers 2025; 30:10-22. [PMID: 39989261 DOI: 10.1080/1354750x.2024.2435867] [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: 09/30/2024] [Accepted: 11/24/2024] [Indexed: 02/25/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a substantial global health burden, with treatment outcomes significantly influenced by the interaction between the immune system and the tumor microenvironment. OBJECTIVE This study aims to investigate the role of peripheral blood immune cell subpopulations, particularly CD8+ CD28+ T cells, in predicting treatment response in metastatic CRC patients receiving bevacizumab combined with chemotherapy. METHODS A cohort of 45 CRC patients was analyzed. Flow cytometry was utilized to assess immune cell subpopulations. RESULTS Higher CD8+ CD28+ T cell counts were associated with better treatment responses, including improved objective response rates. In a murine CRC model, the combination therapy significantly inhibited tumor growth and enhanced immune cell function. CONCLUSION These findings highlight the importance of CD8+ CD28+ T cells as potential biomarkers for predicting treatment outcomes in CRC. They also suggest that bevacizumab, when combined with chemotherapy, can modulate immune function and improve clinical efficacy.
Collapse
Affiliation(s)
- Fei Fei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Peihua Lu
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Jingyi Ni
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| |
Collapse
|
41
|
Wang M, Qi J, Tan Z, Zhou R, Zhuo Q, Deng X, Wang Z, Zhou R, Li F, Xu Y. GABPα targeted by miR-378a-5p inhibits the growth and angiogenesis of colorectal carcinoma. Int J Biochem Cell Biol 2025; 179:106729. [PMID: 39710138 DOI: 10.1016/j.biocel.2024.106729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Considering the high degree of malignancy, recurrence rate and poor prognosis, exploring promising targets is an imperious strategy for colorectal carcinoma therapy. Recent studies have indicated that GABPα plays a role in cancer aggressiveness, but its exact function and regulatory mechanisms in colorectal cancer progression remain unclear. This study aims to explore the biological role of GABPα and its upstream regulator, miR-378a-5p, in modulating cancer progression. The expression levels of GABPα and miR-378a-5p were analyzed through comprehensive data mining and qPCR assays. The functional effects of GABPα were assessed using CCK-8, wound healing, transwell invasion assay, tube formation and xenograft model in nude mice. A co-transfection assay was also performed to investigate the regulatory relationship between miR-378a-5p and GABPα. We found that GABPα expression was significantly downregulated in human colorectal cancer tissues and cell lines. Functional assays revealed that GABPα overexpression suppressed the proliferation, migration, invasion and angiogenesis of colorectal cancer cells, and in vivo experiments further confirmed the inhibitory role of GABPα. Additionally, miR-378a-5p was upregulated in colorectal cancer, and GABPα was identified as a direct target of miR-378a-5p, as confirmed by luciferase reporter assays. Furthermore, overexpression of GABPα partially counteracted the enhanced malignant behaviors of cancer cells induced by miR-378a-5p. Our findings suggest that miR-378a-5p promotes the aggressive progression of colorectal cancer by directly targeting GABPα, highlighting this regulatory axis as a potential therapeutic target for colorectal carcinoma.
Collapse
Affiliation(s)
- Mengyi Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Jiangfa Qi
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhenlin Tan
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Runlong Zhou
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Qing Zhuo
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Xiaotong Deng
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Zhenrong Wang
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Ruijie Zhou
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China
| | - Fan Li
- Wuhan Bio-Raid Biotechnology Co., Ltd., Wuhan, Hubei 430075, China
| | - Yao Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China.
