[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Next Issue
Volume 16, September-2
Previous Issue
Volume 16, August-2
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Cancers, Volume 16, Issue 17 (September-1 2024) – 191 articles

Cover Story (view full-size image): Quadruplet induction regimens for patients with treatment-naïve, newly diagnosed multiple myeloma have shown impressive results in multiple trials. However, trials of 4-drug regimens typically use a 3-drug regimen as their comparator, limiting the ability to determine whether the benefit is from the specific combination or just the addition of more agents. We performed a meta-analysis of all trials that randomized patients into a 4-drug vs. 3-drug induction regimens. Our dataset included 11 trials (representing 6509 unique patients) with a goal of determining the PFS and OS benefit, if any, of a 4-drug combination. We also evaluated the toxicity profile of these regimens, recognizing that the addition of more agents to induction regimens may result in more adverse effects. View this paper
  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
12 pages, 1159 KiB  
Article
Clinical Interest in Exome-Based Analysis of Somatic Mutational Signatures for Non-Small Cell Lung Cancer
by Morgane Peroz, Hugo Mananet, Nicolas Roussot, Courèche Guillaume Kaderbhai, Valentin Derangère, Caroline Truntzer and François Ghiringhelli
Cancers 2024, 16(17), 3115; https://doi.org/10.3390/cancers16173115 - 9 Sep 2024
Viewed by 1189
Abstract
Background: Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related mortality. This study investigates the clinical interest of whole exome sequencing (WES) for analyzing somatic mutational signatures in patients with advanced or metastatic NSCLC treated with the current standard of care. [...] Read more.
Background: Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related mortality. This study investigates the clinical interest of whole exome sequencing (WES) for analyzing somatic mutational signatures in patients with advanced or metastatic NSCLC treated with the current standard of care. Methods: Exome sequencing data and clinical characteristics from 132 patients with advanced or metastatic NSCLC were analyzed. Somatic mutational signatures including single base substitutions (SBSs), double base substitutions (DBSs), and copy number signatures were evaluated. Structural variations including tumor mutational burden (TMB), the number of neoantigens, TCR clonality, homologous recombination deficiency (HRD), copy number alterations (CNAs), and microsatellite instability (MSI) score were determined. The association between these genomic features, NSCLC subtypes, and patient outcomes (progression-free and overall survival) was evaluated. Conclusions: Exome sequencing offers valuable insights into somatic mutational signatures in NSCLC. This study identified specific signatures associated with a poor response to immune checkpoint inhibitor (ICI) therapy and chemotherapy, potentially aiding treatment selection and identifying patients unlikely to benefit from these approaches. Full article
Show Figures

