Background: Hepatocellular carcinoma (HCC) patients with coronavirus disease 2019 (COVID-19) undergoing open surgery show increased adverse events (AEs) and mortality, while the safety of transarterial chemoembolization (TACE) in coinfected patients remains understudied, limiting available evidence. This study aims to investigate the safety of TACE in HCC patients coinfected with COVID-19, and to explore the potential risk factors affecting the occurrence of serious AEs (SAEs), thus providing evidence for clinical treatment strategies in such patients.
Methods: This retrospective study involved HCC patients who underwent TACE with or without COVID-19 infection at our institution from November 2022 to February 2023. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used for the diagnosis of COVID-19. Patients were divided into an infected group (diagnosed with COVID-19 within 2 weeks before or after the procedure) and an uninfected group (tested negative for COVID-19). SAEs were ascertained according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 5.0. Logistic regression analysis of multiple clinical factors in preoperative baseline characteristics was performed to identify risk factors that might predict the occurrence of SAEs.
Results: A total of 118 patients (73 in the infected group, 45 in the uninfected group) were included, of whom 83.9% were male (86.3% in the infected group vs. 80.0% in the uninfected group) and the median age was 55.9±12.4 years (56.8±12.3 vs. 54.5±12.7 years). The clinical spectrum of COVID-19 in the infected group were 80.8% mild, 13.7% moderate, 1.4% severe and 4.1% critical. Sixteen of the 118 patients experienced SAEs (19.2% vs. 4.4%, P=0.046). The predominant SAEs were respiratory system diseases (9.6% vs. 0.0%) and liver damage (2.7% vs. 2.2%). In the univariate analysis, infection status [odds ratio (OR): 5.102, P=0.04, 95% confidence interval (CI): 1.102-23.627], gender (OR: 2.857, P=0.09, 95% CI: 0.862-9.468), age (OR: 1.061, P=0.03, 95% CI: 1.007-1.118) and clinical spectrum of COVID-19 (OR: 4.259, P<0.001, 1.943-9.336) were considered as the potential risk factors of grade ≥3 AEs. In multivariate analysis, younger age (OR: 1.064, P=0.044, 95% CI: 1.002-1.131) and a milder clinical spectrum of COVID-19 (OR: 5.736, P=0.004, 95% CI: 1.772-18.568) were independent factors associated with a lower occurrence of SAEs.
Conclusions: TACE in HCC patients co-infected with COVID-19 was considered relatively safe. Age and clinical spectrum of COVID-19 were associated with SAEs in HCC patients treated with TACE.
{"title":"Age and clinical spectrum of COVID-19 are associated with safety of transarterial chemoembolization in hepatocellular carcinoma: a retrospective cohort study.","authors":"Zizhuo Wang, Tingting Yang, Lijie Zhang, Joyman Makamure, Wei Hong, Bin Liang","doi":"10.21037/jgo-24-527","DOIUrl":"https://doi.org/10.21037/jgo-24-527","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) patients with coronavirus disease 2019 (COVID-19) undergoing open surgery show increased adverse events (AEs) and mortality, while the safety of transarterial chemoembolization (TACE) in coinfected patients remains understudied, limiting available evidence. This study aims to investigate the safety of TACE in HCC patients coinfected with COVID-19, and to explore the potential risk factors affecting the occurrence of serious AEs (SAEs), thus providing evidence for clinical treatment strategies in such patients.</p><p><strong>Methods: </strong>This retrospective study involved HCC patients who underwent TACE with or without COVID-19 infection at our institution from November 2022 to February 2023. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used for the diagnosis of COVID-19. Patients were divided into an infected group (diagnosed with COVID-19 within 2 weeks before or after the procedure) and an uninfected group (tested negative for COVID-19). SAEs were ascertained according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 5.0. Logistic regression analysis of multiple clinical factors in preoperative baseline characteristics was performed to identify risk factors that might predict the occurrence of SAEs.</p><p><strong>Results: </strong>A total of 118 patients (73 in the infected group, 45 in the uninfected group) were included, of whom 83.9% were male (86.3% in the infected group <i>vs.</i> 80.0% in the uninfected group) and the median age was 55.9±12.4 years (56.8±12.3 <i>vs.</i> 54.5±12.7 years). The clinical spectrum of COVID-19 in the infected group were 80.8% mild, 13.7% moderate, 1.4% severe and 4.1% critical. Sixteen of the 118 patients experienced SAEs (19.2% <i>vs.</i> 4.4%, P=0.046). The predominant SAEs were respiratory system diseases (9.6% <i>vs.</i> 0.0%) and liver damage (2.7% <i>vs.</i> 2.2%). In the univariate analysis, infection status [odds ratio (OR): 5.102, P=0.04, 95% confidence interval (CI): 1.102-23.627], gender (OR: 2.857, P=0.09, 95% CI: 0.862-9.468), age (OR: 1.061, P=0.03, 95% CI: 1.007-1.118) and clinical spectrum of COVID-19 (OR: 4.259, P<0.001, 1.943-9.336) were considered as the potential risk factors of grade ≥3 AEs. In multivariate analysis, younger age (OR: 1.064, P=0.044, 95% CI: 1.002-1.131) and a milder clinical spectrum of COVID-19 (OR: 5.736, P=0.004, 95% CI: 1.772-18.568) were independent factors associated with a lower occurrence of SAEs.</p><p><strong>Conclusions: </strong>TACE in HCC patients co-infected with COVID-19 was considered relatively safe. Age and clinical spectrum of COVID-19 were associated with SAEs in HCC patients treated with TACE.</p>","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2642-2655"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732337/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Following tumor resection, imaging recommendation for the follow-up of patients with intrahepatic cholangiocarcinoma (IHCCA) include frequent chest, abdomen and pelvis computed tomography (CT) imaging. The appropriateness of additional imaging studies is usually derived from their clinical utility. The purpose of this work is to determine the value of chest CT imaging in the follow-up of patients with IHCCA.
