[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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

Journals

Article Types

Countries / Regions

Search Results (155)

Search Parameters:
Keywords = mucinous adenocarcinoma

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 781 KiB  
Review
Comprehensive Overview of Molecular, Imaging, and Therapeutic Challenges in Rectal Mucinous Adenocarcinoma
by Mihaela Berar, Andra Ciocan, Emil Moiș, Luminița Furcea, Călin Popa, Răzvan Alexandru Ciocan, Florin Zaharie, Cosmin Puia, Nadim Al Hajjar, Cosmin Caraiani, Ioana Rusu and Florin Graur
Int. J. Mol. Sci. 2025, 26(2), 432; https://doi.org/10.3390/ijms26020432 - 7 Jan 2025
Viewed by 298
Abstract
Rectal cancer is one of the most frequent malignancies worldwide. The most common histological type is adenocarcinoma, followed by mucinous adenocarcinoma. The outcome is less favorable for the mucinous type, yet the treatment course is the same. The aim of this systematic literature [...] Read more.
Rectal cancer is one of the most frequent malignancies worldwide. The most common histological type is adenocarcinoma, followed by mucinous adenocarcinoma. The outcome is less favorable for the mucinous type, yet the treatment course is the same. The aim of this systematic literature review is to assess existing information in order to improve survival in rectal mucinous adenocarcinoma (RMA) and establish a starting point for future research. A systematic search of PubMed, Google Scholar, and Web of Science online libraries was performed in October 2024, evaluating studies regarding clinicopathological and genetic features in connection with targeted treatment and survival outcomes in RMA, using the terms “rectal cancer”, “rectum”, “mucinous adenocarcinoma”, or a combination of the terms. We selected 23 studies, 10 of them regarding the diagnostic implications and 13 discussing the treatment strategies and prognosis of this histological subtype. There were six studies addressing the imaging aspects, highlighting the distinct features of mucinous histology in MRI. The molecular specifics were detailed in four studies, outlining the molecular footprint. The prognosis and treatment course were addressed in 12 studies. The inflammation index prognosis, complete response to neoadjuvant chemotherapy, and surgical aspects were addressed individually in each study. We encapsulated the molecular and clinicopathological characteristics of RMA, as well as diagnostic and treatment approaches, to establish a baseline of references for the benefit of daily practice and further research. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapies of Colorectal Cancer 3.0)
Show Figures

Figure 1

Figure 1
<p>PRISMA flowchart of the included studies.</p>
Full article ">Figure 2
<p>Subject flowchart of the studies included.</p>
Full article ">
15 pages, 38002 KiB  
Article
Differentiating Sinonasal Tumor Entities with Fluorescein-Enhanced Confocal Laser Endomicroscopy: A Step Forward in Precision Diagnostics
by Nina Wenda, Sebastian Wagner, Kai Fruth, Annette Fisseler-Eckhoff and Jan Gosepath
Cancers 2024, 16(24), 4245; https://doi.org/10.3390/cancers16244245 - 20 Dec 2024
Viewed by 356
Abstract
Abstract: Background/Objectives: Sinonasal malignancies are rare and highly diverse cancers that pose significant diagnostic challenges due to their variable histological features and complex anatomical locations. Accurate diagnosis is critical for guiding treatment, yet conventional methods often require multiple biopsies. This study aimed [...] Read more.
Abstract: Background/Objectives: Sinonasal malignancies are rare and highly diverse cancers that pose significant diagnostic challenges due to their variable histological features and complex anatomical locations. Accurate diagnosis is critical for guiding treatment, yet conventional methods often require multiple biopsies. This study aimed to evaluate the potential of confocal laser endomicroscopy (CLE) for real-time imaging of sinonasal tumors to characterize specific features of different entities and improve diagnostic precision. Methods: Ten patients with various sinonasal malignancies, including squamous cell carcinoma, adenocarcinoma, sinonasal undifferentiated carcinoma, olfactory neuroblastoma, sinonasal mucosal melanoma, and endonasal lymphoma, were examined using CLE during diagnostic endoscopy. CLE images were compared descriptively with histopathological cross-sections to identify unique imaging patterns for each tumor type. Results: CLE was feasible across all cases, with high-quality images obtained despite anatomical challenges in some cases. Characteristic features, such as vascular clusters in undifferentiated carcinoma, mucin-filled bubbles in adenocarcinoma, and small round cells in neuroblastoma, were identified and corresponded well with histopathological findings. CLE also helped guide biopsies by revealing areas with diagnostic relevance. Conclusions: CLE demonstrates promise as an adjunct diagnostic tool in sinonasal malignancies, offering real-time imaging that correlates with histopathological findings and aids in targeted biopsies. While this study provides preliminary insights into the utility of CLE, further research with larger cohorts and statistical validation is necessary to establish its diagnostic reliability and broader clinical application. Full article
(This article belongs to the Special Issue Application of Fluorescence Imaging in Cancer)
Show Figures

Figure 1

Figure 1
<p>Intraoperative setup from the surgeon’s perspective, arranged from left to right. The endoscopic image shows the laser probe positioned on the inferior turbinate, electromagnetic navigation system (Stealth Station<sup>®</sup>, Medtronic, Jacksonville, FL, USA), and confocal laser endomicroscope (Cellvizio<sup>®</sup>, Mauna Kea Technologies, Paris, France).</p>
Full article ">Figure 2
<p>Positioning of the laser probe. (<b>a</b>) Examination of an SCC of the right nasal septum. (<b>b</b>) Examination of an SNMM of the left inferior turbinate.</p>
Full article ">Figure 3
<p>Juxtaposition of CLE and histopathological cross-section in healthy mucosa: (<b>a</b>) CLE image of endonasal mucosa of the inferior turbinate; (<b>b</b>) regular endonasal squamous epithelium with hematoxylin and eosin (H&amp;E) staining.</p>
Full article ">Figure 4
<p>Comparison of CLE and histopathological cross-section respiratory epithelium: (<b>a</b>) CLE image of the endonasal respiratory epithelium of the nasal septum with cross-sections of capillaries. (<b>b</b>) Corresponding respiratory epithelium, also with cross-sections of capillaries in hematoxylin and eosin (H&amp;E).</p>
Full article ">Figure 5
<p>Comparison of CLE and histopathological cross-section in SCC: (<b>a</b>) CLE image of an endonasal SCC with blurry cell borders, irregular cell configuration, and inhomogeneous distribution of fluorescein. (<b>b</b>) Corresponding histopathological cross-section H&amp;E staining. (<b>c</b>,<b>d</b>) Highlighted parallels in both modalities: <b>red:</b> cross-section of capillary, <b>yellow:</b> area of tumor necrosis without contrast enhancement, <b>green:</b> tumor cells with contrast enhancement.</p>
Full article ">Figure 6
<p>Comparison of CLE and histopathological cross-section in AC: (<b>a</b>) CLE image of one of the examined endonasal AC with a highly contrasted cluster of tumor cells with surrounding stromal desmoplasia with irregular cellular architecture and fluorescein leakage. (<b>b</b>) Corresponding histopathological cross-section demonstrating the clustered tumor cells as well. (<b>c</b>) CLE image from another area of the tumor with mucin-filled cells (*).</p>
Full article ">Figure 7
<p>Comparison of CLE and histopathological cross-section in SNUC: (<b>a</b>) CLE image of an endonasal SNUC with an increased number of irregularly shaped, cluster-forming capillaries (<b>*</b>) in contrast to less contrasted stromal desmoplasia (white triangles). (<b>b</b>) Corresponding histopathological cross-section H&amp;E staining.</p>
Full article ">Figure 8
<p>Comparison of CLE and histopathological cross-section in ONB: (<b>a</b>) CLE image of ONB with tumor clusters of small spheric cells (white *). (<b>b</b>) Corresponding histopathological cross-section H&amp;E staining showing the typical small round blue appearance of the tumor cells (white *).</p>
Full article ">Figure 9
<p>Comparison of CLE and histopathological cross-section in SNMM: (<b>a</b>) CLE image of one of the included SNMM with small round cells and a cross-section of a capillary (<b>*</b>). (<b>b</b>) Corresponding histopathological cross-section H&amp;E staining showing the highly similar round cellular shape. The brown melanin pigment is not detected in the CLE image.</p>
Full article ">Figure 10
<p>Comparison of CLE and histopathological cross-section in SL: (<b>a</b>) CLE image of SL with characteristically uniform clusters of spherical cells. (<b>b</b>) Corresponding histopathological cross-section with Ki67 immunostaining. (<b>c</b>) Corresponding histopathological cross-section with CD-20 immunostaining displaying the conformity of the modalities.</p>
Full article ">
15 pages, 2659 KiB  
Article
Bacillus amyloliquefaciens Regulates the Keap1/Nrf2 Signaling Pathway to Improve the Intestinal (Caco-2 Cells and Chicken Jejunum) Oxidative Stress Response Induced by Lipopolysaccharide (LPS)
by Xing Chen, Aijuan Zheng, Shuzhen Li, Zedong Wang, Zhimin Chen, Jiang Chen, Zhiheng Zou, Haijun Liang and Guohua Liu
Antioxidants 2024, 13(12), 1550; https://doi.org/10.3390/antiox13121550 - 17 Dec 2024
Viewed by 606
Abstract
This article aims to investigate the mechanism by which Bacillus amyloliquefaciens alleviates lipopolysaccharide (LPS)-induced intestinal oxidative stress. The study involved two experimental subjects: human colorectal adenocarcinoma (Caco-2) cells and Arbor Acres broiler chickens. The experiment involving two samples was designed with the same [...] Read more.
This article aims to investigate the mechanism by which Bacillus amyloliquefaciens alleviates lipopolysaccharide (LPS)-induced intestinal oxidative stress. The study involved two experimental subjects: human colorectal adenocarcinoma (Caco-2) cells and Arbor Acres broiler chickens. The experiment involving two samples was designed with the same treatment groups, specifically the control (CK) group, lipopolysaccharide (LPS) group, Bacillus amyloliquefaciens (JF) group, and JF+LPS group. In the Caco-2 experiment, we administered 2 μg/mL of LPS and 1 × 106 CFU/mL of JF to the LPS and JF groups, respectively. In the broiler experiment, the LPS group (19–21 d) received an abdominal injection of 0.5 mg/kg BW of LPS, whereas the JF group was fed 1 × 107 CFU/g of JF throughout the entire duration of the experiment (1–21 d). The results indicated the following: (1) JF significantly decreased the DPPH free radical clearance rate and hydrogen peroxide levels (p < 0.05). (2) JF significantly enhanced the total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH Px) activity in Caco-2 cells (p < 0.05), while concurrently reducing malondialdehyde (MDA) content (p < 0.05). (3) Compared to the CK group, JF significantly increased the mRNA expression levels of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), SOD, catalase (CAT), GSH-Px, interleukin-4 (IL-4), interleukin-10 (IL-10), Claudin, Occludin1, zonula occludens-1 (ZO-1), and mucin 2 (MUC2) in Caco-2 cells (p < 0.05), while concurrently reducing the mRNA expression of Kelch-like ECH-associated protein 1 (Keap1), tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-8 (IL-8) (p < 0.05). In comparison to the LPS group, the JF+LPS group demonstrated a significant increase in the mRNA expression of Nrf2, SOD, GSH-Px, and IL-4, as well as Occludin1, ZO-1, and MUC2 in Caco-2 cells (p < 0.05), alongside a decrease in the mRNA expression of Keap1, TNF-α, and IL-1β (p < 0.05). (4) In broiler chickens, the JF group significantly elevated the levels of T-AOC, CAT, and GSH-Px in the jejunum while reducing MDA content (p < 0.05). Furthermore, the CAT level in the JF+LPS group was significantly higher than that observed in the LPS group, and the levels of MDA, TNF-α, and IL-1β were significantly decreased (p < 0.05). (5) In comparison to the CK group, the JF group exhibited a significant increase in Nrf2 levels in the jejunum of broiler chickens (p < 0.05). Notably, the mRNA expression levels of IL-4, IL-10, Claudin, Occludin1, ZO-1, and MUC2 were reduced (p < 0.05), while the mRNA expression levels of Keap1, TNF-α, and IL-1β also showed a decrease (p < 0.05). Furthermore, the mRNA expression levels of Nrf2, Occludin1, ZO-1, and MUC2 in the JF+LPS group were significantly elevated compared to those in the LPS group (p < 0.05), whereas the mRNA expression levels of Keap1 and TNF-α were significantly diminished (p < 0.05). In summary, JF can enhance the intestinal oxidative stress response, improve antioxidant capacity and intestinal barrier function, and decrease the expression of inflammatory factors by regulating the Keap1/Nrf2 signaling pathway. Full article
Show Figures

