[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,749)

Search Parameters:
Keywords = skin tumor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 2942 KiB  
Article
Deferred Lateral Margin Control in the Surgical Treatment of Genital Paget’s Disease and Lentiginous Vulvar Melanoma
by Pedro Redondo
J. Clin. Med. 2025, 14(1), 69; https://doi.org/10.3390/jcm14010069 (registering DOI) - 26 Dec 2024
Abstract
Background/Objectives: Some skin tumors can extend beyond their clinical appearance. This presents an additional challenge, especially when the affected area is the genital region, which is more difficult for both the patient and the physician to access and monitor due to its location [...] Read more.
Background/Objectives: Some skin tumors can extend beyond their clinical appearance. This presents an additional challenge, especially when the affected area is the genital region, which is more difficult for both the patient and the physician to access and monitor due to its location and anatomical characteristics. The treatment of these lesions is complex, and literature postulates Mohs surgery as the best therapeutic option. Methods: We describe our experience in two patients with the resection of vulvar lentiginous melanoma and genital extramammary Paget’s disease, using a method of deferred lateral margin control in the surgical treatment. Results: The “spaghetti technique”(ST) initially removing a small strip from all lateral margins of the lesion, which is then closed directly while awaiting the paraffin histological result. In a second stage, the tumors within those margins are removed, and immediate reconstruction is performed. The final oncological and functional result was satisfactory, with no notable side effects. Conclusions: This method is suited for large, poorly defined superficial tumors in the genital, perineal, and perianal regions, where a frozen section study would be slow and burdensome for the patient and surgeon. The ST preserves healthy tissue and can be performed by any surgeon and pathologist without additional training, and is more comfortable for patients, avoiding prolonged open wounds during multiple steps of tumor excision. Full article
(This article belongs to the Section Dermatology)
14 pages, 3619 KiB  
Article
Bioadhesive Chitosan Films Loading Curcumin for Safe and Effective Skin Cancer Topical Treatment
by Seila Tolentino, Mylene M. Monteiro, Felipe Saldanha-Araújo, Marcilio Cunha-Filho, Tais Gratieri, Eliete N. Silva Guerra and Guilherme M. Gelfuso
Pharmaceutics 2025, 17(1), 18; https://doi.org/10.3390/pharmaceutics17010018 - 26 Dec 2024
Abstract
Background/Objectives: This study aimed to evaluate the safety and efficacy of chitosan-based bioadhesive films for facilitating the topical delivery of curcumin in skin cancer treatment, addressing the pharmacokinetic limitations associated with oral administration. Methods: The films, which incorporated curcumin, were formulated [...] Read more.
Background/Objectives: This study aimed to evaluate the safety and efficacy of chitosan-based bioadhesive films for facilitating the topical delivery of curcumin in skin cancer treatment, addressing the pharmacokinetic limitations associated with oral administration. Methods: The films, which incorporated curcumin, were formulated using varying proportions of chitosan, polyvinyl alcohol, Poloxamer® 407, and propylene glycol. These films were assessed for stability, drug release, in vitro skin permeation, cell viability (with and without radiotherapy), and skin irritation. Results: The films demonstrated physical stability and preserved curcumin content at room temperature for 90 days. Drug release was effectively controlled during the first 8 h, with release rates ranging from 51.6 ± 4.8% to 65.6 ± 13.0%. The films also enhanced drug penetration into the skin compared to a curcumin solution used as a control (stratum corneum: 1.3 ± 0.1 to 1.9 ± 0.8 µg/cm²; deeper skin layers: 1.7 ± 0.1 to 2.7 ± 0.2 µg/cm²). A cytotoxicity test on metastatic melanoma cells showed that curcumin at topical doses exerted activity similar to that delivered via the skin. Furthermore, curcumin alone was more effective in inhibiting tumor cells than radiotherapy alone (p < 0.01), with no additional benefit observed when curcumin was combined with radiotherapy. Finally, irritation tests confirmed that the films were safe for topical application. Conclusion: The developed chitosan-based bioadhesive films represent a promising alternative for the topical treatment of skin tumors using curcumin. Full article
Show Figures

Figure 1

Figure 1
<p>Images captured from the F1, F2, and F3 films at the predefined times for the stability study (0, 7, 30, 60, and 90 days) and variation in curcumin content over time. The storage conditions were room temperature (RT) and 40 °C.</p>
Full article ">Figure 2
<p>Curcumin release profiles from films (F1, F2, and F3) over 24 h, with collections at 1, 2, 4, 6, 8, 18, 20, 22, and 24 h and recording photographs of the films at the end of the study.</p>
Full article ">Figure 3
<p>Curcumin recovered from the skin layers (µg/cm<sup>2</sup>) after a 24 h treatment with the films compared to the control. (<b>a</b>) Stratum corneum, (<b>b</b>) remaining skin. The data represent the mean of 5 determinations ± standard deviation. #, values below the limit of quantification; (*), <span class="html-italic">p</span> ≤ 0.05; and (****), <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 4
<p>Effect of curcumin on the viability of MeWo cell line. The Kruskal–Wallis test was employed for cell viability data, and IC<sub>50</sub> values were calculated following nonlinear regression on dose–response curves. The data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 9). (**), <span class="html-italic">p</span> ≤ 0.01; and (****), <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">Figure 5
<p>Effect of curcumin on the viability of MeWo cell line, with or without three different doses of radiotherapy. The data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 9). Statistical analysis was performed using ANOVA. (*), <span class="html-italic">p</span> ≤ 0.05; (**), <span class="html-italic">p</span> ≤ 0.01; and (***); <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 6
<p>Illustrative sequence of photographic records taken during the HET-CAM assay demonstrating the effects with positive control (1.0 mol/L NaOH), negative control (PBS), and solutions containing each of the films (F1, F2, and F3) solubilized in water on the chorioallantoic membrane after 30 s, 2 min, and 5 min of application.</p>
Full article ">Figure 7
<p>Effect of chitosan on the viability of the human keratinocytes (HaCaT cell line). The Kruskal–Wallis test was employed for cell viability data. Due to the sustained cell viability, it was not possible to calculate the IC<sub>50</sub> from the obtained dose–response curve. The data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 9).</p>
Full article ">
21 pages, 501 KiB  
Systematic Review
Sentinel Lymph Node Biopsy: Is There a Role in Non-Melanoma Skin Cancer? A Systematic Review
by Lorenzo Borgognoni, Pietro Susini, Gianni Gerlini, Paola Brandani, Vanni Giannotti and Serena Sestini
Cancers 2024, 16(24), 4279; https://doi.org/10.3390/cancers16244279 - 23 Dec 2024
Abstract
Background/Objectives: Sentinel Lymph Node Biopsy (SLNB) aims at identifying clinically occult nodal metastases. It is the standard staging procedure for patients with T1b to T4 primary cutaneous melanoma. Moreover, it is recommended whenever the risk of a positive SLNB is >5%, according to [...] Read more.
Background/Objectives: Sentinel Lymph Node Biopsy (SLNB) aims at identifying clinically occult nodal metastases. It is the standard staging procedure for patients with T1b to T4 primary cutaneous melanoma. Moreover, it is recommended whenever the risk of a positive SLNB is >5%, according to the National Comprehensive Cancer Network Melanoma guidelines. When considering Non-Melanoma Skin Cancer (NMSC), the SLNB could play a role in tumors that mainly spreads via lymphatics, but strong evidence is missing. In this paper, the hot topics and controversies are reviewed; Methods: A PRISMA systematic review was carried out on the PubMed (MEDLINE) library from 2004–2024, searching for studies on SLNB in NMSC; Results: Seventy articles and 6379 patients undergoing SLNB for Squamous Cell Carcinoma (SCC), Merkel Cell Carcinoma (MCC), and Porocarcinoma were included. Overall, the SLNB positivity rate in these NMSCs was 24.4%, with an SNLB detection rate of 97.6%. Specifically, the SLNB positivity rate was 12.3% for high-risk cutaneous SCC, 24.4% for anogenital SCC, 29.3% for MCC, and 30.6% for Porocarcinoma. Most papers concluded that SLNB is safe, feasible, and significant in these malignancies; Conclusions: SLNB should be discussed and offered to every patient with MCC, and it should be discussed and considered in “high risk” SCC and Porocarcinoma for staging and prognostic purposes, aiming to identify a subgroup of patients who may benefit the most from early treatments. Full article
(This article belongs to the Special Issue Advances in Skin Cancer: Diagnosis, Treatment and Prognosis)
Show Figures

