[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
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (12,511)

Search Parameters:
Keywords = cancer selectivity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 6880 KiB  
Review
Targeting of the 8-oxodG Base Excision Repair Pathway for Cancer Therapy
by Anna Piscone, Francesca Gorini, Susanna Ambrosio, Anna Noviello, Giovanni Scala, Barbara Majello and Stefano Amente
Cells 2025, 14(2), 112; https://doi.org/10.3390/cells14020112 - 14 Jan 2025
Viewed by 234
Abstract
Genomic integrity is critical for cellular homeostasis, preventing the accumulation of mutations that can drive diseases such as cancer. Among the mechanisms safeguarding genomic stability, the Base Excision Repair (BER) pathway plays a pivotal role in counteracting oxidative DNA damage caused by reactive [...] Read more.
Genomic integrity is critical for cellular homeostasis, preventing the accumulation of mutations that can drive diseases such as cancer. Among the mechanisms safeguarding genomic stability, the Base Excision Repair (BER) pathway plays a pivotal role in counteracting oxidative DNA damage caused by reactive oxygen species. Central to this pathway are enzymes like 8-oxoguanine glycosylase 1 (OGG1), which recognize and excise 8-oxo-7,8-dihydro-2′-deoxyguanosine (8-oxodG) lesions, thereby initiating a series of repair processes that restore DNA integrity. BER inhibitors have recently been identified as a promising approach in cancer therapy, increasing the sensitivity of cancer cells to radiotherapy and chemotherapy. By exploiting tumor-specific DNA repair dependencies and synthetic lethal interactions, these inhibitors could be used to selectively target cancer cells while sparing normal cells. This review provides a robust reference for scientific researchers, offering an updated perspective on small-molecule inhibitors targeting the 8-oxodG-BER pathway and highlighting their potential role in expanding cancer treatment strategies. Full article
(This article belongs to the Special Issue DNA Damage and Repair for Targeted Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Scheme of 8-oxodG-BER pathway: upon detecting 8-oxodG, OGG1 removes the damaged base and the APE1 endonuclease processes the resulting AP site, generating a single-strand break (SSB) intermediate. If this intermediate is not immediately repaired, PARP1 may recognize and bind to the SSB. However, PARP1 is not essential for accurate repair if the BER pathway is functioning properly. The BER pathway is completed when the DNA polymerase β (POL β) incorporates a single nucleotide, and DNA Ligase 3 (LIG3), along with the scaffold protein XRCC1, seals the nick to complete the repair. If the BER machinery fails to complete ligation, long patch repair is thought to take over.</p>
Full article ">Figure 2
<p>Schematic representation of the BER pathway, visualized as a balanced framework: damage recognition via DNA glycosylases like OGG1 complements repair synthesis and ligation driven by XRCC1, LIG 3, and POL β. Maintaining balance between these forces is crucial for genome stability, while targeted manipulation, by activating (arrow pointing up) damage recognition and inhibiting (arrow pointing down) the repair synthesis, offers a novel strategy to induce cancer cell death through genomic instability.</p>
Full article ">
15 pages, 3920 KiB  
Article
Ferroptosis Inducers Erastin and RSL3 Enhance Adriamycin and Topotecan Sensitivity in ABCB1/ABCG2-Expressing Tumor Cells
by Lalith Perera, Shalyn M. Brown, Brian B. Silver, Erik J. Tokar and Birandra K. Sinha
Int. J. Mol. Sci. 2025, 26(2), 635; https://doi.org/10.3390/ijms26020635 - 14 Jan 2025
Viewed by 195
Abstract
Acquired resistance to chemotherapeutic drugs is the primary cause of treatment failure in the clinic. While multiple factors contribute to this resistance, increased expression of ABC transporters—such as P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance proteins—play significant roles in the [...] Read more.
Acquired resistance to chemotherapeutic drugs is the primary cause of treatment failure in the clinic. While multiple factors contribute to this resistance, increased expression of ABC transporters—such as P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and multidrug resistance proteins—play significant roles in the development of resistance to various chemotherapeutics. We found that Erastin, a ferroptosis inducer, was significantly cytotoxic to NCI/ADR-RES, a P-gp-expressing human ovarian cancer cell line. Here, we examined the effects of both Erastin and RSL3 (Ras-Selected Ligand 3) on reversing Adriamycin resistance in these cell lines. Our results show that Erastin significantly enhanced Adriamycin uptake in NCI/ADR-RES cells without affecting sensitive cells. Furthermore, we observed that Erastin enhanced Adriamycin cytotoxicity in a time-dependent manner. The selective iNOS inhibitor, 1400W, reduced both uptake and cytotoxicity of Adriamycin in P-gp-expressing NCI/ADR-RES cells only. These findings were also confirmed in a BCRP-expressing human breast cancer cell line (MCF-7/MXR), which was selected for resistance to Mitoxantrone. Both Erastin and RSL3 were found to be cytotoxic to MCF-7/MXR cells. Erastin significantly enhanced the uptake of Hoechst dye, a well-characterized BCRP substrate, sensitizing MCF-7/MXR cells to Topotecan. The effect of Erastin was inhibited by 1400W, indicating that iNOS is involved in Erastin-mediated enhancement of Topotecan cytotoxicity. RSL3 also significantly increased Topotecan cytotoxicity. Our findings—demonstrating increased cytotoxicity of Adriamycin and Topotecan in P-gp- and BCRP-expressing cells—suggest that ferroptosis inducers may be highly valuable in combination with other chemotherapeutics to manage patients’ cancer burden in the clinical setting. Full article
Show Figures

Figure 1

Figure 1
<p>Cytotoxicity of ER (<b>A</b>,<b>B</b>) and RSL3 (<b>C</b>,<b>D</b>) in OVCAR-8 (OV-WT), NCI/ADR-RES (OV-R), MCF-7, and MCF7/MXR cells. Cytotoxicity studies were conducted as described in the methods section. ER cytotoxicity was determined following a 72 h drug treatment while RSL3 cytotoxicity was determined following a 24 h drug treatment.</p>
Full article ">Figure 2
<p>Sensitization of ADR (<b>A</b>) and TPT (<b>E</b>) by ER and RSL3 (<b>C</b>–<b>E</b>) and effects of 1400W (<b>B</b>) on ER-mediated cytotoxicity of ADR in ovarian tumor cells (OV-R and OV-WT). *, **, and *** are <span class="html-italic">p</span> values &lt; 0.05, 0.005 and 0.001 against untreated controls. #, ##, and ### are <span class="html-italic">p</span> values &lt; 0.05, 0.005 and 0.001, respectively, and <span>$</span>, &amp;, and &amp;&amp;&amp; are <span class="html-italic">p</span> values &lt; 0.05, and 0.001, respectively, against matched samples of ADR- and TPT-treated samples alone.</p>