Methods: Data review of radiology reports of baseline post-operative chest, abdominal, and pelvic CT imaging reports following the resection of IHCCA, and of subsequent follow-up exams. The radiology findings were stratified as intrathoracic metastasis only, combined intra-thoracic and intraabdominal metastasis, intra-abdominal metastases without intrathoracic involvement. We assessed the prevalence of intra-thoracic disease progression in comparison to other groups. Descriptive statistical analysis was carried out using John's Macintosh Project (JMP) statistical software.
Results: Eighty-seven patients were included in the study, 6 patients were found to have disease progression in the chest without corresponding disease progression in the abdomen on follow-up CT, accounting for 6.9% of the total. Only four patients had disease progression in the chest with a normal CT chest at baseline (4.6%).
Conclusions: The use of short-interval chest CT for surveillance in IHCCA has limited utility, particularly in patients with disease-free abdominal studies.
背景:肿瘤切除后,肝内胆管癌(IHCCA)患者随访的影像学建议包括频繁的胸部、腹部和骨盆CT成像。额外影像学检查的适当性通常源于其临床应用。本工作的目的是确定胸部CT成像在IHCCA患者随访中的价值。方法:回顾IHCCA切除术后胸部、腹部和骨盆基线CT影像学报告以及随后随访检查的资料。影像学表现分为单胸转移、胸腹合并转移、不累及胸内转移。与其他组相比,我们评估了胸内疾病进展的患病率。采用John’s Macintosh Project (JMP)统计软件进行描述性统计分析。结果:纳入研究的87例患者中,随访CT发现6例患者胸部有疾病进展,腹部无相应疾病进展,占总数的6.9%。只有4例患者在基线时胸部CT显示正常,但胸部疾病进展(4.6%)。结论:在IHCCA中使用短间隔胸部CT进行监测的效用有限,特别是在无疾病腹部研究的患者中。
{"title":"Assessing the value of computed tomography (CT) of the chest in the follow-up of patients with intrahepatic cholangiocarcinoma.","authors":"Moamen Abdelaal, Mahmoud Diab, Maguy Farhat, Milind Javle, Mostafa Shehata, Emad Singer, Yomna Khamis, Manal Hassan, Khaled M Elsayes, Janio Szklaruk","doi":"10.21037/jgo-24-365","DOIUrl":"https://doi.org/10.21037/jgo-24-365","url":null,"abstract":"<p><strong>Background: </strong>Following tumor resection, imaging recommendation for the follow-up of patients with intrahepatic cholangiocarcinoma (IHCCA) include frequent chest, abdomen and pelvis computed tomography (CT) imaging. The appropriateness of additional imaging studies is usually derived from their clinical utility. The purpose of this work is to determine the value of chest CT imaging in the follow-up of patients with IHCCA.</p><p><strong>Methods: </strong>Data review of radiology reports of baseline post-operative chest, abdominal, and pelvic CT imaging reports following the resection of IHCCA, and of subsequent follow-up exams. The radiology findings were stratified as intrathoracic metastasis only, combined intra-thoracic and intraabdominal metastasis, intra-abdominal metastases without intrathoracic involvement. We assessed the prevalence of intra-thoracic disease progression in comparison to other groups. Descriptive statistical analysis was carried out using John's Macintosh Project (JMP) statistical software.</p><p><strong>Results: </strong>Eighty-seven patients were included in the study, 6 patients were found to have disease progression in the chest without corresponding disease progression in the abdomen on follow-up CT, accounting for 6.9% of the total. Only four patients had disease progression in the chest with a normal CT chest at baseline (4.6%).</p><p><strong>Conclusions: </strong>The use of short-interval chest CT for surveillance in IHCCA has limited utility, particularly in patients with disease-free abdominal studies.</p>","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2656-2662"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732355/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006537","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-09DOI: 10.21037/jgo-24-515
Mélodie Cyr, Naim Chabaytah, Joud Babik, Behnaz Behmand, Guillaume St-Jean, Shirin A Enger
Background: Orthotopic models offer a more accurate representation of colorectal cancer (CRC) compared to subcutaneous models. Despite promising results from the reported intra-rectal models, establishing a standardized method for CRC research remains challenging due to model variability, hindering comprehensive studies on CRC pathogenesis and treatment modalities, such as brachytherapy. This study aimed to establish a standardized workflow for an orthotopic intra-rectal animal model to induce the growth of colorectal adenocarcinoma in male and female mice.