Figure 1

Figure 1
<p>Kaplan–Meier curves for progression-free and overall survival according to response status (<b>A</b>,<b>B</b>), actionable mutation pattern (<b>C</b>,<b>D</b>), and type of treatment (<b>E</b>,<b>F</b>).</p>
Full article ">Figure 2
<p>Boxplots representing the distribution of tumor mutational burden (<b>A</b>), the number of neoantigens (<b>B</b>), TCR clones (<b>C</b>), MSI (<b>D</b>), CNA (<b>E</b>), and HRD (<b>F</b>) scores, according to actionable mutation pattern. *: Wilcoxon <span class="html-italic">p</span>-value &lt; 0.05; **: Wilcoxon <span class="html-italic">p</span>-value &lt; 0.01.</p>
Full article ">Figure 3
<p>Kaplan–Meier curves for progression-free survival, according to SBS-associated signatures (<b>A</b>), and according to DBS-associated signatures (<b>B</b>).</p>
Full article ">
11 pages, 980 KiB  
Article
The Real-World Efficacy and Safety of Direct-Acting Antivirals for Chronic Hepatitis C in Patients Active Malignancies
by Maria Dąbrowska, Jerzy Jaroszewicz, Marek Sitko, Justyna Janocha-Litwin, Dorota Zarębska-Michaluk, Ewa Janczewska, Beata Lorenc, Magdalena Tudrujek-Zdunek, Anna Parfieniuk-Kowerda, Jakub Klapaczyński, Hanna Berak, Łukasz Socha, Beata Dobracka, Dorota Dybowska, Włodzimierz Mazur, Łukasz Ważny and Robert Flisiak
Cancers 2024, 16(17), 3114; https://doi.org/10.3390/cancers16173114 - 9 Sep 2024
Viewed by 922
Abstract
Background: Over the past years, the introduction of direct-acting antivirals (DAAs) revolutionized chronic hepatitis C treatment. We aimed to characterize and assess treatment efficacy in three specific groups of patients treated with DAAs: those with active solid malignant tumors (SMTs), hematological diseases (HDs) [...] Read more.
Background: Over the past years, the introduction of direct-acting antivirals (DAAs) revolutionized chronic hepatitis C treatment. We aimed to characterize and assess treatment efficacy in three specific groups of patients treated with DAAs: those with active solid malignant tumors (SMTs), hematological diseases (HDs) and hepatocellular carcinomas (HCCs). Methods: A total of 203 patients with active oncological disease (SMT n = 61, HD = 67, HCC n = 74) during DAA treatment in 2015–2020 selected from the EpiTer-2 database were analyzed retrospectively and compared to 12,983 patients without any active malignancy. Results: Extrahepatic symptoms were more frequent in HD patients (17.2% vs. SMT = 10.3%, HCC = 8.2%, without = 7.8%, p = 0.004). HCC patients characterized with the highest ALT activity (81 IU/L vs. SMT = 59.5 IU/L, HD = 52 IU/L, without = 58 IU/L, p = 0.001) more often had F4 fibrosis as well (86.11% vs. SMT = 23.3%, HD = 28.8%, controls = 24.4%, p = 0.001). A significant majority of subjects in HCC, HD and SMT populations completed the full treatment plan (HCC = 91%; n = 67, HD = 97%; n = 65, SMT = 100%; n = 62). Concerning the treatment efficacy, the overall sustained virologic response, excluding non-virologic failures, was reported in 93.6% HD, 90.16% SMT and 80.6% in HCC patients. Conclusions: As presented in our study, DAA therapy has proven to be highly effective and safe in patients with active SMTs and HDs. However, therapy discontinuations resulting from liver disease progression remain to be the major concern in HCC patients. Full article