Figure 1

Figure 1
<p>Cellular antioxidant capacity in Caco-2 cells. (<b>A</b>) DPPH free radical clearance rate; (<b>B</b>) hydrogen peroxide content; (<b>C</b>) T-AOC, total antioxidant capacity; (<b>D</b>) SOD, superoxide dismutase; (<b>E</b>) MDA, malondialdehyde; (<b>F</b>) GSH-Px, glutathione peroxidase. CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 2
<p>mRNA expression of cellular antioxidant in Caco-2 cells. (<b>A</b>) mRNA relative expression (<span class="html-italic">Keap1</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">Nrf2</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">HO-1</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">SOD</span>/<span class="html-italic">GAPDH</span>); (<b>E</b>) mRNA relative expression (<span class="html-italic">CAT</span>/<span class="html-italic">GAPDH</span>); (<b>F</b>) mRNA relative expression (<span class="html-italic">GSH-Px</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 3
<p>mRNA expression of cellular inflammatory factors in Caco-2 cells. (<b>A</b>) mRNA relative expression (<span class="html-italic">TNF-α</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">IL-1β</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">IL-6</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">IL-8</span>/<span class="html-italic">GAPDH</span>); (<b>E</b>) mRNA relative expression (<span class="html-italic">IL-4</span>/<span class="html-italic">GAPDH</span>); (<b>F</b>) mRNA relative expression (<span class="html-italic">IL-10</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 4
<p>mRNA expression of cellular intestinal barrier function in Caco-2 cells. (<b>A</b>) mRNA relative expression (<span class="html-italic">Claudin</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">Occludin1</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">ZO-1</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">MUC2</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 5
<p>Antioxidant index of 21 d broiler jejunum. (<b>A</b>) T-AOC, total antioxidant capacity; (<b>B</b>) SOD, superoxide dismutase; (<b>C</b>) CAT, catalase; (<b>D</b>) MDA, malondialdehyde; (<b>E</b>) GSH-Px, glutathione peroxidase. CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 6.</p>
Full article ">Figure 6
<p>Inflammatory factors of 21 d broiler jejunum. (<b>A</b>) TNF-α, tumor necrosis factor-alpha; (<b>B</b>) IL-1β, interleukin-1β; (<b>C</b>) IL-6, interleukin-6; (<b>D</b>) IL-8, interleukin-8; (<b>E</b>) IL-4, interleukin-4; (<b>F</b>) IL-10, interleukin-10. CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 6.</p>
Full article ">Figure 7
<p>Expression of antioxidant-related mRNA in 21 d broiler jejunum. (<b>A</b>) mRNA relative expression (Keap1/GAPDH); (<b>B</b>) mRNA relative expression (Nrf2/GAPDH); (<b>C</b>) mRNA relative expression (HO-1/GAPDH). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 8
<p>Expression of inflammatory factor-related mRNA in 21 d broiler jejunum. (<b>A</b>) mRNA relative expression (<span class="html-italic">TNF-α</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">IL-1β</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">IL-4</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">IL-10</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">Figure 9
<p>Expression of intestinal barrier function-related mRNA in 21 d broiler jejunum. (<b>A</b>) mRNA relative expression (<span class="html-italic">Claudin</span>/<span class="html-italic">GAPDH</span>); (<b>B</b>) mRNA relative expression (<span class="html-italic">Occludin1</span>/<span class="html-italic">GAPDH</span>); (<b>C</b>) mRNA relative expression (<span class="html-italic">ZO-1</span>/<span class="html-italic">GAPDH</span>); (<b>D</b>) mRNA relative expression (<span class="html-italic">MUC2</span>/<span class="html-italic">GAPDH</span>). CK group, control check group; LPS group, lipopolysaccharide group; JF group, <span class="html-italic">Bacillus amyloliquefaciens</span> group; JF+LPS group, <span class="html-italic">Bacillus amyloliquefaciens</span> + lipopolysaccharide group. * There was a significant difference between the two treatment groups, where * represents <span class="html-italic">p</span> &lt; 0.05, ** represents <span class="html-italic">p</span> &lt; 0.01. Results are presented as the mean and standard error of the mean (SEM), n = 3.</p>
Full article ">
15 pages, 1839 KiB  
Article
Molecular Pathway and Immune Profile Analysis of IPMN-Derived Versus PanIN-Derived Pancreatic Ductal Adenocarcinomas
by Margaret A. Park, Kristyn Gumpper-Fedus, Somashekar G. Krishna, Maria C. Genilo-Delgado, Stephen Brantley, Phil A. Hart, Mary E. Dillhoff, Maria F. Gomez, Toni L. Basinski, Shaffer R. Mok, Anjuli K. Luthra, Jason B. Fleming, Amir Mohammadi, Barbara A. Centeno, Kun Jiang, Aleksandra Karolak, Daniel Jeong, Dung-Tsa Chen, Paul A. Stewart, Jamie K. Teer, Zobeida Cruz-Monserrate and Jennifer B. Permuthadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2024, 25(23), 13164; https://doi.org/10.3390/ijms252313164 - 7 Dec 2024
Viewed by 794
Abstract
Intraductal papillary mucinous neoplasms (IPMN) are commonly detected pancreatic cysts that may transform into pancreatic ductal adenocarcinoma (PDAC). Predicting which IPMNs will progress to PDAC remains a clinical challenge. Moreover, identifying those clinically evident IPMNs for which a surveillance approach is best is [...] Read more.
Intraductal papillary mucinous neoplasms (IPMN) are commonly detected pancreatic cysts that may transform into pancreatic ductal adenocarcinoma (PDAC). Predicting which IPMNs will progress to PDAC remains a clinical challenge. Moreover, identifying those clinically evident IPMNs for which a surveillance approach is best is a dire clinical need. Therefore, we aimed to identify molecular signatures that distinguished between PDAC with and without clinical evidence of an IPMN to identify novel molecular pathways related to IPMN-derived PDAC that could help guide biomarker development. Data from the Oncology Research Information Exchange Network (ORIEN) multi-institute sequencing project were utilized to analyze 66 PDAC cases from Moffitt Cancer Center and The Ohio State University Wexner Medical Center, for which tumor whole transcriptome sequencing datasets were generated. Cases were classified based on whether a tumor had originated from an IPMN (n = 16) or presumably through the pancreatic intraepithelial neoplasia (PanIN) pathway (n = 50). We then performed differential expression and pathway analysis using Gene-Set Enrichment Analysis (GSEA) and Pathway Analysis with Down-weighted Genes (PADOG) algorithms. We also analyzed immune profiles using the Tumor-Immune Microenvironment Deconvolution web portal for Bulk Transcriptomics (TIMEx). Both GSEA and TIMEx indicate that PanIN-derived PDAC tumors enrich inflammatory pathways (complement, hedgehog signaling, coagulation, inflammatory response, apical surface, IL-2/STAT5, IL-6/STAT3, EMT, KRAS signaling, apical junction, IFN-gamma, allograft rejection) and are comparatively richer in almost all immune cell types than those from IPMN-derived PDAC. IPMN-derived tumors were enriched for metabolic and energy-generating pathways (oxidative phosphorylation, unfolded protein response, pancreas beta cells, adipogenesis, fatty acid metabolism, protein secretion), and the most significantly upregulated genes (padj < 0.001) included mucin 2 (MUC2) and gastrokine-2 (GKN2). Further, the metabolic-linked gene signature enriched in the IPMN-derived samples is associated with a cluster of early-stage and long-survival (top 4th quartile) PDAC cases from The Cancer Genome Atlas (TCGA) expression database. Our data suggest that IPMN-derived and PanIN-derived PDACs differ in the expression of immune profiles and metabolic pathways. These initial findings warrant validation and follow-up to develop biomarker-based strategies for early PDAC detection and treatment. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Flowchart of all eligible samples analyzed. Sample types are denoted in red font. A total of 66 samples were included in the analytic dataset (16 IPMN-derived and 50 PanIN-derived tumors). Abbreviations: FFPE, formalin-fixed paraffin-embedded; QC, quality control; MCC, Moffitt Cancer Center; and OSU, The Ohio State University.</p>
Full article ">Figure 2
<p>Expression profiles differ between IPMN- and PanIN-derived tumors. <a href="#ijms-25-13164-f002" class="html-fig">Figure 2</a> shows a volcano plot of differential expression (x-axis) for IPMN-derived versus PanIN-derived tumors. PanIN-derived tumors were used as the reference and thus have negative fold change values.</p>
Full article ">Figure 3
<p>PanIN-derived tumors are enriched for genes in inflammatory pathways, whereas IPMN-derived tumors are enriched for genes in energy production/metabolism pathways. (<b>A</b>,<b>B</b>) Bar graph of the normalized enrichment score (NES) on the x-axis versus the GSEA “Hallmark” pathways (<b>A</b>) or immune cell signatures (<b>B</b>) on the y-axis. Gene sets with a <span class="html-italic">p</span>-value of &gt;0.05 are excluded.</p>
Full article ">Figure 4
<p>IPMN-derived metabolic gene sets cluster in The Cancer Genome Atlas data with Stage I and low-grade tumors: Heatmaps showing patient samples (y-axis) and genes in IPMN-derived and non-IPMN-derived gene sets (x-axis). Hierarchical clustering was performed on gene sets. Patients were annotated based on disease-specific survival (DSSurvival, based on quartiles), stage (AJCC), and grade (1, 2, 3, 4 or not graded (GX)).</p>
Full article ">
17 pages, 15777 KiB  
Article
Carcinoembryonic Antigen Expression in Human Tumors: A Tissue Microarray Study on 13,725 Tumors
by Kristina Jansen, Lara Kornfeld, Maximilian Lennartz, Sebastian Dwertmann Rico, Simon Kind, Viktor Reiswich, Florian Viehweger, Ahmed Abdulwahab Bawahab, Christoph Fraune, Natalia Gorbokon, Andreas M. Luebke, Claudia Hube-Magg, Anne Menz, Ria Uhlig, Till Krech, Andrea Hinsch, Frank Jacobsen, Eike Burandt, Guido Sauter, Ronald Simon, Martina Kluth, Stefan Steurer, Andreas H. Marx, Till S. Clauditz, David Dum, Patrick Lebok, Sarah Minner and Christian Bernreutheradd Show full author list remove Hide full author list
Cancers 2024, 16(23), 4052; https://doi.org/10.3390/cancers16234052 - 3 Dec 2024
Viewed by 817
Abstract
Background/Objectives: Carcinoembryonic antigen (CEA) is a cell-surface glycoprotein serving as a drug target, diagnostic marker, and serum marker for cancer monitoring. However, prevalence data on CEA expression in cancer tissues vary considerably. This study was designed to determine CEA expression in normal and [...] Read more.
Background/Objectives: Carcinoembryonic antigen (CEA) is a cell-surface glycoprotein serving as a drug target, diagnostic marker, and serum marker for cancer monitoring. However, prevalence data on CEA expression in cancer tissues vary considerably. This study was designed to determine CEA expression in normal and neoplastic tissues. Methods: A tissue microarray containing 13,725 samples from 120 different tumor types, as well as 76 different normal tissue types, was analyzed by immunohistochemistry (IHC). Results: CEA was detectable in 65 (54.2%) of 120 tumor categories, including 49 (40.8%) tumor types with at least one strongly positive case. CEA positivity was most common in colorectal adenomas (100%) and carcinomas (98.7%), other gastrointestinal adenocarcinomas (61.1–80.3%), medullary carcinomas of the thyroid (96.3%), pulmonary adenocarcinoma (73.7%), mucinous carcinomas of the ovary (79.8%) and the breast (43.2%), small-cell carcinomas of the lung (64.3%), and urinary bladder (38.9%). CEA overexpression was linked to high tumor grade and invasive growth (p < 0.0001 each) in urinary bladder cancer, and estrogen and HER2 receptor positivity (p ≤ 0.0158) in invasive breast cancer of no special type. In colorectal adenocarcinomas, reduced CEA expression was associated with mismatch repair deficiency (p < 0.0001). Conclusions: The comprehensive list of CEA-positive human tumor types demonstrates that CEA is expressed in a broad range of epithelial neoplasms, many of which might benefit from CEA serum monitoring and anti-CEA therapies. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

