1
|
Kato T, Hikichi T, Nakamura J, Hashimoto M, Kobashi R, Yanagita T, Takagi T, Suzuki R, Sugimoto M, Asama H, Sato Y, Shioya Y, Kobayakawa M, Ohira H. Visibility of esophageal squamous cell carcinoma under iodine staining on texture and color enhancement imaging. DEN OPEN 2025; 5:e370. [PMID: 38725874 PMCID: PMC11079435 DOI: 10.1002/deo2.370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 05/12/2024]
Abstract
Objective Iodine staining on white light imaging (WLI) is the gold standard for detecting and demarcating esophageal squamous cell carcinoma (ESCC). We examined the effects of texture and color enhancement imaging (TXI) on improving the endoscopic visibility of ESCC under iodine staining. Methods Twenty ESCC lesions that underwent endoscopic submucosal dissection were retrospectively included. The color difference between ESCC and the surrounding mucosa (ΔEe) on WLI, TXI, and narrow-band imaging was assessed, and ΔEe under 1% iodine staining on WLI and TXI. Furthermore, the visibility grade determined by endoscopists was evaluated on each imaging. Result The median ΔEe was greater on TXI than on WLI (14.53 vs. 10.71, respectively; p < 0.005). Moreover, the median ΔEe on TXI under iodine staining was greater than the median ΔEe on TXI and narrow-band imaging (39.20 vs. 14.53 vs. 16.42, respectively; p < 0.005 for both). A positive correlation in ΔEe under iodine staining was found between TXI and WLI (correlation coefficient = 0.61, p < 0.01). Moreover, ΔEe under iodine staining on TXI in each lesion was greater than the corresponding ΔEe on WLI. The visibility grade assessed by endoscopists on TXI was also significantly greater than that on WLI under iodine staining (p < 0.01). Conclusions The visibility of ESCC after iodine staining was greater on TXI than on WLI.
Collapse
Affiliation(s)
- Tsunetaka Kato
- Department of EndoscopyFukushima Medical University HospitalFukushimaJapan
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Takuto Hikichi
- Department of EndoscopyFukushima Medical University HospitalFukushimaJapan
| | - Jun Nakamura
- Department of EndoscopyFukushima Medical University HospitalFukushimaJapan
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Minami Hashimoto
- Department of EndoscopyFukushima Medical University HospitalFukushimaJapan
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Ryoichiro Kobashi
- Department of EndoscopyFukushima Medical University HospitalFukushimaJapan
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Takumi Yanagita
- Department of EndoscopyFukushima Medical University HospitalFukushimaJapan
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Tadayuki Takagi
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Rei Suzuki
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Mitsuru Sugimoto
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Hiroyuki Asama
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Yuki Sato
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Yasuo Shioya
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| | - Masao Kobayakawa
- Department of EndoscopyFukushima Medical University HospitalFukushimaJapan
- Medical Research CenterFukushima Medical UniversityFukushimaJapan
| | - Hiromasa Ohira
- Department of GastroenterologyFukushima Medical University School of MedicineFukushimaJapan
| |
Collapse
|
2
|
Fathi M, Taher HJ, Al-Rubiae SJ, Yaghoobpoor S, Bahrami A, Eshraghi R, Sadri H, Asadi Anar M, Gholamrezanezhad A. Role of molecular imaging in prognosis, diagnosis, and treatment of gastrointestinal cancers: An update on new therapeutic methods. World J Methodol 2024; 14:93461. [DOI: 10.5662/wjm.v14.i4.93461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/31/2024] [Accepted: 07/15/2024] [Indexed: 07/26/2024] Open
Abstract
One of the leading causes of cancer-related death is gastrointestinal cancer, which has a significant morbidity and mortality rate. Although preoperative risk assessment is essential for directing patient care, its biological behavior cannot be accurately predicted by conventional imaging investigations. Potential pathophysiological information in anatomical imaging that cannot be visually identified can now be converted into high-dimensional quantitative image features thanks to the developing discipline of molecular imaging. In order to enable molecular tissue profile in vivo, molecular imaging has most recently been utilized to phenotype the expression of single receptors and targets of biological therapy. It is expected that molecular imaging will become increasingly important in the near future, driven by the expanding range of biological therapies for cancer. With this live molecular fingerprinting, molecular imaging can be utilized to drive expression-tailored customized therapy. The technical aspects of molecular imaging are first briefly discussed in this review, followed by an examination of the most recent research on the diagnosis, prognosis, and potential future clinical methods of molecular imaging for GI tract malignancies.
Collapse
Affiliation(s)
- Mobina Fathi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | | | | | - Shirin Yaghoobpoor
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Ashkan Bahrami
- Faculty of Medicine, Kashan University of Medical Sciences, Kashan 1617768911, Iran
| | - Reza Eshraghi
- Faculty of Medicine, Kashan University of Medical Sciences, Kashan 1617768911, Iran
| | - Hossein Sadri
- Faculty of Medicine, Kashan University of Medical Sciences, Kashan 1617768911, Iran
| | - Mahsa Asadi Anar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Ali Gholamrezanezhad
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| |
Collapse
|
3
|
Sewastjanow-Silva M, Kwiatkowski E, Yamashita K, Abdelhakeem A, Yoshimura K, Vicentini ER, Pizzi MP, Jin J, Fan Y, Zou G, Wang L, Yin F, Dhar SS, Blum Murphy M, Mares JE, Li JJ, Gan Q, Waters RE, Rogers JE, Ajani JA. Three biomarkers (HER2, PD-L1, and microsatellite status) in a large cohort of metastatic gastroesophageal adenocarcinomas: The MD Anderson Cancer Center experience. Int J Cancer 2024; 155:2277-2286. [PMID: 38995150 DOI: 10.1002/ijc.35090] [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: 01/16/2024] [Revised: 05/11/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024]
Abstract
Human epidermal growth factor receptor-2 (HER2), programmed death-ligand 1 (PD-L1), and microsatellite (MS) status are well-established biomarkers in gastroesophageal adenocarcinomas (GEAs). However, it is unclear how the combination of these biomarkers is associated with clinicopathological factors and prognosis. This retrospective study included baseline metastatic GEA patients who were tested for all three biomarkers (HER2, PD-L1, and MS status) at the MD Anderson Cancer Center between 2012 and 2022. Stratification was performed according to the combination of biomarker profiles: triple negative (TN), single positive (SP), and multiple positive (MP). Comparative analyses of clinicopathological factors and survival using combinations of biomarkers were performed. Among the 698 GEA patients analyzed, 251 (36.0%) were classified as TN, 334 (47.9%) as SP, and 113 (16.1%) as MP. The MP group showed a significant association with tumors located in the esophagus (p < .001), well to moderate differentiation (p < .001), and the absence of signet ring cells (p < .001). In the survival analysis, MP group had a significantly longer overall survival (OS) compared to the other groups (MP vs. TN, p < .001 and MP vs. SP, p < .001). Multivariate Cox regression analysis revealed that MP serves as an independent positive prognostic indicator for OS (hazard ratio = 0.63, p < .01). Our findings indicate that MP biomarkers are associated with a favorable prognosis in metastatic GEA. These results are reflective of clinical practice and offer valuable insights into how therapeutics and future biomarkers could influence therapy/prognosis.
Collapse
Affiliation(s)
- Matheus Sewastjanow-Silva
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Evan Kwiatkowski
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kohei Yamashita
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ahmed Abdelhakeem
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Katsuhiro Yoshimura
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ernesto R Vicentini
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Melissa P Pizzi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yibo Fan
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gengyi Zou
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lingzhi Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Feng Yin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shilpa S Dhar
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mariela Blum Murphy
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeannette E Mares
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jenny J Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qiong Gan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rebecca E Waters
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jane E Rogers
- Department of Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Živanović M, Selaković M, Pavić A, Selaković Ž, Šolaja B, Santibanez JF, Srdić-Rajić T. Unveiling the 4-aminoquinoline derivatives as potent agents against pancreatic ductal adenocarcinoma (PDAC) cell lines. Chem Biol Interact 2024; 404:111281. [PMID: 39428053 DOI: 10.1016/j.cbi.2024.111281] [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: 07/31/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Common antimalarials such as artemisinins, chloroquine and their derivatives also possess potent anti-inflamantory, antiviral and anticancer properties. In the search for new therapeutics to combat difficult-to-treat pancreatic carcinomas, we unveiled that 4-aminoquinoline derivatives, with significant antiplasmodial properties and a great safety profile in vivo, have remarkable anticancer activity against pancreatic ductal adenocarcinoma (PDAC) and considerable efficacy in the xenograft model in vivo. The aim of the present study was to further investigate anticancer properties of these compounds in a drug-repurposing manner. The compounds showed profound cytotoxic effects at nanomolar to low micromolar concentration in 2D cultured cells (in vitro) and in the zebrafish PDAC xenograft model (in vivo). A deeper insight into their mechanisms of cytotoxic action showed these compounds induce apoptosis while increasing reactive oxygen species levels along with autophagy inhibition. Additional investigation of the autophagy modulation proved that tested quinoline derivatives cause P62 and LC3-II accumulation in PDAC cells alongside lysosomal alkalinization. Further, in vivo toxicity studies in the zebrafish model showed low toxicity without developmental side effects of the investigated 4-aminoquinolines, while the applied compounds effectively inhibited tumor growth and prevented the metastasis of xenografted pancreatic cells. Taken together, these results highlight the 4-aminoquinolines as privileged structures that ought to be investigated further for potential application in pancreatic carcinoma treatment.
Collapse
Affiliation(s)
- Marija Živanović
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia; Department of Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Milica Selaković
- Innovative Centre of the Faculty of Chemistry in Belgrade, ltd., Studentski Trg 12-16, 11158, Belgrade, Serbia.
| | - Aleksandar Pavić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042, Belgrade, Serbia
| | - Života Selaković
- University of Belgrade - Faculty of Chemistry, Studentski Trg 12-16, 11158, Belgrade, Serbia
| | - Bogdan Šolaja
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11158, Belgrade, Serbia
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Tatjana Srdić-Rajić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia.
| |
Collapse
|
5
|
Sui X, Wang W, Zhang D, Xu J, Li J, Jia Y, Qin Y. Integrated analysis of ferroptosis and stemness based on single-cell and bulk RNA-sequencing data provide insights into the prognosis and treatment of esophageal carcinoma. Gene 2024; 927:148701. [PMID: 38885819 DOI: 10.1016/j.gene.2024.148701] [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: 02/28/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Cancer stem cells (CSCs) play a significant role in the recurrence and drug resistance of esophageal carcinoma (ESCA). Ferroptosis is a promising anticancer therapeutic strategy that effectively targets CSCs exhibiting high tumorigenicity and treatment resistance. However, there is a lack of research on the combined role of ferroptosis-related genes (FRGs) and stemness signature in the prognosis of ESCA. METHODS The cellular compositions were characterized using single-cell RNA sequencing (scRNA-seq) data from 18 untreated ESCA samples. 50 ferroptosis-related stemness genes (FRSGs) were identified by integrating FRGs with stemness-related genes (SRGs), and then the cells were grouped by AUCell analysis. Next, functional enrichment, intercellular communication, and trajectory analyses were performed to characterize the different groups of cells. Subsequently, the stem-ferr-index was calculated using machine learning algorithms based on the expression profiles of the identified risk genes. Additionally, therapeutic drugs were predicted by analyzing the GDSC2 database. Finally, the expression and functional roles of the identified marker genes were validated through in vitro experiments. RESULTS The analysis of scRNA-seq data demonstrates the diversity and cellular heterogeneity of ESCA. Then, we identified 50 FRSGs and classified cells into high or low ferroptosis score stemness cells accordingly. Functional enrichment analysis conducted on the differentially up-regulated genes between these groups revealed predominant enrichment in pathways associated with intercellular communication and cell differentiation. Subsequently, we identified 9 risk genes and developed a prognostic signature, termed stem_ferr_index, based on these identified risk genes. We found that the stem-ferr-index was correlated with the clinical characteristics of patients, and patients with high stem-ferr-index had poor prognosis. Furthermore, we identified four drugs (Navitoclax, Foretinib, Axitinib, and Talazoparib) with potential efficacy targeting patients with a high stem_ferr_index. Additionally, we delineated two marker genes (STMN1 and SLC2A1). Particularly noteworthy, SLC2A1 exhibited elevated expression levels in ESCA tissues and cells. We provided evidence suggesting that SLC2A1 could influence the migration, invasion, and stemness of ESCA cells, and it was associated with sensitivity to Foretinib. CONCLUSION This study constructed a novel ferroptosis-related stemness signature, identified two marker genes for ESCA, and provided valuable insights for developing more effective therapeutic targets targeting ESCA CSCs in the future.
Collapse
Affiliation(s)
- Xin Sui
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Wenjia Wang
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Daidi Zhang
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Jiayao Xu
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Jing Li
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Yongxu Jia
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Yanru Qin
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
Maresova P, Rezny L, Hruska J, Klimova B, Swanstrom LL, Kuca K. Diagnosis and treatment of patients with gastroesophageal reflux disease - a systematic review of cost-effectiveness and economic burden. BMC Health Serv Res 2024; 24:1351. [PMID: 39501242 PMCID: PMC11539747 DOI: 10.1186/s12913-024-11781-8] [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: 01/21/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND This study aims to review the existing knowledge on the cost-effectiveness and item costs related to the diagnosis and treatment of gastroesophageal reflux disease (GERD) patients at different stages. METHODS The study adhered to the PRISMA guidelines. The systematic search involved several steps: finding and identifying relevant articles, filtering them according to the set criteria, and examining the final number of selected articles to obtain the primary information. The number of articles published between 2013 and September 2024 in the Web of Science and PubMed databases was considered. The CHEERS checklist was used for the risk of bias assessment. Ultimately, 36 studies were included. RESULTS Regarding the cost-effectiveness of GERD treatment, Proton pump inhibitors (PPIs) appeared to be the dominant solution for non-refractory patients. However, this might change with the adoption of the novel drug vonoprazan, which is more effective and cheaper. With advancements in emerging technologies, new diagnostic and screening approaches such as Endosheath, Cytosponge, and combined multichannel intraluminal impedance and pH monitoring catheters should be considered, with potential implications for optimal GERD management strategies. DISCUSSION The new diagnostic methods are reliable, safe, and more comfortable than standard procedures. PPIs are commonly used as the first line of treatment for GERD. Surgery, such as magnetic sphincter augmentation or laparoscopic fundoplication, is only recommended for patients with treatment-resistant GERD or severe symptoms. OTHER Advances in emerging technologies for diagnostics and screening may lead to a shift in the entire GERD treatment model, offering less invasive options and potentially improving patients' quality of life.
Collapse
Affiliation(s)
- Petra Maresova
- Betthera s.r.o, Hradec Kralove, Czech Republic
- Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Lukas Rezny
- Betthera s.r.o, Hradec Kralove, Czech Republic
- Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Hruska
- Betthera s.r.o, Hradec Kralove, Czech Republic
- Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Blanka Klimova
- Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | | | - Kamil Kuca
- Betthera s.r.o, Hradec Kralove, Czech Republic.