Full article ">Figure 3
<p>Sensitization of TPT by ER (<b>A</b>,<b>D</b>) and RSL3 (<b>C</b>,<b>E</b>) and effects of 1400W (<b>B</b>,<b>D</b>) on ER-mediated cytotoxicity of TPT in MCF-7 and MCF-7/MXR breast tumor cells. *, **, and *** are <span class="html-italic">p</span> values &lt; 0.05, 0.005 and 0.001 against untreated controls. #, ##, and ### are <span class="html-italic">p</span> values &lt; 0.05, 0.005 and 0.001, respectively, against matched samples of TPT-treated samples alone. <span>$</span><span>$</span> is <span class="html-italic">p</span> values &lt; 0.005 against ER-treated control.</p>
Full article ">Figure 4
<p>Effects of (<b>A</b>) ER on the uptake/retention of Adriamycin in OV-WT and NCI/ADR-RES cells and (<b>B</b>) effects of Verapamil and 1400W on ER-induced uptake of ADR. ***, <span class="html-italic">p</span> values &lt; 0001 compared to the control. ###, <span class="html-italic">p</span> value&lt; 0.001 compared to ADR alone. ## and <span>$</span><span>$</span> are <span class="html-italic">p</span> values &lt; 0.005 compared to ER + ADR. &amp;&amp; is <span class="html-italic">p</span> value &lt; 0.005 against ADR + ER treated cells.</p>
Full article ">Figure 5
<p>Uptake of Hoechst dye in (<b>A</b>) MCF-7 (<b>B</b>) MCF-7/MXR cells following treatment with different concentrations of ER and (<b>C</b>) effects of 1400W on ER-mediated uptake of the dye in MCF-7/MXR cells. ***, <span class="html-italic">p</span> value &lt; 0.001 compared to untreated control. <span>$</span>, <span class="html-italic">p</span> value &lt; 0.05 compared to ER 1.0 µM alone. ##, ###, <span class="html-italic">p</span> value &lt; 0.005 and &lt;0.001 compared to untreated control and ER 5.0 µM alone, respectively, and &amp;&amp;, &amp;&amp;&amp;, <span class="html-italic">p</span> value &lt; 0.005 and &lt;0.001 compared to ER, 5 µM, and 10 µM, respectively and +++ <span class="html-italic">p</span> values &lt; 0.001 compared to ER, 5 µM alone.</p>
Full article ">Figure 6
<p>Western blots (<b>A</b>) for P-170 in OVCAR-8 (WT) and NCI/ADR-RES (R) cells and (<b>B</b>) for BCRP in MCF-7/MXR and MCF-7 cells following treatment with different concentrations of Erastin for 4 h or 24 h.</p>
Full article ">Figure 7
<p>Docking of (<b>A</b>) Erastin and (<b>B</b>) Adriamycin in P-gp (ABCB1, pdb ID: 6C0V.pdb) ER and ADR are shown in solid spheres, and the proteins are represented by ribbon diagrams. Residues of the protein that are in contact or forming H-bonds are shown in righthand panels.</p>
Full article ">Figure 8
<p>Docking of (<b>A</b>) Erastin and (<b>B</b>) Topotecan in BCRP (ABCG2, pdb ID: 6hbu.pdb). ER and TPT are shown in solid spheres, and the proteins are represented by ribbon diagrams. Residues of the protein that are in contact or forming H-bonds are shown in righthand panels.</p>
Full article ">
24 pages, 2153 KiB  
Article
The Genetic Expression Difference of A2058 Cells Treated by Plasma Direct Exposure and Plasma-Treated Medium and the Appropriate Treatment Strategy
by Chao-Yu Chen, Chung-Hsien Chou and Yun-Chien Cheng
Biomedicines 2025, 13(1), 184; https://doi.org/10.3390/biomedicines13010184 - 13 Jan 2025
Viewed by 197
Abstract
Background/Objectives: Cold atmospheric plasma (CAP) has been demonstrated as an adjustable device to generate various combinations of short-lived reactive oxygen and nitrogen species (RONS) and as a promising appliance for cancer therapy. This study investigated the effects of direct and indirect treatments [...] Read more.
Background/Objectives: Cold atmospheric plasma (CAP) has been demonstrated as an adjustable device to generate various combinations of short-lived reactive oxygen and nitrogen species (RONS) and as a promising appliance for cancer therapy. This study investigated the effects of direct and indirect treatments of Argon-based CAP to cancer cells (A2058, A549, U2OS and BCC) and fibroblasts (NIH3T3 and L929) on cell viability. We also aimed to understand whether plasma-generated RONS were involved in this process using genetic evidence. Methods: The intensity of reactive species in the plasma gas and the concentrations of RONS in phosphate-buffered saline (PBS) and cell culture medium were measured. A viability assay was performed after the cells were treated by plasma in PBS and medium with various volumes to realize the lethal effects of plasma under different conditions. Diverse cells were treated in the same solution to compare the sensitivities of different cells to plasma treatments. The gene expression profiles of A2058 cells after the direct and indirect treatments were analyzed by next generation gene sequencing. Accordingly, we discovered the advantages of sequential treatments on cancer therapy. Results: The cumulative concentration of hydroxyterephthalic acid (HTA) revealed that the pre-existing OH radical (•OH) in PBS increased with the treatment durations. However, there was no significant increase in the concentration of HTA in culture medium. HTA was detected in the treatment interface of PBS but not medium, showing the penetration of •OH through PBS. The concentrations of H2O2 and NO2 increased with the treatment durations, but that of NO3 was low. The direct treatments caused stronger lethal effects on cancer cells under certain conditions. The fibroblasts showed higher tolerance to plasma treatments. From gene expression analysis, the initial observations showed that both treatments influenced transcription-related pathways and exhibited shared or unique cellular stress responses. The pre-treatments, especially of direct exposure, revealed better cancer inhibition. Conclusions: The anti-cancer efficiency of plasma could be enhanced by pre-treatments and by adjusting the liquid interfaces to avoid the rapid consumption of short-lived RONS in the medium. To achieve better therapeutic effects and selectivity, more evidence is necessary to find optional plasma treatments. Full article
16 pages, 4528 KiB  
Article
SORL1-Mediated EGFR and FGFR4 Regulation Enhances Chemoresistance in Ovarian Cancer
by Ziyan Jiang, Fangfang Bi, Zhiping Ge, Miranda Mansolf, Tobias M. P. Hartwich, Viktoriia Kolesnyk, Kevin Yang, Wonmin Park, Dongin Kim, Olga Grechukhina, Pei Hui, Sang Wun Kim and Yang Yang-Hartwich
Cancers 2025, 17(2), 244; https://doi.org/10.3390/cancers17020244 - 13 Jan 2025
Viewed by 290
Abstract
Recurrent tumors that are resistant to conventional chemotherapy are a major challenge of ovarian cancer treatment. A better understanding of the underlying molecular mechanisms of chemoresistance is critical for developing more effective targeted therapies for ovarian cancer. In this study, we analyzed the [...] Read more.