(This article belongs to the Special Issue Advances in the Prevention and Treatment of Liver Cancer)
Show Figures

Figure 1

Figure 1
<p>Flow chart showing the selection of patients included in this study.</p>
Full article ">Figure 2
<p>Efficacy of DAAs in patients with active oncological disease: SVR rates in ITT and PP analyses: excluding non-virologic failures. HCC, hepatocellular carcinoma; HD, hemato-oncologic disease; ITT, intention to treat; PP, per protocol; SMT, solid malignant tumor; SVR, sustained virologic response.</p>
Full article ">Figure 3
<p>Percentage of patients completing DAA therapy as recommended. DAA, direct-acting antiviral; HCC, hepatocellular carcinoma; HD, hemato-oncologic disease; SMT, solid malignant tumor.</p>
Full article ">
25 pages, 1718 KiB  
Review
Cellular and Microbial In Vitro Modelling of Gastrointestinal Cancer
by Kristina Žukauskaitė, Melissa Li, Angela Horvath, Sonata Jarmalaitė and Vanessa Stadlbauer
Cancers 2024, 16(17), 3113; https://doi.org/10.3390/cancers16173113 - 9 Sep 2024
Viewed by 1781
Abstract
Human diseases are multifaceted, starting with alterations at the cellular level, damaging organs and their functions, and disturbing interactions and immune responses. In vitro systems offer clarity and standardisation, which are crucial for effectively modelling disease. These models aim not to replicate every [...] Read more.
Human diseases are multifaceted, starting with alterations at the cellular level, damaging organs and their functions, and disturbing interactions and immune responses. In vitro systems offer clarity and standardisation, which are crucial for effectively modelling disease. These models aim not to replicate every disease aspect but to dissect specific ones with precision. Controlled environments allow researchers to isolate key variables, eliminate confounding factors and elucidate disease mechanisms more clearly. Technological progress has rapidly advanced model systems. Initially, 2D cell culture models explored fundamental cell interactions. The transition to 3D cell cultures and organoids enabled more life-like tissue architecture and enhanced intercellular interactions. Advanced bioreactor-based devices now recreate the physicochemical environments of specific organs, simulating features like perfusion and the gastrointestinal tract’s mucus layer, enhancing physiological relevance. These systems have been simplified and adapted for high-throughput research, marking significant progress. This review focuses on in vitro systems for modelling gastrointestinal tract cancer and the side effects of cancer treatment. While cell cultures and in vivo models are invaluable, our main emphasis is on bioreactor-based in vitro modelling systems that include the gut microbiome. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

