Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (186)

Search Parameters:
Keywords = gastrointestinal adenocarcinomas

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 1453 KiB  
Case Report
Exosome-Based Therapy for Skin Complications in Oncology Patients Treated with EGFR Inhibitors: A Case Report Highlighting the Need for Coordinated Dermato-Oncologic Care
by Lidia Majewska, Karolina Dorosz and Jacek Kijowski
Pharmaceuticals 2025, 18(8), 1090; https://doi.org/10.3390/ph18081090 - 23 Jul 2025
Cited by 1 | Viewed by 300
Abstract
Patients undergoing epidermal growth factor receptor inhibitor (EGFRI) therapy frequently experience dermatologic side effects, notably papulopustular rash, which impacts 50–90% of recipients. This rash typically appears on the face, chest, and back within weeks of treatment, resembling acne but stemming from distinct pathophysiological [...] Read more.
Patients undergoing epidermal growth factor receptor inhibitor (EGFRI) therapy frequently experience dermatologic side effects, notably papulopustular rash, which impacts 50–90% of recipients. This rash typically appears on the face, chest, and back within weeks of treatment, resembling acne but stemming from distinct pathophysiological mechanisms, causing significant discomfort and reduced quality of life. Prophylactic measures and symptom-based treatment are recommended, emphasizing patient education, topical agents, and systemic therapies for severe cases. A 41-year-old female with advanced colonic mucinous adenocarcinoma developed severe acneiform rash and pruritus during EGFRI therapy with panitumumab. Initial standard treatment with oral doxycycline was discontinued after two days due to severe gastrointestinal intolerance characterized by intense nausea and dyspepsia. With limited access to dermatological consultation, treatment with rose stem cell-derived exosomes (RSCEs) provided rapid symptom relief. Significant improvement was observed within 24 h, with complete resolution of pruritus and substantial reduction in inflammatory lesions within 72 h. RSCEs demonstrate anti-inflammatory effects through the modulation of pro-inflammatory cytokines including interleukin-6, interleukin-1β, and tumor necrosis factor-α, while promoting fibroblast proliferation and collagen synthesis enhancement. They may represent a possible alternative to corticosteroids, avoiding associated side effects such as skin atrophy, delayed wound healing, and local immunosuppression. This case underscores the potential of innovative treatments like RSCEs in managing EGFRI-induced skin complications when standard therapies are not tolerated, particularly in healthcare systems with limited dermato-oncological resources. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

21 pages, 940 KiB  
Review
Immunotherapy in GI Cancers: Lessons from Key Trials and Future Clinical Applications
by Supriya Peshin, Faizan Bashir, Naga Anvesh Kodali, Adit Dharia, Sajida Zaiter, Sakshi Singal and Nagaishwarya Moka
Antibodies 2025, 14(3), 58; https://doi.org/10.3390/antib14030058 - 11 Jul 2025
Cited by 1 | Viewed by 732
Abstract
Immunotherapy has emerged as a transformative approach in gastrointestinal (GI) cancers, addressing historically poor survival rates in advanced-stage disease. Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis demonstrate remarkable efficacy in colorectal cancer with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), [...] Read more.
Immunotherapy has emerged as a transformative approach in gastrointestinal (GI) cancers, addressing historically poor survival rates in advanced-stage disease. Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis demonstrate remarkable efficacy in colorectal cancer with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), exemplified by trials like NICHE-2 achieving exceptional pathological response rates. However, significant limitations persist, including resistance in some dMMR/MSI-H tumors, minimal efficacy in proficient mismatch repair (pMMR) tumors, and low overall response rates across most GI malignancies due to tumor heterogeneity and immune evasion mechanisms. Predictive biomarkers such as tumor mutational burden (TMB) and PD-L1 expression are crucial for optimizing patient selection, while hypermutated pMMR tumors with POLE mutations represent emerging therapeutic opportunities. In pancreatic adenocarcinoma, where survival remains dismal, combination strategies with chemotherapy and novel approaches like cancer vaccines show promise but lack transformative breakthroughs. Esophagogastric cancers benefit from ICIs combined with chemotherapy, particularly in MSI-H and HER2-positive tumors, while hepatocellular carcinoma has achieved significant progress with combinations like atezolizumab–bevacizumab and durvalumab–tremelimumab surpassing traditional therapies. Biliary tract cancers show modest improvements with durvalumab–chemotherapy combinations. Despite these advances, immunotherapy faces substantial challenges including immune-related adverse events, acquired resistance through cancer immunoediting, and the need for biomarker-driven approaches to overcome tumor microenvironment barriers. This review discusses key clinical trials, therapeutic progress, and emerging modalities including CAR T-cell therapies and combination strategies, emphasizing the critical need to address resistance mechanisms and refine precision medicine approaches to fully realize immunotherapy’s potential in GI malignancies. Full article
(This article belongs to the Section Antibody-Based Therapeutics)
Show Figures