Figure 1
<p>CEA immunostaining of normal tissues. A membranous and cytoplasmic CEA staining of variable intensity is seen in surface epithelial cells of the stomach (<b>A</b>), epithelial cells (predominantly at the surface) of the colon (<b>B</b>), goblet cells of the small intestine (<b>C</b>), respiratory epithelial cells (<b>D</b>), Hassal’s corpuscles of the thymus (<b>E</b>), and in superficial cell layers of the squamous epithelium of the cervix uteri (<b>F</b>). CEA staining is absent in tissues from the epidermis of the skin (<b>G</b>) and in the pancreas (<b>H</b>). Original magnifications 10×, spot size 600 μm.</p>
Full article ">Figure 2
<p>CEA immunostaining in cancer. The panels show a strong CEA staining in an adenocarcinoma of the colon (<b>A</b>), an adenocarcinoma of the esophagus (<b>B</b>), a ductal adenocarcinoma of the pancreas (<b>C</b>), a cholangiocellular carcinoma of the liver (<b>D</b>), a small-cell neuroendocrine carcinoma of the lung (<b>E</b>), and an adenocarcinoma of the lung (<b>F</b>). CEA staining is lacking in a malignant mesothelioma of the pleura (<b>H</b>) and a hepatocellular carcinoma in the liver (<b>G</b>). Original magnifications 10×, spot size 600 μm.</p>
Full article ">Figure 3
<p>Ranking order of CEA immunostaining in cancers. Both the percentage of positive cases (blue dots) and the percentage of strongly positive cases (orange dots) are shown.</p>
Full article ">Figure 4
<p>CEA immunostaining and patient prognosis in invasive breast carcinoma of no special type.</p>
Full article ">Figure 5
<p>Comparison of CEA expression with previous works in the literature. An “X” represents the proportion of CEA-positive cancers in the present study, dots indicate the frequencies reported in the literature for comparison: studies with ≤10 tumors analyzed are marked with red dots, studies with 11 to 25 tumors analyzed are marked with yellow dots, and green dots mark studies with &gt;25 tumors analyzed.</p>
Full article ">
16 pages, 7392 KiB  
Article
Pre-Surgical Endoscopic Biopsies Are Representative of Esophageal and Esophago-Gastric Junction Adenocarcinoma Histologic Classes and Survival Risk
by Alessandro Gambella, Roberto Fiocca, Marialuisa Lugaresi, Antonietta D’Errico, Deborah Malvi, Paola Spaggiari, Anna Tomezzoli, Luca Albarello, Ari Ristimäki, Luca Bottiglieri, Elena Bonora, Kausilia K. Krishnadath, Gian Domenico Raulli, Riccardo Rosati, Uberto Fumagalli Romario, Giovanni De Manzoni, Jari Räsänen, Sandro Mattioli, Federica Grillo and Luca Mastracci
Cancers 2024, 16(23), 4045; https://doi.org/10.3390/cancers16234045 - 2 Dec 2024
Viewed by 618
Abstract
Background and Objectives: The Esophageal Adenocarcinoma Study Group Europe (EACSGE) recently proposed a granular histologic classification of esophageal–esophago-gastric junctional adenocarcinomas (EA-EGJAs) based on the study of naïve surgically resected specimens that, when combined with the pTNM stage, is an efficient indicator of prognosis, [...] Read more.
Background and Objectives: The Esophageal Adenocarcinoma Study Group Europe (EACSGE) recently proposed a granular histologic classification of esophageal–esophago-gastric junctional adenocarcinomas (EA-EGJAs) based on the study of naïve surgically resected specimens that, when combined with the pTNM stage, is an efficient indicator of prognosis, molecular events, and response to treatment. In this study, we compared histologic classes of endoscopic biopsies taken before surgical resection with those of the surgical specimen, to evaluate the potential of the EACSGE classification at the initial diagnostic workup. Methods: A total of 106 EA-EGJA cases with available endoscopic biopsies and matched surgical resection specimens were retrieved from five Italian institutions. Histologic classification was performed on all specimens to identify well-differentiated glandular adenocarcinoma (WD-GAC), poorly differentiated glandular adenocarcinoma (PD-GAC), mucinous muconodular carcinoma (MMC), infiltrative mucinous carcinoma (IMC), diffuse desmoplastic carcinoma, diffuse anaplastic carcinoma (DAC), and mixed subtypes. Related risk subgroups (low-risk versus high-risk) were also assessed. The correlations of histologic classes and risk subgroups between diagnostic biopsies and surgical resection specimens were explored with Spearman’s correlation test. Sensitivity, specificity, accuracy, positive predictive value, negative predictive value, true positives, true negatives, false positives, and false negatives were also calculated. Results: A strong positive correlation between biopsies and surgical specimens occurred for both histologic classes (coefficient: 0.75, p < 0.001) and risk subgroups (coefficient: 0.65, p < 0.001). The highest sensitivities and specificities were observed for MMC, IMC, and DAC (100% and 99% for all), followed by WD-GAC (sensitivity 91%, specificity 79%) and PD-GAC (sensitivity 722%, specificity 86%). The low-risk and high-risk groups presented a sensitivity and specificity of 89% and 76% (low-risk) and 76% and 89% (high-risk). Conclusions: The EACSGE histologic classification of EA-EGJAs and associated prognostic subgroups can be reliably assessed on pre-operative diagnostic biopsies. Further studies on larger and more representative cohorts of EA-EGJAs will allow us to validate our findings and confirm if the EA-EGJA biopsy histomorphology and clinical TNM staging will be as efficient as the surgical specimen histomorphology and pTNM in predicting patient prognoses and tailoring personalized therapeutic approaches. Full article
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)
Show Figures