Recurrent tumors that are resistant to conventional chemotherapy are a major challenge of ovarian cancer treatment. A better understanding of the underlying molecular mechanisms of chemoresistance is critical for developing more effective targeted therapies for ovarian cancer. In this study, we analyzed the transcriptomic profiles of thirteen pairs of matching primary and recurrent ovarian cancers to identify genes that were upregulated in the recurrent tumors. Among these genes, we identified sortilin-related receptor 1 (SORL1) and its role in promoting carboplatin resistance through regulating the stability of epidermal growth factor receptor (EGFR) and fibroblast growth receptor 4 (FGFR4) using ovarian cancer models in vitro and in vivo. We further identified that an anti-SORL1 antibody inhibited the pro-tumor functions of SORL1. Our data showed that a selective inhibitor of FGFR4, FGF401, can improve the therapeutic efficacy of carboplatin in a xenograft mouse model of ovarian cancer. This study has demonstrated the therapeutic potential of targeting the SORL1/FGFR4 pathway to improve the chemoresponse of patients with recurrent and/or resistant ovarian cancer. Full article
(This article belongs to the Special Issue Gynecologic Cancer: From Diagnosis to Treatment)
Show Figures

Figure 1

Figure 1
<p>The increased expression of SORL1 is associated with the resistance to carboplatin in ovarian cancer. (<b>A</b>) A volcano plot of the data generated from the Human Transcriptome Array of the paired recurrent tumors versus primary ovarian cancers. n = 15. The genes of which expression was downregulated in the recurrent tumors (<span class="html-italic">p</span> &lt; 0.01 and 0 &lt; fold change &lt; 1) were labeled as blue dots. The genes of which expression was upregulated (<span class="html-italic">p</span> &lt; 0.01 and fold change &gt; 1) were labeled as red dots. The gray dots indicate genes of which expression was upregulated or downregulated, but the <span class="html-italic">p</span>-value was larger than the cutoff. The genes of which expression was not changed in the recurrent tumors versus the primary tumors (fold change = 1) were not shown in this graph. (<b>B</b>) QPCR of the target genes in five ovarian cancer cell lines (OVCAR8, A2780, KRCH31, R127, and R1820) that were untreated or treated with 100 μM carboplatin for 48 h and recovered for 24 h. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005. (<b>C</b>) A western blot of SORL1 protein in ovarian cancer cell lines that were untreated or treated with 100 μM carboplatin for 48 h and recovered for 24 h. (<b>D</b>) RNA-sequencing data of SORL1 in ovarian cancer samples (n = 418, TCGA dataset) and normal ovary (n = 93, the GTEx dataset). **** <span class="html-italic">p</span> &lt; 0.0001. (<b>E</b>) Kaplan–Meier curves show the comparison of patient survival between SORL1-high (n = 209) and SOLR-low (n = 164) cases of ovarian cancer in the TCGA dataset. (<b>F</b>) A western blot of SORL1 protein in ovarian cancer cell lines transfected with SORL1 or control plasmids. (<b>G</b>) The cell growth curves of ovarian cancer cell lines overexpressing SORL1 or control plasmids. * <span class="html-italic">p</span> &lt; 0.05, # <span class="html-italic">p</span> &lt; 0.001. (<b>H</b>) Caspase-3/7 activity of ovarian cancer cell lines transfected with SORL1 or control plasmids after treatment of carboplatin for 48 h. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>SORL1 knockdown inhibits cell proliferation and improves sensitivity of ovarian cancer cell lines to carboplatin. (<b>A</b>) Western blot of SORL1 protein in OVCAR8 ovarian cancer cells transfected with control esiRNA (control) or SORL1-targeting esiRNAs (esiRNA-SORL1) at 48 h post transfection. (<b>B</b>) Cell growth curves of OVCAR8 cells. (<b>C</b>) Western blot of SORL1 protein in KRCH31 cells transfected with control or SORL1-targeting esiRNAs. (<b>D</b>) Cell growth curves of KRCH31 cells. (<b>E</b>) Cell growth curves of OVCAR8 and KRCH31 cells treated with carboplatin. (<b>F</b>) Distribution of OVCAR8 cells in different phases of cell cycle. (<b>G</b>) Caspase-3/7 activity of OVCAR8 and KRCH31 cells treated with 100 μM carboplatin. Two-way ANOVA, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005, **** <span class="html-italic">p</span> &lt; 0.0001, # <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>SORL1 is involved in regulation of EGFR and FGFR4 signaling. (<b>A</b>,<b>B</b>) Cell viability of OVCAR8 (<b>A</b>) or KRCH31 (<b>B</b>) cells treated with ten different cytokines or growth factors. OVCAR8 or KRCH31 cells stably expressed control shRNA (control-shRNA) or shRNA targeting SORL1 (shRNA-SORL1). NT, untreated control. Red dotted lines indicate levels of cell viability in NT groups. Two-way ANOVA, * <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. (<b>C</b>) Western blot images of OVCAR8 and KRCH31 cells with overexpression of knockdown (shRNA) of SORL1. (<b>D</b>) Co-immunoprecipitation (co-IP) of SORL1 and EGFR or FGFR4 using protein lysate of KRCH31 cells. (<b>E</b>) Proximity Ligation Assay (PLA) of SORL1 and EGFR or FGFR4. Scale bar = 50 μm.</p>
Full article ">Figure 4
<p>Treatment with anti-SORL1 antibody reduces viability of ovarian cancer cells. (<b>A</b>) Schematic description of domain in SORL1 protein and targeting region of anti-SORL1 antibody (AA1220-1337). (<b>B</b>) Western blot of OVCAR8 cells treated with anti-SORL1 antibody (Ab) in comparison to untreated control. (<b>C</b>) Cell viability of OVCAR8 and KRCH31 cells treated with different concentrations of anti-SORL1 Ab. One-way ANOVA, * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001. (<b>D</b>) Cell viability of OVCAR8 and KRCH31 cells treated with anti-SORL1 Ab (10 μg/mL) in combination with carboplatin (40 μM). Two-way ANOVA, * <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; 0.005, **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>SORL1 knockdown in ovarian cancer cells inhibits tumor growth in vivo. (<b>A</b>) Images of tumors dissected from mice injected with KRCH31 cells expressing lentiviral control vector or lentiviral shRNA targeting SORL1 (shRNA-SORL1). (<b>B</b>) Tumor growth curves (n = 5 mice/group). Two-way ANOVA. <span class="html-italic">p</span> &lt; 0.001. (<b>C</b>) Survival curves of mice. (<b>D</b>) Western blot of tumor samples collected from mice. (<b>E</b>) Representative images of H&amp;E staining using tumors from mice. Scale bar = 100 μm.</p>
Full article ">Figure 6
<p>FGFR4 inhibitors have potential to increase sensitivity to carboplatin in ovarian cancer cells. (<b>A</b>) Cell viability of OVCAR8 and KRCH31 cells treated with carboplatin or FGFR4 inhibitors (FGF401 and fisogatinib). (<b>B</b>) Cell viability of OVCAR8 and KRCH31 cells treated with combination or single treatment of carboplatin (50 μM) and/or FGFR4 inhibitors. Two-way ANOVA, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005, **** <span class="html-italic">p</span> &lt; 0.001. ns, not significant. (<b>C</b>) Representative images of tumors collected from mice treated with combination or single treatment of carboplatin (5 mg/kg, IP) and/or FGF401 (3 mg/kg, IP). (<b>D</b>) Tumor growth curves. (n = 5 mice/group).</p>
Full article ">
46 pages, 9390 KiB  
Review
Volatolomics for Anticipated Diagnosis of Cancers with Chemoresistive Vapour Sensors: A Review
by Abhishek Sachan, Mickaël Castro and Jean-François Feller
Chemosensors 2025, 13(1), 15; https://doi.org/10.3390/chemosensors13010015 - 13 Jan 2025
Viewed by 180
Abstract
The anticipated diagnosis of cancers and other fatal diseases from the simple analysis of the volatiles emitted by the body (volatolome) is getting closer and closer from becoming reality. The promises of vapour sensor arrays are to provide a rapid, reliable, non-invasive and [...] Read more.
The anticipated diagnosis of cancers and other fatal diseases from the simple analysis of the volatiles emitted by the body (volatolome) is getting closer and closer from becoming reality. The promises of vapour sensor arrays are to provide a rapid, reliable, non-invasive and ready-to-use method for clinical applications by making an olfactive fingerprint characteristic of people’s health state, to increase their chance of early recovery. However, the different steps of this complex and ambitious process are still paved with difficulties needing innovative answers. The purpose of this review is to provide a statement of the blocs composing the diagnostic chain to identify the improvements still needed. Nanocomposite chemo-resistive transducers have unique prospects to enhance both the selectivity and sensitivity to volatile biomarkers. The variety of their formulations offers multiple possibilities to chemical functionalization and conductive architectures that should provide solutions to discriminations and stability issues. A focus will be made on the protocols for the collection of organic volatile compounds (VOC) from the body, the choice of vapour sensors assembled into an array (e-nose), in particular, chemo-resistive vapour sensors, their principle, fabrication and characteristics, and the way to extract pertinent features and analyse them with suitable algorithms that are able to find and produce a health diagnosis. Full article
20 pages, 732 KiB  
Systematic Review
Hypericin-Mediated Photodynamic Therapy for Head and Neck Cancers: A Systematic Review
by Jakub Fiegler-Rudol, Natalia Zięba, Radosław Turski, Maciej Misiołek and Rafał Wiench
Biomedicines 2025, 13(1), 181; https://doi.org/10.3390/biomedicines13010181 - 13 Jan 2025
Viewed by 222
Abstract
Background: Conventional treatments for cancers of the head and neck region are often associated with high recurrence rates and impaired quality of life. Photodynamic therapy (PDT) has emerged as a promising alternative, leveraging photosensitizers such as hypericin to selectively target tumour cells [...] Read more.