Figure 1

9 pages, 319 KiB  
Case Report
Case Report: A Novel Reconstruction Method (Itihaas’s Anastomosis) for Proximal Gastrectomy
by Birendra Kumar Sah, Zhenjia Yu, Chen Li and Zhenggang Zhu
Healthcare 2025, 13(14), 1663; https://doi.org/10.3390/healthcare13141663 - 10 Jul 2025
Viewed by 264
Abstract
This study introduces a novel reconstruction method (Itihaas’s Anastomosis) for the upper gastrointestinal (UGI) tract following proximal gastrectomy, designed to mitigate the severity of acid reflux syndrome, a frequent postoperative complication. The procedure comprises three side-to-side anastomoses: esophago-gastrostomy, gastro-jejunostomy, and jejuno-jejunostomy. The esophago-gastrostomy [...] Read more.
This study introduces a novel reconstruction method (Itihaas’s Anastomosis) for the upper gastrointestinal (UGI) tract following proximal gastrectomy, designed to mitigate the severity of acid reflux syndrome, a frequent postoperative complication. The procedure comprises three side-to-side anastomoses: esophago-gastrostomy, gastro-jejunostomy, and jejuno-jejunostomy. The esophago-gastrostomy anastomosis aims to prevent direct reflux of gastric contents into the esophagus by creating a fundus-like structure, which also facilitates future endoscopic procedures. The gastro-jejunostomy reduces acid reflux by diverting gastric acid to the jejunum for further neutralization, while the jejuno-jejunostomy prevents bile and pancreatic juice reflux into the stomach. A 75-year-old male with adenocarcinoma of the upper stomach underwent this surgical procedure. Postoperative outcomes showed no major complications, with smooth oral contrast passage and no evidence of anastomotic leaks. The patient was discharged after resuming consumption of semi-solid food and experienced no signs of reflux. Itihaas’s Anastomosis represents a novel technical approach that theoretically may reduce acid reflux following proximal gastrectomy. Initial case experience suggests technical feasibility, though all claimed benefits remain theoretical without objective validation. Long-term outcomes, anti-reflux efficacy, and comparative effectiveness require validation through systematic case series with objective assessments. Full article
Show Figures

Figure 1

15 pages, 1190 KiB  
Article
Risk Factors of Multiple Primary Cancers Among Colorectal Cancer Survivors
by Mulugeta Melku, Oliver G. Best, Jean M. Winter, Lauren A. Thurgood, Muktar Ahmed, Ganessan Kichenadasse, Molla M. Wassie and Erin L. Symonds
Cancers 2025, 17(13), 2145; https://doi.org/10.3390/cancers17132145 - 25 Jun 2025
Viewed by 559
Abstract
Background: Colorectal cancer (CRC) is the most common cancer and the leading cause of cancer-related death globally. While survival improved, CRC patients face the risk of subsequent multiple primary cancers (MPCs). This study aimed to determine the incidence and identify risk factors [...] Read more.
Background: Colorectal cancer (CRC) is the most common cancer and the leading cause of cancer-related death globally. While survival improved, CRC patients face the risk of subsequent multiple primary cancers (MPCs). This study aimed to determine the incidence and identify risk factors associated with metachronous MPCs among CRC survivors. Methods: A retrospective analysis was performed on adults diagnosed with invasive colorectal adenocarcinoma at Flinders Medical Centre from 2011 to 2024, who had at least 6 months of post-CRC follow-up. Sociodemographic factors, clinical information, tumour characteristics, and treatment types were collected. Cumulative incidence function and sub-distribution hazard models were used to estimate the incidence and identify risk factors of developing MPCs. Results: Of the total 554 eligible study participants, 12% developed MPC, with a median follow-up time of 5 years (interquartile range: 2.8–7.6 years) until the diagnosis of MPC. Gastrointestinal, prostate, and haematological malignancies were the most common types of MPCs identified. The cumulative incidence and standardised incidence ratio (SIR) of an MPC were 20.9% (95% CI: 15.3–25.6) and 1.32 (95% CI: 1.03–1.68), respectively. Male sex, older age (>65 y), early-stage cancer, and loss of mismatch repair (MMR) protein expression were associated with an increased risk of developing MPCs. Conclusions: CRC survivors have a higher risk of developing an MPC compared to the general population. Sex, age, cancer stage, and MMR protein expression are factors associated with MPCs. Therefore, tailored surveillance based on the individual’s risk profile should be considered for timely diagnosis of subsequent cancers to improve long-term outcomes. Full article
(This article belongs to the Special Issue Advances in Cancer Data and Statistics: 2nd Edition)
Show Figures