- Faculty of Informatics and Management, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
7
|
Chatzipanagiotou OP, Woldesenbet S, Munir MM, Catalano G, Khalil M, Rashid Z, Altaf A, Pawlik TM. Impact of Contemporary Redlining on Healthcare Disparities Among Patients with Gastrointestinal Cancer: A Mediation Analysis. Ann Surg Oncol 2024:10.1245/s10434-024-16373-8. [PMID: 39485616 DOI: 10.1245/s10434-024-16373-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Historically, housing policies have perpetuated the marginalization and economic disinvestment of redlined neighborhoods. Residential segregation persists nowadays in the form of contemporary redlining, promoting healthcare disparities. The current study sought to assess the effect of redlining on oncological outcomes of patients with gastrointestinal cancer and identify mediators of the association. METHODS Patients with colorectal or hepatobiliary cancer were identified from the linked Surveillance, Epidemiology, and End Results (SEER)-Medicare database (2007-2019). The contemporary redlining index, a measure of mortgage lending bias, was assessed relative to disease stage at diagnosis, receipt of appropriate treatment, textbook outcome, and mortality. Mediation analysis was used to identify socioeconomic, structural, and clinical mediating factors. RESULTS Among 94,988 patients, 32.2% resided in high (n = 23,872) and highest (n = 6,791) redlining census tracts compared with 46.2% in neutral and 21.6% in low redlining tracts. The proportion of Black, Hispanic, and White patients experiencing high and highest redlining was 65.9%, 41.6%, and 27.9%, respectively. Highest redlining was associated with 18.2% higher odds of advanced disease at diagnosis, greater odds of not undergoing surgery for localized disease (adjusted odds ratio [aOR] 1.363, 95% confidence interval [CI] 1.219-1.524) or not receiving chemotherapy for advanced disease (aOR 1.385, 95% CI 1.216-1.577), and 26.7% lower odds of textbook outcome achievement. Mediation analysis for appropriate treatment quantified the proportion of the association driven by socioeconomic status, racial/ethnic minority status, racial/economic segregation, primary care shortage, and housing/transportation. CONCLUSIONS Contemporary redlining contributed both directly, and via downstream factors, to disparities in oncological care and outcomes of patients with gastrointestinal cancer.
Collapse
Affiliation(s)
| | - Selamawit Woldesenbet
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Muhammad Musaab Munir
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Giovanni Catalano
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, University of Verona, Verona, Italy
| | - Mujtaba Khalil
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zayed Rashid
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Abdullah Altaf
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
8
|
He S, Zhang Z, Song G, Wang Z, Li H, Cao M, Yang F, Sun D, Yan X, Zhang S, Teng Y, Li Q, Xia C, Chen W. Personalized starting age of gastric cancer screening based on individuals' risk profiles: a population-based, prospective study. J Natl Cancer Inst 2024; 116:1775-1783. [PMID: 38976632 DOI: 10.1093/jnci/djae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND The current recommended starting age for gastric cancer screening lacks unified guideline and individualized criteria. We aimed to determine the risk-stratified starting age for gastric cancer screening in China based on individuals' risk profiles and to develop an online calculator for clinical application. METHODS In this multicenter, population-based, prospective study, we allocated participants enrolled between 2015 and 2017 (N = 59 771, aged 40-69 years) to screened and unscreened groups and observed them for primary endpoints: gastric cancer occurrence as well as all-cause and gastric cancer-specific death. Median follow-up was 6.07 years. To determine the reference starting age, the effectiveness of gastric cancer screening was assessed by age group after propensity score matching. Further, we categorized the calculated individual risk scores (using well-established risk factors) by quantile. Subsequently, we used age-specific, 10-year cumulative risk curves to estimate the risk-stratified starting age-that is, when the individual's risk level matches the reference starting age risk threshold. RESULTS During follow-up, 475 gastric cancer case patients, 182 gastric cancer-related deaths, and 1860 all-cause deaths occurred. All-cause and gastric cancer-specific mortality decreased among screened individuals 45 years of age and older and 50 to 59 years of age, respectively. Thus, the average population (referent) starting age was set as 50 years. The 10-year cumulative risk of gastric cancer in the average population aged 50 years was 1.147%. We stratified the starting age using 8 risk factors and categorized participants as low-risk, medium-risk, and high-risk individuals whose risk-stratified starting age was 58, 50, and 46 years, respectively. CONCLUSION Although high-risk individuals warrant starting gastric cancer screening 3 to 5 years earlier than for the average population (aged 50 years), low-risk individuals can tolerate delayed screening. Our online, personalized starting age calculator will help with risk-adapted gastric cancer screening (https://web.consultech.com.cn/gastric/#/).
Collapse
Affiliation(s)
- Siyi He
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiyi Zhang
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital, Wuwei, China
| | - Guohui Song
- Department of Epidemiology, Cancer Institute/Hospital of Ci County, Handan, China
| | - Zhenhai Wang
- Department of General Surgery, Linzhou Cancer Hospital, Linzhou, China
| | - He Li
- Office of National Cancer Regional Medical Centre in Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Maomao Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fan Yang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dianqin Sun
- Office of Cancer Screening, 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 Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Xinxin Yan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shaoli Zhang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Teng
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qianru Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changfa Xia
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Zhang Y, Yang Y, Hou Y, Yan W, Zhang X, Huang X, Song Q, He F, Wang J, Sun A, Tian C. ZNF8 promotes progression of gastrointestinal cancers via a p53-dependent mechanism. Cell Signal 2024; 123:111354. [PMID: 39173856 DOI: 10.1016/j.cellsig.2024.111354] [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: 03/20/2024] [Revised: 07/09/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024]
Abstract
p53 is a critical tumor suppressor, and the disruption of its normal function is often a prerequisite for the development or progression of tumors. Our previous works revealed that multiple members of Krüppel-associated box (KRAB) domain zinc-finger proteins (KZFPs) family regulate p53 transcriptional activity by interacting with it. But the tumor biology functions of these members have not been fully elucidated. Here, the pan-cancer analysis related to gastrointestinal cancers (GICs) revealed that ZNF8, a p53-interacting protein, is an unfavorable prognostic factor for patients with malignancies. ZNF8 interacts with p53 and further depresses its transcriptional activity in colon cancer cells. The knockdown of ZNF8 or the overexpression of ZNF8 inhibits or facilitates the in vitro colony formation, migration, invasion, and angiogenesis of p53+/+ colon cancer HCT116 cells, HepG2 cells and EC109 cells rather than p53-/- colon cancer HCT116 cells and p53-knockout HepG2 cells, respectively. Xenograft experiments conducted in vivo also showed that the knockdown of ZNF8 in p53+/+ but not in p53-/- HCT116 cells curbs the tumor growth and metastasis to lung, leading to an extended life span for tumor-bearing mice. Clinically, two independent immunohistochemistry cohorts of colon cancer and esophageal cancer also indicated that ZNF8 is higher expression in carcinoma tissues than adjacent tissues and this is associated with worse overall survival outcomes in patients without harboring p53 mutation. Together, our results provide insight into the p53-specific tumor oncogenic function of ZNF8. ZNF8 may prove to be a potential target for GICs treatment.
Collapse
Affiliation(s)
- Yiming Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Yingchuan Yang
- College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yushan Hou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Wei Yan
- The First Medical Center of Chinese PLA General Hospital, Beijing 100036, China
| | - Xiuyuan Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Xiaofen Huang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; College of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Qin Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; College of Life Sciences, Hebei University, Baoding 071002, Hebei, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Jian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China; College of Life Sciences, Hebei University, Baoding 071002, Hebei, China.
| | - Aihua Sun
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China; College of Life Sciences, Hebei University, Baoding 071002, Hebei, China.
| | - Chunyan Tian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing 102206, China; College of Life Sciences, Hebei University, Baoding 071002, Hebei, China.
| |
Collapse
|
10
|
Sassano M, Seyyedsalehi MS, Siea AC, Boffetta P. Occupational arsenic exposure and digestive and head and neck cancers: A systematic review and meta-analysis. ENVIRONMENTAL RESEARCH 2024; 260:119643. [PMID: 39053758 DOI: 10.1016/j.envres.2024.119643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/08/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Arsenic is a known carcinogen for the lungs, the bladder, and the skin, while systematic evidence on other cancer types is lacking, especially for occupational exposure. Thus, we aimed to systematically summarize current evidence on the association between occupational arsenic exposure and digestive cancers, including head and neck cancer (HNC). We conducted a systematic review on Pubmed, Web of Science, and Embase search engines. We computed pooled relative risks (RRs) and corresponding 95% confidence intervals (CIs) using DerSimonian and Laird random-effects model. Occurrence of publication bias was assessed using contour-enhanced funnel plots and Egger's test. Twenty-two studies on digestive cancers and 11 on HNC were included in the meta-analysis. RRs for the association with occupational exposure to arsenic of 1.23 (95% CI: 1.07-1.40; I2 = 72.3%, p < 0.001) and 1.08 (95% CI: 0.76-1.53; I2 = 76.6%, p < 0.001) for digestive cancer and HNC, respectively, were observed. As for specific cancer types, arsenic was associated with rectal cancer (RR: 1.51; 95% CI: 1.003-2.28; I2 = 37.0%, p = 0.174), but not with other investigated cancer types. No clear evidence of publication bias was found. The results of our study suggest that the observed association between occupational arsenic exposure and digestive cancer might be mainly driven by a positive association for rectal cancer, while arsenic exposure did not appear to be associated with HNC. However, further high-quality studies with detailed assessment of arsenic exposure are warranted to clarify the potential association of arsenic with digestive cancers and HNC.
Collapse
Affiliation(s)
- Michele Sassano
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Monireh Sadat Seyyedsalehi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Andrei Cosmin Siea
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; Stony Brook Cancer Center, Stony Brooke University, Stony Brook, NY, USA; Department of Family, Population and Preventive Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
11
|
Li Y, Jia X, Li C, Sun H, Nie S, Giovannucci EL, Liu L. The Global Incident Gastrointestinal Cancers Attributable to Suboptimal Diets From 1990 to 2018. Gastroenterology 2024; 167:1141-1151. [PMID: 39019406 DOI: 10.1053/j.gastro.2024.07.009] [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: 03/14/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND & AIMS The contribution of suboptimal diets to gastrointestinal (GI) cancer incidence globally remains unquantified, and we aimed to evaluate it. METHODS Comprehensive meta-analyses and rigorous evidence-grading assessment identified the associations between suboptimal diets and 6 GI cancers and their subtypes. A comparative risk assessment model was used to estimate the proportional attributable burden and attributable rate of GI cancers to suboptimal diets by using the corroborative association estimates. In addition, correlation assessments with the Sociodemographic Index were carried out. RESULTS In 2018, 21.5% (95% uncertainty interval, 19.1%-24.5%) of incident GI cancer cases globally were attributable to suboptimal diets, maintaining a relatively stable proportion since 1990 (22.4%; 19.7%-25.6%), whereas the absolute diet-attributable cases doubled from 580,862 (510,658-664,076) in 1990 to 1,039,877 (923,482-1,187,244) in 2018. Excessive processed meat consumption (5.9%; 4.2%-7.9%), insufficient fruit intake (4.8%; 3.8%-5.9%), and insufficient whole grain intake (3.6%; 2.8%-5.1%) were the most significant dietary risk factors in 2018, a shift from 1990 when the third major concern was insufficient nonstarchy vegetable intake. In addition, Central and Eastern Europe and Central Asia experienced the highest attributable burden across regions in both 1990 (31.6%; 27.0%-37.4%) and 2018 (31.6%; 27.3%-36.5%), and a positive correlation (P < .001) between the Sociodemographic Index and the attributable GI cancer incidence was observed. CONCLUSIONS Although the proportional attributable GI incidence remains relatively stable, the doubling of absolute cases from 1990 to 2018, along with the discrepancies among urbanicity and countries/regions, informs dietary priorities and more targeted preventive measures.
Collapse
Affiliation(s)
- Yiting Li
- Department of Epidemiology and Biostatistics, Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Jia
- Department of Epidemiology and Biostatistics, Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caiyu Li
- Department of Epidemiology and Biostatistics, Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoxin Sun
- Department of Epidemiology and Biostatistics, Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaofa Nie
- Department of Epidemiology and Biostatistics, Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Li Liu
- Department of Epidemiology and Biostatistics, Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Colorectal Cancer; Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, Hubei 430030, PR China; Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, Hubei, PR China.
| |
Collapse
|
12
|
Cheng S, Chen J, Li Q, Nie Y, Ni T, Peng C, Luo X, Yasin P, Zhang S, Tang J, Liu Z. Protective effect of folic acid on MNNG-induced proliferation of esophageal epithelial cells via the PI3K/AKT/mTOR signaling pathway. J Nutr Biochem 2024; 133:109702. [PMID: 39025456 DOI: 10.1016/j.jnutbio.2024.109702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 06/19/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Recent research has revealed that N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) constitutes a significant risk factor in the development of esophageal cancer. Several investigations have elucidated the beneficial impact of folic acid (FA) in safeguarding esophageal epithelial cells against MNNG-induced damage. Therefore, we hypothesized that FA might prevent MNNG-induced proliferation of esophageal epithelial cells by interfering with the PI3K/AKT/mTOR signaling pathway. In vivo experiments, we found that FA antagonized MNNG-induced proliferation of rat esophageal mucosal epithelial echinocytes and activation of the PI3K/AKT/mTOR signaling pathway. In our in vitro experiments, it was observed that acute exposure to MNNG for 24 h led to a decrease in proliferative capacity and inhibition of the PI3K/AKT/mTOR signaling pathway in an immortalized human normal esophageal epithelial cell line (Het-1A), which was also ameliorated by supplementation with FA. We successfully established a Het-1A-T-cell line by inducing malignant transformation in Het-1A cells through exposure to MNNG. Notably, the PI3K/AKT2/mTOR pathway showed early suppression followed by activation during this transition. Next, we observed that FA inhibited cell proliferation and activation of the PI3K/AKT2/mTOR signaling pathway in Het-1A-T malignantly transformed cells. We further investigated the impact of 740Y-P, a PI3K agonist, and LY294002, a PI3K inhibitor, on Het-1A-T-cell proliferation. Overall, our findings show that FA supplementation may be beneficial in safeguarding normal esophageal epithelial cell proliferation and avoiding the development of esophageal cancer by decreasing the activation of the MNNG-induced PI3K/AKT2/mTOR signaling pathway.