Figure 1

Figure 1
<p>PRISMA 2020 flow diagram summarizing research results.</p>
Full article ">
11 pages, 1109 KiB  
Article
The Safety and Suitability of DNA Sequencing of Tissue Biopsies Performed on Patients Referred to a Phase I Unit
by Angela Esposito, Edoardo Crimini, Carmen Criscitiello, Carmen Belli, Roberta Scafetta, Raimondo Scalia, Grazia Castellano, Elisa Giordano, Jalissa Katrini, Liliana Ascione, Luca Boscolo Bielo, Matteo Repetto, Antonio Marra, Dario Trapani, Gianluca Maria Varano, Daniele Maiettini, Paolo Della Vigna, Franco Orsi, Elena Guerini Rocco, Nicola Fusco and Giuseppe Curiglianoadd Show full author list remove Hide full author list
Cancers 2024, 16(24), 4252; https://doi.org/10.3390/cancers16244252 - 20 Dec 2024
Viewed by 247
Abstract
Background: Early-phase clinical trials offer a unique opportunity for patients with cancer. These trials often mandate biopsies to collect tumor tissue for research purposes, requiring patients to undergo invasive procedures. Some trials mandate molecular prescreening, but the success of these analyses relies on [...] Read more.
Background: Early-phase clinical trials offer a unique opportunity for patients with cancer. These trials often mandate biopsies to collect tumor tissue for research purposes, requiring patients to undergo invasive procedures. Some trials mandate molecular prescreening, but the success of these analyses relies on the quality and quantity of the tested materials. Additionally, bioptic procedures may result in complications. Methods: We retrospectively examined the records of patients referred to the Early Drug Development (EDD) Unit of the European Institute of Oncology who underwent biopsies for research purposes between January 2014 and December 2022. Our objective was to assess the safety of biopsy procedures and adequacy of the samples for NGS testing. Results: In total, 355 out of 731 patients (48.6%) underwent protocol-mandated biopsies. The most frequent sites of biopsy were the liver, lymph nodes, skin, and breast. Histological diagnosis was achieved in 349 (98%) patients, and NGS testing was successfully conducted in 111/127 (88.4%) cases. Of the 16 unsuccessful NGS attempts, 9 were performed on liver tissue. Unsuccessful NGS testing was attributed to poor sample quality and/or quantity, and the success rate varied significantly based on the specific tests attempted. Complications occurred in a small proportion of patients (4.8%), and none were serious. Conclusions: The non-negligible failure rate of NGS testing highlights the crucial need for implementing specific guidelines and Standard Operating Procedures for samples intended for NGS. With the use of a risk-based biopsy framework to guide clinical decisions, procedure-related complications may be minimized. Full article
(This article belongs to the Special Issue Pre-Clinical Studies of Personalized Medicine for Cancer Research)
Show Figures

Figure 1

Figure 1
<p>Circular grouped and stacked barplot: bars represent the sites of biopsy, grouped by primary tumors (showed inside the circle: Breast, breast cancer; Chol, cholangiocarcinoma; Other, other tumor types). Only tumors that counted at least 5 NGS attempts are reported separately from “Other”. The height of the bars corresponds to the number of biopsies performed at the anatomic site in the specific tumor type. Bars of stacked colors represent the number of each NGS test (see legend). Donut plots report the success rate of each NGS test for the biopsy site and primary tumor. The inner, middle, and outer circles represent other tests, GerSom and Foundation One<sup>TM</sup>, respectively. The color of each test is consistent with the circular barplot. The proportion of failures for each test is reported in red. The tables provide the corresponding data.</p>
Full article ">Figure 2
<p>Schematization of study population and adverse events associated with biopsies. * Biopsies for which NGS results are available.</p>
Full article ">
37 pages, 1965 KiB  
Review
Photon-Based Innovations in Oncology: Precise Diagnostic Techniques and Advanced Therapies
by Emilia Kamizela, Jakub Oberda, Albert Chomątowski, Angelika Masiarz, Kacper Ponikowski, Monika Lejman and Joanna Zawitkowska
Photonics 2024, 11(12), 1201; https://doi.org/10.3390/photonics11121201 - 20 Dec 2024
Viewed by 248
Abstract
In diagnostics, photons are used in basic methods such as computed tomography (CT) and positron emission tomography (PET), which are pivotal tools for high-resolution, non-invasive tumor detection, offering insights into tumor staging and progression. Mentioned techniques facilitate early diagnosis and the planning of [...] Read more.
In diagnostics, photons are used in basic methods such as computed tomography (CT) and positron emission tomography (PET), which are pivotal tools for high-resolution, non-invasive tumor detection, offering insights into tumor staging and progression. Mentioned techniques facilitate early diagnosis and the planning of therapeutic strategies. However, new methods are emerging, enhancing the precision and detail of diagnostics, such as ultra-weak photon emission (UPE) imagining, two-photon fluorescence imaging, photo acoustic imaging, and others. Therapeutically, external beam radiation therapy (EBRT) uses photons to target cancer cells while minimizing harm to healthy tissue. Photodynamic therapy (PDT), which uses light-sensitive compounds activated by specific wavelengths, represents a photon-based treatment applicable to certain malignancies. Other treatments include photo thermal therapy (PTT), radio dynamic therapy (RDT), intensity-modulated radiation therapy (IMRT), volumetric modulated arc therapy (VMAT), and more. These constantly evolving photon-driven technologies can be used to treat a broad spectrum of cancers, such as pancreatic, prostate, breast, and skin cancers. This review article discusses the latest photon-based methods in oncology, focusing on new possibilities, solutions, perspectives, and the potential disadvantages of these approaches. Full article
Show Figures

Figure 1

Figure 1
<p>A scheme of the construction and operation of third-generation (<b>A</b>) and fourth-generation (<b>B</b>) X-ray CT scanners.</p>
Full article ">Figure 2
<p>PET detector ring coincidence imaging (<b>A</b>). After emission, positrons travel a short distance before colliding with an electron in an annihilation event, producing two 511 keV photons that move in opposite directions and are detected by opposing detectors within a coincidence time (<b>B</b>).</p>
Full article ">Figure 3
<p>Reactions of reactive oxygen species with biomolecules lead to the synthesis of excited carbonyl compounds and singlet oxygen from 1,2-dioxetane and tetraoxide [<a href="#B83-photonics-11-01201" class="html-bibr">83</a>,<a href="#B84-photonics-11-01201" class="html-bibr">84</a>,<a href="#B85-photonics-11-01201" class="html-bibr">85</a>].</p>
Full article ">Figure 4
<p>The extensive network of tumor vessels is rich in hemoglobin. The intensive absorption of light at a particular wavelength by hemoglobin enables the detection of the tumor through photoacoustic imaging [<a href="#B104-photonics-11-01201" class="html-bibr">104</a>].</p>
Full article ">
19 pages, 665 KiB  
Review
Treatment and Prevention of HPV-Associated Skin Tumors by HPV Vaccination
by Thomas Meyer and Eggert Stockfleth
Vaccines 2024, 12(12), 1439; https://doi.org/10.3390/vaccines12121439 - 20 Dec 2024
Viewed by 229
Abstract
HPV-associated dermatological diseases include benign lesions like cutaneous warts and external genital warts. In addition, HPV infection is associated with the development of epithelial skin cancers, in particular cutaneous squamous cell carcinoma (cSCC). In contrast to anogenital and oropharyngeal cancers caused by mucosal [...] Read more.
HPV-associated dermatological diseases include benign lesions like cutaneous warts and external genital warts. In addition, HPV infection is associated with the development of epithelial skin cancers, in particular cutaneous squamous cell carcinoma (cSCC). In contrast to anogenital and oropharyngeal cancers caused by mucosal HPV types of genus alpha papillomavirus, cSCC-associated HPV types belong to the genus beta papillomavirus. Currently available HPV vaccines that target mucosal HPV types associated with anogenital cancer and genital warts are type-specific and provide no cross-protection against beta HPV. When implementing vaccination to beta HPV to prevent skin tumors, it must be considered that acquisition of these HPV types occurs early in childhood and that the risk for cSCC increases with growing age and decreasing immune surveillance. Thus, individuals considered for beta HPV vaccination usually have pre-existing infection and are largely immunocompromised. On the other hand, worldwide increasing incidence rates of epithelial skin cancer reflect an urgent need for skin cancer prevention measures. Based on the pathogenic involvement of beta HPV, vaccination may represent a promising prevention strategy. Indeed, various procedures of prophylactic and therapeutic vaccination have been developed, and some of them have shown efficiency in animal models. Thus far, however, none of these vaccine candidates has been approved for application in humans. Full article
(This article belongs to the Section Human Papillomavirus Vaccines)
Show Figures