Figure 1

30 pages, 30383 KiB  
Technical Note
Dataset and AI Workflow for Deep Learning Image Classification of Ulcerative Colitis and Colorectal Cancer
by Joaquim Carreras, Giovanna Roncador and Rifat Hamoudi
Data 2025, 10(7), 99; https://doi.org/10.3390/data10070099 - 24 Jun 2025
Viewed by 435
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract characterized by the deregulation of immuno-oncology markers. IBD includes ulcerative colitis and Crohn’s disease. Chronic active inflammation is a risk factor for the development of colorectal cancer (CRC). This technical [...] Read more.
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract characterized by the deregulation of immuno-oncology markers. IBD includes ulcerative colitis and Crohn’s disease. Chronic active inflammation is a risk factor for the development of colorectal cancer (CRC). This technical note describes a dataset of histological images of ulcerative colitis, CRC (adenocarcinoma), and colon control. The samples were stained with hematoxylin and eosin (H&E), and immunohistochemically analyzed for LAIR1 and TOX2 markers. The methods used for collecting, processing, and analyzing scientific data, including this dataset, using convolutional neural networks (CNNs) and information about the dataset’s use are also described. This article is a companion to the manuscript “Ulcerative Colitis, LAIR1 and TOX2 Expression, and Colorectal Cancer Deep Learning Image Classification Using Convolutional Neural Networks”. Full article
Show Figures

Figure 1

12 pages, 937 KiB  
Article
Antiviral Activity of Brazilian Propolis from Stingless Bees Against Rotavirus
by Guadalupe González-Ochoa, Ana Paola Balderrama-Carmona, Jesús Antonio Erro-Carvajal, José Guadalupe Soñanez-Organis, Luis Alberto Zamora-Álvarez and Marcelo Andrés Umsza Guez
Microorganisms 2025, 13(6), 1424; https://doi.org/10.3390/microorganisms13061424 - 19 Jun 2025
Viewed by 435
Abstract
Group A rotavirus (RV) causes gastrointestinal disease in infants worldwide, and there is currently no specific treatment to eliminate the virus. Due to its chemical properties, propolis is a promising compound for improving gastrointestinal infections. This study aimed to evaluate the action of [...] Read more.
Group A rotavirus (RV) causes gastrointestinal disease in infants worldwide, and there is currently no specific treatment to eliminate the virus. Due to its chemical properties, propolis is a promising compound for improving gastrointestinal infections. This study aimed to evaluate the action of stingless bee propolis against RV. The method involved determining the concentrations of the extracts that do not exhibit cytotoxicity in colon adenocarcinoma cells using the MTT assay and measuring the reduction in infectivity through a focus forming assay. The results showed that stingless bee propolis was non-cytotoxic up to 200 µg/mL. The reduction in RV infectivity exceeded 99% when using propolis from Plebeia droryana and Melipona quadrifasciata. Brazilian stingless bee propolis, whose active components are known for their activity against various viruses, was experimentally tested and demonstrated effective antiviral activity against RV, supporting its potential application as an antiviral agent. Full article
(This article belongs to the Section Microbial Biotechnology)
Show Figures