Figure 1

Figure 1
<p>Representative images of EA-EGJA histologic classes. (<b>A</b>) Well-differentiated glandular adenocarcinoma (WD-GAC) showing well-formed glands (10× original magnification); (<b>B</b>) poorly differentiated glandular adenocarcinoma (PD-GAC) showing loss of glandular structure but preserved cell cohesion; scant glandular structures can still be recognized (10× original magnification); (<b>C</b>) mucinous muconodular carcinoma (MMC) showing mucin lakes with floating glandular structure and cluster of cohesive cells (5× original magnification); (<b>D</b>) infiltrative mucinous carcinoma (IMC) with poorly cohesive tumor cells, isolated or in small aggregates, showing signet ring cell features floating in mucin (10× original magnification); (<b>E</b>) diffuse desmoplastic carcinoma (DDC) showing marked desmoplasia with scant, poorly cohesive isolated cells or in small aggregates (10× original magnification); (<b>F</b>) diffuse anaplastic carcinoma (DAC) characterized by poorly cohesive and highly atypical tumor cells (10× original magnification); (<b>G</b>,<b>H</b>) mixed subtype (MXD) showing two or more distinct histologic components (glandular/tubular/papillary and poorly cohesive/signet ring) (5× original magnification).</p>
Full article ">Figure 2
<p>Sankey diagram highlighting the shift of histologic class from biopsies to surgical specimens. DAC: diffuse anaplastic carcinoma; DDC: diffuse desmoplastic carcinoma; IMC: invasive muconodular carcinoma; MIX: mixed adenocarcinoma; MMC: mucinous muconodular carcinoma; PD-GAC: poorly differentiated glandular adenocarcinoma; WD-GAC: well-differentiated glandular adenocarcinoma.</p>
Full article ">Figure 3
<p>Sankey diagram highlighting the shift of survival risk subgroups between the biopsies and surgical specimens.</p>
Full article ">Figure 4
<p>ROC curve for adequate histologic classification based on the number of biopsies with invasive EA-EGJA. Orange line: reference line (line of no-discrimination with an AUC = 0.5); solid dark blue line: ROC curve for 5 biopsies (AUC = 0.503).</p>
Full article ">
16 pages, 1800 KiB  
Article
Thermal Liquid Biopsy: A Promising Tool for the Differential Diagnosis of Pancreatic Cystic Lesions and Malignancy Detection
by Judith Millastre, Sonia Hermoso-Durán, María Ortiz de Solórzano, Nicolas Fraunhoffer, Guillermo García-Rayado, Sonia Vega, Luis Bujanda, Carlos Sostres, Ángel Lanas, Adrián Velázquez-Campoy and Olga Abian
Cancers 2024, 16(23), 4024; https://doi.org/10.3390/cancers16234024 - 30 Nov 2024
Viewed by 563
Abstract
Pancreatic cystic lesions (PCLs) are a heterogeneous group of lesions with increasing incidence, usually identified incidentally on imaging studies (multidetector computed tomography (MDCT), magnetic resonance imaging (MRI), or endoscopic ultrasound (EUS)) [...] Full article
(This article belongs to the Special Issue Developments in the Management of Gastrointestinal Malignancies)
Show Figures

Figure 1

Figure 1
<p>Normalized thermograms of intracystic fluid (ICF) for each pancreatic cystic lesion (PCL) subtype. This figure displays the means and standard deviations of the normalized thermograms of ICF for various PCL subtypes. The thermograms represent the excess heat capacity (C<sub>P</sub>) in arbitrary units (a.u.) as a function of temperature (°C), showing the variability both between and within individuals with each subtype. The black lines represent the mean C<sub>P</sub> values, while the shaded areas indicate the standard deviations, illustrating the range of thermal responses for each subtype of PCL. This graphical representation helps visualize the distinct thermal profiles associated with each type of cystic lesion. Panel (<b>A</b>) represents cysts with confirmed diagnoses and includes thermograms for Serous Cystadenoma (SCA, n = 4), Pseudocyst (PC, n = 1), Walled-off Necrosis (WON, n = 3), Lymphoepithelial cyst/Lymphangioma (LINF, n = 2), Intraductal Papillary Mucinous Neoplasm (IPMN, n = 2), Mucinous Cystic Neoplasm (MCN, n = 2), Simple Mucinous Cyst with High-Grade Dysplasia (SMC-HGD, n = 1), and Pancreatic Ductal Adenocarcinoma (PDAC, n = 4). Panel (<b>B</b>) represents cysts with highly probable diagnoses and includes thermograms for Serous Cystadenoma (SCA, n = 1), Pseudocyst (PC, n = 6), Intraductal Papillary Mucinous Neoplasm (IPMN, n = 5), and Walled-off Necrosis (WON, n = 1).</p>
Full article ">Figure 2
<p>Mean normalized thermograms by area for each group and results of the iTLB1 model for differentiating thermograms of intracystic fluid from non-mucinous and mucinous lesions from PCLs with confirmed diagnosis. In panel (<b>A</b>), the green line represents non-mucinous PCLs, while in panel (<b>B</b>), the red line represents mucinous PCLs. The grey shading indicates the mean ± standard deviation for each group. Panel (<b>C</b>) displays the mean normalized thermograms by area for each group overlapping, with non-mucinous lesions in green and mucinous lesions in red. Black dots represent the temperatures used in the iTLB1 model, with vertical dashed lines marking the temperature range used to train the model (55–85 °C). After, this figure presents the results of the iTLB model obtained for differentiating the thermograms of intracystic fluid (ICF) from non-mucinous lesions (NM-PCLs) and mucinous lesions (M-PCLs). Panel (<b>D</b>) shows the absolute value of the coefficients of the iTLB1 model for each of the predictive temperature pairs. Panel (<b>E</b>) illustrates the median differences in the iTLB1 model results for each group (non-mucinous in green and mucinous in red), with the horizontal line representing the zero cutoff point. Panel (<b>F</b>) shows the area under the ROC curve (AUC) of the iTLB1 model. Panel (<b>G</b>) presents the contingency table for the prediction results (top) and the performance metrics of the iTLB1 model (bottom). Notes: C<sub>P</sub>: excess heat capacity; a.u.: arbitrary units; AUC: area under the curve; iTLB: intelligence Thermal Liquid Biopsy; Acc: accuracy; Sens: sensitivity; Spec: specificity; PPV: positive predictive value; NPV: negative predictive value.</p>
Full article ">Figure 3
<p>iTLB1 model scores for different cystic lesions in patients with confirmed and highly probable diagnoses. Panel (<b>A</b>) presents the scores by cyst type (non-mucinous in green, non-malignant mucinous in black, and malignant mucinous in red). Panel (<b>B</b>) shows the scores based on malignancy (red for malignant and black for non-malignant mucinous lesions). Notes: iTLB: intelligence Thermal Liquid Biopsy; PC: Pseudocyst; SCA: Serous Cystadenoma; LINF: Lymphoepithelial/Lymphangioma; MCN: Mucinous Cystic Neoplasm; IPMN: Intraductal Papillary Mucinous Neoplasm; mIPMN: malignant Intraductal Papillary Mucinous Neoplasm; SMC-HGD: Simple Mucinous Cyst with High-Grade Dysplasia; PDAC: Pancreatic Ductal Adenocarcinoma; ns: not significant.</p>
Full article ">Figure 4
<p>Mean area-normalized thermograms for mucinous group and results of the iTLB2 model for differentiating thermograms of intracystic fluid from benign mucinous and malignant mucinous lesions. In panel (<b>A</b>), the black line represents benign mucinous pancreatic cystic lesions (bM-PCLs), while in panel (<b>B</b>), the red line represents malignant mucinous pancreatic cystic lesions (mM-PCLs). The grey shading indicates the mean ± standard deviation for each group. In panel (<b>C</b>), the mean thermograms of both groups are superimposed, with vertical dashed lines marking the temperature range used to train the classification model (55–85 °C) and blue dots representing the temperatures used in the iTLB2 model. After, this figure presents the results of the iTLB2 model obtained for differentiating the thermograms of intracystic fluid (ICF) from benign mucinous lesions (bM-PCLs) and malignant mucinous lesions (mM-PCLs). Panel (<b>D</b>) shows the absolute value of the coefficients of the iTLB2 model for each of the predictive temperature pairs. Panel (<b>E</b>) illustrates the median differences in the iTLB2 model results for each group (benign mucinous in black and malignant mucinous in red), with the horizontal line representing the zero cutoff point. Panel (<b>F</b>) shows the area under the ROC curve (AUC) of the iTLB2 model. Panel (<b>G</b>) presents the contingency table for the prediction results (top) and the performance metrics of the iTLB2 model (bottom). Notes: CP: excess heat capacity; a.u.: arbitrary units; AUC: area under the curve; iTLB: intelligence thermal liquid biopsy; Acc: accuracy; Sens: sensitivity; Spec: specificity; PPV: positive predictive value; NPV: negative predictive value; bM-PCL: benign mucinous pancreatic cystic lesion; mM-PCL: malignant mucinous pancreatic cystic lesion.</p>
Full article ">
13 pages, 1576 KiB  
Article
Histological Subtypes Might Help Risk Stratification in Different Morphological Types of IPMNs: Back to the Future?
by Giuseppe Anzillotti, Francesca Vespasiano, Chiara Maria Scandavini, Marco Del Chiaro, Asif Halimi, Alessandro Anselmo, Giuseppe Tisone, Carlos Fernández Moro, Zeeshan Ateeb, Urban Arnelo, J.-Matthias Löhr, Ernesto Sparrelid and Roberto Valente
J. Clin. Med. 2024, 13(22), 6759; https://doi.org/10.3390/jcm13226759 - 10 Nov 2024
Viewed by 844
Abstract
Background: Intraductal papillary mucinous neoplasms (IPMNs) display four histological subtypes: gastric foveolar, pancreaticobiliary, intestinal, and oncocytic. All of these subtypes harbor a different risk of cancer development. The clinical impact of these subtypes concerning the occurrence of high-grade dysplasia (HGD)/cancer (C) in specific [...] Read more.
Background: Intraductal papillary mucinous neoplasms (IPMNs) display four histological subtypes: gastric foveolar, pancreaticobiliary, intestinal, and oncocytic. All of these subtypes harbor a different risk of cancer development. The clinical impact of these subtypes concerning the occurrence of high-grade dysplasia (HGD)/cancer (C) in specific morphological types, such as branch-duct (BD), main-duct (MD), and mixed-type (MT) IPMNs, has been less investigated. Hence, our aim was to investigate the prevalence of histological subtypes and their possible association with HGD/C concerning morphologically different IPMNs. Methods: This was a retrospective review of demographics, risk factors, and histological features in a surgical cohort of patients having undergone resection for suspect malignant IPMNs at a high-volume tertiary center from 2007 to 2017. Results: A total of 273 patients were resected for IPMNs from during the study period, of which 188 were included in the final analysis. With sex- and age-adjusted multivariable logistic regression analysis across the entire cohort, gastric foveolar subtypes were associated with a reduced prevalence of HGD/C (OR = 0.30; 0.11–0.81, 95% CI, 95%CI; p = 0.01). With univariable logistic regression analysis, in the BD-IPMN subgroup, the pancreaticobiliary subtype was associated with an increased prevalence of HGD/C (OR = 18.50, 1.03–329.65, 95% CI; p = 0.04). In MD- and MT-IPMNs, the gastric foveolar subtype was associated with a decreased prevalence of HGD/cancer (OR = 0.30, 0.13–0.69, 95% CI; p = 0.004). Conclusions: In MD and MT-IPMNs, the gastric-foveolar subtype is associated with a lower prevalence of HGD/C, possibly identifying in such a high-risk group, a subgroup with more indolent behavior. In BD-IPMNs, the pancreaticobiliary subtype is associated with a higher prevalence of HGD/C, conversely identifying among those patients, a subgroup deserving special attention. Full article
(This article belongs to the Section General Surgery)
Show Figures