| |
Collapse
|
42
|
Alshenaifi JY, Vetere G, Maddalena G, Yousef M, White MG, Shen JP, Vilar E, Parseghian C, Dasari A, Morris VK, Huey R, Overman MJ, Wolff R, Raghav KP, Willis J, Alfaro K, Futreal A, You YN, Kopetz S. Mutational and co-mutational landscape of early onset colorectal cancer. Biomarkers 2025; 30:64-76. [PMID: 39761813 PMCID: PMC11856746 DOI: 10.1080/1354750x.2024.2447089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Colorectal cancer (CRC) incidence and mortality before 50 have been rising alarmingly in the recent decades. METHODS Using a cohort of 10,000 patients, this study investigates the clinical, mutational, and co-mutational features of CRC in early-onset (EOCRC, < 50 years) compared to late-onset (LOCRC, ≥ 50 years). RESULTS EOCRC was associated with a higher prevalence of Asian and Hispanic patients, rectal or left-sided tumors (72% vs. 59%), and advanced-stage disease. Molecular analyses revealed differences in mutation patterns, with EOCRC having higher frequencies of TP53 (74% vs. 68%, p < 0.01) and SMAD4 (17% vs. 14%, p = 0.015), while BRAF (5% vs. 11%, p < 0.001) and NOTCH1 (2.7% vs. 4.1%, p = 0.01) mutations were more prevalent in LOCRC. Stratification by tumor site and MSI status highlighted significant location- and age-specific molecular differences, such as increased KRAS and CTNNB1 mutations in right-sided EOCRC and higher BRAF prevalence in MSI-H LOCRC (47% vs. 6.7%, p < 0.001). Additionally, co-occurrence analysis revealed unique mutational networks in EOCRC MSS, including significant co-occurrences of FBXW7 with NOTCH3, RB1, and PIK3R1. CONCLUSION This study highlights the significance of age-specific molecular profiling, offering insights into the unique biology of EOCRC and potential clinical applications.
Collapse
Affiliation(s)
- Jumanah Yousef Alshenaifi
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guglielmo Vetere
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Giulia Maddalena
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael G. White
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eduardo Vilar
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine Parseghian
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Van Karlyle Morris
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Wolff
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kanwal P. Raghav
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andy Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y. Nancy You
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
43
|
Dagnaw GG, Dejene H. Colorectal cancer in Ethiopia: Epidemiological trends, diagnostic and laboratory capacities, and challenges. Semin Oncol 2025; 52:19-26. [PMID: 39537473 DOI: 10.1053/j.seminoncol.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 11/16/2024]
Abstract
Colorectal cancer (CRC) refers to cancer that develops in the colon or rectum, parts of the large intestine. It ranks as the third most prevalent form of cancer globally. Colorectal cancer is responsible for the morbidity of millions and the loss of hundreds of thousands of lives worldwide although the incidence varies significantly depending on geographical location. In recent years, CRC has decreased in high-income countries due to technological advancements in diagnosis and treatment. However, there is an increased occurrence of CRC morbidity and mortality in low- and middle-income countries. Colorectal cancer is becoming an emerging public health concern in Ethiopia. We noticed that the incidence rates have been lower compared to more developed countries, but recent years have seen a noticeable increase. This rise is attributed to factors such as changes in diet, lifestyle, and an aging population. Common risk factors include dietary shifts towards processed foods and red meat, physical inactivity, obesity, smoking, and alcohol consumption. Unfortunately, in Ethiopia, screening programs for CRC are not widespread, and limited access to diagnostic facilities, lack of public awareness, and insufficient healthcare infrastructure contribute to late-stage diagnoses or left without diagnosis. Treatment options, including surgery, chemotherapy, and radiotherapy, are available but not uniformly accessible across the country, posing challenges for timely and effective treatment. Addressing colorectal cancer in Ethiopia requires a comprehensive approach to enhance public awareness, improve screening and early detection, expand treatment facilities, and train healthcare professionals to provide effective care.