Figure 1

13 pages, 995 KiB  
Article
Practical Management of Zolbetuximab Administration: The Project VYLOY Initiative
by Yukiya Narita, Taro Mizuno, Takato Suda, Junko Kurono, Yasunobu Ishizuka, Yumi Iida, Akiko Kondo, Kazuhiro Shimomura, Chisato Yamada, Eri Hotta, Koji Kuraishi, Kanae Tozaki, Makiko Kobara, Chihoko Takahata and Kei Muro
Cancers 2025, 17(12), 1996; https://doi.org/10.3390/cancers17121996 - 15 Jun 2025
Viewed by 1836
Abstract
Background: Zolbetuximab, a monoclonal antibody targeting claudin-18.2 (CLDN18.2), which was recently approved as first-line treatment for advanced gastric cancer (AGC), presents unique safety challenges, particularly infusion-related gastrointestinal toxicity and hypoalbuminemia. This study aimed to present our experience with zolbetuximab administration in patients with [...] Read more.
Background: Zolbetuximab, a monoclonal antibody targeting claudin-18.2 (CLDN18.2), which was recently approved as first-line treatment for advanced gastric cancer (AGC), presents unique safety challenges, particularly infusion-related gastrointestinal toxicity and hypoalbuminemia. This study aimed to present our experience with zolbetuximab administration in patients with AGC, focusing on the safety and management effectiveness of our adapted protocol in routine clinical practice. Methods: This study presents our single-institution real-world experience implementing a proactive management protocol (“Project VYLOY”) using zolbetuximab to mitigate these toxicities. We adopted a standardized stepwise infusion protocol and antiemetic premedication to reduce infusion-related nausea and vomiting. Patients with CLDN18.2-positive advanced gastric or gastroesophageal junction adenocarcinoma who received zolbetuximab combined with chemotherapy were included. Results: Twenty-four patients were included. The median infusion duration was 215 min, with an interruption rate of 25.0%. In cycle 1, 62.5% experienced infusion-associated adverse events, primarily grade 1 nausea (54%) and vomiting (25%). Hypoalbuminemia (grade ≥ 2) occurred in 57% of first-line patients, potentially linked to zolbetuximab-induced gastritis and gastrointestinal protein loss. Proactive antiemetic support and infusion rate adjustments substantially reduced infusion interruptions in subsequent cycles (10.9%). Patients without prior gastrectomy had higher nausea and vomiting rates, confirming the stomach’s role in mediating toxicity. Conclusions: Our results suggest that proactive management can improve the safety and tolerability of zolbetuximab, especially by reducing infusion-related toxicity in real-world practice. Full article
(This article belongs to the Special Issue Advances in Drug Delivery for Cancer Therapy)
Show Figures

Figure 1

12 pages, 758 KiB  
Article
Immunohistochemical TTF-1 and Napsin a Expression in Gastrointestinal Adenocarcinomas—Low Frequency but an Important Pitfall
by Petar Noack, Claudia Grosse, Simon Eschemann, Bastian Dislich and Rupert Langer
Diagnostics 2025, 15(12), 1490; https://doi.org/10.3390/diagnostics15121490 - 11 Jun 2025
Viewed by 619
Abstract
Background/Objectives: TTF-1 and Napsin A are immunohistochemical markers that are widely used for the diagnosis of lung adenocarcinomas or thyroid carcinomas, as well as the characterization of metastases. However, several publications have reported the aberrant expression of one or both markers in [...] Read more.
Background/Objectives: TTF-1 and Napsin A are immunohistochemical markers that are widely used for the diagnosis of lung adenocarcinomas or thyroid carcinomas, as well as the characterization of metastases. However, several publications have reported the aberrant expression of one or both markers in extrathoracic malignancies, including gastrointestinal adenocarcinomas. The goal of our study was to determine the frequency of TTF-1- and Napsin A-positive neoplasms in cohorts consisting of esophageal, gastric and colorectal adenocarcinomas. Methods: Buffered formalin-fixed paraffin-embedded tumor tissues from 854 patients with primary resected gastrointestinal and esophageal carcinomas were placed in tissue microarrays (TMAs) for investigation. Between two and six tumor cores were analyzed for each case. For immunohistochemical staining, we used TTF-1 (SPT24 clone) and Napsin A (IP64 clone). Tumors were considered positive if at least 5% of their tumor cells showed weak nuclear (TTF-1) or cytoplasmic (Napsin A) staining. Results: In total, 16 cases showed positive staining for TTF-1, alongside 7 cases for Napsin A. The greatest proportion of TTF-1- and/or Napsin A-positive tumors was found among esophageal adenocarcinomas (5/125 cases; 4%). Co-expression of TTF-1 and Napsin A was found in five cases, including three esophageal and two gastric adenocarcinomas. In colorectal carcinomas, co-expression of these markers was not detected. Conclusions: TTF-1 and Napsin A are useful immunohistochemical makers for establishing the diagnosis of pulmonary adenocarcinoma. Additionally, knowing that a proportion of gastrointestinal neoplasms express these markers can help to avoid diagnostic misinterpretations. Full article
(This article belongs to the Special Issue Histopathology in Cancer Diagnosis and Prognosis—2nd Edition)
Show Figures