Background: Conventional treatments for cancers of the head and neck region are often associated with high recurrence rates and impaired quality of life. Photodynamic therapy (PDT) has emerged as a promising alternative, leveraging photosensitizers such as hypericin to selectively target tumour cells with minimal damage to surrounding healthy tissues. Objectives: We aimed to evaluate the efficacy and underlying mechanisms of hypericin-mediated PDT (HY-PDT) in treating head and neck cancers. Methods: Adhering to PRISMA 2020 guidelines, a systematic search was conducted across PubMed/Medline, Embase, Scopus, and the Cochrane Library for studies published between January 2000 and December 2024. Inclusion criteria encompassed preclinical in vitro and in vivo studies and clinical trials focusing on HY-PDT for head and neck malignancies and its subtypes. Results: A total of 13 studies met the inclusion criteria, comprising both in vitro and in vivo investigations. HY-PDT consistently demonstrated significant cytotoxicity against squamous cell carcinoma cells through apoptotic and necrotic pathways, primarily mediated by ROS generation. Hypericin exhibited selective uptake in cancer cells over normal keratinocytes. Additionally, HY-PDT modulated the tumour microenvironment by altering cytokine profiles, such as by increasing IL-20 and sIL-6R levels, which may enhance antitumor immunity and reduce metastasis. Conclusions: HY-PDT emerges as a highly promising and minimally toxic treatment modality for head and neck cancers, demonstrating efficacy in inducing selective tumour cell death and modulating the immune microenvironment. Despite the encouraging preclinical evidence, significant methodological variability and limited clinical data necessitate further large-scale, standardized and randomized controlled trials. Full article
Show Figures

Figure 1

Figure 1
<p>Prisma 2020 flow diagram [<a href="#B20-biomedicines-13-00181" class="html-bibr">20</a>].</p>
Full article ">
9 pages, 1825 KiB  
Proceeding Paper
NevusCheck: A Dysplastic Nevi Detection Model Using Convolutional Neural Networks
by Andreluis Ingaroca-Torres, Lucía Heredia-Moscoso and Alvaro Aures-García
Eng. Proc. 2025, 83(1), 11; https://doi.org/10.3390/engproc2025083011 - 13 Jan 2025
Viewed by 125
Abstract
Dysplastic nevi are skin lesions that have distinctive clinical features and are considered risk markers for the development of melanoma, the deadliest type of skin cancer. A specific deep learning technique to identify diseases is convolutional neural networks (CNNs) because of their great [...] Read more.
Dysplastic nevi are skin lesions that have distinctive clinical features and are considered risk markers for the development of melanoma, the deadliest type of skin cancer. A specific deep learning technique to identify diseases is convolutional neural networks (CNNs) because of their great capacity to extract features and classify objects. Therefore, the research aims to develop a model to diagnose dysplastic nevi using a deep learning network whose classification is based on the pre-trained architecture EfficientNet-B7, which was selected for its high classification accuracy and low computational complexity. As for the results obtained, an accuracy of 78.33% was achieved in the classification model. Also, the degree of similarity between the detection by a dermatology expert and the proposed model reached an accuracy of 79.69%. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart of the proposed model for the detection of dysplastic nevi.</p>
Full article ">Figure 2
<p>Preprocessing techniques applied to a dysplastic nevus image.</p>
Full article ">Figure 3
<p>Flowchart of the UNet model for image segmentation.</p>
Full article ">Figure 4
<p>Distribution of the samples of 50 people.</p>
Full article ">Figure 5
<p>Comparison of dysplastic nevi detections by a dermatology expert and the proposed model.</p>
Full article ">
28 pages, 3912 KiB  
Review
MoS2–Plasmonic Hybrid Platforms: Next-Generation Tools for Biological Applications
by Nayra A. M. Moussa, Seungah Lee and Seong Ho Kang
Nanomaterials 2025, 15(2), 111; https://doi.org/10.3390/nano15020111 - 13 Jan 2025
Viewed by 583
Abstract
The combination of molybdenum disulfide (MoS2) with plasmonic nanomaterials has opened up new possibilities in biological applications by combining MoS2’s biocompatibility and high surface area with the optical sensitivity of plasmonic metals. These MoS2–plasmonic hybrid systems hold [...] Read more.
The combination of molybdenum disulfide (MoS2) with plasmonic nanomaterials has opened up new possibilities in biological applications by combining MoS2’s biocompatibility and high surface area with the optical sensitivity of plasmonic metals. These MoS2–plasmonic hybrid systems hold great promise in areas such as biosensing, bioimaging, and phototherapy, where their complementary properties facilitate improved detection, real-time visualization, and targeted therapeutic interventions. MoS2’s adjustable optical features, combined with the plasmon resonance of noble metals such as gold and silver, enhance signal amplification, enabling detailed imaging and selective photothermal or photodynamic therapies while minimizing effects on healthy tissue. This review explores various synthesis strategies for MoS2–plasmonic hybrids, including seed-mediated growth, in situ deposition, and heterojunction formation, which enable tailored configurations optimized for specific biological applications. The primary focus areas include highly sensitive biosensors for detecting cancer and infectious disease biomarkers, high-resolution imaging of cellular dynamics, and the development of phototherapy methods that allow for accurate tumor ablation through light-induced thermal and reactive oxygen species generation. Despite the promising advancements of MoS2–plasmonic hybrids, translating these platforms into clinical practice requires overcoming considerable challenges, such as synthesis reproducibility, toxicity, stability in physiological conditions, targeted delivery, and scalable manufacturing. Addressing these challenges is essential for realizing their potential as next-generation tools in diagnostics and targeted therapies. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>An illustration of the different metal coordination geometries and stacking sequences of the three distinct phases of MoS<sub>2</sub>. (<b>Top Row</b>) 1T Phase: Exhibits octahedral coordination of the Mo atoms, represented by purple spheres, with S atoms depicted as blue spheres; 2H Phase: Features trigonal prismatic coordination, where the Mo atoms are surrounded by six S atoms arranged in a prism-like structure; and 3R Phase: Displays trigonal prismatic coordination, similar to the 2H phase but with a different stacking arrangement. (<b>Middle Row</b>) Top view of each phase, showing the arrangement of the Mo and S atoms in the respective structures. Yellow triangles indicate the coordination environment around the Mo atoms. (<b>Bottom Row</b>) Stacking sequence of the Mo and S layers for each phase, showing arrangement of layers in three-dimensional space. Reprinted with permission from Ref. [<a href="#B46-nanomaterials-15-00111" class="html-bibr">46</a>]. Copyright (2017) Royal Society of Chemistry.</p>