Collapse
Affiliation(s)
- Suizhi Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacology, School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Jin Chen
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Qianhui Li
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacology, School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Yuhong Nie
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Ting Ni
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Caiting Peng
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Xi Luo
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Pazilat Yasin
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Shumin Zhang
- Department of Biochemistry, School of Basic Medical Sciences, North Sichuan Medical College, Nanchong 637000, China
| | - Jiancai Tang
- Department of Biochemistry, School of Basic Medical Sciences, North Sichuan Medical College, Nanchong 637000, China
| | - Zhenzhong Liu
- Department of Nutrition and Food Hygiene, School of Public Health, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
13
|
Bohm MS, Ramesh AV, Pierre JF, Cook KL, Murphy EA, Makowski L. Fecal microbial transplants as investigative tools in cancer. Am J Physiol Gastrointest Liver Physiol 2024; 327:G711-G726. [PMID: 39301964 DOI: 10.1152/ajpgi.00171.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/22/2024]
Abstract
The gut microbiome plays a critical role in the development, progression, and treatment of cancer. As interest in microbiome-immune-cancer interactions expands, the prevalence of fecal microbial transplant (FMT) models has increased proportionally. However, current literature does not provide adequate details or consistent approaches to allow for necessary rigor and experimental reproducibility. In this review, we evaluate key studies using FMT to investigate the relationship between the gut microbiome and various types of cancer. In addition, we will discuss the common pitfalls of these experiments and methods for improved standardization and validation as the field uses FMT with greater frequency. Finally, this review focuses on the impacts of the gut and extraintestinal microbes, prebiotics, probiotics, and postbiotics in cancer risk and response to therapy across a variety of tumor types.NEW & NOTEWORTHY The microbiome impacts the onset, progression, and therapy response of certain types of cancer. Fecal microbial transplants (FMTs) are an increasingly prevalent tool to test these mechanisms that require standardization by the field.
Collapse
Affiliation(s)
- Margaret S Bohm
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Arvind V Ramesh
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Joseph F Pierre
- Department of Nutritional Sciences, College of Agriculture and Life Science, The University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Katherine L Cook
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, United States
| | - Liza Makowski
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
14
|
Kotelevets SM. Global strategy for prevention of gastric cancer. World J Clin Cases 2024; 12:6353-6357. [PMID: 39464323 PMCID: PMC11438683 DOI: 10.12998/wjcc.v12.i30.6353] [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: 06/15/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
Global prevention of gastric cancer needs to increase its level of effectiveness. The prevention strategy should include all stages of primary and secondary prevention. The necessary steps to prevent gastric cancer are the following: Maintaining a healthy lifestyle and diet, avoiding smoking and alcohol; serological screening of Helicobacter pylori infections and eradication; serological screening of atrophic gastritis in the population over 45 years of age and identification of severe atrophic gastritis with a high risk of developing gastric cancer; verification of atrophic gastritis and precancerous changes in the gastric mucosa using modern endoscopic (confocal laser endomicroscopy, narrow-spectrum imaging, and magnifying endoscopy) and morphological methods among patients with severe atrophic gastritis who were identified using serological screening; treatment of patients with atrophic gastritis during diagnosis verification; annual endoscopic and morphological monitoring of patients with atrophic gastritis during permanent treatment; annual serological monitoring of patients with atrophic gastritis who refused endoscopic and morphological monitoring; and radical treatment of patients with verified early gastric cancer. Ways to implement the algorithm for the global strategy for the prevention of gastric cancer (protocol of practical recommendations) are: State, government, and municipal programs; departmental programs of health departments; family doctors for patients who have a contract at the initiative of the doctor; family doctors for patients with a contract at the patient's initiative; and within private healthcare system where both doctors and patients can initiate the implementation of algorithm.
Collapse
Affiliation(s)
- Sergey M Kotelevets
- Department of Therapy, North Caucasus State Academy, Cherkessk 369000, Russia
| |
Collapse
|
15
|
Fenu G, Griñán-Lisón C, Pisano A, González-Titos A, Farace C, Fiorito G, Etzi F, Perra T, Sabalic A, Toledo B, Perán M, Solinas MG, Porcu A, Marchal JA, Madeddu R. Unveiling the microRNA landscape in pancreatic ductal adenocarcinoma patients and cancer cell models. BMC Cancer 2024; 24:1308. [PMID: 39448959 PMCID: PMC11515555 DOI: 10.1186/s12885-024-13007-w] [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: 03/07/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge due to late-stage diagnoses resulting from nonspecific early symptoms and the absence of early diagnostic biomarkers. MicroRNAs (miRNAs) play a crucial role in regulating diverse biological processes, and their abnormal expression is observed in various diseases, including cancer. Cancer stem cells (CSCs) are thought to act as a driving force in PDAC spread and recurrence. In pursuing the goal of unravelling the complexities of PDAC and its underlying molecular mechanisms, our study aimed to identify PDAC-associated miRNAs and relate them to disease progression, focusing on their involvement in various PDAC stages in patients and in reliable in vitro models, including pancreatic CSC (PaCSC) models. METHODS The miRNA profiling datasets of serum and solid biopsies of PDAC patients deposited in GEO DataSets were analyzed by REML-based meta-analysis. The panel was then investigated by Real Time PCR in serum and solid biopsies of 37 PDAC patients enrolled in the study, as well as on BxPC-3 and AsPC-1 PDAC cell lines. We extended our focus towards a possible role of PDAC-associated miRNAs in the CSC phenotype, by inducing CSC-enriched pancreatospheres from BxPC-3 and AsPC-1 PDAC cell lines and performed differential miRNA expression analysis between PaCSCs and monolayer-grown PDAC cell lines. RESULTS Meta-analysis showed differentially expressed miRNAs in blood samples and cancerous tissues of PDAC patients, allowing the identification of a panel of 9 PDAC-associated miRNAs. The results emerging from our patients fully confirmed the meta-analysis for the majority of miRNAs under investigation. In vitro tasks confirmed the aberrant expression of the panel of PDAC-associated miRNAs, with a dramatic dysregulation in PaCSC models. Notably, PaCSCs have shown significant overexpression of miR-4486, miR-216a-5p, and miR-216b-5p compared to PDAC cell lines, suggesting the recruitment of such miRNAs in stemness-related molecular mechanisms. Globally, our results showed a dual behaviour of miR-216a-5p and miR-216b-5p in PDAC while miR-4486, miR-361-3p, miR-125a-5p, miR-320d expression changes during the disease suggest they could promote PDAC initiation and progression. CONCLUSIONS This study contributed to an enhanced comprehension of the role of miRNAs in the development and progression of PDAC, shedding new light on the miRNA landscape in PDAC and its intricate interplay with CSCs, and providing specific insights useful in the development of miRNA-based diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Grazia Fenu
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, 18016, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, 18071, Spain
| | - Andrea Pisano
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Aitor González-Titos
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, 18016, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain
| | - Cristiano Farace
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy.
- National Institute of Biostructures and Biosystems, Rome, 00136, Italy.
| | - Giovanni Fiorito
- Clinical Bioinformatics Unit, IRCSS Istituto Giannina Gaslini, Genoa, 16147, Italy
| | - Federica Etzi
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Teresa Perra
- Department of Medicine, Surgery and Pharmacy - Unit of General Surgery, University of Sassari, Sassari, 07100, Italy
| | - Angela Sabalic
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
| | - Belén Toledo
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain
- Department of Health Sciences, University of Jaén, Jaén, 23071, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, 23071, Spain
| | | | - Alberto Porcu
- Department of Medicine, Surgery and Pharmacy - Unit of General Surgery, University of Sassari, Sassari, 07100, Italy
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, 18016, Spain.
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University of Granada, Granada, 18071, Spain.
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18016, Spain.
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18016, Spain.
| | - Roberto Madeddu
- Department of Biomedical Science, University of Sassari, Sassari, 07100, Italy
- National Institute of Biostructures and Biosystems, Rome, 00136, Italy
| |
Collapse
|
16
|
Zhu X, Xue J, Jiang H, Xue D. CAR-NK cells for gastrointestinal cancer immunotherapy: from bench to bedside. Mol Cancer 2024; 23:237. [PMID: 39443938 PMCID: PMC11515662 DOI: 10.1186/s12943-024-02151-3] [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: 07/14/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers represent a significant health burden worldwide. Their incidence continues to increase, and their management remains a clinical challenge. Chimeric antigen receptor (CAR) natural killer (NK) cells have emerged as a promising alternative to CAR-T cells for immunotherapy of GI cancers. Notably, CAR-NK cells offer several advantages, including reduced risk of graft-versus-host disease, lower cytokine release syndrome, and the ability to target cancer cells through both CAR-dependent and natural cytotoxic mechanisms. MAIN BODY This review comprehensively discusses the development and applications of CAR-NK cells in the treatment of GI cancers. We explored various sources of NK cells, CAR design strategies, and the current state of CAR-NK cell therapy for GI cancers, highlighting recent preclinical and clinical trials. Additionally, we addressed existing challenges and propose potential strategies to enhance the efficacy and safety of CAR-NK cell therapy. CONCLUSIONS Our findings highlight the potential of CAR-NK cells to revolutionize GI cancer treatment and pave the way for future clinical applications.
Collapse
Affiliation(s)
- Xingwang Zhu
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Jieyun Xue
- China Medical University, Shenyang, Liaoning Province, 110000, P.R. China
| | - Hongzhou Jiang
- Department of Neurosurgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Dongwei Xue
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China.
| |
Collapse
|
17
|
Kafi L, Abu-Odah H, Xu Q. Experiences and needs of colorectal cancer survivors in resource-limited countries: A qualitative descriptive study in Syria. Eur J Oncol Nurs 2024; 73:102715. [PMID: 39471701 DOI: 10.1016/j.ejon.2024.102715] [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: 02/24/2024] [Revised: 08/16/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
PURPOSE Colorectal cancer (CRC) is one of the three most prevalent cancers globally, with incidence rates projected to increase. CRC survivors face various challenges due to the disease and its treatments. However, there is limited evidence on the experiences and needs of CRC survivors in resource-constrained countries. This study aimed to explore the experiences and needs of CRC survivors in Syria and to identify perceived opportunities for improvement. METHODS A descriptive qualitative study design was employed, using semi-structured in-depth interviews with 12 adult CRC survivors who had completed treatment at least 6 months prior, selected through purposive sampling. Data were analyzed using an inductive thematic analysis approach. RESULTS The interviews revealed 15 major themes: 11 concerning the experiences of CRC survivors and four related to their needs. The survivors' experiences included physical burden, psychological expressions, economic burden, social withdrawal, stigmatization, family and friend supports, cultural and social impact, religion and coping mechanisms, relationship with healthcare professionals, support from healthcare system. Their needs encompassed emotional and psychological, informational, financial, and sexual needs. CONCLUSION This study highlights the unpredictable psychological, economic and physical burdens experienced by Syrian CRC survivors. Personalized care plans that address unmet needs of survivors are crucial to improving their quality of life. Prioritizing the development of comprehensive survivorship care plans that address the multifaceted needs of CRC survivors is crucial. Additionally, further qualitative research is essential to gain a deeper understanding of the challenges faced by these survivors and to inform the design of effective interventions.
Collapse
Affiliation(s)
- Lina Kafi
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu province, 211166, China; Department of Adult Nursing, Faculty of Nursing, Tishreen University, Latakia, Syria.
| | - Hammoda Abu-Odah
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong, 999077, China; WHO Collaborating Centre for Community Health Services (WHOCC), School of Nursing, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Qin Xu
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu province, 211166, China.
| |
Collapse
|
18
|
Li Y, Du J, Du L, Li S, Zhang J. Managements for perioperative anxiety in patients with gastrointestinal cancers. Front Psychiatry 2024; 15:1391403. [PMID: 39497898 PMCID: PMC11533141 DOI: 10.3389/fpsyt.2024.1391403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 10/01/2024] [Indexed: 11/07/2024] Open
Abstract
Gastrointestinal (GI) cancers are the most common malignancies, while surgical intervention remains the sole therapeutic approach offering the possibility of a definite cure for cancer. Perioperative anxiety negatively impacts the recovery of GI cancers. Recently, mounting studies have demonstrated that proper nursing interventions may alleviative perioperative anxious illnesses in patients with GI cancers. We conducted a first comprehensive review to summarize all the current evidence on this topic. After a systematically search in the six common databases, eighteen relevant studies were included for further analysis. The present review highlighted that there is a high prevalence of perioperative anxiety in patients with GI cancers (e.g., colorectal cancer, gastric/stomach cancer, hepatocellular carcinoma, gallbladder cancer, and esophageal cancer), while specific nursing interventions are the reliable methods to reduce postoperative anxiety. These nursing strategies include, but are not limited to, therapeutic listening intervention, implementing perioperative music, predictive nursing, progressive relaxation exercises, psychological interventions in the nursing care, comprehensive nursing, continuous nursing care, video-based nursing education, multidisciplinary cooperative continuous care, accelerated rehabilitation nursing, TCM nursing, evidence-based early warning nursing, target nursing care, and high-quality nursing. Since several limitations existed in the eligible studies as well as in this review, a well-designed multicenter RCT with large sample size is still warranted for the confirmation of nursing intervention for managing perioperative anxiety in patients with GI cancers. Also, future studies should focus on the long-term effects of relevant interventions, specific patient populations, multidisciplinary approaches, technological innovations, and educational programs.
Collapse
Affiliation(s)
- Ying Li
- Department of Anesthesia Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Juan Du
- Health Examination and Oncology Screening Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Li Du
- Educational Administration Department, Chongqing University Cancer Hospital, Chongqing, China
| | - Shan Li
- Department of Anesthesia Surgery, Mianyang Central Hospital, Mianyang, China
| | - Jianping Zhang
- Department of General Surgery, The Affiliated Hospital of Jiujiang University, Jiujiang, China
| |
Collapse
|
19
|
Melchor-Ruan J, Santiago-Ruiz L, Murillo-Ortiz BO, Rivera-Rivera S, Leal-Herrera YA, Suárez-García D, Remes-Troche JM, Grube P, Martínez-Mier G, Ruiz-García E, Ramos-Mayo A, Velarde-Ruiz-Velasco JA, Gamboa-Gutierrez R, Ordoñez-Escalante KG, Cisneros-Garza LE, Leal-Leyte P, Sepúlveda-Delgado J, González-Huezo MS, Arvizu-Castillo R, Urías-Rocha J, Flores-de-la-Torre CB, Carrillo-Mendoza LM, Gámez-del-Castillo JM, Lajous M, Monge A, Zamora-Valdés D. Characteristics of Hepatocellular Carcinoma by Sex in Mexico: A Multi-Institutional Collaboration. Diseases 2024; 12:262. [PMID: 39452505 PMCID: PMC11508022 DOI: 10.3390/diseases12100262] [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: 05/29/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 10/26/2024] Open
Abstract
Liver cancer is the fourth leading cause of cancer-related death worldwide. In Mexico, there is a high burden of liver cancer mortality in rural states, affecting both women and men equally. Thus, we aimed to describe the demographic and clinical characteristics of hepatocellular cancer (HCC) by sex in Mexico. Demographic and clinical information was extracted retrospectively from the medical records of patients with HCC initially treated (2015-2022) at institutions participating in a national survey across the country. The male-to-female ratio was calculated at the national and regional levels, and the results were stratified by sex. Among 697 HCC patients, the age at diagnosis was 65.4 ± 11.9 years and 20% were diagnosed at ≥75 years. The male-to-female ratio was 1.4:1, ranging from 1:1 in the northwestern and southwestern regions, to 2.1:1 in the western region. The proportion of cirrhosis was similar between the sexes; however, the etiology of cirrhosis differed: cryptogenic cirrhosis was higher in women and alcohol consumption was higher in men. Men had a higher proportion of advanced HCC, poor/undifferentiated tumors, and ≥4 nodules than women. HCC in the Mexican population affects both men and women at a 1.4:1 male-to-female ratio. This unique proportion by sex could be explained by the differences in the prevalence of risk factors across our heterogeneous country.