Figure 1

13 pages, 1026 KiB  
Article
A Clinical Validation of a Diagnostic Test for Esophageal Adenocarcinoma Based on a Novel Serum Glycoprotein Biomarker Panel: PromarkerEso
by Jordana Sheahan, Iris Wang, Peter Galettis, David I. Watson, Virendra Joshi, Michelle M. Hill, Richard Lipscombe, Kirsten Peters and Scott Bringans
Proteomes 2025, 13(2), 23; https://doi.org/10.3390/proteomes13020023 - 4 Jun 2025
Viewed by 654
Abstract
Background: Esophageal adenocarcinoma (EAC) diagnosis involves invasive and expensive endoscopy with biopsy, but rising EAC incidence has not been reduced by increased surveillance. This study aimed to develop and clinically validate a novel glycoprotein biomarker blood test for EAC, named PromarkerEso. Methods: Serum [...] Read more.
Background: Esophageal adenocarcinoma (EAC) diagnosis involves invasive and expensive endoscopy with biopsy, but rising EAC incidence has not been reduced by increased surveillance. This study aimed to develop and clinically validate a novel glycoprotein biomarker blood test for EAC, named PromarkerEso. Methods: Serum glycoprotein relative concentrations were measured using a lectin-based magnetic bead array pulldown method, with multiple reaction monitoring mass spectrometry in 259 samples across three independent cohorts. A panel of glycoproteins: alpha-1-antitrypsin, alpha-1-antichymotrypsin, complement C9 and plasma kallikrein, were combined with clinical factors (age, sex and BMI) in an algorithm to categorize the samples by the risk of EAC. Results: PromarkerEso demonstrated a strong discrimination of EAC from the controls (area under the curve (AUC) of 0.91 in the development cohort and 0.82 and 0.98 in the validation cohorts). The test exhibited a high sensitivity for EAC (98% in the development cohort, and 99.9% and 91% in the validation cohorts) and a high specificity (88% in the development cohort, and 86% and 99% in the validation cohorts). PromarkerEso identified individuals with and without EAC (96% and 95% positive and negative predictive values). Conclusions: This less invasive approach for EAC detection with the novel combination of these glycoprotein biomarkers and clinical factors coalesces in a potential step toward improved diagnosis. Full article
Show Figures