Methods: HT-29 colorectal adenocarcinoma cells were injected into the rectal mucosa of female (n=21) and male (n=26) non-obese diabetic severe combined immunodeficiency (NOD SCID) gamma (NSG) mice. Mice were placed on a 45° wedge elevating their pelvis for better visualization of the anus. Tumor growth and localization were monitored using a 7-T magnetic resonance imaging (MRI) scanner with rapid acquisition with relaxation echo (RARE) sequence at weeks 1, 2, and 3 post-cell instillation. Once tumors reached 5-8 mm in diameter, the mice were euthanized. Histopathology and immunohistochemical analyses confirmed the tumors' morphology, including necrosis, vascularity (CD-31) and apoptosis (cleaved caspase-3).
Results: There was a 92% and 95% tumor growth success rate in male and female mice, respectively. Tumors grew to 5-8 mm in diameter within ~20 days. No significant difference in tumor size was observed between genders. Tumor morphology was consistent across cases. Most tumors exhibited a lack of central blood vessels, accompanied by varying degrees of necrosis and apoptosis, whereas external portions were highly vascularized.
Conclusions: An orthotopic intra-rectal model was successfully developed. This model will be used in future studies to evaluate the efficacy of CRC treatments.
{"title":"Establishing a standardized murine orthotopic intra-rectal model for the study of colorectal adenocarcinoma.","authors":"Mélodie Cyr, Naim Chabaytah, Joud Babik, Behnaz Behmand, Guillaume St-Jean, Shirin A Enger","doi":"10.21037/jgo-24-515","DOIUrl":"https://doi.org/10.21037/jgo-24-515","url":null,"abstract":"<p><strong>Background: </strong>Orthotopic models offer a more accurate representation of colorectal cancer (CRC) compared to subcutaneous models. Despite promising results from the reported intra-rectal models, establishing a standardized method for CRC research remains challenging due to model variability, hindering comprehensive studies on CRC pathogenesis and treatment modalities, such as brachytherapy. This study aimed to establish a standardized workflow for an orthotopic intra-rectal animal model to induce the growth of colorectal adenocarcinoma in male and female mice.</p><p><strong>Methods: </strong>HT-29 colorectal adenocarcinoma cells were injected into the rectal mucosa of female (n=21) and male (n=26) non-obese diabetic severe combined immunodeficiency (NOD SCID) gamma (NSG) mice. Mice were placed on a 45° wedge elevating their pelvis for better visualization of the anus. Tumor growth and localization were monitored using a 7-T magnetic resonance imaging (MRI) scanner with rapid acquisition with relaxation echo (RARE) sequence at weeks 1, 2, and 3 post-cell instillation. Once tumors reached 5-8 mm in diameter, the mice were euthanized. Histopathology and immunohistochemical analyses confirmed the tumors' morphology, including necrosis, vascularity (CD-31) and apoptosis (cleaved caspase-3).</p><p><strong>Results: </strong>There was a 92% and 95% tumor growth success rate in male and female mice, respectively. Tumors grew to 5-8 mm in diameter within ~20 days. No significant difference in tumor size was observed between genders. Tumor morphology was consistent across cases. Most tumors exhibited a lack of central blood vessels, accompanied by varying degrees of necrosis and apoptosis, whereas external portions were highly vascularized.</p><p><strong>Conclusions: </strong>An orthotopic intra-rectal model was successfully developed. This model will be used in future studies to evaluate the efficacy of CRC treatments.</p>","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2578-2587"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732354/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006573","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-28DOI: 10.21037/jgo-24-524
Alexandre A Jácome, Maria Cecília Mathias-Machado, Mariana Gil, Thaís M Passarini, Sabrina Cristofaro, Eduardo D Moraes, Laura V W Freitas, Gabriel Prolla, Larissa C Amorim, Rafael D Paes, Bianca Gasparotto, Jorge Canedo, Carlos Gil Ferreira, Bruno Ferrari, Bernardo Garicochea, Roberto Gil, Renata D'Alpino Peixoto
Background: Regorafenib and trifluridine/tipiracil (TFD/TPI) are oral systemic therapies with survival benefit in chemorefractory metastatic colorectal cancer (mCRC) patients, but they are not widely available worldwide. We aimed to evaluate the treatment patterns and outcomes of patients with limited access to these drugs.