Collapse
Affiliation(s)
- Javier Melchor-Ruan
- Departamento de Gastroenterología, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico; (J.M.-R.); (E.R.-G.); (A.R.-M.)
| | - Luis Santiago-Ruiz
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (L.S.-R.); (M.L.)
| | - Blanca Olivia Murillo-Ortiz
- Unidad de Investigación en Epidemiologia Clínica, UMAE No. 1, OOAD, Instituto Mexicano del Seguro Social, Guanajuato 37328, Mexico;
| | - Samuel Rivera-Rivera
- División de Atención Oncológica en Adultos, Coordinación de Atención Oncológica, Instituto Mexicano del Seguro Social, Ciudad de México 03020, Mexico;
| | - Yelda A. Leal-Herrera
- Registro Poblacional de Cáncer Mérida, Unidad de Medicina de Alta Especialidad UMAE Mérida, Instituto Mexicano del Seguro Social, Mérida 97155, Mexico;
- Centro Institucional de Capacitación y Registro de Cáncer, Coordinación de Investigación en Salud, CMN SXXI del IMSS, Ciudad de México 06720, Mexico
| | - David Suárez-García
- Unidad de Medicina de Alta Especialidad UMAE No. 1, Instituto Mexicano del Seguro Social, León de Los Aldama 37328, Mexico;
| | | | - Peter Grube
- Universidad Veracruzana, Veracruz 94294, Mexico; (J.M.R.-T.); (P.G.); (G.M.-M.)
| | - Gustavo Martínez-Mier
- Universidad Veracruzana, Veracruz 94294, Mexico; (J.M.R.-T.); (P.G.); (G.M.-M.)
- Unidad de Medicina de Alta Especialidad UMAE Hospital de Especialidades 14, Instituto Mexicano del Seguro Social, Veracruz 91810, Mexico
| | - Erika Ruiz-García
- Departamento de Gastroenterología, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico; (J.M.-R.); (E.R.-G.); (A.R.-M.)
| | - Alan Ramos-Mayo
- Departamento de Gastroenterología, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico; (J.M.-R.); (E.R.-G.); (A.R.-M.)
| | | | - Ricardo Gamboa-Gutierrez
- División de Oncología, Unidad de Medicina de Alta Especialidad UMAE Mérida, Instituto Mexicano del Seguro Social, Mérida 97155, Mexico;
| | - Karla Gabriela Ordoñez-Escalante
- Laboratorio de Anatomía Patológica, Unidad de Medicina de Alta Especialidad UMAE Mérida, Instituto Mexicano del Seguro Social, Mérida 97155, Mexico;
| | | | | | | | | | - Ricardo Arvizu-Castillo
- Unidad de Quemados, Hospital General de Zona # 14, Instituto Mexicano del Seguro Social, Hermosillo 83120, Mexico; (R.A.-C.); (J.U.-R.)
| | - Jorge Urías-Rocha
- Unidad de Quemados, Hospital General de Zona # 14, Instituto Mexicano del Seguro Social, Hermosillo 83120, Mexico; (R.A.-C.); (J.U.-R.)
| | | | | | | | - Martín Lajous
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (L.S.-R.); (M.L.)
| | - Adriana Monge
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (L.S.-R.); (M.L.)
| | - Daniel Zamora-Valdés
- Hepatobiliary Sciences and Liver Transplantation, KASCH, KAMC, MNGHA, Ar Rimayah, Riyadh 14611, Saudi Arabia;
| |
Collapse
|
20
|
Li Y, Gu X, Yang L, Wang X, Wang Q, Xu X, Zhang A, Yue M, Wang M, Cong M, Ren J, Ren W, Shi G. Prediction of lymphovascular invasion in esophageal squamous cell carcinoma by computed tomography-based radiomics analysis: 2D or 3D ? Cancer Imaging 2024; 24:141. [PMID: 39420415 PMCID: PMC11488362 DOI: 10.1186/s40644-024-00786-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND To compare the performance between one-slice two-dimensional (2D) and whole-volume three-dimensional (3D) computed tomography (CT)-based radiomics models in the prediction of lymphovascular invasion (LVI) status in esophageal squamous cell carcinoma (ESCC). METHODS Two hundred twenty-four patients with ESCC (158 LVI-absent and 66 LVI-present) were enrolled in this retrospective study. The enrolled patients were randomly split into the training and testing sets with a 7:3 ratio. The 2D and 3D radiomics features were derived from the primary tumors' 2D and 3D regions of interest (ROIs) using 1.0 mm thickness contrast-enhanced CT (CECT) images. The 2D and 3D radiomics features were screened using inter-/intra-class correlation coefficient (ICC) analysis, Wilcoxon rank-sum test, Spearman correlation test, and the least absolute shrinkage and selection operator, and the radiomics models were built by multivariate logistic stepwise regression. The performance of 2D and 3D radiomics models was assessed by the area under the receiver operating characteristic (ROC) curve. The actual clinical utility of the 2D and 3D radiomics models was evaluated by decision curve analysis (DCA). RESULTS There were 753 radiomics features from 2D ROIs and 1130 radiomics features from 3D ROIs, and finally, 7 features were retained to construct 2D and 3D radiomics models, respectively. ROC analysis revealed that in both the training and testing sets, the 3D radiomics model exhibited higher AUC values than the 2D radiomics model (0.930 versus 0.852 and 0.897 versus 0.851, respectively). The 3D radiomics model showed higher accuracy than the 2D radiomics model in the training and testing sets (0.899 versus 0.728 and 0.788 versus 0.758, respectively). In addition, the 3D radiomics model has higher specificity and positive predictive value, while the 2D radiomics model has higher sensitivity and negative predictive value. The DCA indicated that the 3D radiomics model provided higher actual clinical utility regarding overall net benefit than the 2D radiomics model. CONCLUSIONS Both 2D and 3D radiomics features can be employed as potential biomarkers to predict the LVI in ESCC. The performance of the 3D radiomics model is better than that of the 2D radiomics model for the prediction of the LVI in ESCC.
Collapse
Affiliation(s)
- Yang Li
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| | - Xiaolong Gu
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| | - Li Yang
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| | - Xiangming Wang
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China.
| | - Qi Wang
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| | - Xiaosheng Xu
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| | - Andu Zhang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Meng Yue
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mingbo Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mengdi Cong
- Department of Radiology, The Hebei Children's Hospital, Shijiazhuang, Hebei Province, China
| | | | - Wei Ren
- GE Healthcare China, Beijing, China
| | - Gaofeng Shi
- Department of Computed Tomography and Magnetic Resonance Imaging, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050011, China
| |
Collapse
|
21
|
Liu S, Ju R, Zhang Z, Jiang Z, Cui J, Liu W, Han B, Wang S. Temperature-sensitive injectable chitosan-based hydrogel for endoscopic submucosal dissection. Int J Biol Macromol 2024; 282:136566. [PMID: 39414205 DOI: 10.1016/j.ijbiomac.2024.136566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Endoscopic submucosal dissection (ESD) is an effective treatment for polyps and early gastrointestinal cancers, but requires a high level of operator skill. Injecting submucosal materials (SIM) helps create a fluid cushion between the mucosal and muscular layers, making the procedure easier and reducing associated risks. However, SIMs commonly used in current clinical practice tend to spread quickly and fail to provide long-lasting submucosal fluid cushions (SFC). Thus, there is a critical need for a material that is easy to inject while also maintaining a durable barrier. We prepared succinylated hydroxybutyl chitosan (HBC-SA) by adding succinic anhydride (SA) to hydroxybutyl chitosan (HBC). The hydrogel had excellent temperature-sensitive properties and was able to be injected via an endoscopic injection needle even after gel formation. In vitro and in vivo studies showed that it has satisfactory biocompatibility. Functional experiments showed that the submucosal lifting properties of this hydrogel were significantly better than that of normal saline (NS) and sodium hyaluronate (SH), two commonly used clinical materials. In addition, the hydrogel possessed excellent hemostatic properties. Based on these results, HBC-SA is a promising candidate for submucosal injection during ESD.
Collapse
Affiliation(s)
- Shourui Liu
- College of Marine Life Sciences, Ocean University of China, 266003, PR China
| | - Ruibao Ju
- College of Marine Life Sciences, Ocean University of China, 266003, PR China
| | - Zhenguo Zhang
- College of Marine Life Sciences, Ocean University of China, 266003, PR China
| | - Zhen Jiang
- College of Marine Life Sciences, Ocean University of China, 266003, PR China
| | - Jingzhao Cui
- College of Marine Life Sciences, Ocean University of China, 266003, PR China
| | - Wanshun Liu
- College of Marine Life Sciences, Ocean University of China, 266003, PR China
| | - Baoqin Han
- College of Marine Life Sciences, Ocean University of China, 266003, PR China.
| | - Shuo Wang
- College of Marine Life Sciences, Ocean University of China, 266003, PR China.
| |
Collapse
|
22
|
Chen DS, Chen ZP, Zhu DZ, Guan LX, Zhu Q, Lou YC, He ZP, Chen HN, Sun HC. Burden landscape of hepatobiliary and pancreatic cancers in Chinese young adults: 30 years’ overview and forecasted trends. World J Gastrointest Oncol 2024; 16:4177-4193. [DOI: 10.4251/wjgo.v16.i10.4177] [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: 06/16/2024] [Revised: 08/18/2024] [Accepted: 08/28/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Hepatobiliary and pancreatic (HBP) cancers impose a considerable burden on young populations (aged 15 to 49 years), resulting in a substantial number of new cases and fatalities each year. In young populations, the HBP cancers shows extensive variance worldwide and the updated data in China is lacking.
AIM To investigate the current status, trends, projections, and underlying risk factors of HBP cancers among young populations in China.
METHODS The Global Burden of Disease Study 2019 provided data on the annual incidence, mortality, disability-adjusted life years (DALYs), age-standardized incidence rate (ASIR), mortality rate (ASMR), and DALYs rate (ASDR) of HBP cancers in young Chinese adults between 1990 and 2019. Temporal trends were assessed using estimated annual percentage change and hierarchical clustering. Sex-specific mortality and DALYs caused by various risks were analyzed across China and other regions, with future trends until 2035 projected using the Bayesian age-period-cohort model.
RESULTS From 1990 to 2019, incident cases, deaths, DALYs, ASIR, ASMR, and ASDR for liver cancer (LC) in young Chinese individuals decreased, classified into 'significant decrease' group. Conversely, cases of gallbladder and biliary tract cancer and pancreatic cancer rose, categorized as either 'significant increase' or 'minor increase' groups. The contribution of risk factors to mortality and DALYs for HBP tumors increased to varying degrees. Healthy lifestyle behaviors, such as tobacco control, weight management, alcohol moderation, and drug avoidance, could lower HBP cancers incidence. Moreover, except for LC in females, which is likely to initially decline slightly and then rise, the forecasting model predicted that the ASIR and ASMR for all HPB cancers subtypes by gender will increase among young adults.
CONCLUSION HBP cancers burden among young adults in China is expected to increase until 2035, necessitating lifestyle interventions and targeted treatment strategies to mitigate the public health impact of these cancers.
Collapse
Affiliation(s)
- De-Sheng Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ze-Ping Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Dong-Zi Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lv-Xin Guan
- Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong Province, China
| | - Qi Zhu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yi-Chao Lou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ze-Ping He
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hao-Nan Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Hong-Cheng Sun
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
23
|
Shigematsu Y, Kanda H, Takahashi Y, Takeuchi K, Inamura K. Relationships between tumor CD147 expression, tumor-infiltrating lymphocytes, and oncostatin M in hepatocellular carcinoma. Virchows Arch 2024:10.1007/s00428-024-03939-w. [PMID: 39395054 DOI: 10.1007/s00428-024-03939-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/02/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
In hepatocellular carcinoma (HCC), CD147 expression contributes to tumor malignancy; however, its relationship with the tumor-immune microenvironment (TIME) remains unclear. This study aimed to elucidate the clinicopathological characteristics associated with CD147 expression in HCC and investigate its association with the TIME, specifically its association with tumor-infiltrating lymphocytes (TILs) and oncostatin M (OSM). Using 397 HCC specimens from patients undergoing curative-intent resection, we assessed CD147 expression in tumor cells and quantified OSM-positive cells and various TILs (CD8+, CD4+, FOXP3+, and CD20+ cells) in the TIME. Using tissue microarrays, these assessments were performed through immunohistochemical analysis. We investigated the associations between CD147 expression status, the density of OSM-positive cells, and the densities of various TILs. High CD147 expression, found in 332 specimens (83.6%), was associated with advanced clinical stage (P = 0.029), fibrosis (P = 0.036), and higher densities of FOXP3+ cells (P = 0.0039), CD4+ cells (P = 0.0012), and OSM-positive cells (P = 0.0017). In CD147-high tumors, OSM-positive cell density was associated with all assessed TIL subsets (CD8+, CD4+, FOXP3+, and CD20+ cells; all Ps < 0.001), whereas in CD147-low tumors, OSM-positive cell density was associated only with FOXP3+ cells (P = 0.0004). In HCC, CD147 expression is associated with an immunosuppressive TIME, characterized by increased FOXP3+ regulatory T cells and a correlation with OSM-positive cells. These results elucidate the potential mechanisms through which CD147 facilitates tumor-immune evasion, suggesting the CD147 - OSM axis as a promising target for therapeutic intervention in HCC.
Collapse
Affiliation(s)
- Yasuyuki Shigematsu
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research (JFCR), 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
- Division of Pathology, Cancer Institute, JFCR, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Hiroaki Kanda
- Department of Pathology, Saitama Cancer Center, 780 Komuro, Ina, Kita-adachi-gun, Saitama, 362-0806, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, JFCR, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Kengo Takeuchi
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research (JFCR), 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Division of Pathology, Cancer Institute, JFCR, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
- Pathology Project for Molecular Targets, Cancer Institute, JFCR, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Kentaro Inamura
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research (JFCR), 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
- Division of Pathology, Cancer Institute, JFCR, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
- Division of Tumor Pathology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
24
|
Wan M, Gan A, Dai J, Lin F, Wang R, Wu B, Yan T, Jia Y. Rhein induces apoptosis of AGS and MGC803 cells by regulating the Ras/PI3K/AKT and p38/MAPK signaling pathway. J Pharm Pharmacol 2024:rgae115. [PMID: 39393789 DOI: 10.1093/jpp/rgae115] [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: 03/08/2024] [Accepted: 08/18/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES Rhein is one of the main bioactive compounds in the Polygonaceae plant, and has been proven to have anti-cancer activity in some reports. But the mechanism of Rhein in the treatment of gastric cancer (GC) is limited reported. In this research, network pharmacology combined with in vitro experiments was used for systematically studying the mechanism of Rhein. METHODS Network pharmacology confirmed the major effect signaling pathway and key targets of Rhein in the treatment of GC. Cell viability assay, colony formation assay, fluorescence probe assay, apoptosis assay, western blot and qRT-PCR verified the mechanism of Rhein in the treatment of GC cells (AGS and MGC803 cells). KEY FINDINGS The results showed that Rhein significantly induced the apoptosis process of AGS and MGC803 cells by regulating the Ras/phosphoinositide-3 kinase (PI3K)/protein kinase B (AKT) and the p38/mitogen-activated protein kinase signaling pathways. The AKT activator (SC79) and p38 inhibitor (SB202190) inhibited Rhein-induced apoptosis. CONCLUSIONS All results proved that Rhein could be recognized as a potential natural drug for the treatment of GC.