Figure 1

Figure 1
<p>Inclusion flow-chart according to CONSORT.</p>
Full article ">Figure 2
<p>Prevalence of histological subtypes in the entire cohort (in %).</p>
Full article ">Figure 3
<p>Prevalence of histological subtypes in BD-IPMNs.</p>
Full article ">Figure 4
<p>Prevalence of histological subtypes in MD/MT-IPMNs.</p>
Full article ">
11 pages, 1827 KiB  
Article
Targeting Human Pancreatic Cancer with a Fluorophore-Conjugated Mucin 4 (MUC4) Antibody: Initial Characterization in Orthotopic Cell Line Mouse Models
by Sunidhi Jaiswal, Kristin E. Cox, Siamak Amirfakhri, Aylin Din Parast Saleh, Keita Kobayashi, Thinzar M. Lwin, Sumbal Talib, Abhijit Aithal, Kavita Mallya, Maneesh Jain, Aaron M. Mohs, Robert M. Hoffman, Surinder K. Batra and Michael Bouvet
J. Clin. Med. 2024, 13(20), 6211; https://doi.org/10.3390/jcm13206211 - 18 Oct 2024
Viewed by 919
Abstract
Background/Objectives: Pancreatic cancer is the third leading cause of death related to cancer. The only possible cure presently is complete surgical resection; however, this is limited by difficulty in clearly defining tumor margins. Enhancement of the visualization of pancreatic ductal adenocarcinoma (PDAC) tumor [...] Read more.
Background/Objectives: Pancreatic cancer is the third leading cause of death related to cancer. The only possible cure presently is complete surgical resection; however, this is limited by difficulty in clearly defining tumor margins. Enhancement of the visualization of pancreatic ductal adenocarcinoma (PDAC) tumor margins using near-infrared dye-conjugated tumor-specific antibodies was pioneered by using anti-CEA, anti-CA19.9, and anti-MUC5AC in orthotopic mouse models of pancreatic cancer. Recently, an antibody to Mucin 4 (MUC4) conjugated to a fluorescent probe has shown promise in targeting colon tumors in orthotopic mouse models. Methods: In the present study, we targeted pancreatic cancer using an anti-MUC4 antibody conjugated to IRDye800 (anti-MUC4-IR800) in orthotopic mouse models. Two pancreatic cancer human cell lines were used, SW1990 and CD18/HPAF. Results: Anti-MUC4-IR800 targeted the two pancreatic cancer cell line tumors in orthotopic mouse models with high tumor-to-pancreas ratios and high tumor-to-liver ratios, with greater targeting seen in SW1990. Conclusions: The present results suggest anti-MUC4-IR800’s potential to be used in fluorescence-guided surgical resection of pancreatic cancer. Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Treatment of Pancreatic Cancer)
Show Figures

Figure 1

Figure 1
<p>Characterization of anti-MUC4-IR800 conjugates. (<b>A</b>) Excitation and emission spectra of the anti-MUC4-IR800 conjugate. (<b>B</b>) SDS gel showing a fluorescent band below 10 kDa for the free IRDye800 and a ~150 kDa band for anti-MUC4-IR800. The dye/protein ratio is 1.42.</p>
Full article ">Figure 2
<p>Orthotopic models of the SW1990 (left panel) and CD18/HPAF human pancreatic cancer cell lines (right panel). (<b>A</b>) NIR and (<b>A’</b>) bright-light images of SW1990 orthotopic nude mouse models labeled with 50 µg anti-MUC4-IR800. (<b>B</b>) Non-specific NIR labeling with 50 µg IgG-IR800 and (<b>B’</b>) bright-light images. (<b>C</b>) Average TPR and TLR of mice treated with 50 µg anti-MUC4-IR800, n = 5. (<b>D</b>) NIR and (<b>D’</b>) bright-light images of CD18/HPAF orthotopic nude mouse models labeled with 50 µg anti-MUC4-IR800. (<b>E</b>) Non-specific NIR labeling with 50 µg IgG-IR800 and (<b>E’</b>) bright light images. (<b>F</b>) Average TPR and TLR of mice treated with 50 µg anti-MUC4-IR800, n = 5. Scale bar: 1 cm. White arrow: pancreatic tumor; blue arrow: normal pancreas; yellow arrow: liver. Error bars represent the standard error.</p>
Full article ">Figure 3
<p>Fluorescence biodistribution of anti-MUC4-IR800 in different organs in (<b>A</b>) SW1990, n = 5 (anti-MUC4-IR800), n = 4 (IgG-IR800), and (<b>B</b>) CD18/HPAF tumor-bearing mice, n = 5 (anti-MUC4-IR800), n = 5 (IgG-IR800), at 72 h post-administration. Error bars represent the standard error. ** <span class="html-italic">p</span>-value = 0.0008. *** <span class="html-italic">p</span>-value = 0.0077.</p>
Full article ">Figure 4
<p>Immunohistochemical staining of MUC4 and IgG isotype control in orthotopic xenografts of pancreatic cancer cell lines: (<b>A</b>) 10× images of SW1990 and CD18/HPAF tumor sections showing MUC4 expression (brown staining); (<b>B</b>) 10× images of SW1990 and CD18/HPAF tumor sections treated with the isotype control, IgG antibody.</p>
Full article ">
17 pages, 6948 KiB  
Article
The Formation of Stable Lung Tumor Spheroids during Random Positioning Involves Increased Estrogen Sensitivity
by Balkis Barkia, Viviann Sandt, Daniela Melnik, José Luis Cortés-Sánchez, Shannon Marchal, Bjorn Baselet, Sarah Baatout, Jayashree Sahana, Daniela Grimm, Markus Wehland, Herbert Schulz, Manfred Infanger, Armin Kraus and Marcus Krüger
Biomolecules 2024, 14(10), 1292; https://doi.org/10.3390/biom14101292 - 12 Oct 2024
Viewed by 1276
Abstract
The formation of tumor spheroids on the random positioning machine (RPM) is a complex and important process, as it enables the study of metastasis ex vivo. However, this process is not yet understood in detail. In this study, we compared the RPM-induced spheroid [...] Read more.
The formation of tumor spheroids on the random positioning machine (RPM) is a complex and important process, as it enables the study of metastasis ex vivo. However, this process is not yet understood in detail. In this study, we compared the RPM-induced spheroid formation of two cell types of lung carcinoma (NCI-H1703 squamous cell carcinoma cells and Calu-3 adenocarcinoma cells). While NCI-H1703 cells were mainly present as spheroids after 3 days of random positioning, Calu-3 cells remained predominantly as a cell layer. We found that two-dimensional-growing Calu-3 cells have less mucin-1, further downregulate their expression on the RPM and therefore exhibit a higher adhesiveness. In addition, we observed that Calu-3 cells can form spheroids, but they are unstable due to an imbalanced ratio of adhesion proteins (β1-integrin, E-cadherin) and anti-adhesion proteins (mucin-1) and are likely to disintegrate in the shear environment of the RPM. RPM-exposed Calu-3 cells showed a strongly upregulated expression of the estrogen receptor alpha gene ESR1. In the presence of 17β-estradiol or phenol red, more stable Calu-3 spheroids were formed, which was presumably related to an increased amount of E-cadherin in the cell aggregates. Thus, RPM-induced tumor spheroid formation depends not solely on cell-type-specific properties but also on the complex interplay between the mechanical influences of the RPM and, to some extent, the chemical composition of the medium used during the experiments. Full article
Show Figures