Full article ">Figure 2
<p>Band structure plots of the electron levels of bulk, multilayer, and monolayer MoS<sub>2</sub>. Green and blue lines denote the conduction and valence bands, respectively, while the red dashed lines indicate the Fermi level. The small bold arrows represent the bandgap values for each system. Reprinted with permission from Ref. [<a href="#B58-nanomaterials-15-00111" class="html-bibr">58</a>]. Copyright (2011) American Physical Society.</p>
Full article ">Figure 3
<p>(<b>A</b>) The reflection difference observed from an ultrathin MoS<sub>2</sub> layer on a quartz substrate is proportional to the MoS<sub>2</sub> absorption coefficient. The absorption peaks at 1.85 eV (670 nm) and 1.98 eV (627 nm) can be attributed to the direct excitonic transitions of A<sub>1</sub> and B<sub>1</sub>, resulting from energy splitting owing to valence band spin–orbit coupling. The inset shows the bulk MoS<sub>2</sub> band structure, excluding the comparatively weak spin–orbit coupling, which exhibits an indirect bandgap of approximately 1 eV and a single higher-energy direct excitonic transition near the K point, indicated by an arrow. (<b>B</b>) A pronounced photoluminescence (PL) peak is observed at the energies of direct excitonic transitions in monolayer MoS<sub>2</sub>, whereas the indirect bandgap bulk MoS<sub>2</sub> sample lacks such luminescence. (<b>C</b>) PL and Raman spectra of monolayer, bilayer, hexalayer, and bulk MoS<sub>2</sub>. Various Raman peaks correspond to the vibrational modes of MoS<sub>2</sub> and silicon. The Raman signal from the MoS<sub>2</sub> monolayer is faint owing to the limited material being stimulated. However, the PL intensity is most pronounced in monolayer MoS<sub>2</sub> despite the reduced material. (<b>D</b>) PL spectra normalized to Raman intensity for MoS<sub>2</sub> layers of varying thicknesses, demonstrating a substantial increase in luminescence efficiency for the MoS<sub>2</sub> monolayer. Reprinted with permission from Ref. [<a href="#B65-nanomaterials-15-00111" class="html-bibr">65</a>]. Copyright (2010) American Chemical Society.</p>
Full article ">Figure 4
<p>(<b>A</b>) Schematic illustration of nanotag preparation (TMB/Ab2/AuNPs) and (<b>B</b>) the construction process of the EC/SERS dual-mode immunosensor. (<b>C</b>) SERS spectra of PSA samples at varying concentrations in PBS and (<b>D</b>) the corresponding plot of PSA concentrations versus SERS intensities at the Raman shift of 1602 cm<sup>−1</sup> in PBS. Reprinted with permission from Ref. [<a href="#B143-nanomaterials-15-00111" class="html-bibr">143</a>]. Copyright (2024) Elsevier B.V.</p>
Full article ">Figure 5
<p>(<b>A</b>) Two-photon cell imaging using AuNBPs@MoS<sub>2</sub>. The two-photon confocal laser scanning microscopy images show HeLa cancer cells incubated with (<b>B</b>) MoS<sub>2</sub>, (<b>C</b>) AuNBPs, and (<b>D</b>) AuNBPs@MoS<sub>2</sub>–PEG (50 μg/mL) for 12 h. Reprinted with permission from Ref. [<a href="#B146-nanomaterials-15-00111" class="html-bibr">146</a>]. Copyright (2018) American Chemical Society.</p>
Full article ">Figure 6
<p>(<b>A</b>) Preparation process of MAHP and its drug release mechanism under a synergistic photothermal effect. (<b>B</b>) DOX release from MAHP under varying pH conditions and NIR laser irradiation. (<b>C</b>) Cytotoxicity assessment of MAHP–DOX in MCF-7 cells with and without laser irradiation at pH 6.0. Reprinted with permission from Ref. [<a href="#B149-nanomaterials-15-00111" class="html-bibr">149</a>]. Copyright (2023) Elsevier B.V.</p>
Full article ">
16 pages, 302 KiB  
Review
Nuclear Medicine and Molecular Imaging in Urothelial Cancer: Current Status and Future Directions
by Sam McDonald, Kevin G. Keane, Richard Gauci and Dickon Hayne
Cancers 2025, 17(2), 232; https://doi.org/10.3390/cancers17020232 - 13 Jan 2025
Viewed by 257
Abstract
Background: The role of molecular imaging in urothelial cancer is less defined than other cancers, and its utility remains controversial due to limitations such as high urinary tracer excretion, complicating primary tumour assessment in the bladder and upper urinary tract. This review [...] Read more.
Background: The role of molecular imaging in urothelial cancer is less defined than other cancers, and its utility remains controversial due to limitations such as high urinary tracer excretion, complicating primary tumour assessment in the bladder and upper urinary tract. This review explores the current landscape of PET imaging in the clinical management of urothelial cancer, with a special emphasis on potential future advancements including emerging novel non-18F FDG PET agents, PET radiopharmaceuticals, and PET-MRI applications. Methods: We conducted a comprehensive literature search in the PubMed database, using keywords such as “PET”, “PET-CT”, “PET-MRI”, “FDG PET”, “Urothelial Cancer”, and “Theranostics”. Studies were screened for relevance, focusing on imaging modalities and advances in PET tracers for urothelial carcinoma. Non-English language, off-topic papers, and case reports were excluded, resulting in 80 articles being selected for discussion. Results: 18F FDG PET-CT has demonstrated superior sensitivity over conventional imaging, such as contrast-enhanced CT and MRI, for detecting lymph node metastasis and distant disease. Despite these advantages, FDG PET-CT is limited for T-staging of primary urothelial tumours due to high urinary excretion of the tracer. Emerging evidence supports the role of PETC-CT in assessing response to neoadjuvant chemotherapy and in identifying recurrence, with a high diagnostic accuracy reported in several studies. Novel PET tracers, such as 68Ga-labelled FAPI, have shown promising results in targeting cancer-associated fibroblasts, providing higher tumour-to-background ratios and detecting lesions missed by traditional imaging. Antibody-based PET tracers, like those targeting Nectin-4, CAIX, and uPAR, are under investigation for their diagnostic and theranostic potential, and initial studies indicate that these agents may offer advantages over conventional imaging and FDG PET. Conclusions: Molecular imaging is a rapidly evolving field in urothelial cancer, offering improved diagnostic and prognostic capabilities. While 18F FDG PET-CT has shown utility in staging, further prospective research is needed to establish and refine standardised protocols and validate new tracers. Advances in theranostics and precision imaging may revolutionise urothelial cancer management, enhancing the ability to tailor treatments and improve patient outcomes. Full article
(This article belongs to the Special Issue Advances in Management of Urothelial Cancer)
12 pages, 739 KiB  
Review
iPSC Technology Revolutionizes CAR-T Cell Therapy for Cancer Treatment
by Jiepu Zong and Yan-Ruide Li
Bioengineering 2025, 12(1), 60; https://doi.org/10.3390/bioengineering12010060 - 13 Jan 2025
Viewed by 306
Abstract
Chimeric Antigen Receptor (CAR)-engineered T (CAR-T) cell therapy represents a highly promising modality within the domain of cancer treatment. CAR-T cell therapy has demonstrated notable efficacy in the treatment of hematological malignancies, solid tumors, and various infectious diseases. However, current CAR-T cell therapy [...] Read more.