Collapse
Affiliation(s)
| | - Haileyesus Dejene
- Department of Epidemiology and Public Health, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
44
|
Lan YZ, Wu Z, Chen WJ, Yu XN, Wu HT, Liu J. Sine oculis homeobox homolog family function in gastrointestinal cancer: Progression and comprehensive analysis. World J Clin Oncol 2025; 16:97163. [PMID: 39867730 PMCID: PMC11528897 DOI: 10.5306/wjco.v16.i1.97163] [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: 05/25/2024] [Revised: 09/20/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024] Open
Abstract
The sine oculis homeobox homolog (SIX) family, a group of transcription factors characterized by a conserved DNA-binding homology domain, plays a critical role in orchestrating embryonic development and organogenesis across various organisms, including humans. Comprising six distinct members, from SIX1 to SIX6, each member contributes uniquely to the development and differentiation of diverse tissues and organs, underscoring the versatility of the SIX family. Dysregulation or mutations in SIX genes have been implicated in a spectrum of developmental disorders, as well as in tumor initiation and progression, highlighting their pivotal role in maintaining normal developmental trajectories and cellular functions. Efforts to target the transcriptional complex of the SIX gene family have emerged as a promising strategy to inhibit tumor development. While the development of inhibitors targeting this gene family is still in its early stages, the significant potential of such interventions holds promise for future therapeutic advances. Therefore, this review aimed to comprehensively explore the advancements in understanding the SIX family within gastrointestinal cancers, focusing on its critical role in normal organ development and its implications in gastrointestinal cancers, including gastric, pancreatic, colorectal cancer, and hepatocellular carcinomas. In conclusion, this review deepened the understanding of the functional roles of the SIX family and explored the potential of utilizing this gene family for the diagnosis, prognosis, and treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Yang-Zheng Lan
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Zheng Wu
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Wen-Jia Chen
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Xin-Ning Yu
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Hua-Tao Wu
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jing Liu
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
45
|
Singer M, Valerin J, Zhang Z, Zhang Z, Dayyani F, Yaghmai V, Choi A, Imagawa D, Abi-Jaoudeh N. Promising Cellular Immunotherapy for Colorectal Cancer Using Classical Dendritic Cells and Natural Killer T Cells. Cells 2025; 14:166. [PMID: 39936958 PMCID: PMC11817869 DOI: 10.3390/cells14030166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related morbidity and mortality around the world. Despite advances in surgery, chemotherapy, and targeted therapies, the prognosis for patients with metastatic or advanced CRC remains poor. Immunotherapies comprising immune checkpoint inhibitors showed disappointing responses in metastatic CRC (mCRC). However, cellular immunotherapy, specifically using classical dendritic cells (cDCs), may hold unique promise in immune recognition for CRC antigens. cDCs are substantial players in immune recognition and are instrumental in orchestrating innate and adaptive immune responses by processing and presenting tumor antigens to effector cells. Natural killer T (NKT) cells are insufficiently studied but unique effector cells because of their ability to bridge innate and adaptive immune reactions and the crosstalk with dendritic cells in cancer. This review explores the therapeutic potential of using both cDCs and NKT cells as a synergistic therapy in CRC, focusing on their biological roles, strategies for harnessing their capabilities, clinical applications, and the challenges within the tumor microenvironment. Both cDCs and NKT cells can be used as a new effective approach for cell-based therapies in cancers to provide a new hope for CRC patients that are challenging to treat.
Collapse
Affiliation(s)
- Mahmoud Singer
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Jennifer Valerin
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - Zhuoli Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Zigeng Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Farshid Dayyani
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - Vahid Yaghmai
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - April Choi
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - David Imagawa
- Department of Surgery, University of California Irvine, Orange, CA 92697, USA
| | - Nadine Abi-Jaoudeh
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| |
Collapse
|
46
|
Zhang X, Jin M, Chu Y, Liu F, Qu H, Chen C. PRMT6 promotes colorectal cancer progress via activating MYC signaling. J Transl Med 2025; 23:74. [PMID: 39819457 PMCID: PMC11736931 DOI: 10.1186/s12967-025-06097-y] [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: 09/09/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
Colorectal cancer (CRC) remains a major global health challenge, with high rates of incidence and mortality. This study investigates the role of protein arginine methyltransferase 6 (PRMT6) as an oncogene in CRC and its mechanistic involvement in tumor progression. We found that PRMT6 is significantly overexpressed in CRC tissues compared to adjacent normal tissues and is associated with poorer patient survival. Functional assays demonstrated that PRMT6 promotes CRC cell proliferation, migration, and invasion. Mechanistically, PRMT6 enhances MYC signaling by stabilizing c-MYC through mono-methylation at arginine 371, which inhibits c-MYC poly-ubiquitination and subsequent degradation. This post-translational modification is crucial for PRMT6-induced cancer cell proliferation. Xenograft models further validated that PRMT6 knockdown results in reduced tumor growth and decreased c-MYC levels. Our findings highlight PRMT6 as a key regulator of c-MYC stability and CRC progression, suggesting that targeting PRMT6 or its effects on c-MYC could offer a promising strategy for CRC treatment.