Figure 1

Figure 1
<p>Interventions available and under development for mucosal and cutaneous HPV infections. Prophylactic vaccination with licensed L1-VLPs and screening by cytology and HPV tests are established methods to prevent or monitor mucosal HPV infection. Although not approved, L1-VLPs may also be used to treat pre-existing infection. Prophylactic vaccination targeting beta HPVs and therapeutic vaccination against mucosal and cutaneous HPVs are still under development. Antiviral compounds against HPV proteins are presently not on hand and the development is still in an experimental stage.</p>
Full article ">
21 pages, 4656 KiB  
Article
Synergistic Anti-Cancer Effects of Curcumin and Thymoquinone Against Melanoma
by Hana Mohd and Bozena Michniak-Kohn
Antioxidants 2024, 13(12), 1573; https://doi.org/10.3390/antiox13121573 - 20 Dec 2024
Viewed by 379
Abstract
Combining anti-cancer agents in cancer therapies is becoming increasingly common because of their improved efficacy, reduced toxicity, and decreased risk of resistance development. Melanoma, a highly aggressive form of skin cancer characterized by limited treatment options due to chemoresistance, poses a considerable challenge [...] Read more.
Combining anti-cancer agents in cancer therapies is becoming increasingly common because of their improved efficacy, reduced toxicity, and decreased risk of resistance development. Melanoma, a highly aggressive form of skin cancer characterized by limited treatment options due to chemoresistance, poses a considerable challenge for effective management. Here, we test the hypothesis that dietary supplements such as thymoquinone (TQ) and curcumin (CU) cooperatively modulate cancer-associated cellular mechanisms to inhibit melanoma progression. Through a series of in vitro experiments utilizing the A375 melanoma cell line, including assessments of cell viability, apoptosis, multicellular tumor spheroid models, reactive oxygen species (ROS) quantification, metabolomics analysis, and RNA sequencing, we established that the combined application of TQ and CU exhibited superior anti-tumor effects compared to their individual use. Our results indicate that the combination treatment significantly inhibited cell viability and induced apoptosis more effectively than either agent alone, with optimal synergy observed at concentrations of 25 µM CU and 10 µM TQ against A375 cells. Additionally, the combination treatment markedly elevated ROS levels, selectively activating the mitochondrial apoptotic pathway via caspase-9. Differential gene expression analysis further revealed a unique synergistic effect of the combination treatment, with enhanced regulation of genes related to oxidative stress and apoptosis. Notably, pathways such as mitochondrial apoptotic signaling and redox homeostasis were more effectively influenced by the combination, with genes such as GPX3, CYP4F11, and HSPB8 cooperatively regulated. Overall, the findings suggest that, in combination, TQ and CU acts synergistically against melanoma; however, further experimental and clinical studies are required to confirm its therapeutic potential. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of thymoquinone, curcumin, and their combination on the viability of human (A375) metastatic melanoma cells and the two healthy human keratinocyte (HaCaT) and human dermal fibroblast (HDfa) cell lines. The values are presented as the percentage of cell death. The data represent the mean ± SD of three independent experiments conducted in triplicate (vs. control, * <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 2
<p>Synergism between TQ and CU in A375 cells. Bliss and HSA synergy scores (SynergyFinder.com) were calculated to predict the potential synergism of TQ and CU.</p>
Full article ">Figure 3
<p>(<b>A</b>) Representative histograms obtained using flow cytometry on an apoptosis assay from various groups are shown (quadrant: upper left—dead cells; upper right—late apoptosis; lower left—live cells; lower right—early apoptosis); (<b>B</b>) percentage of viable, dead, early apoptotic, and late apoptotic cells in the control and compound-treated groups. <span class="html-italic">n</span> = 3; value are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control cells; ** <span class="html-italic">p</span> &lt; 0.05 combination vs. 25 μM CU; *** <span class="html-italic">p</span> &lt; 0.05 combination vs. 10 μM TQ.</p>
Full article ">Figure 4
<p>(<b>A</b>) Calcein-AM/PI dual-stained A375 multicellular tumor spheroids after 24 h of treatment with CU, TQ, or their combination. A red signal indicates dead cells, and a green signal indicates viable cells. Images obtained using an ECHO Revolve Microscope (Model RVL-100-M, Serial #: M-00395-RVL) (20×/0.4 objective). Scale bar: 180 µM. (<b>B</b>) Effect of thymoquinone, curcumin, and their combination on the viability of human (A375) metastatic melanoma cells in an MCTS model using a CellTiter-Glo 3D assay.<span class="html-italic">p</span>-Values are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control.</p>
Full article ">Figure 4 Cont.
<p>(<b>A</b>) Calcein-AM/PI dual-stained A375 multicellular tumor spheroids after 24 h of treatment with CU, TQ, or their combination. A red signal indicates dead cells, and a green signal indicates viable cells. Images obtained using an ECHO Revolve Microscope (Model RVL-100-M, Serial #: M-00395-RVL) (20×/0.4 objective). Scale bar: 180 µM. (<b>B</b>) Effect of thymoquinone, curcumin, and their combination on the viability of human (A375) metastatic melanoma cells in an MCTS model using a CellTiter-Glo 3D assay.<span class="html-italic">p</span>-Values are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control.</p>
Full article ">Figure 5
<p>(<b>A</b>) Intracellular ROS levels in A375 cells after treatment with CU, TQ, and their combination. After the indicated treatment for 24 h, cells were incubated with 10 μM DCFH-DA for 30 min and then immediately subjected to a flow cytometry analysis. The results are expressed as a ratio of the relative fluorescence intensity compared to the control group. (<b>B</b>) The DCFH-DA spectrum represents the fluorescence intensities of the probe. DCFH-DA (10 μM) was incubated with the cell culture for 30 min. (<b>C</b>) Caspases-9 and -8 were measured using Caspase-Glo 8 Assay and Caspase-Glo 9 Assay kits. <span class="html-italic">p</span>-Values were recorded as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control cells; ** <span class="html-italic">p</span> &lt; 0.05 combination vs. 25 μM CU; *** <span class="html-italic">p</span> &lt; 0.05 combination vs. 10 μM TQ.</p>
Full article ">Figure 5 Cont.
<p>(<b>A</b>) Intracellular ROS levels in A375 cells after treatment with CU, TQ, and their combination. After the indicated treatment for 24 h, cells were incubated with 10 μM DCFH-DA for 30 min and then immediately subjected to a flow cytometry analysis. The results are expressed as a ratio of the relative fluorescence intensity compared to the control group. (<b>B</b>) The DCFH-DA spectrum represents the fluorescence intensities of the probe. DCFH-DA (10 μM) was incubated with the cell culture for 30 min. (<b>C</b>) Caspases-9 and -8 were measured using Caspase-Glo 8 Assay and Caspase-Glo 9 Assay kits. <span class="html-italic">p</span>-Values were recorded as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control cells; ** <span class="html-italic">p</span> &lt; 0.05 combination vs. 25 μM CU; *** <span class="html-italic">p</span> &lt; 0.05 combination vs. 10 μM TQ.</p>
Full article ">Figure 6
<p>Significantly differentially expressed genes were clustered by their gene ontology, and the enrichment of gene ontology terms was tested using Fisher’s exact test (GeneSCF v1.1-p2): (<b>A</b>) significantly enriched gene ontology terms with adjusted <span class="html-italic">p</span>-values less than 0.05 in the differentially expressed gene sets (up to 40 terms) between 10 μM TQ+ 25 μM CU and the control; (<b>B</b>) mRNA expression of highly regulated genes, measured by RT-PCR and shown as the fold change compared to the control group. <span class="html-italic">p</span>-Values are presented as the mean ± SD. * <span class="html-italic">p</span> &lt; 0.05 vs. control.</p>
Full article ">Figure 7
<p>A bi-clustering heatmap for the combination vs. the control, which visualizes the expression profiles of the top 30 differentially expressed genes sorted by their adjusted <span class="html-italic">p</span>-values by plotting their log2-transformed expression values in the samples.</p>
Full article ">Figure 8
<p>Metabolomics of TQ, CU, and their combination on treated melanoma cell lines: (<b>A</b>) PCA analysis of three groups showing that samples in each group are clustered away from one another; (<b>B</b>) heatmap of pairwise correlation values of 120 metabolites and depiction of the major metabolic pathways in A375 cells; (<b>C</b>) identified pathways altered by the combination treatment vs. the control treatment in A375 cell lines.</p>
Full article ">Figure 8 Cont.
<p>Metabolomics of TQ, CU, and their combination on treated melanoma cell lines: (<b>A</b>) PCA analysis of three groups showing that samples in each group are clustered away from one another; (<b>B</b>) heatmap of pairwise correlation values of 120 metabolites and depiction of the major metabolic pathways in A375 cells; (<b>C</b>) identified pathways altered by the combination treatment vs. the control treatment in A375 cell lines.</p>
Full article ">Figure 8 Cont.
<p>Metabolomics of TQ, CU, and their combination on treated melanoma cell lines: (<b>A</b>) PCA analysis of three groups showing that samples in each group are clustered away from one another; (<b>B</b>) heatmap of pairwise correlation values of 120 metabolites and depiction of the major metabolic pathways in A375 cells; (<b>C</b>) identified pathways altered by the combination treatment vs. the control treatment in A375 cell lines.</p>
Full article ">
7 pages, 1703 KiB  
Interesting Images
It Looks Like a Zebra but Is Not: [18F]FDG PET/CT in a Giant Cutaneous Malignant Melanoma Mimicking Squamous Cell Carcinoma
by Ilaria Proietti, Giulia Azzella, Diana Dirzu, Claudio Di Cristofano, Oreste Bagni, Concetta Potenza and Luca Filippi
Diagnostics 2024, 14(24), 2860; https://doi.org/10.3390/diagnostics14242860 - 19 Dec 2024
Viewed by 212
Abstract
Cutaneous malignant melanoma (MM) is the most aggressive form of skin cancer, associated with high mortality and rising incidence rates in Europe despite prevention efforts. Nodular MM, the most aggressive subtype, often mimics other skin tumors, complicating diagnosis. We present the case of [...] Read more.
Cutaneous malignant melanoma (MM) is the most aggressive form of skin cancer, associated with high mortality and rising incidence rates in Europe despite prevention efforts. Nodular MM, the most aggressive subtype, often mimics other skin tumors, complicating diagnosis. We present the case of a 66-year-old woman with a large, ulcerated tumor beneath the left scapula, along with multiple nodular lesions on the left arm and chest. Initially suspected to be an aggressive squamous cell carcinoma, the diagnosis was confirmed as invasive cutaneous MM with a BRAF(V600) mutation via biopsy. Staging with PET/CT revealed extensive glucose metabolism in the tumors and surrounding tissues, as well as metastatic lymphadenopathy. The disease was classified as stage IV (T4bN3cM1a0). Neoadjuvant systemic therapy with BRAF and MEK inhibitors (Dabrafenib and Trametinib) was initiated to reduce tumor size. Remarkable regression was observed within a week, with further reduction in tumor size after one month. A follow-up PET/CT after 3 months showed significant decreases in tracer uptake and lesion size, with a ΔSUVmax of 51.9%, a ΔMTV of 74.5%, and a ΔTLG of 83.5%, indicating an excellent response to targeted therapy. Full article
(This article belongs to the Special Issue Diagnostic Insights for Skin Melanoma)
Show Figures