Collapse
Affiliation(s)
- Meiqi Wan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Anna Gan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Jun Dai
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Fei Lin
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Ruixuan Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Bo Wu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| |
Collapse
|
25
|
Santana PT, de Lima IS, da Silva e Souza KC, Barbosa PHS, de Souza HSP. Persistent Activation of the P2X7 Receptor Underlies Chronic Inflammation and Carcinogenic Changes in the Intestine. Int J Mol Sci 2024; 25:10874. [PMID: 39456655 PMCID: PMC11507540 DOI: 10.3390/ijms252010874] [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: 09/18/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Aberrant signaling through damage-associated molecular patterns (DAMPs) has been linked to several health disorders, attracting considerable research interest over the last decade. Adenosine triphosphate (ATP), a key extracellular DAMP, activates the purinergic receptor P2X7, which acts as a danger sensor in immune cells and is implicated in distinct biological functions, including cell death, production of pro-inflammatory cytokines, and defense against microorganisms. In addition to driving inflammation mediated by immune and non-immune cells, the persistent release of endogenous DAMPs, including ATP, has been shown to result in epigenetic modifications. In intestinal diseases such as inflammatory bowel disease (IBD) and colorectal cancer (CRC), consequent amplification of the inflammatory response and the resulting epigenetic reprogramming may impact the development of pathological changes associated with specific disease phenotypes. P2X7 is overexpressed in the gut mucosa of patients with IBD, whereas the P2X7 blockade prevents the development of chemically induced experimental colitis. Recent data suggest a role for P2X7 in determining gut microbiota composition. Regulatory mechanisms downstream of the P2X7 receptor, combined with signals from dysbiotic microbiota, trigger intracellular signaling pathways and inflammasomes, intensify inflammation, and foster colitis-associated CRC development. Preliminary studies targeting the ATP-P2X7 pathway have shown favorable therapeutic effects in human IBD and experimental colitis.
Collapse
Affiliation(s)
- Patricia Teixeira Santana
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
- D’Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro 30, Botafogo, Rio de Janeiro 22281-100, Brazil
| | - Isadora Schmukler de Lima
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
| | - Karen Cristina da Silva e Souza
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
| | - Pedro Henrique Sales Barbosa
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
| | - Heitor Siffert Pereira de Souza
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
- D’Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro 30, Botafogo, Rio de Janeiro 22281-100, Brazil
| |
Collapse
|
26
|
Wang Y, Zhu J, Li H, Wang L, Zhu C, Li X, Wang S, Du L. Effectiveness of Risk-Adapted Upper Gastrointestinal Cancer Screening in China: Prospective Cohort Study. JMIR Public Health Surveill 2024; 10:e62864. [PMID: 39388714 PMCID: PMC11486449 DOI: 10.2196/62864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 10/12/2024] Open
Abstract
Background Previous studies have proved the effectiveness of endoscopic screening in rural areas; however, long-term, high-quality evidence regarding the effectiveness of risk-adapted upper gastrointestinal cancer (UGC) sequential screening strategies in resource-rich regions is currently lacking. Objective The objectives were to validate the effectiveness of risk-adapted sequential screening strategies in UGC prevention and control and assess the potential of sequential screening to lower mortality rates. Methods Based on the Cancer Screening Program in Urban China, a prospective, large-scale cohort study based on population was conducted to recruit individuals from 4 cities in China from 2013-2019. Those identified as having a high risk of UGC according to a validated risk-score model were advised to undergo endoscopy tests. Follow-up outcomes were tracked until June 2021. Incidence of UGC, UGC-related mortality, and all-cause mortality were evaluated between the screened and nonscreened cohorts. Results The study included 153,079 participants at baseline. In total, 113,916 (74.42%) of the participants were designated as low risk of UGC. The remaining 39,163 (25.68%) participants were deemed to be at high risk of UGC and were offered gastroscopy tests. Among the high-risk participants, 9627 (compliance rate 24.6%) adhered to the gastroscopy tests. Over a median follow-up of 6.05 (IQR 3.06-7.06) years, 622 UGC cases, 180 UGC deaths, and 1958 all-cause death cases were traced. The screened cohort exhibited the highest cumulative incidence of UGC (119.2 per 100,000 person-years), followed by the nonscreened and low-risk cohorts. Obvious reductions in both all-cause mortality and UGC mortality were observed between those who undertook screening (153.7 and 4.7 per 100,000 person-years, respectively) and the nonscreened group (245.3 and 27 per 100,000 person-years, respectively). The screening population showed a significant 36% and 82% reduction in both all-cause mortality (hazard ratio [HR] 0.64, 95% CI 0.49-0.83, P<.001) and UGC mortality (HR 0.18, 95% CI 0.04-0.74, P=.02), respectively, compared to the nonscreened group. Reductions of 35% in all-cause mortality (HR 0.65, 95% CI 0.49-0.86, P=.003) and 81% in UGC mortality (HR 0.19, 95% CI 0.05-0.80, P=.02) were observed in participants aged older than 55 years in the screened group compared to the nonscreened group. The reductions in all-cause mortality and UGC mortality were statistically significant in males (all-cause mortality: HR 0.64, 95% CI 0.47-0.88, P=.005; UGC mortality: HR 0.10, 95% CI 0.01-0.72, P=.02), but significant reductions were not observed in females (all P values were >.05). Conclusions Our study suggests the significance of one-off risk-adapted UGC screening in reducing both all-cause mortality and UGC mortality, particularly among high-risk individuals, indicating its effectiveness in UGC prevention and management.
Collapse
Affiliation(s)
- Youqing Wang
- Department of Cancer Prevention, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Juan Zhu
- Department of Cancer Prevention, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Huizhang Li
- Department of Cancer Prevention, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Le Wang
- Department of Cancer Prevention, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Chen Zhu
- Department of Cancer Prevention, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xue Li
- Department of Cancer Prevention, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shi Wang
- Department of Endoscopy, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lingbin Du
- Department of Cancer Prevention, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
27
|
Wang L, Zhu Y, Zhang N, Xian Y, Tang Y, Ye J, Reza F, He G, Wen X, Jiang X. The multiple roles of interferon regulatory factor family in health and disease. Signal Transduct Target Ther 2024; 9:282. [PMID: 39384770 PMCID: PMC11486635 DOI: 10.1038/s41392-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024] Open
Abstract
Interferon Regulatory Factors (IRFs), a family of transcription factors, profoundly influence the immune system, impacting both physiological and pathological processes. This review explores the diverse functions of nine mammalian IRF members, each featuring conserved domains essential for interactions with other transcription factors and cofactors. These interactions allow IRFs to modulate a broad spectrum of physiological processes, encompassing host defense, immune response, and cell development. Conversely, their pivotal role in immune regulation implicates them in the pathophysiology of various diseases, such as infectious diseases, autoimmune disorders, metabolic diseases, and cancers. In this context, IRFs display a dichotomous nature, functioning as both tumor suppressors and promoters, contingent upon the specific disease milieu. Post-translational modifications of IRFs, including phosphorylation and ubiquitination, play a crucial role in modulating their function, stability, and activation. As prospective biomarkers and therapeutic targets, IRFs present promising opportunities for disease intervention. Further research is needed to elucidate the precise mechanisms governing IRF regulation, potentially pioneering innovative therapeutic strategies, particularly in cancer treatment, where the equilibrium of IRF activities is of paramount importance.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanghui Zhu
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yali Xian
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Tang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Ye
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fekrazad Reza
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Wen
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
Liu J, Hong A, Zeng J, Liang X. The efficacy of ciprofol versus propofol on anesthesia in patients undergoing endoscopy: a systematic review and meta-analysis of randomized controlled trials. BMC Anesthesiol 2024; 24:359. [PMID: 39379828 PMCID: PMC11460030 DOI: 10.1186/s12871-024-02721-4] [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: 10/15/2023] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION Ciprofol is a new intravenous anesthetic with a similar chemical structure to propofol. We aimed to compare the incidence of adverse actions like injection pain and time indexes of ciprofol versus propofol on anesthesia in patients undergoing endoscopy. We also compared anesthetists' satisfaction during the procedure. METHODS Two independent researchers (Liu and Zeng) searched the Cochrane Library, Embase databases, and PubMed for controlled clinical trials. This meta-analysis of randomized controlled trials (RCTs) was performed with the Review Manager, Stata and the Cochrane Risk-of-Bias 2 tool to evaluate methodological quality. Relative risks with 95% confidence interval (CI) were calculated for outcomes. RESULTS Ten trials, including 1545 patients, were examined in the current meta-analysis. During anesthesia in patients undergoing endoscopy, the incidence of injection pain was significantly reduced in the research group. Compared with propofol, the pooled risk difference (RD) with the use of ciprofol for injection pain for all the procedures was - 0.34 (95% confidence interval [CI], -0.48 to 0.19), and RR for hypotension was 0.73(95% CI:0.58 to 0.92). GRADE showed this meta-analysis has moderate or low confidence. Trial sequential analysis for mortality indicated insufficient sample size for a definitive judgment for lower incidence of hypotension. CONCLUSION In painless endoscopy, compared with propofol, ciprofol exhibited non-inferiority anesthesia/sedation in patients, and had a good safety profile with a lower incidence of pain on injection and may reduce the chance of hypotension. Trial sequential analysis suggested the need for more cases, and GRADE highlighted moderate certainty, emphasizing the necessity for further targeted RCTs. SYSTEMATIC REVIEW REGISTRATION PROSPERO, CRD42023433627.
Collapse
Affiliation(s)
- Jikai Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Aonan Hong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinfang Zeng
- Department of Anesthesiology, Jiangnan University Medical Center (Wuxi No. 2 People's Hospital, Wuxi Clinical College of Nantong University), No.68, Zhongshan Road, Wuxi, 214002, China.
| | - Xiao Liang
- Department of Anesthesiology, Jiangnan University Medical Center (Wuxi No. 2 People's Hospital, Wuxi Clinical College of Nantong University), No.68, Zhongshan Road, Wuxi, 214002, China.
| |
Collapse
|
29
|
Kazi IA, Jahagirdar V, Kabir BW, Syed AK, Kabir AW, Perisetti A. Role of Imaging in Screening for Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:3400. [PMID: 39410020 PMCID: PMC11476228 DOI: 10.3390/cancers16193400] [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: 08/31/2024] [Revised: 09/22/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Primary liver cancer is among the most common cancers globally. It is the sixth-most common malignancy encountered and the third-most common cause of cancer-related death. Hepatocellular carcinoma (HCC) is the most common primary liver malignancy, accounting for about 90% of primary liver cancers. The majority of HCCs occur in patients with underlying cirrhosis, which results from chronic liver diseases such as fatty liver, hepatitis B and hepatitis C infections, and chronic alcohol use, which are the leading causes. The obesity pandemic has led to an increased prevalence of nonalcoholic fatty liver disease (NAFLD), which leads to nonalcoholic steatohepatitis and could progress to cirrhosis. As HCC is among the most common cancers and occurs in the setting of chronic liver disease in most patients, screening the population at risk could help in early diagnosis and management, leading to improved survival. Screening for HCC is performed using biochemical marker testing such as α-fetoprotein (AFP) and cross-sectional imaging. It is critical to emphasize that HCC could potentially occur in patients without cirrhosis (non-cirrhotic HCC), which can account for almost 20% of all HCCs. The lack of cirrhosis can cause a delay in surveillance, which could potentially lead to diagnosis at a later stage, worsening the prognosis for such patients. In this article, we discuss the diagnosis of cirrhosis in at-risk populations with details on the different modalities available for screening HCC in patients with cirrhosis, emphasizing the role of abdominal ultrasounds, the primary imaging modality in HCC screening.
Collapse
Affiliation(s)
- Irfan A. Kazi
- Department of Radiology, University of Missouri Columbia, Columbia, MO 65212, USA;
| | - Vinay Jahagirdar
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Bareen W. Kabir
- Department of Internal Medicine, University of Missouri Columbia, Columbia, MO 65212, USA;
| | - Almaan K. Syed
- Blue Valley Southwest High School, Overland Park, KS 6622, USA;
| | | | - Abhilash Perisetti
- Division of Gastroenterology and Hepatology, Kansas City Veteran Affairs, Kansas City, MO 64128, USA
| |
Collapse
|
30
|
Kolahi Sadeghi L, Vahidian F, Eterafi M, Safarzadeh E. Gastrointestinal cancer resistance to treatment: the role of microbiota. Infect Agent Cancer 2024; 19:50. [PMID: 39369252 PMCID: PMC11453072 DOI: 10.1186/s13027-024-00605-3] [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: 06/15/2024] [Accepted: 08/20/2024] [Indexed: 10/07/2024] Open
Abstract
The most common illnesses that adversely influence human health globally are gastrointestinal malignancies. The prevalence of gastrointestinal cancers (GICs) is relatively high, and the majority of patients receive ineffective care since they are discovered at an advanced stage of the disease. A major component of the human body is thought to be the microbiota of the gastrointestinal tract and the genes that make up the microbiome. The gut microbiota includes more than 3000 diverse species and billions of microbes. Each of them has benefits and drawbacks and been demonstrated to alter anticancer medication efficacy. Treatment of GIC with the help of the gut bacteria is effective while changes in the gut microbiome which is linked to resistance immunotherapy or chemotherapy. Despite significant studies and findings in this field, more research on the interactions between microbiota and response to treatment in GIC are needed to help researchers provide more effective therapeutic strategies with fewer treatment complication. In this review, we examine the effect of the human microbiota on anti-cancer management, including chemotherapy, immunotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Leila Kolahi Sadeghi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Fatemeh Vahidian
- Faculty of Medicine, Laval University, Quebec, Canada
- Centre de Recherche de I'Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Quebec, Canada
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students' Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology, and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
31
|
Ren X, Hu R, Zhang H. A Mendelian analysis of the causality between inflammatory cytokines and digestive tract cancers. Postgrad Med J 2024:qgae132. [PMID: 39362654 DOI: 10.1093/postmj/qgae132] [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: 07/04/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE In this study, we performed a two-sample Mendelian randomization (MR) analysis to assess the causality between inflammatory cytokines and the risk of digestive tract cancers (DTCs). Furthermore, we conducted a molecular docking study to predict the therapeutic mechanisms of traditional Chinese medicine (TCM) compounds in the treatment of DTCs. METHODS In our MR analysis, genetic variations associated with eight types of DTCs were utilized, which were sourced from a large publicly available genome-wide association study dataset (7929 cases and 1 742 407 controls of European ancestry) and inflammatory cytokines data from a genome-wide association study summary of 8293 European participants. Inverse-variance weighted method, MR-Egger, and weighted median were performed to analyze and strengthen the final results. We investigated the effects of 41 inflammatory molecules on 8 types of DTCs. Subsequently, the effect of DTCs on positive inflammatory factors was analyzed by means of inverse MR. Molecular docking was exploited to predict therapeutic targets with TCM compounds. RESULTS Interleukin-7, interleukin-16, macrophage colony-stimulating factor, monokine induced by interferon-gamma, and vascular endothelial growth factor may be significantly associated with various types of DTCs. Five TCM compounds (baicalin, berberine, curcumin, emodin, and salidroside) demonstrated better binding energies to both interleukin-7 and vascular endothelial growth factor than carboplatin. CONCLUSION This study provides strong evidence to support the potential causality of some inflammatory cytokines on DTCs and indicates the potential molecular mechanism of TCM compounds in the treatment of DTCs. Key message What is already known on this topic The increasing evidence indicates that inflammatory cytokines are implicated in the pathogenesis of digestive tract cancers (DTCs). Nevertheless, the causal relationship between inflammatory cytokines and DTCs remains indistinct. Additionally, certain traditional Chinese medicine compounds have been demonstrated to treat DTCs by influencing inflammatory factors, yet their underlying potential mechanisms remain ambiguous. What this study adds In this study, Mendelian randomization analysis was performed for the first time regarding the causality between human inflammatory cytokines and eight types of DTCs, which revealed that inflammatory factors may play different roles in different types of DTCs. Moreover, molecular docking of key inflammatory factors was implemented, indicating the targets for drug actions. How this study might affect research, practice, or policy This research has the potential to reveal the causality between 41 inflammatory factors and 8 DTCs, offering novel perspectives for the prevention and management strategies of DTCs. Additionally, it indicates the targets for the actions of traditional Chinese medicine on the key inflammatory factors of these cancers.