Figure 1
<p>Graphical representation of the structure of the small intestine epithelium. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>. The illustration is partially based on [<a href="#B7-cancers-16-03113" class="html-bibr">7</a>].</p>
Full article ">Figure 2
<p>Brief overview of the primary characteristics of single-vessel bioreactors. <a href="#app1-cancers-16-03113" class="html-app">Supplementary Table S1</a> contains a more comprehensive description and relevant references. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>Brief overview of the primary characteristics of multi-vessel bioreactors. <a href="#app1-cancers-16-03113" class="html-app">Supplementary Table S1</a> contains a more comprehensive description and relevant references. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 4
<p>Brief overview of the primary characteristics of high-throughput systems. <a href="#app1-cancers-16-03113" class="html-app">Supplementary Table S1</a> contains a more comprehensive description and relevant references. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
17 pages, 295 KiB  
Perspective
Immunotherapy and Radiotherapy for Older Patients with Locally Advanced Non-Metastatic Non-Small-Cell Lung Cancer Who Are Not Candidates for or Decline Surgery and Chemotherapy: A Practical Proposal by the International Geriatric Radiotherapy Group
by Nam P. Nguyen, Brandi R. Page, Huan Giap, Zineb Dahbi, Vincent Vinh-Hung, Olena Gorobets, Mohammad Mohammadianpanah, Micaela Motta, Maurizio Portaluri, Meritxell Arenas, Marta Bonet, Pedro Carlos Lara, Lyndon Kim, Fabien Dutheil, Elena Natoli, Gokoulakrichenane Loganadane, David Lehrman, Satya Bose, Sarabjot Kaur, Sergio Calleja Blanco and Alexander Chiadd Show full author list remove Hide full author list
Cancers 2024, 16(17), 3112; https://doi.org/10.3390/cancers16173112 - 9 Sep 2024
Cited by 1 | Viewed by 1578
Abstract
The standard of care for locally advanced non-small-cell lung cancer (NSCLC) is either surgery combined with chemotherapy pre- or postoperatively or concurrent chemotherapy and radiotherapy. However, older and frail patients may not be candidates for surgery and chemotherapy due to the high mortality [...] Read more.
The standard of care for locally advanced non-small-cell lung cancer (NSCLC) is either surgery combined with chemotherapy pre- or postoperatively or concurrent chemotherapy and radiotherapy. However, older and frail patients may not be candidates for surgery and chemotherapy due to the high mortality risk and are frequently referred to radiotherapy alone, which is better tolerated but carries a high risk of disease recurrence. Recently, immunotherapy with immune checkpoint inhibitors (ICIs) may induce a high response rate among cancer patients with positive programmed death ligand 1 (PD-L1) expression. Immunotherapy is also well tolerated among older patients. Laboratory and clinical studies have reported synergy between radiotherapy and ICI. The combination of ICI and radiotherapy may improve local control and survival for NSCLC patients who are not candidates for surgery and chemotherapy or decline these two modalities. The International Geriatric Radiotherapy Group proposes a protocol combining radiotherapy and immunotherapy based on the presence or absence of PD-L1 to optimize the survival of those patients. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
25 pages, 1897 KiB  
Review
The Role of Predictive and Prognostic MRI-Based Biomarkers in the Era of Total Neoadjuvant Treatment in Rectal Cancer
by Sebastian Curcean, Andra Curcean, Daniela Martin, Zsolt Fekete, Alexandru Irimie, Alina-Simona Muntean and Cosmin Caraiani
Cancers 2024, 16(17), 3111; https://doi.org/10.3390/cancers16173111 - 9 Sep 2024
Cited by 1 | Viewed by 1560
Abstract
The role of magnetic resonance imaging (MRI) in rectal cancer management has significantly increased over the last decade, in line with more personalized treatment approaches. Total neoadjuvant treatment (TNT) plays a pivotal role in the shift from traditional surgical approach to non-surgical approaches [...] Read more.
The role of magnetic resonance imaging (MRI) in rectal cancer management has significantly increased over the last decade, in line with more personalized treatment approaches. Total neoadjuvant treatment (TNT) plays a pivotal role in the shift from traditional surgical approach to non-surgical approaches such as ‘watch-and-wait’. MRI plays a central role in this evolving landscape, providing essential morphological and functional data that support clinical decision-making. Key MRI-based biomarkers, including circumferential resection margin (CRM), extramural venous invasion (EMVI), tumour deposits, diffusion-weighted imaging (DWI), and MRI tumour regression grade (mrTRG), have proven valuable for staging, response assessment, and patient prognosis. Functional imaging techniques, such as dynamic contrast-enhanced MRI (DCE-MRI), alongside emerging biomarkers derived from radiomics and artificial intelligence (AI) have the potential to transform rectal cancer management offering data that enhance T and N staging, histopathological characterization, prediction of treatment response, recurrence detection, and identification of genomic features. This review outlines validated morphological and functional MRI-derived biomarkers with both prognostic and predictive significance, while also exploring the potential of radiomics and artificial intelligence in rectal cancer management. Furthermore, we discuss the role of rectal MRI in the ‘watch-and-wait’ approach, highlighting important practical aspects in selecting patients for non-surgical management. Full article
(This article belongs to the Special Issue Application of Advanced Biomedical Imaging in Cancer Treatment)
Show Figures