Figure 1

Figure 1
<p>Cutaneous malignant melanoma (MM) is the most aggressive form of skin cancer, associated with a high mortality rate and accounting for over 80% of skin cancer deaths [<a href="#B1-diagnostics-14-02860" class="html-bibr">1</a>]. In Europe, despite the implementation of primary and secondary prevention methods, its incidence continues to rise [<a href="#B2-diagnostics-14-02860" class="html-bibr">2</a>]. Nodular MM is the most aggressive subtype of MM, and its clinical presentation can sometimes be misleading due to its resemblance to other skin tumors such as seborrheic keratosis, squamous cell carcinoma, nodular basal cell carcinoma, or pyogenic granuloma [<a href="#B3-diagnostics-14-02860" class="html-bibr">3</a>,<a href="#B4-diagnostics-14-02860" class="html-bibr">4</a>,<a href="#B5-diagnostics-14-02860" class="html-bibr">5</a>]. Here, we present the case of a 66-year-old woman who was referred to our clinic with a large, round, ulcerated tumor mass measuring 13 cm in diameter located beneath the left scapula along with multiple nodular skin lesions of varying sizes on the left arm and left side of the chest. ((<b>A</b>), arrow) Voluminous cutaneous tumor in the left scapula; ((<b>B</b>), circles) multiple nodular skin lesions of varying sizes on the left arm and left side of the chest: specifically, in the upper lateral and medial quadrants of the breast and in the axilla. The patient reported that the lesions appeared two years ago and have been growing progressively since then. In recent months, the primary lesion has enlarged significantly, and satellite lesions have begun to emerge. During physical examination, we observed multiple enlarged lymph nodes in the left axilla, which were fixed to the surrounding soft tissue. Based on the clinical characteristics, an aggressive form of squamous cell carcinoma with satellitosis was initially hypothesized. She was submitted to incisional biopsy, positive for ulcerated MM, positive for BRAF(V600) mutation.</p>
Full article ">Figure 2
<p>A tissue fragment measuring 1.5 × 1 × 0.3 cm was excised. Microscopic examination revealed a neoplasm composed of atypical melanocytes, organized in nests and solid cords, with a proliferative index (Ki67) of 20% and positivity for immunohistochemical markers typical of melanoma (S100+, MelanA+, HMB45+, and panCKAE1/AE3-) [<a href="#B6-diagnostics-14-02860" class="html-bibr">6</a>]. Due to the limited size of the sample, the Clark level and Breslow thickness could not be determined. A positron mission computed tomography (PET/CT) with <sup>18</sup>F-fluoro-deoxyglucose ([<sup>18</sup>F]-FDG) was requested as a staging procedure.</p>
Full article ">Figure 3
<p>Whole-body [<sup>18</sup>F]FDG PET/CT showed increased glucose metabolism in the cutaneous and subcutaneous tissue surrounding the tumors, as well as in several lymph node stations (right axillary lymph nodes and, on the left side, axillary, supraclavicular, and along the internal mammary chain) ((<b>A</b>), arrows). High tracer uptake in the cutaneous lesion of the left scapula was clearly visible in the fused coronal ((<b>B</b>), arrow) and axial ((<b>C</b>), arrow) views, as well as in the fused coronal slice of the left breast region ((<b>D</b>), arrow). Furthermore, coronal images showed pericardial effusion ((<b>D</b>), yellow bordered arrow). Quantitative PET-derived parameters for the largest scapular region were as follows: maximum standardized uptake value (SUVmax), 10.4; metabolic tumor volume (MTV), 52 cc; and total lesion glycolysis (TLG), 267 g/mL. No visceral or skeletal metastases were detected. Subsequent cerebral magnetic resonance imaging (MRI) did not identify any brain metastases. According to the 8th edition of the AJCC TNM Classification, the disease stage was determined to be T4bN3cM1a0, corresponding to stage IV [<a href="#B7-diagnostics-14-02860" class="html-bibr">7</a>]. Given the locally advanced and large primary tumor along with the presence of multiple metastatic lymphadenopathies, we opted for neoadjuvant systemic treatment. Our aim was to reduce the tumor size to make resection feasible, thereby potentially improving the patient’s quality of life. The patient underwent combined targeted therapy with BRAF and MEK inhibitors: Dabrafenib 150 mg b.i.d. plus Trametinib 2 mg q.d. After only one week, the outcome was remarkable, with partial regression of both primary tumor and metastases, presence of necrotic areas, and size reduction, confirmed by further regression of the lesions upon objective evaluation at 1 month.</p>
Full article ">Figure 4
<p>After 1 month of treatment, the primary tumor significantly shrank, reaching 5 cm in diameter ((<b>A</b>), arrow) as well the other localizations in the left breast region ((<b>B</b>), circles).</p>
Full article ">Figure 5
<p>Follow-up PET/CT scan after 3 months, confirming response to treatment. Whole-body PET/CT showed a marked regression in the previously described foci of increased tracer uptake (<b>A</b>). Fused axial coronal ((<b>B</b>), arrow) and axial ((<b>C</b>), arrow) images demonstrated a significant reduction in tracer uptake and lesion size, clearly visible on both visual and quantitative analysis (SUVmax: 5, MTV: 14 cc, TLG: 44 g/mL), with a ΔSUVmax of 51.9%, a ΔMTV of 74.5%, and a ΔTLG of 83.5%. In addition, the fused coronal image demonstrated regression of the hypermetabolic lesions in the left breast region, as well as disappearance of the pericardial effusion ((<b>D</b>), arrow). After an additional three months, the patient underwent excision of the skin tumor in the left scapular region. Six additional months have now passed since the surgical excision, and the patient continues to be under active follow-up. The physical examination of the post-surgical scar has not shown any macroscopic signs of loco-regional recurrence so far, supporting the radicality of the intervention. She remains on molecularly targeted therapy, which is currently well tolerated. Despite the relatively short follow-up period, no signs of resistance to treatment have emerged to date. While we acknowledge the limitation related to the duration of follow-up, we believe this case highlights the potential of BRAF/MEK-targeted therapy in the neoadjuvant setting for managing a particularly large and aggressive melanoma. Cutaneous MM remains one of the most aggressive forms of skin cancer, with high rates of metastasis and poor prognosis, particularly in advanced stages [<a href="#B8-diagnostics-14-02860" class="html-bibr">8</a>]. The complexity in diagnosing and managing MM stems from its highly variable clinical presentation, which can often be misleading. Nodular melanoma, a notably aggressive subtype, is frequently mistaken for other skin lesions, such as seborrheic keratosis, basal cell carcinoma, Merkel cell carcinoma, or pyogenic granulomas [<a href="#B9-diagnostics-14-02860" class="html-bibr">9</a>]. This diagnostic challenge underscores the value of advanced imaging techniques in enhancing diagnostic accuracy and informing effective disease management [<a href="#B10-diagnostics-14-02860" class="html-bibr">10</a>]. Giant nodular melanomas are exceedingly rare but represent significant clinical and surgical challenges due to their destructive nature and poor prognosis [<a href="#B11-diagnostics-14-02860" class="html-bibr">11</a>]. Despite their high metastatic potential, a few cases in the literature have shown unexpectedly benign courses. To our knowledge, this is the first reported case of a metastatic giant cutaneous melanoma demonstrating an early remarkable response after targeted treatment, assessed both clinically and by [<sup>18</sup>F]FDG PET/CT scan. In a similar case described by Kruijff et al., a patient with stage IV cutaneous melanoma (T4N2M1c1) underwent palliative surgery, but despite intervention, the outcome was fatal within 12 weeks [<a href="#B12-diagnostics-14-02860" class="html-bibr">12</a>]. With the introduction of immunotherapies such as Ipilimumab in 2011, followed by PD-1, BRAF, and MEK inhibitors, long-term survival prospects for melanoma patients have improved significantly [<a href="#B13-diagnostics-14-02860" class="html-bibr">13</a>]. Adjuvant therapy has become the standard approach in advanced melanoma cases [<a href="#B14-diagnostics-14-02860" class="html-bibr">14</a>]. However, in our case, the rapid clinical response highlights the potential of targeted therapy as a primary approach. Monitoring treatment efficacy through visible changes in cutaneous lesions has provided critical insights into disease progression and response to therapy. Additionally, the atypical presentation of MM in this case—mimicking squamous cell carcinoma—emphasizes the need to consider a broad spectrum of differential diagnoses, encouraging vigilance among healthcare providers when confronted with unusual clinical features. This also reinforces the importance of personalized treatment approaches and showcases the efficacy of targeted therapies as neoadjuvant treatments in advanced MM management. Neoadjuvant therapy is a promising approach for patients with advanced melanoma [<a href="#B15-diagnostics-14-02860" class="html-bibr">15</a>,<a href="#B16-diagnostics-14-02860" class="html-bibr">16</a>]. However, due to a lack of randomized clinical trials definitively proving its superiority over traditional surgery with postoperative adjuvant treatment, neoadjuvant therapies have not yet become standard practice. A recently published study showed the potential of Dabrafenib plus Vemurafenib as neoadjuvant therapy, reporting a high rate of complete pathological responses (around 50%) and sustained disease control post-surgery, with relapse rates of around 17% [<a href="#B16-diagnostics-14-02860" class="html-bibr">16</a>]. The effectiveness of neoadjuvant therapy may be attributed to heightened immune system activity in response to a large primary tumor, promoting a stronger tumor-specific immune response. Excision of the primary tumor may subsequently alter its microenvironment, possibly diminishing the local immune response. However, it should be underlined that, while the rapid and impressive therapeutic response