Collapse
Affiliation(s)
- Xing Ren
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rong Hu
- Institute of Science, Technology and Humanities, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
32
|
Chiu V, Yee C, Main N, Stevanovski I, Watt M, Wilson T, Angus P, Roberts T, Shackel N, Herath C. Oncogenic plasmid DNA and liver injury agent dictates liver cancer development in a mouse model. Clin Sci (Lond) 2024; 138:1227-1248. [PMID: 39254423 PMCID: PMC11427747 DOI: 10.1042/cs20240560] [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: 04/07/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/11/2024]
Abstract
Primary liver cancer is an increasing problem worldwide and is associated with significant mortality. A popular method of modeling liver cancer in mice is plasmid hydrodynamic tail vein injection (HTVI). However, plasmid-HTVI models rarely recapitulate the chronic liver injury which precedes the development of most human liver cancer. We sought to investigate how liver injury using thioacetamide contributes to the pathogenesis and progression of liver cancer in two oncogenic plasmid-HTVI-induced mouse liver cancer models. Fourteen-week-old male mice received double-oncogene plasmid-HTVI (SB/AKT/c-Met and SB/AKT/NRas) and then twice-weekly intraperitoneal injections of thioacetamide for 6 weeks. Liver tissue was examined for histopathological changes, including fibrosis and steatosis. Further characterization of fibrosis and inflammation was performed with immunostaining and real-time quantitative PCR. RNA sequencing with pathway analysis was used to explore novel pathways altered in the cancer models. Hepatocellular and cholangiocellular tumors were observed in mice injected with double-oncogene plasmid-HTVI models (SB/AKT/c-Met and SB/AKT/NRas). Thioacetamide induced mild fibrosis and increased alpha smooth muscle actin-expressing cells. However, the combination of plasmids and thioacetamide did not significantly increase tumor size, but increased multiplicity of small neoplastic lesions. Cancer and/or liver injury up-regulated profibrotic and proinflammatory genes while metabolic pathway genes were mostly down-regulated. We conclude that the liver injury microenvironment can interact with liver cancer and alter its presentation. However, the effects on cancer development vary depending on the genetic drivers with differing active oncogenic pathways. Therefore, the choice of plasmid-HTVI model and injury agent may influence the extent to which injury promotes liver cancer development.
Collapse
Affiliation(s)
- Vincent Chiu
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Christine Yee
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Nathan Main
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Igor Stevanovski
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Matthew Watt
- School of Biomedical Sciences, University of Melbourne, Victoria, Australia
| | - Trevor Wilson
- Hudson Institute of Medical Research, Monash University, Victoria, Australia
| | - Peter Angus
- Department of Gastroenterology and Hepatology, Austin Health, Heidelberg, Victoria, Australia
| | - Tara Roberts
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Nicholas Shackel
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
| | - Chandana Herath
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- South Western Sydney Clinical School, UNSW Sydney, Liverpool, New South Wales, Australia
- Department of Medicine, Austin Health, University of Melbourne, Victoria, Australia
| |
Collapse
|
33
|
Farinati F, Pelizzaro F. Gastric cancer screening in Western countries: A call to action. Dig Liver Dis 2024; 56:1653-1662. [PMID: 38403513 DOI: 10.1016/j.dld.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Gastric cancer is a major cause of cancer-related death worldwide, despite the reduction in its incidence. The disease is still burdened with a poor prognosis, particularly in Western countries. The main risk factor is the infection by Helicobacter pylori, classified as a class I carcinogen by the IARC, and It is well-known that primary prevention of gastric cancer can be achieved with the eradication of the infection. Moreover, non-invasive measurement of pepsinogens (PGI and PGI/PGII ratio) allows the identification of patients that should undergo upper gastrointestinal (GI) endoscopy. Gastric non-cardia adenocarcinoma is indeed preceded by a well-defined precancerous process that involves consecutive stages, described for the first time by Correa et al. more than 40 years ago, and patients with advance stages of gastric atrophy/intestinal metaplasia and with dysplastic changes should be followed-up periodically with upper GI endoscopies. Despite these effective screening and surveillance methods, national-level screening campaigns have been adopted only in few countries in eastern Asia (Japan and South Korea). In this review, we describe primary and secondary preventive measures for gastric cancer, discussing the need to introduce screening also in Western countries. Moreover, we propose a simple algorithm for screening that could be easily applied in clinical practice.
Collapse
Affiliation(s)
- Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, Padova 35128, Italy; Gastroenterology Unit, Azienda Ospedale-Università di Padova, Via Giustiniani 2, Padova 35128, Italy.
| | - Filippo Pelizzaro
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, Padova 35128, Italy; Gastroenterology Unit, Azienda Ospedale-Università di Padova, Via Giustiniani 2, Padova 35128, Italy
| |
Collapse
|
34
|
Voutsadakis IA. CDX2-Suppressed Colorectal Cancers Possess Potentially Targetable Alterations in Receptor Tyrosine Kinases and Other Colorectal-Cancer-Associated Pathways. Diseases 2024; 12:234. [PMID: 39452477 PMCID: PMC11506651 DOI: 10.3390/diseases12100234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Colorectal cancer, a prevalent gastrointestinal carcinoma, has a high risk for recurrence when locally advanced and remains lethal when in an advanced stage. Prognostic biomarkers may help in better delineating the aggressiveness of this disease in individual patients and help to tailor appropriate therapies. CDX2, a transcription factor of gastrointestinal differentiation, has been proposed as a biomarker for good outcomes and could also be a marker of specific sub-types amenable to targeted therapies. METHODS Colorectal cancers from The Cancer Genome Atlas (TCGA) colorectal cohort and colon cancers from the Sidra-LUMC AC-ICAM cohort were categorized according to their expressions of CDX2 mRNA. Groups with CDX2 suppression were compared with cancers showing no suppression regarding their clinical and genomic characteristics. RESULTS CDX2-suppressed colorectal cancers showed a high prevalence of Microsatellite Instability (MSI) and a lower prevalence of chromosomal Instability (CIN) compared to non-CDX2-suppressed cancers. In addition, CDX2-suppressed cancers had a higher prevalence of mutations in several receptor tyrosine kinase genes, including EGFR, ERBB3, ERBB4, RET, and ROS1. In contrast, CDX2-suppressed cancers displayed lower mutation frequencies than non-CDX2-suppressed cancers in the genes encoding for the two most frequently mutated tumor suppressors, APC and TP53, and the most frequently mutated colorectal cancer oncogene, KRAS. However, CDX2-suppressed colorectal cancers had a higher prevalence of mutations in alternative genes of the WNT/APC/β-catenin and KRAS/BRAF/MEK pathways. In addition, they showed frequent mutations in DNA damage response (DDR) genes, such as BRCA2 and ATM. CONCLUSION CDX2-suppressed colorectal cancers constitute a genomically distinct subset of colon and rectal cancers that have a lower prevalence of KRAS, APC, and TP53 mutations, but a high prevalence of mutations in less commonly mutated colorectal cancer genes. These alterations could serve as targets for personalized therapeutics in this subset.
Collapse
Affiliation(s)
- Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON P6B 0A8, Canada; or
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
35
|
Przystupski D, Baczyńska D, Rossowska J, Kulbacka J, Ussowicz M. Calcium ion delivery by microbubble-assisted sonoporation stimulates cell death in human gastrointestinal cancer cells. Biomed Pharmacother 2024; 179:117339. [PMID: 39216448 DOI: 10.1016/j.biopha.2024.117339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Ultrasound-mediated cell membrane permeabilization - sonoporation, enhances drug delivery directly to tumor sites while reducing systemic side effects. The potential of ultrasound to augment intracellular calcium uptake - a critical regulator of cell death and proliferation - offers innovative alternative to conventional chemotherapy. However, calcium therapeutic applications remain underexplored in sonoporation studies. This research provides a comprehensive analysis of calcium sonoporation (CaSP), which combines ultrasound treatment with calcium ions and SonoVue microbubbles, on gastrointestinal cancer cells LoVo and HPAF-II. Initially, optimal sonoporation parameters were determined: an acoustic wave of 1 MHz frequency with a 50 % duty cycle at intensity of 2 W/cm2. Subsequently, various cellular bioeffects, such as viability, oxidative stress, metabolism, mitochondrial function, proliferation, and cell death, were assessed following CaSP treatment. CaSP significantly impaired cancer cell function by inducing oxidative and metabolic stress, evidenced by increased mitochondrial depolarization, decreased ATP levels, and elevated glucose uptake in a Ca2+ dose-dependent manner, leading to activation of the intrinsic apoptotic pathway. Cellular response to CaSP depended on the TP53 gene's mutational status: colon cancer cells were more susceptible to CaSP-induced apoptosis and G1 phase cell cycle arrest, whereas pancreatic cancer cells showed a higher necrotic response and G2 cell cycle arrest. These promising results encourage future research to optimize sonoporation parameters for clinical use, investigate synergistic effects with existing treatments, and assess long-term safety and efficacy in vivo. Our study highlights CaSP's clinical potential for improved safety and efficacy in cancer therapy, offering significant implications for the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- Dawid Przystupski
- Department of Paediatric Bone Marrow Transplantation, Oncology and Haematology, Wroclaw Medical University, Borowska 213, Wroclaw 50-556, Poland.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, Wroclaw 50-556, Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, Wroclaw 53-114, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, Wroclaw 50-556, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių 5, Vilnius 08410, Lithuania
| | - Marek Ussowicz
- Department of Paediatric Bone Marrow Transplantation, Oncology and Haematology, Wroclaw Medical University, Borowska 213, Wroclaw 50-556, Poland
| |
Collapse
|
36
|
Loukopoulou C, Nikolouzakis T, Koliarakis I, Vakonaki E, Tsiaoussis J. Telomere Length and Telomerase Activity as Potential Biomarkers for Gastrointestinal Cancer. Cancers (Basel) 2024; 16:3370. [PMID: 39409990 PMCID: PMC11482595 DOI: 10.3390/cancers16193370] [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: 08/13/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Gastrointestinal (GI) cancers, such as colorectal and gastric cancers, pose significant global health challenges due to their high rates of incidence and mortality. Even with advancements in treatment and early detection, many patients still face poor outcomes, highlighting the critical need for new biomarkers and therapeutic targets. Telomere length (TL) and telomerase activity (TA) have gained attention in this context. Telomeres, protective nucleotide sequences at chromosome ends, shorten with each cell division, leading to cellular aging. Telomerase, a ribonucleoprotein enzyme, counteracts this shortening by adding telomeric repeats, a process tightly regulated in normal cells but often dysregulated in cancer. This review critically evaluates the role of TL and TA in the pathogenesis of GI cancers, examining their potential as diagnostic, prognostic, and predictive biomarkers. It explores how alterations in telomere biology contribute to the initiation and progression of GI tumors and assesses the therapeutic implications of targeting telomerase. By integrating findings from diverse studies, this review aims to elucidate the intricate relationship between telomere dynamics and gastrointestinal carcinogenesis, offering insights into how TL and TA could be leveraged to enhance the early detection, treatment, and prognosis of GI cancers.
Collapse
Affiliation(s)
- Christina Loukopoulou
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| | - Taxiarchis Nikolouzakis
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| | - Ioannis Koliarakis
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| | - Elena Vakonaki
- Department of Forensic Sciences and Toxicology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - John Tsiaoussis
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| |
Collapse
|
37
|
Onwuka JU, Wuraola FO, Owoade IA, Ogunyemi YF, Di Bernardo M, Dare AJ, Mohammed TO, Sheikh M, Olasehinde O, Kingham TP, Robbins HA, Alatise OI. Delays in Presentation, Diagnosis, and Treatment Among Patients With GI Cancer in Southwest Nigeria. JCO Glob Oncol 2024; 10:e2400060. [PMID: 39418630 DOI: 10.1200/go.24.00060] [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: 02/07/2024] [Revised: 06/15/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
PURPOSE The incidence of GI cancers is increasing in sub-Saharan African countries. We described the oncological care pathway and assessed presentation, diagnosis, and treatment intervals and delays among patients with GI cancer who presented to the Obafemi Awolowo University Teaching Hospitals Complex in Ile-Ife, Nigeria. METHODS We analyzed data from 545 patients with GI cancer in the African Research Group for Oncology (ARGO) database. We defined presentation interval as the interval between symptom onset and presentation to tertiary hospital, diagnostic interval as between presentation and diagnosis, and treatment interval as between diagnosis and initiation of treatment. We considered >3 months, >1 month, and >1 month to be presentation, diagnosis, and treatment delays, respectively. We compared lengths of intervals using Mann-Whitney U tests and logistic regression. RESULTS The most frequent cancer types were pancreatic (32%) and colorectal (28%). Most patients presented at stages III (38%) and IV (30%). The median presentation interval was 84 days (IQR, 56-191), and 49% presented after 3 months or longer. The median diagnosis and treatment intervals were 0 (IQR, 0-8) and 7 (IQR, 0-23) days, respectively. There was no relationship between age, sex, education, or distance to tertiary hospital and presentation delay, but patients with stage III to IV versus I to II had higher odds of presentation delay (odds ratio [OR], 1.68 [95% CI, 1.13 to 2.50]). Among patients with pancreatic cancer, older patients were less likely to have a diagnosis delay (OR, 0.50 [95% CI, 0.25 to 0.98]). CONCLUSION About half of patients with GI cancer in Ile-Ife, Nigeria, did not present to tertiary hospitals until more than 90 days after noticing symptoms. Efforts are warranted to improve public knowledge of GI cancer symptoms and to strengthen health systems for prompt diagnosis and referral to specialty care.