Collapse
Affiliation(s)
- Xin Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, JiNan, 250012, China
| | - Mingxin Jin
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, JiNan, 250012, China
| | - Yali Chu
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, JiNan, 250012, China
| | - Fengjun Liu
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, JiNan, 250012, China
| | - Hui Qu
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, JiNan, 250012, China
| | - Cheng Chen
- Department of General Surgery, Qilu Hospital of Shandong University, 107 West Wenhua Road, JiNan, 250012, China.
| |
Collapse
|
47
|
Tao S, Wang H, Ji Q, Yang Y, Wei G, Li R, Zhou B. Integration of Metabolomics and Transcriptomics to Reveal the Antitumor Mechanism of Dendrobium officinale Polysaccharide-Based Nanocarriers in Enhancing Photodynamic Immunotherapy in Colorectal Cancer. Pharmaceutics 2025; 17:97. [PMID: 39861745 PMCID: PMC11769008 DOI: 10.3390/pharmaceutics17010097] [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: 11/02/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Background: The mechanism of Dendrobium officinale polysaccharide-based nanocarriers in enhancing photodynamic immunotherapy in colorectal cancer (CRC) remains poorly understood. Methods: The effects of TPA-3BCP-loaded cholesteryl hemisuccinate-Dendrobium officinale polysaccharide nanoparticles (DOP@3BCP NPs) and their potential molecular mechanism of action in a tumor-bearing mouse model of CRC were investigated using non-targeted metabolomics and transcriptomics. Meanwhile, a histopathological analysis (H&E staining, Ki67 staining, and TUNEL assay) and a qRT-PCR analysis revealed the antitumor effects of DOP@3BCP NPs with and without light activation. Results: Through metabolomics and transcriptomics analysis, we found an alteration in the metabolome and functional pathways in the examined tumor tissues. The metabolic analysis showed 69 and 60 differentially expressed metabolites (DEMs) in positive- and negative-ion modes, respectively, in the treated samples compared to the Control samples. The transcriptomics analysis showed that 1352 genes were differentially expressed among the three groups. The differentially regulated functional pathways were primally related to the antitumor immune response. The results of the pathological histology assay and qRT-PCR analysis verified the findings of the integrated metabolomics and transcriptomics analysis. Conclusions: Overall, our findings elucidate the potential antitumor mechanisms of the D. officinale polysaccharide-based nanocarrier in enhancing photodynamic immunotherapy in CRC.
Collapse
Affiliation(s)
- Shengchang Tao
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Huan Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (H.W.); (G.W.)
| | - Qiufeng Ji
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yushan Yang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Gang Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (H.W.); (G.W.)