observed may suggest a potential contribution of immune modulation, it is important to note that, in the context of BRAF/MEK-targeted therapy, the role of the tumor microenvironment remains less well defined and less certain compared to its established importance in immunotherapy, though emerging evidence indicates it could play a supportive role [<a href="#B17-diagnostics-14-02860" class="html-bibr">17</a>]. In our case, imaging studies, particularly [<sup>18</sup>F]FDG PET/CT, were essential for an accurate assessment of disease extent. We opted for [<sup>18</sup>F]FDG PET/CT over total-body CT in our case due to its superior sensitivity and specificity in detecting non-pulmonary metastases in advanced melanoma [<a href="#B18-diagnostics-14-02860" class="html-bibr">18</a>]. This imaging modality provides enhanced metabolic imaging to complement anatomical details, which is particularly valuable in a setting where early and accurate staging can significantly influence therapeutic decisions. Moreover, [<sup>18</sup>F]FDG PET/CT entails a lower radiation burden than total-body contrast-enhanced CT, which is particularly relevant in patients requiring repeated imaging during treatment and follow-up. In our patient, the PET scan revealed elevated glucose metabolism in the primary tumor and in multiple regional lymph nodes, confirming metastatic disease. This finding significantly influenced our decision to initiate neoadjuvant therapy, aiming to reduce tumor size and improve surgical outcomes. PET/CT imaging is invaluable for staging and prognosis in melanoma patients [<a href="#B19-diagnostics-14-02860" class="html-bibr">19</a>,<a href="#B20-diagnostics-14-02860" class="html-bibr">20</a>]. Its ability to assess metabolic activity and identify regions of increased glucose uptake is critical for determining disease extent and informing accurate staging. This is particularly relevant in cases like ours, where clinical examination alone may not fully reveal metastatic spread. For instance, in our patient, [<sup>18</sup>F]FDG PET/CT revealed both the primary tumor and metastatic involvement in multiple lymph node stations, guiding our treatment strategy. Moreover, PET imaging serves as a powerful prognostic tool. Recent studies have indicated that percentage changes, the so-called “delta” (Δ), in PET-derived quantitative parameters (e.g., SUVmax, MTV, and TLG) correlate strongly with patient outcome in several tumors, including MM [<a href="#B21-diagnostics-14-02860" class="html-bibr">21</a>,<a href="#B22-diagnostics-14-02860" class="html-bibr">22</a>]. In our case, the significant Δ values obtained after 3 months of neoadjuvant therapy correlated significantly with the patient’s clinical objectivity and support the utility of PET imaging and PET-derived quantitative parameters in the more aggressive cases of melanoma, not only for staging but also for the early follow-up and prognostic stratification of patients.</p>
Full article ">
22 pages, 8152 KiB  
Article
Is Silver a Precious Metal for G-Quadruplex Stabilization Mediated by Porphyrins?
by Nuno M. M. Moura, Sofia Guedes, Diana Salvador, Helena Oliveira, M. Graça P. M. S. Neves and Catarina I. V. Ramos
Int. J. Mol. Sci. 2024, 25(24), 13556; https://doi.org/10.3390/ijms252413556 - 18 Dec 2024
Viewed by 263
Abstract
Cancer is a leading cause of death, so continuous efforts into cancer therapy are imperative. In tumor cells, telomerase and oncogene activity are key points for uncontrolled cell growth. Targeting these processes with ligands that inhibit telomerase and/or reduce oncogene expression has been [...] Read more.
Cancer is a leading cause of death, so continuous efforts into cancer therapy are imperative. In tumor cells, telomerase and oncogene activity are key points for uncontrolled cell growth. Targeting these processes with ligands that inhibit telomerase and/or reduce oncogene expression has been identified as a promising cancer therapy. This study evaluated the selectivity and affinity of the silverII complex of 5,10,15,20-tetrakis(N-methyl-4-pyridinium)porphyrin (AgTMPyP) to stabilize DNA sequences capable of forming G4 structures mimicking the telomeric and oncogene regions, using spectroscopic, biochemical methods and in vitro assays. The tetracationic silver complex was compared with the free base, H2TMPyP, and the zincII complex, ZnTMPyP. The results obtained from UV-Vis and fluorescence methods pointed to a great affinity and good selectivity of AgTMPyP to G4 structures, especially for the oncogene MYC. In general, an increase in the ability of the studied ligands for 1O2 generation when interacting with oncogenic and telomeric G4 sequences was found. The results of the PCR stop assays proved that AgTMPyP has the ability to inhibit Taq polymerase. Additionally, in vitro assays demonstrated that the silverII complex exhibits low cytotoxicity against HaCaT— an immortalized, non-tumorigenic, skin keratinocytes cell line—and, although nonexclusive, AgTMPyP shows nuclear co-localization. Full article
(This article belongs to the Collection Feature Paper Collection in Biochemistry)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of cationic metalloporphyrins <b>AgTMPyP</b> and <b>ZnTMPyP,</b> and their free-base counterpart <b>H<sub>2</sub>TMPyP.</b></p>
Full article ">Figure 2
<p>UV–Vis spectra obtained from the titration of <b>AgTMPyP</b> (2 × 10<sup>−6</sup> M) in PBS (10 mM KH<sub>2</sub>PO<sub>4</sub>, 10 mM K<sub>2</sub>HPO<sub>4</sub> and 100 mM KCl; pH 6.8), with 0.12 to 2.5 equiv. of (<b>A</b>) G4 Tel, (<b>B</b>) <span class="html-italic">MYC</span>, (<b>C</b>) <span class="html-italic">KRAS</span>, and (<b>D</b>) ds26.</p>
Full article ">Figure 3
<p>UV–Vis spectra obtained from the titration of <b>ZnTMPyP</b> (2 × 10<sup>−6</sup> M) in PBS (10 mM KH<sub>2</sub>PO<sub>4</sub>, 10 mM K<sub>2</sub>HPO<sub>4</sub>, and 100 mM KCl; pH 6.8), with increasing amounts (0.125 to 2.5 equiv.) of (<b>A</b>) G4 Tel, (<b>B</b>) <span class="html-italic">MYC</span>, (<b>C</b>) <span class="html-italic">KRAS</span>, and (<b>D</b>) ds26.</p>
Full article ">Figure 4
<p>Fluorescence spectra obtained in the titrations of <b>AgTMPyP</b> (2 × 10<sup>−6</sup> M) in PBS (10 mM KH<sub>2</sub>PO<sub>4</sub>, 10 mM K<sub>2</sub>HPO<sub>4</sub>, and 100 mM KCl; pH 6.8), with 0.125 to 2.5 equivalents of (<b>A</b>) G4 Tel, (<b>B</b>) <span class="html-italic">MYC</span>, (<b>C</b>) <span class="html-italic">KRAS</span>, and (<b>D</b>) ds26.</p>
Full article ">Figure 5
<p>Fluorescence spectra obtained in the titrations of <b>ZnTMPyP</b> (2 × 10<sup>−6</sup> M) in PBS (10 mM KH<sub>2</sub>PO<sub>4</sub>, 10 mM K<sub>2</sub>HPO<sub>4</sub>, and 100 mM KCl; pH 6.8), with 0.125 to 2.5 equivalents of (<b>A</b>) G4 Tel, (<b>B</b>) <span class="html-italic">MYC</span>, (<b>C</b>) <span class="html-italic">KRAS</span>, and (<b>D</b>) ds26.</p>
Full article ">Figure 6
<p>Results from FID experiments for (<b>A</b>) <b>AgTMPyP</b> and (<b>B</b>) <b>ZnTMPyP</b>. TO (3.5 µM)–DNA (1 µM) adduct solutions prepared in PBS (10 mM KH<sub>2</sub>PO<sub>4</sub>, 10 mM K<sub>2</sub>HPO<sub>4</sub>, and 100 mM KCl; pH 6.8).</p>
Full article ">Figure 7
<p>Time-dependent photodecomposition of DPiBF at 50 × 10<sup>−6</sup> M in the presence of (<b>A</b>) <b>AgTMPyP, ZnTMPyP,</b> and <b>H<sub>2</sub>TMPyP</b> (0.5 × 10<sup>−6</sup> M); (<b>B</b>) comparison between <b>AgTMPyP</b> and <b>AgTMPyP</b>–DNA adducts; (<b>C</b>) comparison between <b>ZnTMPyP</b> and <b>ZnTMPyP</b>–DNA adducts; (<b>D</b>) comparison between <b>H<sub>2</sub>TMPyP</b> and <b>H<sub>2</sub>TMPyP</b>–DNA adducts.</p>
Full article ">Figure 8
<p>(<b>A</b>) Polyacrylamide gel electrophoresis of Pu77 (left) and Pu77-mut (right) PCR amplification products in the presence of increased amounts (0 to 16 equivalents) of <b>AgTMPyP</b> (1 equivalent = 4 × 10<sup>−7</sup> M). (<b>B</b>) Representative analysis of the PCR products. The same colors code is represented in both figures.</p>
Full article ">Figure 9
<p>Viability profile of HaCaT cell culture upon 24 h of incubation with 5 to 100 × 10<sup>−6</sup> M of <b>AgTMPyP</b>, <b>ZnTMPyP,</b> and <b>H<sub>2</sub>TMPyP</b>. Results are expressed as the mean ± SD, where ∗ indicates statistically significant differences compared to the control with <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 10
<p>Confocal images of HaCaT cells treated for 24 h with <b>AgTMPyP</b> and <b>H<sub>2</sub>TMPyP</b>, both at a concentration of 50 × 10<sup>−6</sup> M. DAPI was used as a reference for nucleus co-localization.</p>
Full article ">
18 pages, 939 KiB  
Review
Targeting Matrix Metalloproteinases and Their Inhibitors in Melanoma
by Orest Szczygielski, Emilia Dąbrowska, Sylwia Niemyjska, Andrzej Przylipiak and Monika Zajkowska
Int. J. Mol. Sci. 2024, 25(24), 13558; https://doi.org/10.3390/ijms252413558 - 18 Dec 2024
Viewed by 243
Abstract
Malignant melanoma is one of the most important dermatological neoplasms. The high mortality rate associated with this skin disease is primarily due to the occurrence of metastases, while the diagnosis and treatment of melanoma in its early stages has a favorable prognosis. Early [...] Read more.
Malignant melanoma is one of the most important dermatological neoplasms. The high mortality rate associated with this skin disease is primarily due to the occurrence of metastases, while the diagnosis and treatment of melanoma in its early stages has a favorable prognosis. Early detection is crucial because the success of treatment is directly related to the depth of cancerous growth. The family of matrix metalloproteinases (MMPs) plays a critical role in the initiation and progression of melanoma. Prominent MMPs, including MMP-1, MMP-2, MMP-3, MMP-9, MMP-13, and MMP-14, have been shown to significantly contribute to the development of melanoma. The tumor microenvironment, particularly the extracellular matrix (ECM), has emerged as a critical factor in modulating cancer progression. This review focuses on the role of matrix metalloproteinases and their inhibitors in ECM degradation and the subsequent progression of melanoma, as well as their potential as therapeutic targets. Full article
Show Figures