Methods: Retrospective study involving 510 patients with mCRC who were treated at five different centers in Brazil, from January 2011 to December 2019. We analyzed the characteristics and clinical outcomes of patients who were exposed to third-line and beyond systemic therapy (N=148).
Results: A total of 148 (29%) and 73 (14%) patients received third- and fourth-line therapies, respectively. Most patients had left-sided tumors (80%) and had undergone primary tumor resection (87%) and metastasectomy (62%). From the initiation of third-line therapy, median follow-up was 17.8 months and median overall survival (OS) was 13.1 months (95% confidence interval: 8.1-18.0) (58% of deaths). The most adopted therapies were reexposure to previous chemotherapy regimens (85% third-line and 71% fourth-line), followed by regorafenib (13% and 25%) and TFD/TPI (2% and 4%). In multivariable analysis, male patients and right-sided tumors were significant unfavorable prognostic factors for OS.
Conclusions: In a real-world setting with barriers to accessing the standard of care of chemorefractory mCRC, reexposure to chemotherapy was the main therapeutic strategy, with potential negative influence on OS. Our study highlights how barriers hamper the achievement of health equity in cancer care and increases the awareness of patients and stakeholders, contributing to engaging them in ensuring equitable access to high-quality cancer care.
{"title":"Later lines of systemic therapy in patients with metastatic colorectal cancer: real-world data from a setting with barriers to access cancer therapies.","authors":"Alexandre A Jácome, Maria Cecília Mathias-Machado, Mariana Gil, Thaís M Passarini, Sabrina Cristofaro, Eduardo D Moraes, Laura V W Freitas, Gabriel Prolla, Larissa C Amorim, Rafael D Paes, Bianca Gasparotto, Jorge Canedo, Carlos Gil Ferreira, Bruno Ferrari, Bernardo Garicochea, Roberto Gil, Renata D'Alpino Peixoto","doi":"10.21037/jgo-24-524","DOIUrl":"10.21037/jgo-24-524","url":null,"abstract":"<p><strong>Background: </strong>Regorafenib and trifluridine/tipiracil (TFD/TPI) are oral systemic therapies with survival benefit in chemorefractory metastatic colorectal cancer (mCRC) patients, but they are not widely available worldwide. We aimed to evaluate the treatment patterns and outcomes of patients with limited access to these drugs.</p><p><strong>Methods: </strong>Retrospective study involving 510 patients with mCRC who were treated at five different centers in Brazil, from January 2011 to December 2019. We analyzed the characteristics and clinical outcomes of patients who were exposed to third-line and beyond systemic therapy (N=148).</p><p><strong>Results: </strong>A total of 148 (29%) and 73 (14%) patients received third- and fourth-line therapies, respectively. Most patients had left-sided tumors (80%) and had undergone primary tumor resection (87%) and metastasectomy (62%). From the initiation of third-line therapy, median follow-up was 17.8 months and median overall survival (OS) was 13.1 months (95% confidence interval: 8.1-18.0) (58% of deaths). The most adopted therapies were reexposure to previous chemotherapy regimens (85% third-line and 71% fourth-line), followed by regorafenib (13% and 25%) and TFD/TPI (2% and 4%). In multivariable analysis, male patients and right-sided tumors were significant unfavorable prognostic factors for OS.</p><p><strong>Conclusions: </strong>In a real-world setting with barriers to accessing the standard of care of chemorefractory mCRC, reexposure to chemotherapy was the main therapeutic strategy, with potential negative influence on OS. Our study highlights how barriers hamper the achievement of health equity in cancer care and increases the awareness of patients and stakeholders, contributing to engaging them in ensuring equitable access to high-quality cancer care.</p>","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2543-2551"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732344/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-18DOI: 10.21037/jgo-24-755
Ahmed Elnahla, Carlos E Guerra-Londono
{"title":"Transforming hyperthermic intraperitoneal chemotherapy: using computer simulation to improve HIPEC treatments.","authors":"Ahmed Elnahla, Carlos E Guerra-Londono","doi":"10.21037/jgo-24-755","DOIUrl":"https://doi.org/10.21037/jgo-24-755","url":null,"abstract":"","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2745-2747"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732341/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-28DOI: 10.21037/jgo-24-624
Adrianna Glisan, Eric Nielsen, Taylor Billion, Ali Bin Abdul Jabbar, Aditya Avula, Mohsin Mirza, Abubakar Tauseef
Background: Colorectal cancer (CRC) screening via colonoscopy is now advised for most adults beginning at age 45 years, an update from the earlier recommendation of age 50 years. With the increase in CRC screening rates, it is crucial to examine how mortality rates have changed over time. The aim of this study is to identify trends and regional differences in CRC mortality over the last two decades, specifically in individuals within the CRC recommended screening age group.
Methods: This study used the Center for Disease Control's Wide-ranging Online Data for Epidemiologic Research (CDC WONDER) system to collect data on CRC mortality for people ages 45-84 years in the United States from 1999-2022.