Collapse
Affiliation(s)
| | - Funmilola Olanike Wuraola
- Department of Surgery, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
- African Research Group for Oncology, Ile-Ife, Nigeria
| | | | | | | | - Anna J Dare
- Department of Surgery, Li Ka Shing Knowledge Institute, University of Toronto, Toronto, Canada
| | | | - Mahdi Sheikh
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Olalekan Olasehinde
- Department of Surgery, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - T Peter Kingham
- African Research Group for Oncology, Ile-Ife, Nigeria
- Department of Surgery, and Global Cancer Disparities Initiative, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hilary A Robbins
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Olusegun Isaac Alatise
- Department of Surgery, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
- African Research Group for Oncology, Ile-Ife, Nigeria
| |
Collapse
|
38
|
Lee YN, Sung MK, Hwang DW, Park Y, Kwak BJ, Lee W, Song KB, Lee JH, Yoo C, Kim KP, Chang HM, Ryoo BY, Kim SC. Clinical Outcomes of Surgery after Neoadjuvant Chemotherapy in Locally Advanced Pancreatic Ductal Adenocarcinoma. Cancer Res Treat 2024; 56:1240-1251. [PMID: 38901824 PMCID: PMC11491246 DOI: 10.4143/crt.2023.977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
PURPOSE Clinical outcomes of surgery after neoadjuvant chemotherapy have not been investigated for locally advanced pancreatic cancer (LAPC), despite well-established outcomes in borderline resectable pancreatic cancer (BRPC). This study aimed to investigate the clinical outcomes of patients with LAPC who underwent curative resection following neoadjuvant chemotherapy. MATERIALS AND METHODS We retrospectively reviewed the records of patients diagnosed with pancreatic adenocarcinoma between January 2017 and December 2020. RESULTS Among 1,358 patients, 260 underwent surgery following neoadjuvant chemotherapy. Among 356 LAPC patients, 98 (27.5%) and 147 (35.1%) of 418 BRPC patients underwent surgery after neoadjuvant chemotherapy. Compared to resectable pancreatic cancer (resectable PC) with upfront surgery, both LAPC and BRPC exhibited higher rates of venous resection (28.6% vs. 49.0% vs. 4.0%), arterial resection (30.6% vs. 6.8% vs. 0.5%) and greater estimated blood loss (260.5 vs. 213.1 vs. 70.4 mL). However, hospital stay, readmission rates, and postoperative pancreatic fistula rates (grade B or C) did not differ significantly between LAPC, BRPC, and resectable PC. Overall and relapse-free survival did not differ significantly between LAPC and BRPC patients. The median overall survival was 37.3 months for LAPC and 37.0 months for BRPC. The median relapse-free survival was 22.7 months for LAPC and 26.0 months for BRPC. CONCLUSION Overall survival time and postoperative complications in LAPC patients who underwent curative resection following neoadjuvant chemotherapy showed similar results to those of BRPC patients. Further research is needed to identify specific sub-populations of LAPC patients who benefit most from conversion surgery and to minimize postoperative complications.
Collapse
Affiliation(s)
- Yoo Na Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Kyu Sung
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Wook Hwang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yejong Park
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bong Jun Kwak
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Woohyung Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki Byung Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hoon Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Heung-Moon Chang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Baek-Yeol Ryoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
39
|
Li Q, Xie W, Wang Y, Qin K, Huang M, Liu T, Chen Z, Chen L, Teng L, Fang Y, Ye L, Chen Z, Zhang J, Li A, Yang W, Liu S. A Deep Learning Application of Capsule Endoscopic Gastric Structure Recognition Based on a Transformer Model. J Clin Gastroenterol 2024; 58:937-943. [PMID: 38457410 DOI: 10.1097/mcg.0000000000001972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/26/2023] [Indexed: 03/10/2024]
Abstract
BACKGROUND Gastric structure recognition systems have become increasingly necessary for the accurate diagnosis of gastric lesions in capsule endoscopy. Deep learning, especially using transformer models, has shown great potential in the recognition of gastrointestinal (GI) images according to self-attention. This study aims to establish an identification model of capsule endoscopy gastric structures to improve the clinical applicability of deep learning to endoscopic image recognition. METHODS A total of 3343 wireless capsule endoscopy videos collected at Nanfang Hospital between 2011 and 2021 were used for unsupervised pretraining, while 2433 were for training and 118 were for validation. Fifteen upper GI structures were selected for quantifying the examination quality. We also conducted a comparison of the classification performance between the artificial intelligence model and endoscopists by the accuracy, sensitivity, specificity, and positive and negative predictive values. RESULTS The transformer-based AI model reached a relatively high level of diagnostic accuracy in gastric structure recognition. Regarding the performance of identifying 15 upper GI structures, the AI model achieved a macroaverage accuracy of 99.6% (95% CI: 99.5-99.7), a macroaverage sensitivity of 96.4% (95% CI: 95.3-97.5), and a macroaverage specificity of 99.8% (95% CI: 99.7-99.9) and achieved a high level of interobserver agreement with endoscopists. CONCLUSIONS The transformer-based AI model can accurately evaluate the gastric structure information of capsule endoscopy with the same performance as that of endoscopists, which will provide tremendous help for doctors in making a diagnosis from a large number of images and improve the efficiency of examination.
Collapse
Affiliation(s)
- Qingyuan Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Weijie Xie
- School of Biomedical Engineering
- Department of Information, Guangzhou First People's Hospital, School of Medicine, South China University of Technology
| | - Yusi Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Kaiwen Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Mei Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | | | | | - Lu Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Lan Teng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Yuxin Fang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Liuhua Ye
- Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University
| | - Zhenyu Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Jie Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
| | - Wei Yang
- School of Biomedical Engineering
- Pazhou Lab, Guangzhou, Guangdong
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital
- Pazhou Lab, Guangzhou, Guangdong
- Department of Gastroenterology, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| |
Collapse
|
40
|
Lin Y, Li R, Li T, Zhao W, Ye Q, Dong C, Gao Y. A prognostic model for hepatocellular carcinoma patients based on polyunsaturated fatty acid-related genes. ENVIRONMENTAL TOXICOLOGY 2024; 39:4649-4668. [PMID: 38682322 DOI: 10.1002/tox.24273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/19/2024] [Accepted: 03/23/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVE Polyunsaturated fatty acids (PUFAs) have attracted increasing attention for their role in liver cancer development. The objective of this study is to develop a prognosis prediction model for patients with liver cancer based on PUFA-related metabolic gene characteristics. METHOD Transcriptome data and clinical data were obtained from public databases, while gene sets related to PUFAs were acquired from the gene set enrichment analysis (GSEA) database. Univariate Cox analysis was conducted on the training set, followed by LASSO logistic regression and multivariate Cox analysis on genes with p < .05. Subsequently, the stepwise Akaike information criterion method was employed to construct the model. The high- and low-risk groups were divided based on the median score, and the model's survival prediction ability, diagnostic efficiency, and risk score distribution of clinical features were validated. The above procedures were also validated in the validation set. Immune infiltration levels were evaluated using four algorithms, and the immunotherapeutic potential of different groups was explored. Significant enrichment pathways among different groups were selected based on the GSEA algorithm, and mutation analyses were conducted. Nomogram prognostic models were constructed by incorporating clinical factors and risk scores using univariate and multivariate Cox regression analysis, validated through calibration curves and clinical decision curves. Additionally, sensitivity analysis of drugs was performed to screen potential targeted drugs. RESULTS We constructed a prognostic model comprising eight genes (PLA2G12A, CYP2C8, ABCCI, CD74, CCR7, P2RY4, P2RY6, and YY1). Validation across multiple datasets indicated the model's favorable prognostic prediction ability and diagnostic efficiency, with poorer grading and staging observed in the high-risk group. Variations in mutation status and pathway enrichment were noted among different groups. Incorporating Stage, Grade, T.Stage, and RiskScore into the nomogram prognostic model demonstrated good accuracy and clinical decision benefits. Multiple immune analyses suggested greater benefits from immunotherapy in the low-risk group. We predicted multiple targeted drugs, providing a basis for drug development. CONCLUSION Our study's multifactorial prognostic model across multiple datasets demonstrates good applicability, offering a reliable tool for personalized therapy. Immunological and mutation-related analyses provide theoretical foundations for further research. Drug predictions offer important insights for future drug development and treatment strategies. Overall, this study provides comprehensive insights into tumor prognosis assessment and personalized treatment planning.
Collapse
Affiliation(s)
- Yun Lin
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Tong Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Wenrong Zhao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qianling Ye
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
41
|
Ye S, Hu J, Zhang D, Zhao S, Shi X, Li W, Wang J, Guan W, Yan L. Strategies for Preventing Esophageal Stenosis After Endoscopic Submucosal Dissection and Progress in Stem Cell-Based Therapies. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:522-529. [PMID: 38243787 DOI: 10.1089/ten.teb.2023.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
Endoscopic submucosal dissection (ESD) has been widely used in the early neoplasia of the esophagus. However, postoperative esophageal stenosis is a big problem, particularly when a large circumferential proportion of esophageal mucosa is resected. Currently, there are several methods available to prevent esophageal stenosis after ESD, including steroid administration, esophageal stent implantation, and endoscopic balloon dilation (EBD). However, the therapeutic effects of these are not yet satisfactory. Stem cell-based therapies has shown promising potential in reconstructing tissue structure and restoring tissue function. In this study, we discussed the current strategies for preventing esophageal stenosis after ESD and perspectives of stem cell-based therapies for the prevention of esophageal stenosis.
Collapse
Affiliation(s)
- Shujun Ye
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jingjing Hu
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Daxu Zhang
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Shuo Zhao
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiaonan Shi
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Weilong Li
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jingyi Wang
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Weiping Guan
- Department of Geriatric Neurology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Li Yan
- Department of Geriatric Gastroenterology, the Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
42
|
Li Q, Xia C, Li H, Yan X, Yang F, Cao M, Zhang S, Teng Y, He S, Cao M, Chen W. Disparities in 36 cancers across 185 countries: secondary analysis of global cancer statistics. Front Med 2024; 18:911-920. [PMID: 39167345 DOI: 10.1007/s11684-024-1058-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/19/2023] [Indexed: 08/23/2024]
Abstract
Cancer is a major public health problem and represents substantial disparities worldwide. This study reported estimates for 36 cancers across 185 countries by incidence, mortality, 5-year prevalence, mortality-to-prevalence ratio (MPR), and mortality-to-incidence ratio (MIR) to examine its association with human development index (HDI) and gross national income (GNI). Data were collected from the GLOBOCAN 2020. MPR and MIR were calculated by sex, age group, country, and cancer type and then summarized into totals. Segi's population and global cancer spectrum were used to calculate age- and type-standardized ratios. Correlation analyses were conducted to assess associations. Results showed that breast cancer was the most diagnosed cancer globally. Low- and middle-income countries had high MPR and MIR. Cancers of esophagus, pancreas, and liver had the highest ratios. Males and the older population had the highest ratios. HDI and GNI were positively correlated with incidence and mortality but negatively correlated with MPR/MIR. Substantial disparities in cancer burden were observed among 36 cancer types across 185 countries. Socioeconomic development may contribute to narrowing these disparities, and tailored strategies are crucial for regional- and country-specific cancer control.
Collapse
Affiliation(s)
- Qianru Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Changfa Xia
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Li
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinxin Yan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fan Yang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mengdi Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shaoli Zhang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yi Teng
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Siyi He
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Maomao Cao
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
43
|
Zandavi SM, Kim C, Goodwin T, Thilakanathan C, Bostanara M, Akon AC, Al Mouiee D, Barisic S, Majeed A, Kemp W, Chu F, Smith M, Collins K, Wong VWS, Wong GLH, Behary J, Roberts SK, Ng KKC, Vafaee F, Zekry A. AI-powered prediction of HCC recurrence after surgical resection: Personalised intervention opportunities using patient-specific risk factors. Liver Int 2024; 44:2724-2737. [PMID: 39046171 DOI: 10.1111/liv.16050] [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: 03/18/2024] [Revised: 06/18/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) recurrence following surgical resection remains a significant clinical challenge, necessitating reliable predictive models to guide personalised interventions. In this study, we sought to harness the power of artificial intelligence (AI) to develop a robust predictive model for HCC recurrence using comprehensive clinical datasets. METHODS Leveraging data from 958 patients across multiple centres in Australia and Hong Kong, we employed a multilayer perceptron (MLP) as the optimal classifier for model generation. RESULTS Through rigorous internal cross-validation, including a cohort from the Chinese University of Hong Kong (CUHK), our AI model successfully identified specific pre-surgical risk factors associated with HCC recurrence. These factors encompassed hepatic synthetic function, liver disease aetiology, ethnicity and modifiable metabolic risk factors, collectively contributing to the predictive synergy of our model. Notably, our model exhibited high accuracy during cross-validation (.857 ± .023) and testing on the CUHK cohort (.835), with a notable degree of confidence in predicting HCC recurrence within accurately classified patient cohorts. To facilitate clinical application, we developed an online AI digital tool capable of real-time prediction of HCC recurrence risk, demonstrating acceptable accuracy at the individual patient level. CONCLUSION Our findings underscore the potential of AI-driven predictive models in facilitating personalised risk stratification and targeted interventions to mitigate HCC recurrence by identifying modifiable risk factors unique to each patient. This model aims to aid clinicians in devising strategies to disrupt the underlying carcinogenic network driving recurrence.
Collapse
Affiliation(s)
- Seid Miad Zandavi
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
- UNSW Data Science Hub, University of New South Wales, Sydney, New South Wales, Australia
| | - Christy Kim
- St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, New South Wales, Australia
| | - Thomas Goodwin
- Department of Gastroenterology and Hepatology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Cynthuja Thilakanathan
- St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, New South Wales, Australia
| | - Maryam Bostanara
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Anna Camille Akon
- St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, New South Wales, Australia
| | - Daniel Al Mouiee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
- The Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Sasha Barisic
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
- School of Computer Science and Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | - Ammar Majeed
- Department of Gastroenterology and Hepatology, The Alfred Hospital, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - William Kemp
- Department of Gastroenterology and Hepatology, The Alfred Hospital, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Francis Chu
- Department of Liver Surgery, St George Hospital, University of New South Wales, Sydney, New South Wales, Australia
| | - Marty Smith
- Department of Hepatobiliary Surgery, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Kate Collins
- Department of Gastroenterology and Hepatology, The Austin Hospital, Melbourne, Victoria, Australia
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Lai-Hung Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Jason Behary
- St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, New South Wales, Australia
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, The Alfred Hospital, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Kelvin K C Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
- UNSW Data Science Hub, University of New South Wales, Sydney, New South Wales, Australia
| | - Amany Zekry
- St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology and Hepatology, St George Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
44
|
Gunay M, Leone C, Albayati O, Mohamed A, Anyadike N. Metastatic Small Cell Carcinoma of the Stomach. ACG Case Rep J 2024; 11:e01540. [PMID: 39421734 PMCID: PMC11484663 DOI: 10.14309/crj.0000000000001540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Primary gastric small cell carcinoma (GSCC) is an extremely rare type of small cell carcinoma. Its aggressive nature with early widespread metastasis and late detection gives it a poor prognosis with overall survival of <12 months. GSCC is a type of neuroendocrine tumor, and because of its histopathological similarity to small cell lung carcinoma (SCLC), treatment regimen of GSCC includes the same chemotherapy agents as SCLC. We report a case of a 57-year-old man who presented with signs of partial gastrointestinal obstruction and was found to have a primary stage IV GSCC with metastasis to the liver.