Figure 1

Figure 1
<p>Roles of MMPs in tumor progression, invasion, and metastases.</p>
Full article ">Figure 2
<p>Schematic mechanism of action of various MMPIs in melanoma.</p>
Full article ">
16 pages, 29464 KiB  
Review
Molecular Subtypes of Vulvar Squamous Cell Carcinoma: The Significance of HPV-Independent/p53 Wild Type
by Lars-Christian Horn, Christine E. Brambs, Blake Gilks, Lien Hoang, Naveena Singh, Grit Gesine Ruth Hiller, Kathrin Hering, Jessica N. McAlpine, Amy Jamieson, Mona Alfaraidi, Bahriye Aktas, Nadja Dornhöfer and Anne Kathrin Höhn
Cancers 2024, 16(24), 4216; https://doi.org/10.3390/cancers16244216 - 18 Dec 2024
Viewed by 276
Abstract
Vulvar carcinoma is a rare disease, meeting the criteria for a “rare cancer”, but its incidence is increasing, especially in women <60 years of age. Squamous cell carcinoma (VSCC) accounts for the overwhelming majority of vulvar carcinomas and is the focus of this [...] Read more.
Vulvar carcinoma is a rare disease, meeting the criteria for a “rare cancer”, but its incidence is increasing, especially in women <60 years of age. Squamous cell carcinoma (VSCC) accounts for the overwhelming majority of vulvar carcinomas and is the focus of this review. As with many cancers, the increased understanding of molecular events during tumorigenesis has led to the emergence of the molecular subclassification of VSCC, which is subclassified into tumors that arise secondary to high-risk human papillomavirus infection (HPV-associated, or HPVa) and those that arise independently of HPV (HPVi), most commonly in the setting of a chronic inflammatory condition of the vulvar skin. This latter group of HPVi VSCC arises in most cases secondary to mutations in TP53, but recently, attention has focused on the uncommon TP53 wild-type HPVi VSCC. These three molecular subtypes of VSCC (HPVa, HPVi p53 abnormal, and HPVi p53 wild type), as well as their precursor lesions, cannot be diagnosed based on a routine histopathological examination or immunostaining for p53 and p16 as surrogate markers for TP53 mutation and high-risk HPV infection, respectively, are required. The molecular subtyping of VSCC shows high reproducibility and provides important prognostic information. HPVa VSCC has the most favorable prognosis, while HPVi VSCC with TP53 mutations (p53abn) has the worst prognosis, and HPVi VSCC with wild-type TP53 (p53wt) has an intermediate prognosis. In this review, we discuss the evidence supporting this molecular subclassification and its implications for the diagnosis and treatment of VSCC and its precursors. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Subdivision of squamous cell carcinomas of the lower female genital tract based on HPV association according to the recommendations of the WHO classification 2020 [<a href="#B28-cancers-16-04216" class="html-bibr">28</a>]; (<b>b</b>): Classification of VSCC from morphology-based to morpho-molecular classification, depending on the etiology of the disease, using p16 and p53 immunohistochemistry [<a href="#B10-cancers-16-04216" class="html-bibr">10</a>,<a href="#B12-cancers-16-04216" class="html-bibr">12</a>,<a href="#B14-cancers-16-04216" class="html-bibr">14</a>,<a href="#B20-cancers-16-04216" class="html-bibr">20</a>].</p>
Full article ">Figure 2
<p>Summary of the recently described three-tiered molecular classification of VSCC and its precursors (please see text) (uVIN = usual VIN; dVIN = differentiated VIN; vaVIN = verrucous VIN).</p>
Full article ">Figure 3
<p>Patterns of p16 overexpression in non-keratinizing (<b>a</b>) and keratinizing (<b>b</b>) VSCC, so-called block-like staining. Note that the keratinized areas stain negative for a p16 pattern (asterisk; see text).</p>
Full article ">Figure 4
<p>An algorithmic approach for the three-tiered molecular subclassification of VSCC using p16 and p53 immunohistochemistry [<a href="#B14-cancers-16-04216" class="html-bibr">14</a>]. ISH = in situ hybridization.</p>
Full article ">Figure 5
<p>Immunohistochemical classification of VSCC into its molecular subtypes (for details, please see the text and <a href="#cancers-16-04216-f004" class="html-fig">Figure 4</a> and <a href="#cancers-16-04216-f006" class="html-fig">Figure 6</a> and <a href="#cancers-16-04216-t001" class="html-table">Table 1</a>).</p>
Full article ">Figure 6
<p>Algorithmic approach in routine pathology workup for the three-tiered molecular classification of VSCC [<a href="#B10-cancers-16-04216" class="html-bibr">10</a>,<a href="#B14-cancers-16-04216" class="html-bibr">14</a>,<a href="#B40-cancers-16-04216" class="html-bibr">40</a>].</p>
Full article ">Figure 7
<p>A 57-year-old woman with a poorly differentiated squamous cell carcinoma ((<b>a</b>); FIGO stage IVA) with p16 block-type staining overexpression (<b>b</b>) and p53 wild-type expression staining pattern (scattered; (<b>c</b>)) on immunohistochemistry, treated using hypofractionated radiation with a total dose of 38 Gy and sequential chemotherapy consisting of 5 cycles of carboplatin AUC 5 and paclitaxel 175 mg/m<sup>2</sup>. (<b>d</b>) Prior treatment: 25 cm × 10 cm exophytic superficially ulcerated tumor. (<b>e</b>) Partial clinical response after three months of treatment. (<b>f</b>) Complete local clinical response after 10 months. There was no local recurrence within 363 months of follow-up.</p>
Full article ">Figure 8
<p>Summary of the three-tiered molecular classification of VSCC.</p>
Full article ">
10 pages, 12001 KiB  
Case Report
Endocrine Mucin-Producing Sweat Gland Carcinoma (EMPSGC) in a Dog: Immunohistochemical Characterization
by Warisraporn Tangchang, Gi-young Jung, Jun-yeop Song, Poornima Kumbukgahadeniya, Dae-hyun Kim, Hyo-jung Kwon and Hwa-young Son
Animals 2024, 14(24), 3637; https://doi.org/10.3390/ani14243637 - 17 Dec 2024
Viewed by 248
Abstract
A seven-year-old spayed female dog presented with multiple, small oval-shaped masses on the abdominal skin. The excised tissue was submitted for histopathological evaluation. Routine histology revealed basaloid nodules with stippled chromatin and pleomorphic nuclei. Morphologically, the tumor resembled a solid papillary carcinoma of [...] Read more.
A seven-year-old spayed female dog presented with multiple, small oval-shaped masses on the abdominal skin. The excised tissue was submitted for histopathological evaluation. Routine histology revealed basaloid nodules with stippled chromatin and pleomorphic nuclei. Morphologically, the tumor resembled a solid papillary carcinoma of the breast or an endocrine mucin-producing sweat gland carcinoma (EMPSGC). EMPSGC is a rare neuroendocrine-differentiated neoplasm. This study aimed to compare different immunohistochemical markers for distinguishing between canine sweat gland carcinoma (SGC) and mammary gland carcinoma (MGC). CK19 and Sox9 were useful in identifying SGC. Additionally, the tumor was positive for CK5, AE1/AE3+CK8/18, p63, vimentin, E-cadherin, and synaptophysin, while estrogen receptor staining was negative. The final diagnosis of canine EMPSGC was based on the tumor’s morphological and neuroendocrine immunohistopathological features. Our report provides the first detailed description of a canine EMPSGC, an exceedingly rare tumor in dogs. Full article
(This article belongs to the Section Veterinary Clinical Studies)
Show Figures