| | - Ruiming Li
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Benjie Zhou
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
48
|
Valciukiene J, Lastauskiene E, Laurinaviciene A, Jakubauskas M, Kryzauskas M, Valkiuniene RB, Augulis R, Garnelyte A, Kavoliunas J, Silinskaite U, Poskus T. Interaction of human gut microbiota and local immune system in progression of colorectal adenoma (MIMICA-1): a protocol for a prospective, observational cohort study. Front Oncol 2025; 14:1495635. [PMID: 39834942 PMCID: PMC11743970 DOI: 10.3389/fonc.2024.1495635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION The current understanding of colorectal carcinogenesis is based on the adenoma-carcinoma sequence, where genetics, intestinal microbiota changes and local immunity shifts seem to play the key roles. Despite the emerging evidence of dysbiotic intestinal state and immune-cell infiltration changes in patients with colorectal adenocarcinoma, early and advanced adenoma as precursors of colorectal cancer, and carcinoma in situ as the following progression, are rather less studied. The newly colon-site adapted AI-based analysis of immune infiltrates is able to predict long-term outcomes of colon carcinoma. Though it could also facilitate the pathologic evaluation of precancerous lesion's potential to progress. Therefore, the purpose of this prospective cohort study (MIMICA-1) is, firstly, to identify the intestinal microbiota and immune infiltration patterns around the normal bowel tissue, early and advanced adenoma, carcinoma in situ, and adenocarcinoma, and secondly, to analyze the immune - microbiome interplay along the steps of conventional colorectal tumorigenesis. METHODS AND ANALYSES This study aims to prospectively recruit 40 patients (10 per group) with confirmed colorectal dysplasia undergoing endoscopic polypectomy, endoscopic mucosal resection for colorectal small (≤1cm), and large (>1cm) adenoma or carcinoma in situ, or biopsy and subsequent colon resection for invasive colorectal cancer, and 10 healthy patients undergoing screening colonoscopy. Stool samples will be collected prior to bowel preparation for the analysis of fecal (luminal) microbiota composition. Biopsy specimens will be taken from the terminal ileum, right colon, left colon, and a pathological lesion in the colon (if present) to assess mucosa-associated microbiota composition and intestinal immunity response. DNA will be extracted from all samples and sequenced using the Illumina MiSeq platform. Unifrac and Bray-Curtis methods will be used to assess microbial diversity. The intestinal immune system response will be examined using digital image analysis where primarily immunohistochemistry procedures for CD3, CD8, CD20 and CD68 immune cell markers will be performed. Thereafter, the count, density and distribution of immunocompetent cells in epithelial and stromal tissue compartments will be evaluated using AI-based platform. The interaction between the microbial shifts and intestinal immune system response in adenoma-carcinoma sequence and the healthy patients will be examined. In addition, fecal samples will be explored for gut microbiota's composition, comparing fecal- and tissue-derived bacterial patterns in healthy gut and along the adenoma-carcinoma sequence. DISCUSSION We hypothesize that changes within the human gut microbiota led to detectable alterations of the local immune response and correlate with the progression from normal mucosa to colorectal adenoma and invasive carcinoma. It is expectable to find more severe gut immune infiltration at dysplasia site, though analyzing invasive colorectal cancer we expect to detect broader mucosa-associated and luminal microbiota changes with subsequent local immune response at near-lesion site and possibly throughout the entire colon. We believe that specific compositional differences detected around premalignant colorectal lesions are critically important for its primary role in initiation and acceleration of colorectal carcinogenesis. Thus, these microbial patterns could potentially supplement fecal immunohistochemical tests for the early non-invasive detection of colorectal adenoma. Moreover, AI-based analysis of immune infiltrates could become additional diagnostic and prognostic tool in precancerous lesions prior to the development of colorectal cancer. REGISTRATION The study is registered at the Australian New Zealand Clinical Trials Registry (ACTRN12624000976583) https://www.anzctr.org.au/.