Collapse
Affiliation(s)
- Mirac Gunay
- Department of Internal Medicine, Morristown Medical Center, Morristown, NJ
| | - Celia Leone
- Department of Internal Medicine, Morristown Medical Center, Morristown, NJ
| | - Oksana Albayati
- Department of Internal Medicine, Morristown Medical Center, Morristown, NJ
| | - Abdalla Mohamed
- Department of Gastroenterology, Morristown Medical Center, Morristown, NJ
| | - Nnaemeka Anyadike
- Department of Gastroenterology, Morristown Medical Center, Morristown, NJ
| |
Collapse
|
45
|
Perremans P, Van Herpe F, Rasschaert G, Van Ongeval J, Decaestecker J, Topal B, Bislenghi G, Wolthuis A, Topal H, Deroose C, Van Cutsem E, Dekervel J. Salvage Surgery for Unifocal Progressive Metastatic Mismatch Repair-Deficient GI Cancer Responding to Immune Checkpoint Inhibition. JCO Precis Oncol 2024; 8:e2400176. [PMID: 39393035 DOI: 10.1200/po.24.00176] [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: 03/22/2024] [Revised: 07/16/2024] [Accepted: 09/06/2024] [Indexed: 10/13/2024] Open
Abstract
Case series describing excellent outcomes for patients with dMMR GI cancer after resection of a single progressive lesion under immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Baki Topal
- University Hospital Gasthuisberg and University of Leuven (KUL), Leuven, Belgium
| | | | - Albert Wolthuis
- University Hospital Gasthuisberg and University of Leuven (KUL), Leuven, Belgium
| | - Halit Topal
- University Hospital Gasthuisberg, Leuven, Belgium
| | - Christophe Deroose
- University Hospital Gasthuisberg and University of Leuven (KUL), Leuven, Belgium
| | - Eric Van Cutsem
- University Hospital Gasthuisberg and University of Leuven (KUL), Leuven, Belgium
| | - Jeroen Dekervel
- University Hospital Gasthuisberg and University of Leuven (KUL), Leuven, Belgium
| |
Collapse
|
46
|
Vaghari-Tabari M, Qujeq D, Hashemzadeh MS. Long noncoding RNAs as potential targets for overcoming chemoresistance in upper gastrointestinal cancers. Biomed Pharmacother 2024; 179:117368. [PMID: 39214010 DOI: 10.1016/j.biopha.2024.117368] [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: 05/27/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
In the last decade, researchers have paid much attention to the role of noncoding RNA molecules in human diseases. Among the most important of these molecules are LncRNAs, which are RNA molecules with a length of more than 200 nucleotides. LncRNAs can regulate gene expression through various mechanisms, such as binding to DNA sequences and interacting with miRNAs. Studies have shown that LncRNAs may be valuable therapeutic targets in treating various cancers, including upper-gastrointestinal cancers. Upper gastrointestinal cancers, mainly referring to esophageal and gastric cancers, are among the deadliest gastrointestinal cancers. Despite notable advances, traditional chemotherapy remains a common strategy for treating these cancers. However, chemoresistance poses a significant obstacle to the effective treatment of upper gastrointestinal cancers, resulting in a low survival rate. Chemoresistance arises from various events, such as the enhancement of efflux and detoxification of chemotherapy agents, reduction of drug uptake, alteration of drug targeting, reduction of prodrug activation, strengthening of EMT and stemness, and the attenuation of apoptosis in cancerous cells. Tumor microenvironment also plays an important role in chemoresistance. Interestingly, a series of studies have revealed that LncRNAs can influence important mechanisms associated with some of the aforementioned events and may serve as promising targets for mitigating chemoresistance in upper gastrointestinal cancers. In this review paper, following a concise overview of chemoresistance mechanisms in upper gastrointestinal cancers, we will review the most intriguing findings of these investigations in detail.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | | |
Collapse
|
47
|
Cao Z, Wang W, Yang Z, Liu Y, Sun L, Zhang L, Li Z. Discovery of the FXR/CES2 dual modulator LE-77 for the treatment of irinotecan-induced delayed diarrhea. Bioorg Chem 2024; 153:107852. [PMID: 39362081 DOI: 10.1016/j.bioorg.2024.107852] [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: 08/12/2024] [Revised: 09/11/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Irinotecan (CPT-11) is a widely utilized topoisomerase I inhibitor in the treatment of colorectal cancer and other malignant tumors. However, severe and even life-threatening dose-limiting toxicity-delayed diarrhea affects the clinical application of CPT-11. The standard treatment for CPT-11-induced delayed diarrhea is prompt use of loperamide (LPA), however LPA can also cause constipation, diarrhea and even intestinal obstruction and has a high failure rate. Carboxylesterase 2 (CES2) is the main enzyme in the intestinal transformation of CPT-11, which can convert CPT-11 into toxic metabolite SN-38 and produce intestinal toxicity. Inhibiting CES2 activity can block the hydrolysis process of CPT-11 in the intestine and reduce SN-38 accumulation. Additionally, Farnesoid X receptor (FXR) agonists have anti-inflammatory, anti-secretory, and protective functions on intestinal barrier integrity that could potentially alleviate diarrhea. In this study, we investigated for the first time the anti-delayed diarrhea effect of FXR agonists, and the first time identified LE-77 as a potent dual modulator that activates FXR and inhibits CES2 through high-throughput screening. In the CPT-11-induced delayed diarrhea model, LE-77 demonstrated a dual modulator mechanism by activating FXR and inhibiting CES2, thereby reducing the accumulation of SN-38 in the intestine, alleviating intestinal inflammation, preserving intestinal mucosal integrity, and ultimately alleviating delayed diarrhea.
Collapse
Affiliation(s)
- Zhijun Cao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Wenxin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zhongcheng Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yuxia Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lidan Sun
- Department of Pharmaceutics, Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, PR China.
| | - Luyong Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Key Laboratory of New Drug Discovery and Evaluation of the Guangdong Provincial Education Department, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangzhou, 510006, PR China.
| |
Collapse
|
48
|
Yousefpour N, Mahmoodzadeh H, Mahdavi R, Fattahi MR, Jalaeefar A, Ataee H, Ameli F, Hajighasemi F, Mokhtari Dowlatabad H, Mansouri S, Nabavian O, Miri SR, Abdolahad M. Electrical Tumor Detection Probe Calibrated to Diagnose Gastrointestinal Cancer Mass in Real-Time. J Clin Med 2024; 13:5823. [PMID: 39407883 PMCID: PMC11477054 DOI: 10.3390/jcm13195823] [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: 06/02/2024] [Revised: 06/24/2024] [Accepted: 06/29/2024] [Indexed: 10/20/2024] Open
Abstract
Background: The primary objective of this research is to propose an intra-operative tumor detection probe calibrated on human models of gastrointestinal (G.I.) cancers, enabling real-time scanning of dissected masses. Methods: Electrical Gastrointestinal Cancer Detection (EGCD) measures impedimetric characteristics of G.I. masses using a handpiece probe and a needle-based head probe. Impedance Phase Slope (IPS) and impedance magnitude (Z1kHz) are extracted as the classification parameters. EGCD was tested on palpable G.I. masses and compared to histopathology results. Results: Calibration was carried out on 120 GI mass samples. Considering pathological results as the gold standard, most cancer masses showed Z1kHz between 100 Ω and 2500 Ω while their IPS was between -15 and -1. The EGCD total sensitivity and specificity of this categorization in G.I. cancer patients with palpable tumors were 86.4% and 74.4%, respectively (p-value < 0.01). Conclusion: EGCD scoring can be used for 3D scanning of palpable tumors in G.I. tumors during surgery, which can help clarify the tumors' pathological response to neoadjuvant chemotherapy or the nature of intra-operative newly found G.I. tumors for the surgeon to manage their surgical procedure better.
Collapse
Affiliation(s)
- Narges Yousefpour
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran 1439957131, Iran; (N.Y.); (R.M.); (H.A.); (F.H.); (H.M.D.)
| | - Habibollah Mahmoodzadeh
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
| | - Reihane Mahdavi
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran 1439957131, Iran; (N.Y.); (R.M.); (H.A.); (F.H.); (H.M.D.)
| | - Mohammad Reza Fattahi
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
- School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1968917313, Iran
| | - Amirmohsen Jalaeefar
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
| | - Hossein Ataee
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran 1439957131, Iran; (N.Y.); (R.M.); (H.A.); (F.H.); (H.M.D.)
| | - Fereshteh Ameli
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
| | - Farzane Hajighasemi
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran 1439957131, Iran; (N.Y.); (R.M.); (H.A.); (F.H.); (H.M.D.)
| | - Hadi Mokhtari Dowlatabad
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran 1439957131, Iran; (N.Y.); (R.M.); (H.A.); (F.H.); (H.M.D.)
| | - Sepideh Mansouri
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
| | - Omid Nabavian
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
| | - Seyed Rouhollah Miri
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
- UT&TUMS Cancer Electronics Research Center, University of Tehran, Tehran 1417935840, Iran
| | - Mohammad Abdolahad
- Nano Bioelectronics Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran 1439957131, Iran; (N.Y.); (R.M.); (H.A.); (F.H.); (H.M.D.)
- Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran 1419733141, Iran; (H.M.); (M.R.F.); (A.J.); (F.A.); (S.M.); (O.N.)
- UT&TUMS Cancer Electronics Research Center, University of Tehran, Tehran 1417935840, Iran
| |
Collapse
|
49
|
Sreeram A, Stroobant EE, Laszkowska M, Guilford P, Shimada S, Nishimura M, Shah S, Vardhana S, Tang LH, Strong VE. Disappearing Signet Ring Cell Adenocarcinoma in Gastric Cancer Patients. Ann Surg Oncol 2024:10.1245/s10434-024-16117-8. [PMID: 39343820 DOI: 10.1245/s10434-024-16117-8] [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: 05/07/2024] [Accepted: 08/16/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND The incidence of diffuse-type gastric cancer is increasing steadily in the United States, Europe, and Asia. This subtype is known for aggressive clinical characteristics and transmural invasion. However, T1a diffuse-type cancers have been observed to have a better 5-year, disease-specific mortality than stage-matched intestinal tumors, supporting a clinical difference in these early-stage cancers. METHODS Data on all living patients with T1a gastric adenocarcinoma with a finding of signet ring cell morphology on pathology and ≥1 year of follow-up from 2013 to 2023 at Memorial Sloan Kettering Cancer Center (MSK) was collected from a prospectively maintained database. Patients with known CDH1 or CTNNA1 mutations were excluded. RESULTS In 7 of 30 patients, sporadic pathologically confirmed T1a signet ring cell (diffuse) cancer identified on initial biopsy was no longer detectable upon subsequent biopsy or resection with mean follow-up of 50 months. CONCLUSIONS These cases allude to the distinct pathways of carcinogenesis in T1a signet ring cell cancers. Potential factors that may underlie the spontaneous regression of these T1a cancers include complete removal at initial biopsy, immune clearance, and lack of survival advantage conferred by signet ring cell genetic alterations in these cases. Given their more indolent behavior at an earlier stage, we suggest that these lesions can be closely followed by endoscopy in select circumstances with thorough disease assessment and an experienced care team.
Collapse
Affiliation(s)
- Aravind Sreeram
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emily E Stroobant
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Monika Laszkowska
- Department of Medicine, Gastroenterology, Hepatology, and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parry Guilford
- Department of Biochemistry, Cancer Genetics Laboratory, Centre for Translational Cancer Research (Te Aho Matatu), University of Otago, Dunedin, New Zealand
| | - Shoji Shimada
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Makoto Nishimura
- Department of Medicine, Gastroenterology, Hepatology, and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sohrab Shah
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Santosha Vardhana
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura H Tang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vivian E Strong
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
50
|
Lei M, Xiao M, Long Z, Lin T, Ding R, Quan Q. Prognosis of colorectal cancer, prognostic index of immunogenic cell death associated genes in response to immunotherapy, and potential therapeutic effects of ferroptosis inducers. Front Immunol 2024; 15:1458270. [PMID: 39372411 PMCID: PMC11449742 DOI: 10.3389/fimmu.2024.1458270] [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: 07/10/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction This study leverages bioinformatics and medical big data to integrate datasets from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA), providing a comprehensive overview of immunogenic cell death (ICD)-related gene expression in colorectal cancer (CRC). The research aims to elucidate the molecular pathways and gene networks associated with ICD in CRC, with a focus on the therapeutic potential of cell death inducers, including ferroptosis agents, and their implications for precision medicine. Methods We conducted differential expression analysis and utilized advanced bioinformatic techniques to analyze ICD-related gene expression in CRC tissues. Unsupervised consensus clustering was applied to categorize CRC patients into distinct ICD-associated subtypes, followed by an in-depth immune microenvironment analysis and single-cell RNA sequencing to investigate immune responses and cell infiltration patterns. Experimental validation was performed to assess the impact of cell death inducers on ICD gene expression and their interaction with ferroptosis inducers in combination with other clinical drugs. Results Distinct ICD gene expression profiles were identified in CRC tissues, revealing molecular pathways and intricate gene networks. Unsupervised consensus clustering refined the CRC cohort into unique ICD-associated subtypes, each characterized by distinct clinical and immunological features. Immune microenvironment analysis and single-cell RNA sequencing revealed significant variations in immune responses and cell infiltration patterns across these subtypes. Experimental validation confirmed that cell death inducers directly affect ICD gene expression, highlighting their therapeutic potential. Additionally, combinatorial therapies with ferroptosis inducers and clinical drugs were shown to influence drug sensitivity and resistance in CRC. Discussion Our findings underscore the importance of ICD-related genes in CRC prognosis and therapeutic targeting. The study provides actionable insights into the efficacy of cell death-inducing therapies, particularly ferroptosis inducers, and their regulatory mechanisms in CRC. These discoveries support the development of precision medicine strategies targeting ICD genes and offer valuable guidance for translating these therapies into clinical practice, with the potential to enhance CRC treatment outcomes and patient survival.
Collapse
Affiliation(s)
- Mengjie Lei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Meihua Xiao
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Zhiqing Long
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Taolin Lin
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ran Ding
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Qi Quan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of the VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|