Figure 1

Figure 1
<p>Gross finding figure. (<b>a</b>) The skin shows bluish discoloration, and numerous nodules form irregular mass that protrude into the skin of case 1. (<b>b</b>) Removed tumor mass from case 1; the mass is firmly adhered to the skin and subcutaneous connective tissue. (<b>c</b>) A removed mass (7 cm × 6 cm) in the right 2nd mammary gland from case 2.</p>
Full article ">Figure 2
<p>Histological examination of both cases. (<b>a</b>) The tumor displayed a thickened stroma of lobular. The multilobular tumor exhibits mucin secretion in the cystic and solid areas, as observed in case 1, Mag. = ×200. (<b>b</b>) Within the lobules, peripheral palisading was identified in some areas. Nuclei were bland with moderate pleomorphism and diffusely stippled chromatin from case 1, Mag. = ×400. (<b>c</b>) The irregular proliferation of small glands of case 2, Mag. = ×100. (<b>d</b>) The tubules were lined by a single layer of cuboidal or columnar cell hyperplasia with atypia of case 2, Mag. = ×400, hematoxylin and eosin (HE).</p>
Full article ">Figure 3
<p>Immunoreactivity of cytokeratin (CK). Case 1 (sweat gland carcinoma), (<b>a</b>) luminobasal tumor cells were positive for CK5, (<b>b</b>) luminal tumor cells were positive for AE1/AE3+CK8/18, (<b>c</b>) CK19 was cytoplasmic, and cell membrane expression. Case 2 (mammary gland carcinoma), (<b>d</b>) CK5 was positive, (<b>e</b>) AE1/AE3+CK8/18 and (<b>f</b>) CK19 showed no immunoreactivity, immunohistochemistry (IHC).</p>
Full article ">Figure 4
<p>Immunoreactivity of myoepithelial and other markers. Case 1 (sweat gland carcinoma), (<b>a</b>) nuclear was positive for p63, (<b>b</b>) cytoplasmic areas were positive for vimentin, (<b>c</b>) Sox9 found nuclear expression. Case 2 (mammary gland carcinoma), (<b>d</b>) p63 and (<b>e</b>) vimentin were positive in myoepithelial cells, and (<b>f</b>) Sox9 was negative expression, immunohistochemistry (IHC).</p>
Full article ">Figure 5
<p>Immunoreactivity of endocrine and other markers. Case 1 (sweat gland carcinoma), (<b>a</b>) membranous and cytoplasmic areas were positive for E-cadherins, (<b>b</b>) cytoplasmic expression of synaptophysin, (<b>c</b>) nuclear was negative for estrogen. Case 2 (mammary gland carcinoma), (<b>d</b>) E-cadherins showed weak expression, (<b>e</b>) synaptophysin was cytoplasmic expression, and (<b>f</b>) nuclear cell showed positive for estrogen, immunohistochemistry (IHC).</p>
Full article ">
18 pages, 2161 KiB  
Article
Syndecan-1 and E-Cadherin Expression in Canine Cutaneous Squamous Cell Carcinoma
by Rita Files, Cláudia Cardoso, Justina Prada, Filipe Silva and Isabel Pires
Vet. Sci. 2024, 11(12), 652; https://doi.org/10.3390/vetsci11120652 - 14 Dec 2024
Viewed by 404
Abstract
Cutaneous squamous cell carcinoma (CSCC) in dogs is a locally invasive tumor that typically occurs in areas of poorly pigmented skin due to sun exposure. Identifying new biomarkers, such as syndecan-1 (CD138) and E-cadherin, is fundamental for tumor diagnosis and prognosis. Dysregulation of [...] Read more.
Cutaneous squamous cell carcinoma (CSCC) in dogs is a locally invasive tumor that typically occurs in areas of poorly pigmented skin due to sun exposure. Identifying new biomarkers, such as syndecan-1 (CD138) and E-cadherin, is fundamental for tumor diagnosis and prognosis. Dysregulation of syndecan-1, expressed in epithelial tissue, fibroblasts, and plasma cells, is associated with poor prognosis in several types of cancer. Similarly, E-cadherin, which plays a crucial role in cell adhesion and epithelial functionality, is also linked to adverse outcomes. This study evaluated the expression of syndecan-1 and E-cadherin in 47 cases of canine cutaneous squamous cell carcinoma. The results showed that the intensity of syndecan-1 decreased with increasing tumor aggressiveness, and its presence in the stroma was significantly associated with tumor grade. E-cadherin also demonstrated a decrease in intensity with increasing malignancy. However, the association between syndecan-1 and E-cadherin was not statistically significant. E-cadherin reduction and stromal syndecan-1 positivity seem to be associated with tumor aggressiveness in canine cutaneous squamous cell carcinoma. Further studies are needed to explore their roles in tumor progression. Full article
Show Figures