Figure 1

11 pages, 223 KiB  
Article
Exploring the Role of SIRT1 Polymorphisms in Colorectal Cancer Risk: A Case–Control Study
by Justyna Klusek, Piotr Lewitowicz, Grażyna Nowak-Starz, Bartosz Witczak, Ruslan Oblap, Dorota Kozieł, Anna Nasierowska-Guttmejer, Jolanta Klusek, Artur Jóźwik, Tomasz Rogula, Kamila Kocańda and Stanisław Głuszek
J. Clin. Med. 2025, 14(11), 3912; https://doi.org/10.3390/jcm14113912 - 2 Jun 2025
Viewed by 477
Abstract
Background: Colorectal cancer (CRC), the most common malignancy of the gastrointestinal tract, is the second leading cause of cancer-related deaths worldwide. In this context, investigating low-penetrance gene variants associated with the increased risk of CRC represents a novel and crucial approach to enhancing [...] Read more.
Background: Colorectal cancer (CRC), the most common malignancy of the gastrointestinal tract, is the second leading cause of cancer-related deaths worldwide. In this context, investigating low-penetrance gene variants associated with the increased risk of CRC represents a novel and crucial approach to enhancing prevention strategies and clinical surveillance. By focusing on these genetic variants, there is potential for more accurate prediction of individual CRC risk, which could contribute to the refinement of current screening and prophylactic programs. The aim of this case–control study was to explore the association between SIRT1 polymorphisms and CRC risk. Methods: We analyzed three SNPs—rs12778366 (T/C), rs3758391 (C/T), and rs7895833 (A/G)—in the promoter region of the SIRT1 gene, which may influence SIRT1 expression and thus play a role in cancer development. Our study included 200 patients with colorectal adenocarcinoma and 115 controls. Genomic DNA was extracted from blood samples, and SIRT1 SNP analysis was performed using the qPCR method and endpoint genotyping. Results: Univariate regression analysis revealed a slightly increased risk of developing CRC in individuals with minor alleles of the analyzed polymorphisms; however, the observed differences were not statistically significant. Conclusions: Although our findings did not reveal statistically significant differences in SIRT1 gene polymorphism frequencies between the CRC group and the control group, we observed a tendency that suggests further investigation in larger cohorts is warranted. This research underscores the importance of understanding low-penetrance genetic factors in CRC, highlighting their potential to inform more personalized and effective prevention strategies. Full article
(This article belongs to the Section Oncology)
16 pages, 803 KiB  
Review
The Role of Microbiota in Upper Gastrointestinal Cancers
by Giovanni Marasco, Luigi Colecchia, Daniele Salvi, Angelo Bruni, Cecilia Capelli, Elton Dajti, Maria Raffaella Barbaro, Cesare Cremon, Vincenzo Stanghellini and Giovanni Barbara
Cancers 2025, 17(10), 1719; https://doi.org/10.3390/cancers17101719 - 21 May 2025
Viewed by 862
Abstract
The gut microbiota significantly impacts the development and progression of upper gastrointestinal (GI) cancers, including esophageal and gastric cancers. Microbial dysbiosis contributes to carcinogenesis through mechanisms such as inflammation, immune modulation, and direct DNA damage. Techniques for sampling oral, esophageal, and gastric microbiota [...] Read more.
The gut microbiota significantly impacts the development and progression of upper gastrointestinal (GI) cancers, including esophageal and gastric cancers. Microbial dysbiosis contributes to carcinogenesis through mechanisms such as inflammation, immune modulation, and direct DNA damage. Techniques for sampling oral, esophageal, and gastric microbiota vary, with standardization being essential for reliable results. Barrett’s esophagus (BE) and esophageal adenocarcinoma (EAC) are associated with an enrichment of Gram-negative bacteria, promoting inflammation and cancer progression. Esophageal squamous cell carcinoma (ESCC) also shows distinct microbial patterns, with reduced diversity and increased harmful bacteria like Porphyromonas gingivalis and Fusobacterium nucleatum. In gastric cancer (GC), Helicobacter pylori (HP) and non-HP gastric microbiota play significant roles, with diverse microbial communities contributing to cancer development through nitrate reduction, immune modulation, and inflammation. Emerging evidence highlights the role of non-HP bacteria in promoting carcinogenesis, with specific taxa like Fusobacterium nucleatum and Lactobacillus influencing tumor growth and immune evasion. Further research is needed to elucidate the complex interactions between gut microbiota and upper GI cancers, paving the way for novel diagnostic and therapeutic approaches. Understanding these microbial dynamics offers potential for microbiota-based interventions, improving the early detection, prognosis, and treatment of upper GI cancers. This comprehensive review summarizes the available evidence on the role of microbiota in upper GI oncology and the need for continued exploration in this field. Full article
(This article belongs to the Special Issue Developments in the Management of Gastrointestinal Malignancies)
Show Figures