Results: During the study period, the overall age-adjusted mortality rate (AAMR) for CRC decreased across all census regions. The Midwest consistently had the highest AAMR, while the West had the lowest. Among genders, males in the Northeast had the highest rates, whereas females in the West had the lowest. Black or African Americans in the Midwest experienced the highest AAMR among racial groups, while Asian or Pacific Islanders in the Midwest had the lowest AAMR.
Conclusions: The mortality rate of CRC has been decreasing since 1999, but differences between regional groups have persisted. Disparities in outcomes still exist, showing that vulnerable subgroups require targeted interventions such as improved screening and follow-up access to increase early-stage diagnoses and potential for curative treatments.
{"title":"Regional trends in colorectal cancer mortality in people aged 45-84 years in the US, 1999-2022.","authors":"Adrianna Glisan, Eric Nielsen, Taylor Billion, Ali Bin Abdul Jabbar, Aditya Avula, Mohsin Mirza, Abubakar Tauseef","doi":"10.21037/jgo-24-624","DOIUrl":"10.21037/jgo-24-624","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC) screening via colonoscopy is now advised for most adults beginning at age 45 years, an update from the earlier recommendation of age 50 years. With the increase in CRC screening rates, it is crucial to examine how mortality rates have changed over time. The aim of this study is to identify trends and regional differences in CRC mortality over the last two decades, specifically in individuals within the CRC recommended screening age group.</p><p><strong>Methods: </strong>This study used the Center for Disease Control's Wide-ranging Online Data for Epidemiologic Research (CDC WONDER) system to collect data on CRC mortality for people ages 45-84 years in the United States from 1999-2022.</p><p><strong>Results: </strong>During the study period, the overall age-adjusted mortality rate (AAMR) for CRC decreased across all census regions. The Midwest consistently had the highest AAMR, while the West had the lowest. Among genders, males in the Northeast had the highest rates, whereas females in the West had the lowest. Black or African Americans in the Midwest experienced the highest AAMR among racial groups, while Asian or Pacific Islanders in the Midwest had the lowest AAMR.</p><p><strong>Conclusions: </strong>The mortality rate of CRC has been decreasing since 1999, but differences between regional groups have persisted. Disparities in outcomes still exist, showing that vulnerable subgroups require targeted interventions such as improved screening and follow-up access to increase early-stage diagnoses and potential for curative treatments.</p>","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2533-2542"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732362/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-28DOI: 10.21037/jgo-2024-950
Xiuwei Mi, Huihui Yao, Yang Lu, Mei Yang, Yi Yang, Dong Fang, Songbing He
Background: Chemoresistance is a major cause of treatment failure in advanced colorectal cancer (CRC), severely impacting patient survival and quality of life. While conventional chemotherapy regimens can somewhat control tumor progression, their effectiveness is frequently compromised by the development of drug resistance in cancer cells. The aim of this study is to verify and elucidate the specific mechanisms by which leptin enhances chemosensitivity in CRC, providing valuable insights for the development of new combination chemotherapy options.
Methods: We examined the link between CRC chemoresistance and fatty-acid metabolism driven by the high expression of carnitine palmitoyltransferase-1b (CPT1B) through an integrated approach combining bioinformatics and clinical sample analysis. In vitro and in vivo experiments were conducted to evaluate the effect of leptin, an adipocyte-derived cytokine, on CRC cells' response to cisplatin.
Results: Leptin significantly enhanced CRC cells' chemosensitivity to cisplatin by downregulating CPT1B expression, thereby disrupting the fatty-acid oxidation pathways that support drug resistance. In mouse models, the coadministration of leptin and cisplatin resulted in notable reductions in tumor size and weight compared to cisplatin alone, underscoring leptin's potential to enhance chemotherapy efficacy.
Conclusions: These findings indicate that leptin, through modulation of CPT1B, may serve as a promising adjunct to chemotherapy for CRC, addressing the challenge of chemoresistance and improving therapeutic outcomes. The leptin-CPT1B axis may be potential therapeutic target, providing new avenues for CRC treatment strategies aimed at overcoming drug resistance.