Figure 1

Figure 1
<p>On the RPM, two-dimensional cultures of different lung carcinoma cell types resulted in different subpopulations. (<b>A</b>) While NCI-H1703 squamous cell carcinoma cells (yellow line, top) mainly formed 3D aggregates after 3 days of rotation (some of the few adherent cells are indicated by yellow arrows), Calu-3 adenocarcinoma cells (orange line, bottom) remained almost completely as a two-dimensional cell layer. (<b>B</b>) Microscopic images of the cell culture after 3 days of dynamic culture on the RPM. The bar chart shows the number of spheroids per microscopic image. Scale bar: 200 µm. Non-parametric Mann–Whitney U test *** <span class="html-italic">p</span> ≤ 0.001. Parts of the figure were drawn using pictures from Biorender.com and Servier Medical Art.</p>
Full article ">Figure 2
<p>Gene expression differences of cell adhesion molecules in two-dimensional cell cultures of NCI-H1703 (orange) and Calu-3 cells (yellow). (<b>A</b>) Relative expression differences in Calu-3 vs. NCI-H1703 cells in a static cell culture after 24 h (<span class="html-italic">n</span> = 5). (<b>B</b>) Expression changes in a dynamic 2D RPM cell culture after 24 h and 72 h (<span class="html-italic">n</span> = 4–5). (<b>C</b>) Immunofluorescence of mucin-1 in a 3-day culture (<span class="html-italic">n</span> = 5 for each condition; one representative picture is shown). Outlines of the nuclei as indicated using DAPI staining depicted as dashed lines. The averaged mean values of mucin-1 fluorescence per cell for each microscopic image are shown in the bar graphs. Scale bar: 10 µm. Non-parametric Mann–Whitney U test * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, <sup>ns</sup> non-significant. Parts of the figure were drawn using pictures from Biorender.com and Servier Medical Art.</p>
Full article ">Figure 3
<p>(<b>A</b>) Spheroid formation of NCI-H1703 and Calu-3 cells in low-adhesion plates within 7 days. The two cell lines show a different spheroid morphology. Mechanical influences (such as pipetting) led to the disintegration of 7-day Calu-3 tumor spheroids. Scale bar: 200 µm. (<b>B</b>) Gene expression changes in spheroid cells compared to 2D culture on RPM after 3 days (<span class="html-italic">n</span> = 5). (<b>C</b>) Immunofluorescence of laminin, β<sub>1</sub>-integrin, mucin-1, E-cadherin, focal adhesion kinase (FAK) and F-actin combination in a 3-day culture (<span class="html-italic">n</span> = 5 for each condition; one representative picture is shown). Nuclei are stained with DAPI (gray). Scale bar: 10 µm. (<b>D</b>) Typical mucin-1 distribution in a Calu-3 spheroid structure around the subaggregates (yellow arrows). Scale bar: 10 µm. Non-parametric Mann–Whitney U test ** <span class="html-italic">p</span> ≤ 0.01, <sup>ns</sup> non-significant. Parts of the figure were drawn using pictures from Biorender.com and Servier Medical Art.</p>
Full article ">Figure 4
<p>(<b>A</b>) Protein levels in the supernatant of static cell cultures of NCI-H1703 and Calu-3 after 24 and 72 h. (<b>B</b>) Effects of RPM cell culture on the content of secreted proteins. (<b>C</b>) Expression changes in NFκB target genes during RPM cell culture (<span class="html-italic">n</span> = 5). The small diagrams in the gray box describe the changes in gene expression in tumor spheroid cells. (<b>D</b>) Immunofluorescence of RelA, STAT3 and p38 in a 3-day culture (<span class="html-italic">n</span> = 4–5 for each condition; one representative picture is shown). Outlines of the nuclei as indicated using DAPI staining depicted as dashed lines. Scale bar: 10 µm. (<b>E</b>) Expression changes in <span class="html-italic">SPP1</span> during RPM cell culture (<span class="html-italic">n</span> = 4–5). Non-parametric Mann–Whitney U test * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, <sup>ns</sup> non-significant. Parts of the figure were drawn using pictures from Biorender.com and Servier Medical Art.</p>
Full article ">Figure 5
<p>(<b>A</b>) Effect of phenol red on the spheroid formation of Calu-3 cells under static and RPM conditions after 72 h. The bar chart shows the spheroid size on the microscopic images in phenol-red-free medium (w/o), medium with phenol red (PR) and medium with 10 nM 17β-estradiol (E2). Scale bar: 200 µm. (<b>B</b>) STITCH v5.0 interaction network of phenol red in human cells. (<b>C</b>) Expression of <span class="html-italic">EGFR</span> and <span class="html-italic">ESR1</span> in two-dimensional-growing Calu-3 cells (left) and in Calu-3 spheroids (right) on the RPM (<span class="html-italic">n</span> = 5). n.e., not expressed. (<b>D</b>) Immunofluorescence of E-cadherin and β<sub>1</sub>-integrin in a 3-day culture (<span class="html-italic">n</span> = 5 for each condition; one representative picture is shown). Nuclei are stained with DAPI (gray). The increased formation of integrin clusters is indicated by yellow arrows. The averaged mean values of E-cadherin fluorescence per cell for each microscopic image are shown in the bar graph. Scale bar: 10 µm. (<b>E</b>) Adherent cell layer of Calu-3 cells after 3 days on an RPM without and with 10 nM 17β-estradiol. Scale bar: 200 µm. (<b>F</b>) Expression of <span class="html-italic">MUC1</span> in Calu-3 cells on the RPM in the presence of phenol red (PR) or 10 nM 17β-estradiol (E2) (<span class="html-italic">n</span> = 3–4). AKT1, Rho family-alpha serine/threonine-protein kinase; ALB, albumin; ESR1, estrogen receptor 1; ERα, estrogen receptor alpha; UGT2B15, UDP glucuronosyltransferase 2 family polypeptide B15. Non-parametric Mann–Whitney U test * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001, <sup>ns</sup> non-significant. Parts of the figure were drawn using pictures from Biorender.com and Servier Medical Art.</p>
Full article ">Figure 6
<p>The current model of spheroid formation on a random positioning machine. (<b>A</b>) The aggregation of a loose accumulation of suspension cells into a compact, stable spheroid takes place in various stages, each of which focuses on different structural proteins. (<b>B</b>) The easy detachment of the cells from the adherent cell layer is dependent on the expression of anti-adhesive mucin-1 (blue). (<b>C</b>) Presumed molecular biological effects of ERα signaling on the formation of spheroids. Arrows indicate activation/increase, T-arrows inhibition/decrease. Dashed lines indicate the relationships that lead to few unstable Calu-3 spheroids in the RPM cell culture. Solid lines indicate the processes that lead to more and more stable Calu-3 spheroids in the presence of phenol red/estrogen on the RPM. ECM, extracellular matrix; ER, estrogen receptor; GFR, growth factor receptor. Parts of the figure were drawn using pictures from Biorender.com and Servier Medical Art.</p>
Full article ">
13 pages, 1417 KiB  
Article
MUC16 Retention after Neoadjuvant Chemotherapy in Pancreatic Ductal Adenocarcinoma
by Kathryn M. Muilenburg, Evie G. Ehrhorn, Madeline T. Olson, Carly C. Isder, Kelsey A. Klute, Geoffrey A. Talmon, Mark A. Carlson, Quan P. Ly and Aaron M. Mohs
Cancers 2024, 16(20), 3439; https://doi.org/10.3390/cancers16203439 - 10 Oct 2024
Viewed by 1021
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Currently, surgical resection is the only potentially curative treatment. Unfortunately, less than 20% of PDAC patients are eligible for surgical resection at diagnosis. In the past few decades, neoadjuvant chemotherapy treatment (NCT) has [...] Read more.
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Currently, surgical resection is the only potentially curative treatment. Unfortunately, less than 20% of PDAC patients are eligible for surgical resection at diagnosis. In the past few decades, neoadjuvant chemotherapy treatment (NCT) has been investigated as a way to downstage PDAC tumors for surgical resection. Fluorescence-guided surgery (FGS) is a technique that can aid in increasing complete resection rates by enhancing the tumor through passive or active targeting of a contrast agent. In active targeting, a probe (e.g., antibody) binds a protein differentially upregulated in the tumor compared to normal tissue. Mucin 16 (MUC16), a transmembrane glycoprotein, has recently been explored as an FGS target in preclinical tumor models. However, the impact of chemotherapy on MUC16 expression is unknown. Methods: To investigate this issue, immunohistochemistry was performed on PDAC patient samples. Results: We found that MUC16 expression was retained after NCT in patient samples (mean expression = 5.7) with minimal change in expression between the matched diagnostic (mean expression = 3.66) and PDAC NCT patient samples (mean expression = 4.5). Conclusions: This study suggests that MUC16 is a promising target for FGS and other targeted therapies in PDAC patients treated with NCT. Full article
(This article belongs to the Special Issue Enhancing Cancer Treatments through Fluorescence-Guided Surgery)
Show Figures