Chimeric Antigen Receptor (CAR)-engineered T (CAR-T) cell therapy represents a highly promising modality within the domain of cancer treatment. CAR-T cell therapy has demonstrated notable efficacy in the treatment of hematological malignancies, solid tumors, and various infectious diseases. However, current CAR-T cell therapy is autologous, which presents challenges related to high costs, time-consuming manufacturing processes, and the necessity for careful patient selection. A potential resolution to this restriction could be found by synergizing CAR-T technology with the induced pluripotent stem cell (iPSC) technology. iPSC technology has the inherent capability to furnish an inexhaustible reservoir of T cell resources. Experimental evidence has demonstrated the successful generation of various human CAR-T cells using iPSC technology, showcasing high yield, purity, robustness, and promising tumor-killing efficacy. Importantly, this technology enables the production of clinical-grade CAR-T cells, significantly reducing manufacturing costs and time, and facilitating their use as allogeneic cell therapies to treat multiple cancer patients simultaneously. In this review, we aim to elucidate essential facets of current cancer therapy, delineate its utility, enumerate its advantages and drawbacks, and offer an in-depth evaluation of a novel and pragmatic approach to cancer treatment. Full article
(This article belongs to the Section Cellular and Molecular Bioengineering)
Show Figures

Figure 1

Figure 1
<p>Comparison between autologous and allogeneic CAR-T cell therapy. Autologous and allogeneic CAR-T cell therapies differ primarily in their source of T cells and their implications for treatment logistics, safety, and efficacy. Autologous CAR-T cell therapy involves harvesting T cells from the patient’s own body, genetically engineering them to express a CAR, and then reinfusing them into the same patient. This personalized approach reduces the risk of immune rejection but presents challenges in terms of time, cost, and variability in T cell quality, especially in patients with weakened immune systems. In contrast, allogeneic CAR-T cell therapy uses T cells from healthy donors or stem cells such as iPSCs, which are engineered and prepared as an off-the-shelf product. This approach enables faster treatment delivery and the potential for mass production, making it more scalable. However, allogeneic CAR-T cells carry a higher risk of complications, such as graft-versus-host disease (GvHD) and immune rejection, which require additional genetic modifications, such as the disruption of the T cell receptor (TCR) to mitigate these risks. Additionally, strategies such as the ablation of HLA molecules and the overexpression of NK cell inhibitory ligands have been applied to allogeneic CAR-T cells to mitigate host cell-mediated allorejection and enhance the in vivo persistence and antitumor efficacy of the CAR-T cells.</p>
Full article ">
16 pages, 5765 KiB  
Article
Investigating the Role of Osteopontin (OPN) in the Progression of Breast, Prostate, Renal and Skin Cancers
by Gautam Kundu and Selvakumar Elangovan
Biomedicines 2025, 13(1), 173; https://doi.org/10.3390/biomedicines13010173 - 13 Jan 2025
Viewed by 275
Abstract
Background/Objectives: Cancer is caused by disruptions in the homeostatic state of normal cells, which results in dysregulation of the cell cycle, and uncontrolled growth and proliferation in affected cells to form tumors. Successful development of tumorous cells proceeds through the activation of [...] Read more.
Background/Objectives: Cancer is caused by disruptions in the homeostatic state of normal cells, which results in dysregulation of the cell cycle, and uncontrolled growth and proliferation in affected cells to form tumors. Successful development of tumorous cells proceeds through the activation of pathways promoting cell development and functionality, as well as the suppression of immune signaling pathways; thereby providing these cells with proliferative advantages, which subsequently metastasize into surrounding tissues. These effects are primarily caused by the upregulation of oncogenes, of which SPP1 (secreted phosphoprotein 1), a non-collagenous bone matrix protein, is one of the most well-known. Methods: In this study, we conducted a further examination of the transcriptomic expression profile of SPP1 (Osteopontin) during the progression of cancer in four human tissues, breast, prostate, renal and skin, in order to understand the circumstances conducive to its activation and dysregulation, the biological pathways and other mechanisms involved as well as differences in its splicing patterns influencing its expression and functionality. Results: A significant overexpression of SPP1, as well as a set of other highly correlated genes, was seen in most of these tissues, indicating their extensive implication in cancer. Increased expression was observed with higher tumor stages, especially in renal and skin cancer, while applying therapeutic modalities targeting these genes dampened this effect in breast, prostate and skin cancer. Pathway analyses showed gene signatures related to cell growth and development enriched in tumorigenic conditions and earlier cancer stages, while later stages of cancer showed pathways associated with weakened immune response, in all cancers studied. Moreover, the utilization of therapeutic methods showed the activation of immunogenic pathways in breast, prostate and skin cancer, thereby confirming their viability. Further analyses of differential transcript expression levels in these oncogenes showed their exonic regions to be selectively overexpressed similarly in tumorigenic samples in all cancers studied, while also displaying significant differences in exon selectivity between constituent transcripts, providing a basis for their high degree of multifunctionality in cancer. Conclusions: Overall, this study corroborates the entrenched role of SPP1 in the progression of these four types of cancer, as confirmed by its overexpression and activation of related oncogenes, their co-involvement in key cellular pathways, and predisposition to exhibit differential splicing between their transcripts, while the above effects were found to be highly inhibitable through treatment methods, thereby highlighting its promising role in therapeutic development. Full article
(This article belongs to the Special Issue Progress in Protein Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Pipeline for transcriptomic data analyses.</p>
Full article ">Figure 2
<p>Significant genes using differential gene expression (DEG) analyses in (<b>A</b>) breast cancer; (<b>B</b>–<b>D</b>) prostate cancer; (<b>E</b>) renal cancer; and (<b>F</b>) skin cancer datasets. Colors indicate significance categories: Genes showing no significant change in expression (gray), genes with nominal <span class="html-italic">p</span>-value &lt; 0.05 (orange), Benjamini–Hochberg (FDR) corrected <span class="html-italic">p</span>-value &lt; 0.05 (green), Bonferroni corrected <span class="html-italic">p</span>-value &lt; 0.05 (purple). Dashed lines indicate cutoff values for significance: Absolute log2FC &gt; 0.585 (corresponding to FC &gt; 1.5), and nominal <span class="html-italic">p</span>-value &lt; 0.05.</p>
Full article ">Figure 3
<p>Results from GSEA analyses: (<b>A</b>): significantly enriched signatures in a prostate cancer dataset; (<b>B</b>) top signature in NFYA (short isoform) expressing cells v/s controls; (<b>C</b>) signatures in two renal cancer datasets; (<b>D</b>,<b>E</b>) top signatures in RCC cells v/s controls, and IgA nephropathy v/s normal tissue, respectively; (<b>F</b>) significantly enriched signatures with increasing tumor grades in RCC tissue; (<b>G</b>) top signature in Grade 4 RCC v/s Grade 2. Colors in bar plots indicate the total number of signatures (blue) and number of signatures containing SPP1 and at least one of its six top interacting genes (orange). Enrichment plots from GSEA show the enrichment score (ES) obtained per gene in the signature, with the location of the peak ES value indicating the direction of overall enrichment for the signature. Red and blue colors indicate positive and negative enrichment respectively.</p>
Full article ">Figure 4
<p>Results from Metascape analyses showing pathways involving significant DEGs, in (<b>A</b>): breast cancer; (<b>B</b>,<b>C</b>): prostate cancer; (<b>D</b>,<b>E</b>): renal cancer; and (<b>F</b>): skin cancer datasets. The top 20 pathways are shown in descending order of significance, ranked by <span class="html-italic">p</span>-value, with darker-colored bars indicating pathways with greater significance.</p>
Full article ">Figure 5
<p>Results from DEU analyses showing exonic regions present selectively across transcripts for (<b>A</b>): CXCL12 in breast cancer; (<b>B</b>) S100A4 in prostate cancer; (<b>C</b>) SPP1; (<b>D</b>) S100A4 and (<b>E</b>) CXCL12, in renal cancer (RCC); and (<b>F</b>) SPP1 in another renal cancer dataset (IgA nephropathy). Graphs show expression levels in exonic regions for each pair of conditions (colored red and blue) per gene, and lines beneath denote the primary gene and its constituent transcripts (spliced isoforms). Boxes within each line indicate exonic regions per transcript, and are colored by significance categories: exons with non-significant DEU (gray), exons absent in the primary gene but present in any transcript and with significant DEU (white) and those with presence in the primary gene and significant DEU (pink).</p>
Full article ">
15 pages, 2843 KiB  
Article
MSEANet: Multi-Scale Selective Edge Aware Network for Polyp Segmentation
by Botao Liu, Changqi Shi and Ming Zhao
Algorithms 2025, 18(1), 42; https://doi.org/10.3390/a18010042 - 12 Jan 2025
Viewed by 423
Abstract
The colonoscopy procedure heavily relies on the operator’s expertise, underscoring the importance of automated polyp segmentation techniques in enhancing the efficiency and accuracy of colorectal cancer diagnosis. Nevertheless, achieving precise segmentation remains a significant challenge due to the high visual similarity between polyps [...] Read more.