{"title":"Leptin increases chemosensitivity by inhibiting <i>CPT1B</i> in colorectal cancer cells.","authors":"Xiuwei Mi, Huihui Yao, Yang Lu, Mei Yang, Yi Yang, Dong Fang, Songbing He","doi":"10.21037/jgo-2024-950","DOIUrl":"https://doi.org/10.21037/jgo-2024-950","url":null,"abstract":"<p><strong>Background: </strong>Chemoresistance is a major cause of treatment failure in advanced colorectal cancer (CRC), severely impacting patient survival and quality of life. While conventional chemotherapy regimens can somewhat control tumor progression, their effectiveness is frequently compromised by the development of drug resistance in cancer cells. The aim of this study is to verify and elucidate the specific mechanisms by which leptin enhances chemosensitivity in CRC, providing valuable insights for the development of new combination chemotherapy options.</p><p><strong>Methods: </strong>We examined the link between CRC chemoresistance and fatty-acid metabolism driven by the high expression of carnitine palmitoyltransferase-1b (<i>CPT1B</i>) through an integrated approach combining bioinformatics and clinical sample analysis. <i>In vitro</i> and <i>in vivo</i> experiments were conducted to evaluate the effect of leptin, an adipocyte-derived cytokine, on CRC cells' response to cisplatin.</p><p><strong>Results: </strong>Leptin significantly enhanced CRC cells' chemosensitivity to cisplatin by downregulating <i>CPT1B</i> expression, thereby disrupting the fatty-acid oxidation pathways that support drug resistance. In mouse models, the coadministration of leptin and cisplatin resulted in notable reductions in tumor size and weight compared to cisplatin alone, underscoring leptin's potential to enhance chemotherapy efficacy.</p><p><strong>Conclusions: </strong>These findings indicate that leptin, through modulation of <i>CPT1B</i>, may serve as a promising adjunct to chemotherapy for CRC, addressing the challenge of chemoresistance and improving therapeutic outcomes. The leptin-<i>CPT1B</i> axis may be potential therapeutic target, providing new avenues for CRC treatment strategies aimed at overcoming drug resistance.</p>","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2507-2520"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732356/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Gastrointestinal (GI) cancers, particularly pancreatic cancer, are characterized by a dense stromal tumor microenvironment where cancer-associated fibroblasts (CAFs) predominate. CAFs comprise highly heterogeneous subpopulations with different functions, which can be both tumor-promoting and tumor-restraining. This systematic review and meta-analysis aims to comprehensively assess the impact of the CAF marker fibroblast-activation protein (FAP) expression on clinical outcomes in GI cancers.
Methods: Adhering to PRISMA guidelines, we systematically searched PubMed/MEDLINE, Web of Science, Cochrane Library, and ClinicalTrials.gov for relevant articles. Inclusion criteria involved studies comparing GI cancer patients with and without FAP overexpression. Meta-analysis evaluated overall survival (OS), histological differentiation, local tumor invasion, lymph node metastases, and distant metastases. For each observational study, the risk of bias was assessed using the risk of bias in non-randomized studies of exposure (ROBINS-E) tool.
Results: The meta-analysis included 31 cohort studies from six countries, comprising 3,976 patients. Patients without FAP overexpression exhibited a favorable OS [hazard ratio (HR) =1.74; 95% confidence interval (CI): 1.51-2.01]. Subgroup analyses revealed consistent results across esophageal, pancreatic, colorectal, and gastric cancers. While one-year survival rates showed no significant difference, subsequent years displayed lower rates for FAP-overexpressing groups. Lymph node metastases were more frequent in FAP-overexpressing patients, whereas distant metastases did not differ. None of 31 studies systematically controlled confounding and adjusted data so that all studies were categorized as "high risk of bias" for the domain "risk of bias due to confounding". For domains "risk of bias arising from measurement of exposure", "risk of bias due to post-exposure interventions", "risk of bias arising from measurement of outcomes", and "risk of bias in selection of the reported result", all studies were categorized as "low risk of bias".
Conclusions: This meta-analysis underscores the potential adverse prognostic significance of FAP expression in GI cancers. Limitations include heterogeneity in FAP expression cutoffs and definitions. Future research should focus on delineating the precise roles and clinical implications of FAP in GI cancers.
背景:胃肠道(GI)癌症,尤其是胰腺癌,其特征是致密的间质肿瘤微环境,其中癌症相关成纤维细胞(CAFs)占主导地位。CAFs由具有不同功能的高度异质亚群组成,既可以促进肿瘤,也可以抑制肿瘤。本系统综述和荟萃分析旨在全面评估CAF标志物成纤维细胞激活蛋白(FAP)表达对胃肠道癌症临床结局的影响。方法:按照PRISMA指南,系统检索PubMed/MEDLINE、Web of Science、Cochrane Library和ClinicalTrials.gov等网站的相关文章。纳入标准包括比较FAP过表达和非FAP过表达的胃肠道癌患者的研究。荟萃分析评估了总生存期(OS)、组织学分化、局部肿瘤侵袭、淋巴结转移和远处转移。对于每项观察性研究,使用非随机暴露研究的偏倚风险(ROBINS-E)工具评估偏倚风险。结果:荟萃分析包括来自6个国家的31项队列研究,包括3,976名患者。无FAP过表达的患者表现出良好的OS[风险比(HR) =1.74;95%置信区间(CI): 1.51-2.01]。亚组分析在食管癌、胰腺癌、结直肠癌和胃癌中显示一致的结果。虽然一年生存率无显著差异,但fap过表达组随后几年的生存率较低。淋巴结转移在fap过表达的患者中更常见,而远处转移则没有差异。31项研究均未对混杂因素进行系统控制并调整数据,因此在“混杂因素所致偏倚风险”领域,所有研究均被归类为“高偏倚风险”。对于“暴露测量引起的偏倚风险”、“暴露后干预引起的偏倚风险”、“结果测量引起的偏倚风险”和“报告结果选择的偏倚风险”等领域,所有研究都被归类为“低偏倚风险”。结论:该荟萃分析强调了FAP表达在胃肠道肿瘤中的潜在不良预后意义。限制包括FAP表达截止和定义的异质性。未来的研究应集中于描述FAP在胃肠道肿瘤中的确切作用和临床意义。