Figure 1

Figure 1
<p>Mucin 16 (MUC16) is expressed in both NCT and non-NCT patient samples. (<b>a</b>) Dot plot with all samples comparing the mean immunoreactive score (IRS) of the NCT, non-NCT, and normal pancreas specimens. Data were analyzed using the Mann–Whitney test. <span class="html-italic">p</span> &lt; 0.05; * 0.01 &lt; <span class="html-italic">p</span> &lt; 0.05; ** 0.001 &lt; <span class="html-italic">p</span> &lt; 0.01. (<b>b</b>) Representative images of the mean MUC16 expression in each group were taken at 20× magnification. Scale bar = 50 µm.</p>
Full article ">Figure 2
<p>MUC16 retention in PDAC patient samples after chemotherapy treatment. (<b>a</b>) Distribution of MUC16 stain intensity in matched diagnostic and NCT samples. Matched samples are indicated by the same color and shape. (<b>b</b>) Representative images of the distribution of MUC16 stain intensity in the matched diagnostic and NCT samples. Scale bar = 50 µm.</p>
Full article ">Figure 3
<p>Differential MUC16 expression between PDAC and matched adjacent tissues. (<b>a</b>–<b>c</b>) MUC16 stain intensity in treated samples compared to matched adjacent tissue. The matched data points are indicated with the same color and shape. The mean line is plotted in the dot plot. Data were analyzed using the Wilcoxon matched pairs test. <span class="html-italic">p</span> &lt; 0.05; *** 0.0001 &lt; <span class="html-italic">p</span> &lt; 0.001. Differential MUC16 expression in (<b>a</b>) all tumors and matched adjacent tissues. (<b>b</b>) NCT samples and matched adjacent tissues. (<b>c</b>) non-NCT samples and matched adjacent tissues. (<b>d</b>) Representative images of the differential MUC16 expression. Images taken at 20× magnification. Scale bar = 50 µm.</p>
Full article ">
17 pages, 7840 KiB  
Article
Expression of Trefoil Factor 1 (TFF1) in Cancer: A Tissue Microarray Study Involving 18,878 Tumors
by Florian Lutz, Soo-Young Han, Seyma Büyücek, Katharina Möller, Florian Viehweger, Ria Schlichter, Anne Menz, Andreas M. Luebke, Ahmed Abdulwahab Bawahab, Viktor Reiswich, Martina Kluth, Claudia Hube-Magg, Andrea Hinsch, Sören Weidemann, Maximilian Lennartz, David Dum, Christian Bernreuther, Patrick Lebok, Guido Sauter, Andreas H. Marx, Ronald Simon, Till Krech, Christoph Fraune, Natalia Gorbokon, Eike Burandt, Sarah Minner, Stefan Steurer, Till S. Clauditz and Frank Jacobsenadd Show full author list remove Hide full author list
Diagnostics 2024, 14(19), 2157; https://doi.org/10.3390/diagnostics14192157 - 28 Sep 2024
Cited by 1 | Viewed by 1101
Abstract
Background/Objectives: Trefoil factor 1 (TFF1) plays a role in the mucus barrier. Methods: To evaluate the prevalence of TFF1 expression in cancer, a tissue microarray containing 18,878 samples from 149 tumor types and 608 samples of 76 normal tissue types was analyzed through [...] Read more.
Background/Objectives: Trefoil factor 1 (TFF1) plays a role in the mucus barrier. Methods: To evaluate the prevalence of TFF1 expression in cancer, a tissue microarray containing 18,878 samples from 149 tumor types and 608 samples of 76 normal tissue types was analyzed through immunohistochemistry (IHC). Results: TFF1 staining was detectable in 65 of 149 tumor categories. The highest rates of TFF1 positivity were found in mucinous ovarian carcinomas (76.2%), colorectal adenomas and adenocarcinomas (47.1–75%), breast neoplasms (up to 72.9%), bilio-pancreatic adenocarcinomas (42.1–62.5%), gastro-esophageal adenocarcinomas (40.4–50.0%), neuroendocrine neoplasms (up to 45.5%), cervical adenocarcinomas (39.1%), and urothelial neoplasms (up to 24.3%). High TFF1 expression was related to a low grade of malignancy in non-invasive urothelial carcinomas of the bladder (p = 0.0225), low grade of malignancy (p = 0.0003), estrogen and progesterone receptor expression (p < 0.0001), non-triple negativity (p = 0.0005) in invasive breast cancer of no special type, and right-sided tumor location (p = 0.0021) in colorectal adenocarcinomas. Conclusions: TFF1 IHC has only limited utility for the discrimination of different tumor entities given its expression in many tumor entities. The link between TFF1 expression and parameters of malignancy argues for a relevant biological role of TFF1 in cancer. TFF1 may represent a suitable therapeutic target due to its expression in only a few normal cell types. Full article
(This article belongs to the Collection Biomarkers in Medicine)
Show Figures

Figure 1

Figure 1
<p>TFF1 immunostaining of normal tissues. The panels show a cytoplasmic staining of surface epithelial cells but not of glands in the stomach (<b>A</b>), subsets of goblet cells in the duodenum (<b>B</b>) and the colon (<b>C</b>), a subset of mucinous cells in the submandibulary gland (<b>D</b>), a subset of luminal epithelial cells and intraluminal mucus in the breast (<b>E</b>), a small subset of urothelial cells (mostly umbrella cells) in the renal pelvis (<b>F</b>), a large subset of urothelial cells in an inflamed urinary bladder (<b>G</b>), and (focally) in epithelial cells of the gallbladder (<b>H</b>).</p>
Full article ">Figure 2
<p>TFF1 immunostaining in cancer. The panels show TFF1 positivity in a mucinous carcinoma of the ovary (<b>A</b>), a colorectal adenocarcinoma (<b>B</b>), a pancreatic adenocarcinoma (<b>C</b>), a gastric adenocarcinoma (<b>D</b>), an adenocarcinoma of the cervix (<b>E</b>), urothelial carcinoma of the bladder (<b>F</b>), and a neuroendocrine tumor of the lung (<b>G</b>). TFF1 staining is absent in a hepatocellular carcinoma (<b>H</b>).</p>
Full article ">Figure 3
<p>Ranking order of TFF1 immunostaining in tumors. Both the percentage of positive cases (blue dots) and the percentage of strongly positive cases (orange dots) are shown.</p>
Full article ">Figure 4
<p>Comparison of TFF1 and MUC5AC immunostaining.</p>
Full article ">Figure 5
<p>Comparison with previous TFF1 literature. An “X” indicates the fraction of TFF1 positive cancers in the present study, dots indicate the reported frequencies from the literature for comparison: red dots mark studies with ≤10 analyzed tumors, yellow dots mark studies with 11 to 25 analyzed tumors, and green dots mark studies with &gt;25 analyzed tumors.</p>
Full article ">
14 pages, 3317 KiB  
Article
Proinflammatory Microenvironment in Adenocarcinoma Tissue of Colorectal Carcinoma
by Slobodan Todorović, Miljan S. Ćeranić, Borislav Tošković, Miloš Diklić, Olivera Mitrović Ajtić, Tijana Subotički, Milica Vukotić, Teodora Dragojević, Emilija Živković, Svetlana Oprić, Miodrag Stojiljkovic, Jasna Gačić, Nataša Čolaković, Bogdan Crnokrak, Vladan P. Čokić and Dragoslava Đikić
Int. J. Mol. Sci. 2024, 25(18), 10062; https://doi.org/10.3390/ijms251810062 - 19 Sep 2024
Viewed by 1005
Abstract
Cancer-promoting proinflammatory microenvironment influences colorectal cancer (CRC) development. We examined the biomarkers of inflammation, intestinal differentiation, and DNA activity correlated with the clinical parameters to observe progression and prognosis in the adenocarcinoma subtype of CRC. Their immunohistology, immunoblotting, and RT-PCR analyses were performed [...] Read more.
Cancer-promoting proinflammatory microenvironment influences colorectal cancer (CRC) development. We examined the biomarkers of inflammation, intestinal differentiation, and DNA activity correlated with the clinical parameters to observe progression and prognosis in the adenocarcinoma subtype of CRC. Their immunohistology, immunoblotting, and RT-PCR analyses were performed in the adenocarcinoma and neighboring healthy tissues of 64 patients with CRC after routine colorectal surgery. Proinflammatory nuclear factor kappa B (NFκB) signaling as well as interleukin 6 (IL-6) and S100 protein levels were upregulated in adenocarcinoma compared with nearby healthy colon tissue. In contrast to nitrotyrosine expression, the oxidative stress marker 8-Hydroxy-2′-deoxyguanosine (8-OHdG) was increased in adenocarcinoma tissue. Biomarkers of intestinal differentiation β-catenin and mucin 2 (MUC2) were inversely regulated, with the former upregulated in adenocarcinoma tissue and positively correlated with tumor marker CA19-9. Downregulation of MUC2 expression correlated with the increased 2-year survival rate of patients with CRC. Proliferation-related mammalian target of rapamycin (mTOR) signaling was activated, and Ki67 frequency was three-fold augmented in positive correlation with metastasis and cancer stage, respectively. Conclusion: We demonstrated a parallel induction of oxidative stress and inflammation biomarkers in adenocarcinoma tissue that was not reflected in the neighboring healthy colon tissue of CRC. The expansiveness of colorectal adenocarcinoma was confirmed by irregular intestinal differentiation and elevated proliferation biomarkers, predominantly Ki67. The origin of the linked inflammatory factors was in adenocarcinoma tissue, with an accompanying systemic immune response. Full article
Show Figures