Figure 1

24 pages, 31663 KiB  
Review
The Many Faces of Intestinal Tumors in Adults, Including the Primary Role of CT Imaging in Emergencies and the Important Role of Cross-Sectional Imaging: A Pictorial Review
by Barbara Brogna, Francesca Maccioni, Dolores Sgambato, Fabiana Capuano, Lorenzo Iovine, Salvatore Guarino, Lorenzo Di Libero, Alfonso Amendola, Lorenzo Faggioni and Dania Cioni
Healthcare 2025, 13(9), 1071; https://doi.org/10.3390/healthcare13091071 - 6 May 2025
Viewed by 835
Abstract
Background/Objectives: Small bowel tumors (SBTs) encompass a diverse range of tumor types, with benign tumors being the most prevalent. However, the incidence of malignant SBTs is increasing, particularly small bowel adenocarcinoma; this poses a diagnostic challenge for clinicians and radiologists due to the [...] Read more.
Background/Objectives: Small bowel tumors (SBTs) encompass a diverse range of tumor types, with benign tumors being the most prevalent. However, the incidence of malignant SBTs is increasing, particularly small bowel adenocarcinoma; this poses a diagnostic challenge for clinicians and radiologists due to the varied and nonspecific clinical and radiological presentations associated with SBTs. In fact, SBTs can present differently in emergencies, often mimicking inflammatory diseases or manifesting as complications such as intussusception, small bowel obstruction (SBO), intestinal ischemia, perforation, gastrointestinal bleeding, or metastatic disease. These tumors can remain asymptomatic for extended periods. Methods: We present a pictorial review on the role of imaging in evaluating SBTs, focusing on the emergency setting where diagnosis can be incidental. We also include some representative cases that may be useful for radiologists and residents in clinical practice. Results: Despite these challenges, contrast-enhanced computed tomography (CECT) is usually the best modality to use in emergencies for evaluating SBTs, and in some cases, a diagnosis can be made incidentally. However, when possible, multimodal imaging through cross-sectional imaging remains crucial for the non-invasive diagnosis of SBTs in stable patients, as endoscopic procedures may also be impractical. A complementary CT study with distension using negative oral contrast media, such as water, polyethylene glycol, or mannitol solutions, can improve the characterization of SBTs and rule out multiple SBT locations, particularly in small bowel neuroendocrine tumor (NET) and gastrointestinal tumor (GIST) localization. Positive water-soluble iodine-based oral contrast, such as Gastrografin (GGF), can be used to evaluate and monitor the intestinal lumen during the nonsurgical management of small bowel obstruction (SBO) or in suspected cases of small bowel perforations or the presence of fistulas. Magnetic resonance enterography (MRE) can aid in improving the characterization of SBTs through a multiplanar and multisequence study. Positron emission tomography combined with CT is generally an essential modality in evaluating metastatic disease and staging and assessing tumor prognosis, but it has limitations for indolent lymphoma and small NETs. Conclusions: Therefore, the integration of multiple imaging modalities can improve patient management and provide a preoperative risk assessment with prognostic and predictive indicators. In the future, radiomics could potentially serve as a “virtual biopsy” for SBTs, allowing for better diagnosis and more personalized management in precision medicine. Full article
Show Figures

Figure 1

19 pages, 15521 KiB  
Article
Small Bowel Tumors: A 7-Year Study in a Tertiary Care Hospital
by Sergiu Marian Cazacu, Dan Cârțu, Mihai Popescu, Liliana Streba, Bogdan Silviu Ungureanu, Vlad Florin Iovănescu, Mihai Cimpoeru, Cecil Sorin Mirea, Valeriu Marian Surlin, Stelian Mogoantă and Mirela Marinela Florescu
Cancers 2025, 17(9), 1465; https://doi.org/10.3390/cancers17091465 - 27 Apr 2025
Viewed by 525
Abstract
(1) Background: Tumors of the small bowel represent 3–6% of gastrointestinal neoplasms and 3–6% of GI malignancies. The difficulties regarding the diagnosis are associated with larger tumors at the moment of the diagnosis and with advanced forms of malignant tumors, associated with a [...] Read more.
(1) Background: Tumors of the small bowel represent 3–6% of gastrointestinal neoplasms and 3–6% of GI malignancies. The difficulties regarding the diagnosis are associated with larger tumors at the moment of the diagnosis and with advanced forms of malignant tumors, associated with a dismal prognosis. (2) Methods: We performed an observational, retrospective, cohort study that included patients with small bowel tumors admitted to the Craiova County Emergency Clinic Hospital between 1 January 2017 and 31 December 2023. The data were collected from the analysis of the patient’s discharge documents from the Hippocrates computer system of the hospital and the evaluation of endoscopy databases. Patients under 16 years of age, those with no pathological confirmation of the malignancy, and those with insufficient data were excluded. (3) Results: A total of 80 cases of small bowel tumors were diagnosed; 72.5% were malignant, of which 10.3% were metastases. The most frequent primary malignant small bowel tumor was adenocarcinoma; two squamous cell carcinomas were noted. CT scans and upper digestive endoscopy represent the most frequent imaging methods for the diagnosis. The prognosis for malignant tumors was poor, with a 41% 5-year survival rate. (4) Conclusions: Small bowel tumors are rarely encountered, with 72.5% being malignant, and were diagnosed at large dimensions and in advanced stages for malignant tumors, with a dismal prognosis. Full article
Show Figures