The colonoscopy procedure heavily relies on the operator’s expertise, underscoring the importance of automated polyp segmentation techniques in enhancing the efficiency and accuracy of colorectal cancer diagnosis. Nevertheless, achieving precise segmentation remains a significant challenge due to the high visual similarity between polyps and their backgrounds, blurred boundaries, and complex localization. To address these challenges, a Multi-scale Selective Edge-Aware Network has been proposed to facilitate polyp segmentation. The model consists of three key components: (1) an Edge Feature Extractor (EFE) that captures polyp edge features with precision during the initial encoding phase, (2) the Cross-layer Context Fusion (CCF) block designed to extract and integrate multi-scale contextual information from diverse receptive fields, and (3) the Selective Edge Aware (SEA) module that enhances sensitivity to high-frequency edge details during the decoding phase, thereby improving edge preservation and segmentation accuracy. The effectiveness of our model has been rigorously validated on the Kvasir-SEG, Kvasir-Sessile, and BKAI datasets, achieving mean Dice scores of 91.92%, 82.10%, and 92.24%, respectively, on the test sets. Full article
(This article belongs to the Special Issue Artificial Intelligence Algorithms for Medicine (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Overall architecture of the proposed MSEANet. (<b>b</b>) Architecture of EFE; Dconv denotes depthwise separable convolution.</p>
Full article ">Figure 2
<p>(<b>a</b>) Structure of Cross-layer Context Fusion (CCF) module. (<b>b</b>) Structure of Multi-scale Selective Fusion (MSF).</p>
Full article ">Figure 3
<p>Structure of Selective Edge Aware (SEA) module.</p>
Full article ">Figure 4
<p>Image refers to sample images from the Kvasir-SEG and BKAI datasets. GT represents the ground truth annotations of polyps; Edge mask is the ground truth of polyp edges generated using the Canny operator; Edge pred denotes the predicted polyp edge maps generated by the EFE module.</p>
Full article ">Figure 5
<p>Comparison results of MSEANet with other advanced models on the Kvasir-SEG.</p>
Full article ">
13 pages, 1344 KiB  
Article
Neoadjuvant Chemotherapy Followed by Concurrent Chemoradiation Versus Adjuvant Chemotherapy Following Concurrent Chemoradiation for Locally Advanced Cervical Cancer: A Network Meta-Analysis
by Young Ju Suh, Dae Hyung Lee, Hee Joong Lee and Banghyun Lee
Cancers 2025, 17(2), 223; https://doi.org/10.3390/cancers17020223 - 11 Jan 2025
Viewed by 349
Abstract
Background/Objectives: Neoadjuvant chemotherapy followed by concurrent chemoradiation therapy (NACT + CCRT) and adjuvant chemotherapy following CCRT (CCRT + ACT) have inconsistent effects on the survival of women with locally advanced cervical cancer (LACC) compared to CCRT. Moreover, the effects of NACT + CCRT [...] Read more.
Background/Objectives: Neoadjuvant chemotherapy followed by concurrent chemoradiation therapy (NACT + CCRT) and adjuvant chemotherapy following CCRT (CCRT + ACT) have inconsistent effects on the survival of women with locally advanced cervical cancer (LACC) compared to CCRT. Moreover, the effects of NACT + CCRT and CCRT + ACT have not been clearly compared. This study compared the effects of NACT + CCRT and CCRT + ACT on survival using a network meta-analysis to select the optimal treatment in women with LACC. Methods: The PubMed, Medline, and Embase databases were searched, and six randomized controlled trials assessing the progression-free survival (PFS) and overall survival (OS) in women with newly diagnosed LACC treated with NACT + CCRT, CCRT + ACT, or CCRT alone (controls) were identified. A network meta-analysis was conducted. Results: Indirect comparisons showed no significant differences in PFS and OS between NACT + CCRT and CCRT + ACT. Direct comparisons also showed similar PFS and OS between NACT + CCRT and CCRT and between CCRT + ACT and CCRT. CCRT + ACT exhibited the highest surface under the cumulative ranking curve (SUCRA) value as a better treatment option for the PFS and OS (CCRT + ACT vs. NACT + CCRT vs. CCRT: 72% vs. 26.8% vs. 51.2% in PFS and 64.3% vs. 45.1% vs. 40.7% in OS). Conclusions: In women with LACC, NACT + CCRT had no different effects on the PFS and OS compared to CCRT + ACT, despite the relatively higher SUCRA value observed for CCRT + ACT. Further studies are warranted to clarify the effects of these strategies. Full article
(This article belongs to the Section Systematic Review or Meta-Analysis in Cancer Research)
Show Figures

Figure 1

Figure 1
<p>Flow chart of study selection.</p>
Full article ">Figure 2
<p>Network plots of treatments for the PFS and OS. (<b>A</b>) PFS; (<b>B</b>) OS. The size of the three nodes (treatments) increased as the number of studies included in the corresponding nodes increased, and the lines connecting two nodes were thickened as the number of studies comparing the two treatments increased [<a href="#B22-cancers-17-00223" class="html-bibr">22</a>]. CCRT, concurrent chemoradiation therapy; CCRT + ACT, adjuvant chemotherapy following concurrent chemoradiation therapy; NACT + CCRT, neoadjuvant chemotherapy followed by concurrent chemoradiation therapy; OS, overall survival; and PFS, progression-free survival.</p>
Full article ">Figure 3
<p>League tables of the treatments for the PFS and OS. (<b>A</b>) PFS; (<b>B</b>) OS. The hazard ratio (HR) of the upper left treatment (intervention) vs. lower right (comparator) was estimated. CCRT, concurrent chemoradiation therapy; CCRT + ACT, adjuvant chemotherapy following concurrent chemoradiation therapy; CI, confidence interval; NACT + CCRT, neoadjuvant chemotherapy followed by concurrent chemoradiation therapy; OS, overall survival; and PFS, progression-free survival.</p>
Full article ">Figure 4
<p>Forest plots of the treatments for the PFS and OS. (<b>A</b>) PFS, (<b>B</b>) OS. CCRT, concurrent chemoradiation therapy; CCRT + ACT, adjuvant chemotherapy following concurrent chemoradiation therapy; CI, confidence interval; NACT + CCRT, neoadjuvant chemotherapy followed by concurrent chemoradiation therapy; HR, hazard ratio; OS, overall survival; PFS, progression-free survival.</p>
Full article ">Figure 5
<p>SUCRA curves of the treatments for the PFS and OS. The treatments for the PFS: (<b>A</b>) CCRT + ACT, (<b>B</b>) NACT + CCRT, and (<b>C</b>) CCRT; the treatments for the OS: (<b>D</b>) CCRT + ACT, (<b>E</b>) NACT + CCRT, and (<b>F</b>) CCRT. CCRT, concurrent chemoradiation therapy; CCRT + ACT, adjuvant chemotherapy following concurrent chemoradiation therapy; NACT + CCRT, neoadjuvant chemotherapy followed by concurrent chemoradiation therapy; OS, overall survival; PFS, progression-free survival; and SUCRA, the estimated surface under the cumulative ranking probabilities.</p>
Full article ">
12 pages, 1520 KiB  
Article
Incidence and Prevalence of Bone Metastases in Different Solid Tumors Determined by Natural Language Processing of CT Reports
by Niamh Long, David Woodlock, Robert D’Agostino, Gary Nguyen, Natalie Gangai, Varadan Sevilimedu and Richard Kinh Gian Do
Cancers 2025, 17(2), 218; https://doi.org/10.3390/cancers17020218 - 11 Jan 2025
Viewed by 307
Abstract
Background/Objectives: Improved survival due to advances in medical therapy has resulted in increasing numbers of cancer patients living with bone metastases; however, our understanding of the prognostic implications of bone metastases requires larger population-based studies outlining their incidence and prevalence in different primary [...] Read more.