Figure 1

Figure 1
<p>Markers of intestinal differentiation in healthy and adenocarcinoma tissues of the colon. The immunohistochemistry images correspond to the results of antigen frequency of (<b>A</b>) β-catenin in healthy (n = 4) and adenocarcinoma (n = 12) tissues of the epithelium and stroma of the colon/rectum; (<b>B</b>) mucin 2 (MUC2) in healthy (n = 4) and adenocarcinoma (n = 20) tissues of the colon/rectum; and (<b>C</b>) MUC2 in healthy (H1–H3, n = 4) and adenocarcinoma (C1–C3, n = 20) tissues of the colon/rectum. The scale bars in the lower right corner of the microscopic images correspond to a size of 20 μm. The values are the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. healthy tissue.</p>
Full article ">Figure 2
<p>DNA susceptibility and activity in healthy and adenocarcinoma tissues of the colon. The immunohistochemistry images correspond to the results of antigen frequency of (<b>A</b>) the proliferation marker Ki67 in healthy (n = 6) and adenocarcinoma (n = 16) tissues of the colon/rectum; (<b>B</b>) the marker of oxidative DNA damage 8-hydroxydeoxyguanosine (8-OHdG) in healthy (n = 4) and adenocarcinoma (n = 10) tissues of the colon/rectum; and (<b>C</b>) 8-OHdG in healthy (H1–H3) and adenocarcinoma (C1–C3) tissues of the colon/rectum. The scale bars in the lower right corner of the microscopic images correspond to a size of 20 μm. The values are the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 vs. healthy tissue.</p>
Full article ">Figure 3
<p>Inflammation markers in healthy and adenocarcinoma tissues of the colon. The immunohistochemistry images correspond to the results of antigen frequency of (<b>A</b>) IL-6 in healthy (n = 4) and adenocarcinoma (n = 13) tissues of the colon/rectum; (<b>B</b>) S100 in healthy (n = 4) and adenocarcinoma (n = 13) tissues of the colon/rectum; and (<b>C</b>) S100 in healthy (H1–H3, n = 4) and adenocarcinoma (C1–C3, n = 13) tissues of the colon/rectum. The scale bars in the lower right corner of the microscopic images correspond to a size of 20 μm. The values are the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. healthy tissue.</p>
Full article ">Figure 4
<p>Inflammation- and cancer-related gene expression profiles in cancer and nearby healthy tissues of colorectal adenocarcinoma. qPCR detection of mRNA levels of (<b>A</b>) inflammation-related genes: receptor for advanced glycation end products (RAGE), S100A4, S100A9, S100A12, IL-6, nuclear factor kappa B (NFκB), and mammalian target of rapamycin (mTOR) in healthy (n = 10) and adenocarcinoma (n = 10) tissues of colon/rectum; (<b>B</b>) cancer-related genes: D-Glucuronyl C5-epimerase (GLCE), β- and γ-catenin, matrix metallopeptidase 9 (MMP-9), CD133, Mucin-5AC (MUC5AC), and MUC2 in healthy (n = 10) and adenocarcinoma (n = 10) tissues of colon/rectum; and (<b>C</b>) immunoblotting detection of protein levels of GLCE (n = 30), nitrotyrosine (n = 18), phospho mTOR (pmTOR)/mTOR ratio (n = 22), and pNFκB/NFκB ratio (n = 14) in healthy and cancer tissues of colon/rectum of patients with colorectal adenocarcinoma. (<b>D</b>) Densitometry revealed protein expression, as determined by immunoblotting, and is presented as a ratio to β-actin vs. total protein levels or phosphorylated vs. total proteins. Values are mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 *** <span class="html-italic">p</span> &lt; 0.001 vs. healthy tissue from same patient with colorectal adenocarcinoma.</p>
Full article ">
14 pages, 3364 KiB  
Article
B7H3 Immune Checkpoint Overexpression Is Associated with Decreased Complete Response Rates to Neoadjuvant Therapy in Locally Advanced Rectal Cancer
by Sebastian Curcean, Raluca Maria Hendea, Rares Buiga, Alexandru Tipcu, Andra Curcean, Catalin Vlad, Zsolt Fekete, Alina-Simona Muntean, Daniela Martin and Alexandru Irimie
Diagnostics 2024, 14(18), 2023; https://doi.org/10.3390/diagnostics14182023 - 12 Sep 2024
Viewed by 1001
Abstract
Background and Objectives: Rectal cancer accounts for approximately one-third of colorectal cancers, with over 340,000 deaths globally in 2022. Despite advancements in treatment, the five-year overall survival for locally advanced rectal cancer (LARC) remains at 74%, with significant morbidity. B7H3 (CD276), an immune [...] Read more.
Background and Objectives: Rectal cancer accounts for approximately one-third of colorectal cancers, with over 340,000 deaths globally in 2022. Despite advancements in treatment, the five-year overall survival for locally advanced rectal cancer (LARC) remains at 74%, with significant morbidity. B7H3 (CD276), an immune checkpoint protein, plays a role in tumor progression and resistance to therapy, and correlates with poor prognosis in various cancers, including colorectal cancer. This study aims to evaluate the expression of B7H3 in LARC and its impact on overall complete response (oCR) rates to neoadjuvant therapy. Methods: A retrospective study was conducted on 60 patients with LARC who received neoadjuvant chemoradiation (nCRT) followed by total mesorectal excision (TME). B7H3 expression was assessed using immunohistochemistry on surgical specimens. Expression levels were categorized as high or low based on a composite score, and their association with oCR rates was analyzed. Results: High B7H3 expression was observed in 60% of patients, with 73.5% showing expression in more than 50% of tumor cells. Patients who achieved oCR had significantly lower B7H3 expression compared to those with residual disease (p < 0.001). No nuclear expression of B7H3 was detected. No significant correlation was found between B7H3 expression and other clinicopathological variables, except for a higher likelihood of non-restorative surgery in patients with elevated B7H3 levels (p = 0.049). Mucinous adenocarcinoma had high expression of B7H3. Conclusions: Elevated B7H3 expression is associated with reduced oCR rates in LARC, highlighting its potential role as a prognostic biomarker. Further studies with larger cohorts are warranted to validate these findings and explore B7H3-targeted therapies as a treatment strategy for LARC. Full article
(This article belongs to the Special Issue Diagnosis and Management of Colorectal Lesions)
Show Figures

Figure 1

Figure 1
<p>Roles of B7H3 in cancer (NK, natural killer T lymphocytes) [<a href="#B14-diagnostics-14-02023" class="html-bibr">14</a>,<a href="#B17-diagnostics-14-02023" class="html-bibr">17</a>,<a href="#B18-diagnostics-14-02023" class="html-bibr">18</a>,<a href="#B22-diagnostics-14-02023" class="html-bibr">22</a>,<a href="#B23-diagnostics-14-02023" class="html-bibr">23</a>,<a href="#B24-diagnostics-14-02023" class="html-bibr">24</a>].</p>
Full article ">Figure 2
<p>Representative expression patterns for B7H3 expression in rectal tumors. (<b>A</b>) No cytoplasmic/membrane staining on diagnostic biopsy tissue in a patient who achieved a pCR (scale bar 100 um at 200×). (<b>B</b>) Low cytoplasmic/membrane staining in a patient who received upfront surgery (validation cohort; scale bar 50 um at 400×). (<b>C</b>) Moderate cytoplasmic/membrane staining on a surgical specimen, which underwent nCRT (test cohort; scale bar 50 um at 400×). (<b>D</b>) High cytoplasmic/membrane staining on a patient with mucinous adenocarcinoma of the rectum, who underwent upfront surgery (validation cohort; scale bar 50 um at 400×).</p>
Full article ">Figure 3
<p>B7H3 expression on cytoplasm/membrane and tumor stroma. (<b>A</b>) Percentage of positive stained tumor cells. (<b>B</b>) Distribution of stained area intensity. (<b>C</b>) Distribution of composite score; low expression = composite score &lt;4, high expression = composite score ≥4, composite score = expression level × membranal intensity. (<b>D</b>) Distribution of tumor stroma staining.</p>
Full article ">Figure 4
<p>B7H3 expression in overall responders, incomplete responders, and validation cohort.</p>
Full article ">Figure 5
<p>(<b>A</b>) Median composite score is significantly lower in patients with oCR compared to patients with residual disease. (<b>B</b>) Distribution of percentage positive cells according to oCR.</p>
Full article ">
14 pages, 1688 KiB  
Article
Prognostic and Predictive Significance of Primary Tumor Localization and HER2 Expression in the Treatment of Patients with KRAS Wild-Type Metastatic Colorectal Cancer: Single-Centre Experience from Serbia
by Jelena Radić, Ivan Nikolić, Ivana Kolarov-Bjelobrk, Tijana Vasiljević, Aleksandar Djurić, Vladimir Vidović and Bojana Kožik
J. Pers. Med. 2024, 14(8), 879; https://doi.org/10.3390/jpm14080879 - 20 Aug 2024
Cited by 1 | Viewed by 865
Abstract
The treatment of patients with metastatic colorectal cancer (mCRC) is complex and is impacted by the location of the primary tumor (LPT). Our study aims to emphasize the importance of LPT as a prognostic and predictive marker as well as to examine the [...] Read more.
The treatment of patients with metastatic colorectal cancer (mCRC) is complex and is impacted by the location of the primary tumor (LPT). Our study aims to emphasize the importance of LPT as a prognostic and predictive marker as well as to examine the significance of HER2 overexpression in patients with mCRC, particularly in relation to the response to Epidermal Growth Factor Receptor Antibody treatment (anti-EGFR therapy). In this study, 181 patients with Kirsten RAS (KRAS) wild-type mCRC who received anti-EGFR therapy were included. Among them, 101 had left colon cancer (LCC) and 80 had right colon cancer (RCC). Results demonstrated that patients with KRAS wild-type LCC had better median overall survival (OS) (43 vs. 33 months, p = 0.005) and progression-free survival (PFS) (6 vs. 3 months, p < 0.001) compared to those with RCC. Multivariate analysis identified mucinous adenocarcinoma (p < 0.001), RCC location (p = 0.022), perineural invasion (p = 0.034), and tumors at the resection margin (p = 0.001) as independent predictors of OS, while mucinous adenocarcinoma (p = 0.001) and RCC location (p = 0.004) independently correlated with significantly shorter PFS. In addition, human epidermal growth factor receptor 2 (HER2) positive expression was significantly associated with worse PFS compared to HER2 negative results (p < 0.001). In conclusion, LPT is an important marker for predicting outcomes in the treatment of wild-type mCRC using anti-EGFR therapy, since patients with RCC have a statistically significantly shorter PFS and OS. Further investigation is needed to understand the role of HER2 overexpression in wild-type mCRC, as these patients also exhibit shorter survival. Full article
(This article belongs to the Section Disease Biomarker)
Show Figures

Figure 1

Figure 1
<p>Study design and inclusion criteria for patients with KRAS wt mCRC.</p>
Full article ">Figure 2
<p>Overall survival (<b>a</b>) and progression-free survival (<b>b</b>) of patients with wt KRAS mCRC according to localization of the primary tumor. OS—overall survival; PFS—progression-free survival; LCC—left colon cancer; RCC—right colon cancer.</p>
Full article ">Figure 3
<p>Immunohistochemical evaluation of HER2 antibody in colorectal cancer: (<b>A</b>)—negative tumor cells (Score 0); (<b>B</b>)—weak positivity of cellular membranes (Score 1+); (<b>C</b>)—incomplete membrane staining positivity in more than 10% of tumor cells in some tumor areas (Score 2+); (<b>D</b>)—complete membrane staining in more than 10% of colorectal tumor cells.</p>
Full article ">Figure 4
<p>Overall survival (<b>a</b>) and progression-free survival (<b>b</b>) of patients with wt KRAS mCRC according to the HER2 expression status. OS—overall survival; PFS—progression-free survival.</p>
Full article ">
Back to TopTop