Figure 1

17 pages, 260 KiB  
Review
Evolution of Therapeutics for Locally Advanced Upper Gastrointestinal Adenocarcinoma
by Jenny J. Li, Jane E. Rogers, Rebecca E. Waters, Qiong Gan, Mariela Blum Murphy and Jaffer A. Ajani
Cancers 2025, 17(8), 1307; https://doi.org/10.3390/cancers17081307 - 12 Apr 2025
Cited by 1 | Viewed by 751
Abstract
Upper gastrointestinal (GI) malignancies, including esophageal, gastroesophageal junction (GEJ), and gastric adenocarcinomas, remain a major global health concern, with poor overall survival and high recurrence rate despite aggressive treatment. Patients with very early tumors (cT1a) can benefit from endoscopic therapy. However, patients with [...] Read more.
Upper gastrointestinal (GI) malignancies, including esophageal, gastroesophageal junction (GEJ), and gastric adenocarcinomas, remain a major global health concern, with poor overall survival and high recurrence rate despite aggressive treatment. Patients with very early tumors (cT1a) can benefit from endoscopic therapy. However, patients with locally advanced disease require multimodal therapies that may combine surgery, radiation, and systemic therapies. This review provides a comprehensive overview of recent advancements in the treatment of locally advanced upper GI adenocarcinomas. Surgical resection remains the cornerstone of curative treatment, with perioperative chemotherapy emerging as the standard of care. While preoperative chemoradiation has demonstrated some benefits in esophageal and GEJ cancers, recent data suggest a more limited role for radiation going forward. Immunotherapy has shown some promise in both the adjuvant and perioperative settings but has yet to establish definitive survival benefit. The integration of HER2-targeted therapies into treatment regimens for HER2-positive locally advanced gastroesophageal cancers has not yielded significant improvements, underscoring the need for more effective strategies. Ongoing research focuses on better predictive biomarkers, personalized treatment approaches, and potential organ preservation strategies for patients achieving a clinical complete response. Continued advancements in treatment modalities and precision medicine are critical to improving survival for patients with locally advanced upper GI adenocarcinomas. Full article
(This article belongs to the Special Issue Developments in the Management of Gastrointestinal Malignancies)
8 pages, 28576 KiB  
Case Report
An Incidental Finding of Appendiceal Gastrointestinal Stromal Tumor with Abundant Skeinoid Fibers: A Rare Case Report with Insights from a Comprehensive Literature Review
by Yu Liu, Yaomin Chen, Asra Feroze and Zhiyan Fu
Diagnostics 2025, 15(7), 924; https://doi.org/10.3390/diagnostics15070924 - 3 Apr 2025
Viewed by 597
Abstract
Background and Clinical Significance: Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract but are exceedingly rare in the appendix, with only 20 cases reported in the literature. Due to their rarity, clinical behavior, histopathologic features, and management [...] Read more.
Background and Clinical Significance: Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract but are exceedingly rare in the appendix, with only 20 cases reported in the literature. Due to their rarity, clinical behavior, histopathologic features, and management of appendiceal GISTs remain poorly understood. Case Presentation: We present the case of a 74-year-old man who underwent a right hemicolectomy for colonic adenocarcinoma, during which an incidental 1.2 cm appendiceal GIST was discovered. Histopathological examination revealed spindle cell morphology with abundant skeinoid fibers (SF), minimal mitotic activity (<1/50 HPF), and no nuclear atypia. Immunohistochemical staining confirmed positivity for CD117, DOG1, and CD34. The tumor was classified as low risk based on its size and mitotic count, and the patient remained recurrence-free at a 4-month follow-up. Conclusions: Our case expands the limited literature on appendiceal GISTs by demonstrating their histopathological and immunohistochemical features, favorable prognostic outcomes, and potential for incidental detection during surgeries for unrelated conditions. However, additional studies are needed to further elucidate their molecular characteristics and overall clinical behavior. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Back to TopTop