Background/Objectives: Improved survival due to advances in medical therapy has resulted in increasing numbers of cancer patients living with bone metastases; however, our understanding of the prognostic implications of bone metastases requires larger population-based studies outlining their incidence and prevalence in different primary cancer types, including those with lower incidence. This study aimed to evaluate the incidence and prevalence of bone metastases in solid organ tumors by analyzing reports of staging CT studies with natural language processing (NLP). Methods: In this retrospective study, 639,470 reports representing 129,326 unique patients were analyzed; 6279 randomly selected reports were manually annotated and labeled for the presence or absence of bone metastases. From these data, a BERT-based NLP model was developed and applied to the patient database. The cumulative incidence at 5 years and prevalence of bone metastases in each cancer type were calculated. Results: The accuracy of the NLP model on a validation set was 97.1%, with a positive predictive value (precision) of 88.0% and a sensitivity (recall) of 86.3%. The 5-year incidence rate of bone metastases was highest in prostate, breast, head and neck, and lung cancer (52%, 41%, 36%, 33%). Incidence was lowest in central nervous system cancer and testicular cancer (8%, 5%). Prevalence was highest in prostate, breast, and lung cancer (32%, 25% and 23%), and lowest in central nervous system cancer and testicular cancer (4%, 4%). Conclusions: NLP was utilized to demonstrate patterns of bone metastases in a broad range of cancer types and is a valuable tool in population-based assessment of bone metastases. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart detailing the sequential steps in the development of the natural language processing model. There were 639,470 consecutive CT reports first identified between 2009 and 2021. Manual curation was performed on about 1% of reports (6279 report), which were split into training (80%) and testing sets (20%) to build a BERT-based natural language processing model. The final model was evaluated on a validation set of 448 reports and applied to the remaining unlabeled reports. Rule-based labelling was used on a subset of records where the default language (e.g., “unremarkable”) was used in our structured reports.</p>
Full article ">Figure 2
<p>Bone metastases from primary cancers with the highest incidence rates. (<b>A</b>) Sclerotic pelvic metastases in a 76-year-old male patient with prostate cancer. (<b>B</b>) Lytic metastasis in the left third rib in a 60-year-old female patient with breast cancer. (<b>C</b>) Lytic metastasis left pubic bone in a 48-year-old female patient with thyroid cancer. (<b>D</b>) Sclerotic metastasis in an L1 vertebral body in a 59-year-old male patient with adenoid cystic carcinoma of the tongue. (<b>E</b>) Lytic metastases in the left sacral ala and left ilium in a 65-year-old male patient with poorly differentiated lung adenocarcinoma. (<b>F</b>) Multiple sclerotic pelvic bone metastases in a 57-year-old male with a lung carcinoid tumor. (<b>G</b>) Sclerotic right iliac metastasis in a 63-year-old female patient with melanoma. (<b>H</b>) Lytic left sacral metastasis in a 55-year-old female patient with melanoma. (<b>I</b>) Lytic left second rib metastasis in a 60-year-old female patient with hepatocellular carcinoma. (<b>J</b>) Multiple lumbar spine sclerotic metastases in a 92-year-old male patient with hepatocellular carcinoma. (<b>K</b>) Lytic left iliac metastasis in a 67-year-old male patient with esophageal cancer. (<b>L</b>) Follow-up demonstrating interval sclerosis of the left iliac metastasis representing treatment effect.</p>
Full article ">Figure 3
<p>Bone metastases from primary cancers with the highest incidence rates. (<b>A</b>) Sclerotic pelvic metastases in a 76-year-old male patient with prostate cancer. (<b>B</b>) Left 5th rib lytic metastasis in an 84-year-old male with colorectal cancer. (<b>C</b>) Lytic metastasis T5 vertebra in a 64-year-old female patient with ovarian cancer. (<b>D</b>) Sclerotic metastasis right acetabulum in a 62-year-old male patient with urothelial cancer. (<b>E</b>) Lytic metastasis left ilium in a 54-year-old female patient with renal cell cancer. (<b>F</b>) Lytic metastasis left ilium in a 44-year-old male patient with renal cell carcinoma. (<b>G</b>) Multiple sclerotic lumbar spine metastases in a 54-year-old male patient with urothelial carcinoma of the renal pelvis. (<b>H</b>) Sagittal CT lumbar spine of multifocal sclerotic osseous metastases in a 35-year-old female patient with HER2-negative gastric cancer.</p>
Full article ">Figure 4
<p>Kaplan–Meier curves of eight primary tumors: prostate, pancreas, ovary, lung, hepatobiliary, colorectal, stomach, and central nervous system (CNS). 50% of patients with prostate cancer developed bone metastases within five years (dashed lines).</p>
Full article ">
23 pages, 2145 KiB  
Article
Optimized Deep Learning Model for Predicting Liver Metastasis in Colorectal Cancer Patients
by Molan Wang, Jiaqing Chen and Yuqi Liu
Symmetry 2025, 17(1), 103; https://doi.org/10.3390/sym17010103 - 11 Jan 2025
Viewed by 244
Abstract
Colorectal cancer is a leading type of cancer worldwide and a major contributor to cancer fatalities, and liver metastasis is the most likely distant metastasis in colorectal cancer patients. Classifying and predicting whether liver metastasis occurs in colorectal cancer patients can help doctors [...] Read more.
Colorectal cancer is a leading type of cancer worldwide and a major contributor to cancer fatalities, and liver metastasis is the most likely distant metastasis in colorectal cancer patients. Classifying and predicting whether liver metastasis occurs in colorectal cancer patients can help doctors timely determine the progress of the disease and form a more reasonable treatment plan, which results in a better prognosis for patients. In this paper, using the Surveillance, Epidemiology, and End Results database, selecting both symmetric and asymmetric features, we extracted the disease-related data of 40,870 patients who were pathologically diagnosed with colorectal cancer from 2010 to 2015 and classified and modeled whether the patients developed liver metastasis to show the symmetry of this study. A total of six deep learning models were utilized, and hyperparameter optimization was performed on the models using the Crested Porcupine Optimizer. The best-performing model was selected and model interpretation was performed to explore the features that affect whether patients develop liver metastasis. Among the six deep learning models selected, the FT-Transformer model, which was hyperparameter optimized by the Crested Porcupine Optimizer, performed the best, with an accuracy of 0.945, with a 95% confidence interval (CI) of [0.942, 0.952], and an AUC of 0.949, with a 95% CI of [0.942, 0.957]. This study can help doctors make medical decisions, detect patients with liver metastases of colorectal cancer earlier, monitor the indicators that have a significant impact on the occurrence of liver metastasis in patients, and use timely surgical treatment, radiotherapy, chemotherapy, and other corresponding therapeutic interventions to improve the survival rate of patients. Full article
(This article belongs to the Section Mathematics)
Back to TopTop