Collapse
Affiliation(s)
- Jurate Valciukiene
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Egle Lastauskiene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aida Laurinaviciene
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Matas Jakubauskas
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Marius Kryzauskas
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ruta Barbora Valkiuniene
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Renaldas Augulis
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ausra Garnelyte
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Justinas Kavoliunas
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Tomas Poskus
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
49
|
Zhang Y, Cai X, Ma X, Yan H, Wu Q, Tong H, Zheng Z. Delavinone elicits oxidative stress and triggers ferroptosis in colorectal cancer by inhibiting PKCδ-mediated phosphorylation of Nrf2. Chem Biol Interact 2025; 405:111312. [PMID: 39551424 DOI: 10.1016/j.cbi.2024.111312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Ferroptosis is a potential therapeutic approach for colorectal cancer (CRC). Studies have shown that peimine and its analogs exhibit anti-cancer potential; however, the intricate relationship between ferroptosis and their efficacy in fighting CRC remains unclear. In this study, we attempted to assess the therapeutic impact of peimine and its analogs on CRC and unravel the underlying mechanisms. CRC cells and a DSS/AOM-induced CRC mouse model were employed for in vitro and in vivo experiments, molecular interactions and co-immunoprecipitation were used to identify target proteins. Among the compounds, delavinone significantly inhibited CRC cell proliferation and increased cellular lipid ROS levels, MDA accumulation, and GSH depletion; the ferroptosis inhibitors DFO and Fer-1 ameliorated delavinone-induced cell death. Mechanistically, delavinone impedes PKCδ-mediated Nrf2 phosphorylation by inhibiting the kinase activity of PKCδ, thereby decreasing Nrf2 nuclear translocation and downstream GSH synthesis-related gene expression. overexpression of GPX4 weakened the anticancer effect of delavinone, underscoring delavinone's inhibition of the PKCδ/Nrf2/GPX4 signaling axis and induction of ferroptosis in CRC cells. Consistent with in vitro findings, delavinone notably hindered AOM/DSS-induced colorectal carcinogenesis, exhibiting a pronounced pro-ferroptosis effect on CRC. This study delineates that delavinone exerts its anticancer activity by inducing ferroptosis through PKCδ inhibition, consequently reducing Nrf2 phosphorylation. These findings position delavinone as a promising candidate for CRC treatment.
Collapse
Affiliation(s)
- Ya Zhang
- Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China; Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Xiexiao Cai
- Gastrointestinal Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Xiaojing Ma
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huanjuan Yan
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Qifang Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing, 100700, China.
| | - Zhihai Zheng
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
50
|
Xia J, Zhou C, Zhao H, Zhang J, Chai X. LINC01614 Accelerates CRC Progression via STAT1/LINC01614/miR-4443/PFKFB3-Mediated Aerobic Glycolysis. Dig Dis Sci 2025; 70:215-232. [PMID: 39641899 DOI: 10.1007/s10620-024-08756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is an aggressive malignancy among malignant tumours, with a high incidence globally. LINC01614, a long non-coding RNA, has been identified as an essential regulator in multiple cancer types. However, its biological functions and underlying molecular mechanisms in CRC remain largely unknown. METHODS In this study, we employed RT-qPCR to assess the expression levels of LINC01614 in CRC samples. In vitro, glucose metabolism experiments were conducted to evaluate glucose metabolism in cells. The binding relationship between miR-4443, PFKFB3, and LINC01614 was confirmed through fluorescence reporter gene detection. The subcellular localization of LINC01614 in CRC cells was determined using FISH and subcellular fractionation experiments. Additionally, a mouse subcutaneous tumor model was established for in vivo experiments. RESULTS Our findings reveal that LINC01614 is upregulated in CRC tissues. Silencing of LINC01614 suppresses the malignant behaviors of CRC cells, including cell proliferation, invasion, migration, and aerobic glycolysis. Furthermore, we discovered that LINC01614 promotes the expression of PFKFB3. Additional experiments demonstrated that LINC01614 binds to miR-4443, leading to the upregulation of PFKFB3 expression. Further experiments confirmed that the LINC01614/miR-4443/PFKFB3 axis promotes CRC cell malignancy by enhancing aerobic glycolysis. Additionally, we found that STAT1 promotes the transcription of LINC01614. CONCLUSION These findings uncover a novel regulatory pathway wherein STAT1-induced LINC01614 enhances PFKFB3 expression by sponging miR-4443, thereby accelerating CRC development. This understanding may lead to novel therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Jiangyan Xia
- Department of Anesthesiology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
| | - Chenglin Zhou
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China
| | - Heng Zhao
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China
| | - Jun Zhang
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China
| | - Xiaoming Chai
- Department of Anesthesiology, People's Hospital of Xuyi County, Xuyi, Huaian, Jiangsu, China.
| |
Collapse
|