Figure 1

Figure 1
<p>Histological grade of malignancy.</p>
Full article ">Figure 2
<p>Syndecan-1 labeling intensity in tumors with different histological grades of malignancy.</p>
Full article ">Figure 3
<p>Syndecan-1 stromal immunoexpression in tumors with different histological grades of malignancy.</p>
Full article ">Figure 4
<p>Syndecan-1 immunoexpression in canine cutaneous squamous cell carcinoma. (<b>A</b>) Membranous labeling with moderate intensity (2) in a well-differentiated tumor (Grade I); (<b>B</b>) Membranous and cytoplasmatic expression with moderate intensity (2) in a well-differentiated tumor (Grade I); (<b>C</b>) Week cytoplasmatic and nuclear labeling in moderately differentiated tumor (Grade II); (<b>D</b>) Stroma immunoexpression in a poorly differentiated tumor (Grade III).</p>
Full article ">Figure 5
<p>E-cadherin labeling intensity in tumors with different histological grades of malignancy.</p>
Full article ">Figure 6
<p>E-cadherin immunoexpression in canine cutaneous squamous cell carcinoma. (<b>A</b>) Membranous labeling with strong intensity (3) in a well-differentiated tumor (Grade I); (<b>B</b>) Membranous and cytoplasmatic expression with strong intensity (3) in a moderately differentiated tumor (Grade II); (<b>C</b>) Moderate membranous labeling in poorly differentiated tumor (Grade III); (<b>D</b>) Weak membranous labeling in poorly differentiated tumor (Grade III).</p>
Full article ">
10 pages, 2890 KiB  
Article
Tualang Honey Has a Protective Effect Against Photodamage and Skin Cancer: An In Vivo Study
by Mohammed Asif Sherwani, Erin M. Burns, Israr Ahmad, Ahmed Omar Jasser, Ariq Chandra and Nabiha Yusuf
Nutrients 2024, 16(24), 4314; https://doi.org/10.3390/nu16244314 - 13 Dec 2024
Viewed by 370
Abstract
Background/Objective: Ultraviolet (UV) B radiation leads to DNA damage by generating cyclobutane pyrimidine dimers (CPDs). UVB-induced CPDs can also result in immune suppression, which is a major risk factor for non-melanoma skin cancer (NMSC). UVB-induced CPDs are repaired by nucleotide repair mechanisms (NER) [...] Read more.
Background/Objective: Ultraviolet (UV) B radiation leads to DNA damage by generating cyclobutane pyrimidine dimers (CPDs). UVB-induced CPDs can also result in immune suppression, which is a major risk factor for non-melanoma skin cancer (NMSC). UVB-induced CPDs are repaired by nucleotide repair mechanisms (NER) mediated by xeroderma pigmentosum complementation group A (XPA). The purpose of this study was to investigate the use of TH as a chemopreventive agent against the development of skin cancer. Method: SKH-1 hairless mice were exposed were fed with TH (0.1% v/v) for two weeks and exposed to a single dose of UVB (180 mJ/cm2). Dorsal skin was harvested 24 h post-UVB exposure for evaluation of DNA damage and repair. Lymph nodes were also harvested to prepare single cell suspension for flow cytometric evaluation. For carcinogenesis experiments, SKH-1 hairless mice were given TH (0.1% v/v) ad libitum and exposed to UVB (180 mJ/cm2) thrice a week for 30 weeks. Results: Feeding SKH-1 hairless mice with TH (0.1% v/v) for two weeks prior to a single dose of UVB (180 mJ/cm2) led to a significant increase in XPA in skin and DNA repair cytokines IL-12 and IL-23 in draining lymph nodes. Furthermore, when subjected to the photocarcinogenesis protocol; mice fed with TH developed significantly fewer tumors in comparison to mice fed on drinking water. Conclusions: Our data demonstrate that TH has a protective effect against UVB-induced DNA damage, immune suppression, and skin cancer. Future studies will further investigate the potential of TH as a preventive treatment for NMSC. Full article
(This article belongs to the Special Issue Dietary Supplements and Cancer Prevention)
Show Figures

Figure 1

Figure 1
<p>TH 0.1% augments the repair of UVB-induced CPD. (<b>A</b>) Representative images showing CPD+ cells (in green) in frozen skin sections post-UVB exposure. (<b>B</b>) ELISA quantification of CPDs. (<b>C</b>) Real-time PCR analysis of XPA mRNA expression. Data represent mean ± SD for five animals per group (*** <span class="html-italic">p</span> &lt; 0.001, ns: not significant). CPD, cyclobutane pyrimidine dimer; TH, Tualang honey; UVB, ultraviolet B. Scale = 50 µM.</p>
Full article ">Figure 2
<p>TH 0.1% increases IL-12p35- and IL-23p19-producing CD11c cells in UVB-irradiated mice. Percentages of IL-12p35- and IL-23p19-producing CD11c+ cells in draining lymph nodes were analyzed by flow cytometry 24 h post-UVB exposure. Data represent mean ± SD for five animals per group (* <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, ns: not significant). TH, tualang honey; UVB, ultraviolet B.</p>
Full article ">Figure 3
<p>TH 0.1% decreases inflammation and CD11b+Gr-1+ myeloid cell infiltration in UVB-irradiated mice. (<b>A</b>) Representative H&amp;E-stained skin sections showing reduced inflammation in TH-treated mice compared to controls. (<b>B</b>) Flow cytometry analysis of CD11b+Gr-1+ myeloid cells in lymph nodes. Data represent mean ± SD for five animals per group (*** <span class="html-italic">p</span> &lt; 0.001, ns: not significant). TH, tualang honey; UVB, ultraviolet B. Scale = 50 µM.</p>
Full article ">Figure 4
<p>TH 0.1% inhibits tumor development in UVB-irradiated mice. SKH-1 hairless mice were exposed to UVB (180 mJ/cm<sup>2</sup>) thrice a week for 30 weeks. Panels of mice received 0.1% TH or plain drinking water ad libitum throughout the course of experiment. Tumor number/tumor burden were monitored weekly. Mice that received TH had significantly fewer tumors and a lower tumor burden in comparison to the mice receiving plain drinking water. Experiments were conducted in 10 mice per group. (*** <span class="html-italic">p</span> &lt; 0.001). TH, tualang honey; UVB, ultraviolet B.</p>
Full article ">
22 pages, 838 KiB  
Article
MediScan: A Framework of U-Health and Prognostic AI Assessment on Medical Imaging
by Sibtain Syed, Rehan Ahmed, Arshad Iqbal, Naveed Ahmad and Mohammed Ali Alshara
J. Imaging 2024, 10(12), 322; https://doi.org/10.3390/jimaging10120322 - 13 Dec 2024
Viewed by 789
Abstract
With technological advancements, remarkable progress has been made with the convergence of health sciences and Artificial Intelligence (AI). Modern health systems are proposed to ease patient diagnostics. However, the challenge is to provide AI-based precautions to patients and doctors for more accurate risk [...] Read more.
With technological advancements, remarkable progress has been made with the convergence of health sciences and Artificial Intelligence (AI). Modern health systems are proposed to ease patient diagnostics. However, the challenge is to provide AI-based precautions to patients and doctors for more accurate risk assessment. The proposed healthcare system aims to integrate patients, doctors, laboratories, pharmacies, and administrative personnel use cases and their primary functions onto a single platform. The proposed framework can also process microscopic images, CT scans, X-rays, and MRI to classify malignancy and give doctors a set of AI precautions for patient risk assessment. The proposed framework incorporates various DCNN models for identifying different forms of tumors and fractures in the human body i.e., brain, bones, lungs, kidneys, and skin, and generating precautions with the help of the Fined-Tuned Large Language Model (LLM) i.e., Generative Pretrained Transformer 4 (GPT-4). With enough training data, DCNN can learn highly representative, data-driven, hierarchical image features. The GPT-4 model is selected for generating precautions due to its explanation, reasoning, memory, and accuracy on prior medical assessments and research studies. Classification models are evaluated by classification report (i.e., Recall, Precision, F1 Score, Support, Accuracy, and Macro and Weighted Average) and confusion matrix and have shown robust performance compared to the conventional schemes. Full article
Show Figures

Figure 1

Figure 1
<p>Graphical scheme of the system architecture.</p>
Full article ">Figure 2
<p>Graphical scheme of use cases in the proposed framework.</p>
Full article ">Figure 3
<p>Graphical illustration of proposed AI bones fracture detection model.</p>
Full article ">Figure 4
<p>Graphical illustration of the proposed AI lung cancer detection model.</p>
Full article ">Figure 5
<p>Graphical illustration of the proposed AI brain tumor detection model.</p>
Full article ">Figure 6
<p>Graphical illustration of the proposed AI skin cancer detection model.</p>
Full article ">Figure 7
<p>Graphical illustration of the proposed AI kidney malignancy detection model.</p>
Full article ">Figure 8
<p>Graphical illustration of the proposed GPT-4 model system integration.</p>
Full article ">Figure 9
<p>Graphical illustration of the confusion matrix for Bone Fracture recognition; Lung Tumor recognition; Brain Tumor detection; Skin Lesion identification; and Renal Malignancy recognition AI model.</p>
Full article ">Figure 10
<p>Graphical illustration of accuracy graph for Bone Fracture recognition; Lung Tumor recognition; Brain Tumor detection; Skin Lesion identification; and Renal Malignancy recognition AI model.</p>
Full article ">
Back to TopTop