{"title":"Systematic review and meta-analysis of the prognostic role of fibroblast-activation protein in gastrointestinal cancers.","authors":"Yoshiaki Sunami, Rubing Bai, Juliane Friedrichs, Ulrich Ronellenfitsch, Jörg Kleeff, Artur Rebelo","doi":"10.21037/jgo-24-374","DOIUrl":"https://doi.org/10.21037/jgo-24-374","url":null,"abstract":"<p><strong>Background: </strong>Gastrointestinal (GI) cancers, particularly pancreatic cancer, are characterized by a dense stromal tumor microenvironment where cancer-associated fibroblasts (CAFs) predominate. CAFs comprise highly heterogeneous subpopulations with different functions, which can be both tumor-promoting and tumor-restraining. This systematic review and meta-analysis aims to comprehensively assess the impact of the CAF marker fibroblast-activation protein (FAP) expression on clinical outcomes in GI cancers.</p><p><strong>Methods: </strong>Adhering to PRISMA guidelines, we systematically searched PubMed/MEDLINE, Web of Science, Cochrane Library, and ClinicalTrials.gov for relevant articles. Inclusion criteria involved studies comparing GI cancer patients with and without FAP overexpression. Meta-analysis evaluated overall survival (OS), histological differentiation, local tumor invasion, lymph node metastases, and distant metastases. For each observational study, the risk of bias was assessed using the risk of bias in non-randomized studies of exposure (ROBINS-E) tool.</p><p><strong>Results: </strong>The meta-analysis included 31 cohort studies from six countries, comprising 3,976 patients. Patients without FAP overexpression exhibited a favorable OS [hazard ratio (HR) =1.74; 95% confidence interval (CI): 1.51-2.01]. Subgroup analyses revealed consistent results across esophageal, pancreatic, colorectal, and gastric cancers. While one-year survival rates showed no significant difference, subsequent years displayed lower rates for FAP-overexpressing groups. Lymph node metastases were more frequent in FAP-overexpressing patients, whereas distant metastases did not differ. None of 31 studies systematically controlled confounding and adjusted data so that all studies were categorized as \"high risk of bias\" for the domain \"risk of bias due to confounding\". For domains \"risk of bias arising from measurement of exposure\", \"risk of bias due to post-exposure interventions\", \"risk of bias arising from measurement of outcomes\", and \"risk of bias in selection of the reported result\", all studies were categorized as \"low risk of bias\".</p><p><strong>Conclusions: </strong>This meta-analysis underscores the potential adverse prognostic significance of FAP expression in GI cancers. Limitations include heterogeneity in FAP expression cutoffs and definitions. Future research should focus on delineating the precise roles and clinical implications of FAP in GI cancers.</p>","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2447-2459"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732333/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/jgo-24-757
Sofie Seghers, Andreas Domen, Hans Prenen
{"title":"Challenges and prospects of LAG-3 inhibition in advanced gastric and gastroesophageal junction cancer: insights from the RELATIVITY-060 trial.","authors":"Sofie Seghers, Andreas Domen, Hans Prenen","doi":"10.21037/jgo-24-757","DOIUrl":"https://doi.org/10.21037/jgo-24-757","url":null,"abstract":"","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2735-2738"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732350/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006543","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-31Epub Date: 2024-12-26DOI: 10.21037/jgo-24-792
Jinglong Shi, Zehui Hou, Ludi Fan, Chen Hu, Ning Ma, Enmin Huang
<p><strong>Background: </strong>Cellular senescence is considered a new marker of cancer. It has been suggested that long non-coding RNA (lncRNA) can be used to predict the prognosis of cancers. However, it remains to be seen whether the lncRNAs associated with cellular senescence can be used to predict the prognosis of gastric cancer (GC). The present study aimed to develop a novel senescence-related lncRNA signature (SenLncSig) to predict GC prognosis. The SenLncSig model holds promise for enhancing patient stratification, enabling more precise prognostic predictions and facilitating immunotherapy strategies.</p><p><strong>Methods: </strong>Senescence-associated lncRNAs were identified from RNA expression profiles in The Cancer Genome Atlas (TCGA) database through the construction of a co-expression network linking senescence genes and lncRNAs. A prognostic signature for GC (334 patients from TCGA-STAD data set), comprising the senescence-related lncRNAs, was developed through univariate and multivariate Cox proportional hazards regression analyses. By using the median SenLncSig risk score, the GC patients were categorized into high- and low-risk groups. A Kaplan-Meier analysis and gene set enrichment analysis were conducted, and immune infiltration, the tumor mutation burden (TMB), and pharmacological treatments were compared between the high- and low-risk groups. We used an independent GC cohort (an external cohort of 30 pairs of tumor and non-tumor tissues from the GC patients) and three GC cell lines to conduct a quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis to validate the results.</p><p><strong>Results: </strong>We established a SenLncSig, a prognostic risk model comprising the following five senescence-associated lncRNAs; AP000695.2, LINC02381, AC005586.1, AP003392.1, and AP001528.2. According to the SenLncSig, high-risk scores were associated with poor overall survival (multivariate Cox proportional hazard ratio: 1.498, 95% confidence interval: 1.294-1.735; P<0.001). The time-dependent receiver operating characteristic curve indicated that the model performed (area under the curve: 0.711). We developed a nomogram incorporating age, gender, grade, stage, T stage, M stage, N stage, and SenLncSig risk score to estimate 1-year, 3-year, and 5-year survival rates. Further, according to the results of the mutation analysis, patients with a high TMB in the high-risk group had the worst prognosis. Interestingly, the high-risk group had a stronger infiltration of regulatory T cells (P<0.001) and M2 macrophage cells (P<0.001), as well as higher tumor immune dysfunction and exclusion scores than the low-risk group. These results might explain why the high-risk group had a worse prognosis. Finally, the qRT-PCR validation revealed that the AP000695.2 and AP003392.1 expression levels were significantly higher in the tumor tissues and GC cell lines than the normal tissues and normal human gastric epithelial cell line,
{"title":"Development and experimental validation of a senescence-related long non-coding RNA signature for prognostic prediction and immune microenvironment characterization in gastric cancer patients.","authors":"Jinglong Shi, Zehui Hou, Ludi Fan, Chen Hu, Ning Ma, Enmin Huang","doi":"10.21037/jgo-24-792","DOIUrl":"https://doi.org/10.21037/jgo-24-792","url":null,"abstract":"<p><strong>Background: </strong>Cellular senescence is considered a new marker of cancer. It has been suggested that long non-coding RNA (lncRNA) can be used to predict the prognosis of cancers. However, it remains to be seen whether the lncRNAs associated with cellular senescence can be used to predict the prognosis of gastric cancer (GC). The present study aimed to develop a novel senescence-related lncRNA signature (SenLncSig) to predict GC prognosis. The SenLncSig model holds promise for enhancing patient stratification, enabling more precise prognostic predictions and facilitating immunotherapy strategies.</p><p><strong>Methods: </strong>Senescence-associated lncRNAs were identified from RNA expression profiles in The Cancer Genome Atlas (TCGA) database through the construction of a co-expression network linking senescence genes and lncRNAs. A prognostic signature for GC (334 patients from TCGA-STAD data set), comprising the senescence-related lncRNAs, was developed through univariate and multivariate Cox proportional hazards regression analyses. By using the median SenLncSig risk score, the GC patients were categorized into high- and low-risk groups. A Kaplan-Meier analysis and gene set enrichment analysis were conducted, and immune infiltration, the tumor mutation burden (TMB), and pharmacological treatments were compared between the high- and low-risk groups. We used an independent GC cohort (an external cohort of 30 pairs of tumor and non-tumor tissues from the GC patients) and three GC cell lines to conduct a quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis to validate the results.</p><p><strong>Results: </strong>We established a SenLncSig, a prognostic risk model comprising the following five senescence-associated lncRNAs; AP000695.2, LINC02381, AC005586.1, AP003392.1, and AP001528.2. According to the SenLncSig, high-risk scores were associated with poor overall survival (multivariate Cox proportional hazard ratio: 1.498, 95% confidence interval: 1.294-1.735; P<0.001). The time-dependent receiver operating characteristic curve indicated that the model performed (area under the curve: 0.711). We developed a nomogram incorporating age, gender, grade, stage, T stage, M stage, N stage, and SenLncSig risk score to estimate 1-year, 3-year, and 5-year survival rates. Further, according to the results of the mutation analysis, patients with a high TMB in the high-risk group had the worst prognosis. Interestingly, the high-risk group had a stronger infiltration of regulatory T cells (P<0.001) and M2 macrophage cells (P<0.001), as well as higher tumor immune dysfunction and exclusion scores than the low-risk group. These results might explain why the high-risk group had a worse prognosis. Finally, the qRT-PCR validation revealed that the AP000695.2 and AP003392.1 expression levels were significantly higher in the tumor tissues and GC cell lines than the normal tissues and normal human gastric epithelial cell line, ","PeriodicalId":15841,"journal":{"name":"Journal of gastrointestinal oncology","volume":"15 6","pages":"2413-2436"},"PeriodicalIF":2.0,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11732332/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143006467","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}