Figure 1

Figure 1
<p>Axial T2-weighted MRI illustrating rectal tumour infiltrating all layers of the rectal wall, with evidence of mesorectal fascia involvement (orange arrow).</p>
Full article ">Figure 2
<p>T2-weighted MRI in three planes illustrating a low rectal tumour infiltrating all layers of the rectal wall, with evidence of extramural venous invasion (EMVI, red arrow), tumour deposits (green arrow), and malignant lymph nodes (blue arrow). (<b>A</b>) Coronal T2WI showing extramural venous invasion (red arrow) with increased vessel calibre, maintaining contact with the primary tumour; tumour deposits (green arrow) located superiorly tapering into a vessel but discontinuous from the primary tumour. (<b>B</b>) Axial T2WI showing increased vessel calibre and loss of the normal flow void in the mesorectal vein adjacent to the tumour, suggestive of EMVI (red arrow). (<b>C</b>) Comparison between tumour deposits (green arrow), which are inseparable from the course of veins, and lymph nodes (blue arrow), which are isolated by surrounding fat.</p>
Full article ">Figure 3
<p>Axial T2-weighted MRI demonstrates malignant morphological features of mesorectal lymph nodes, including a round shape and heterogeneous internal signal (green arrow), irregular margins (blue arrow), and mucinous component (orange arrow).</p>
Full article ">Figure 4
<p>T2-weighted MRI in axial and sagittal planes of a rectal tumour demonstrating extensive high-T2 signal mucinous component.</p>
Full article ">
15 pages, 1754 KiB  
Article
Rhabdomyosarcoma of the Biliary Tract in Children: Analysis of Single Center Experience
by Ewa Święszkowska, Dorota Broniszczak, Piotr Kaliciński, Marek Szymczak, Marek Stefanowicz, Wiesława Grajkowska and Bożenna Dembowska-Bagińska
Cancers 2024, 16(17), 3110; https://doi.org/10.3390/cancers16173110 - 9 Sep 2024
Viewed by 971
Abstract
Rhabdomyosarcoma (RMS) of the biliary tract is a rare tumor in children, constituting 0.5–0.8% of all pediatric RMS. Still, it is the most common malignancy in this location in children. Due to its rarity and location, it may cause diagnostic and treatment difficulties. [...] Read more.
Rhabdomyosarcoma (RMS) of the biliary tract is a rare tumor in children, constituting 0.5–0.8% of all pediatric RMS. Still, it is the most common malignancy in this location in children. Due to its rarity and location, it may cause diagnostic and treatment difficulties. Above all, there are no therapeutic guidelines specific for this tumor location. The aim of the study was to present an analysis of our experience with the treatment of children with biliary tract rhabdomyosarcoma (RMS) and discuss clinical recommendations for this specific location published in the literature. A retrospective analysis of medical records of eight children with biliary tree RMS treated in one center between 1996–2022 was performed. Records of eight children, five boys and three girls aged 2 yrs 6 mo to 16 yrs 9 mo (median—6 yrs) were analyzed. All patients presented with jaundice as the first symptom. In two patients, initial diagnosis of a tumor was established. For the remaining six, the primary diagnoses were as follows: choledochal cyst—one, malformation of the biliary ducts—one, choledocholithiasis—one, cholangitis—three. In four patients, the extrahepatic bile ducts were involved; in four patients, both the intrahepatic and extrahepatic bile ducts were involved. Embryonal RMS was diagnosed in seven patients (three botryoides type). Alveolar RMS was found in one patient. Biopsy (three surgical, four during endoscopic retrograde cholangiopancreatography (ERCP)) was performed in seven patients. One child underwent primary partial tumor resection (R2). Seven patients received neoadjuvant chemotherapy, followed by delayed resection in five, including liver transplantation in one (five were R0). Two patients did not undergo surgery. Radiotherapy was administered in four patients (two in first-line treatment, two at relapse/progression). Six patients (75%) are alive with no evidence of disease, with follow-up ranging from 1.2 yrs to 27 yrs (median 11 yrs. and 4 mo.). Two patients died from disease, 2 y 9 mo and 3 y 7 mo from diagnosis. Children presenting with obstructive jaundice should be evaluated for biliary tract RMS. The treatment strategy should include biopsy and preoperative chemotherapy, followed by tumor resection and radiotherapy for residual disease and in case of relapse. Full article
(This article belongs to the Section Pediatric Oncology)
Show Figures

Figure 1

Figure 1
<p>Ultrasound and computed tomography in a patient with biliary tract RMS who was incorrectly diagnosed with a bile duct cyst—cystic dilation of the common bile duct up to 2 cm, filled with thick bile, and dilation of intrahepatic bile ducts.</p>
Full article ">Figure 2
<p>Scintigraphy in a patient with biliary tract RMS who was incorrectly diagnosed with a bile duct cyst—no bile passage to the intestines, impaired liver cell function and no cystic dilation seen in the liver hilum with tracer accumulation.</p>
Full article ">Figure 3
<p>Endoscopic Retrograde Cholangiopancreatography in a patient with suspected biliary tract RMS. During this procedure, tissue samples were collected for histopathological examination.</p>
Full article ">Figure 4
<p>Magnetic resonance imaging of a patient with intrahepatic and extrahepatic RMS showing persistent intrahepatic, extrahepatic, and pancreatic head infiltrations despite chemotherapy. The patient underwent a pancreatoduodenectomy and complete hepatectomy with liver transplantation.</p>
Full article ">
11 pages, 1597 KiB  
Review
Minimal Requirements for Cancer Initiation: A Comparative Consideration of Three Prototypes of Human Leukemia
by Toshiyuki Hori
Cancers 2024, 16(17), 3109; https://doi.org/10.3390/cancers16173109 - 9 Sep 2024
Viewed by 1561
Abstract
Even if its completed form is complex, cancer originates from one or two events that happened to a single cell. A simplified model can play a role in understanding how cancer initiates at the beginning. The pathophysiology of leukemia has been studied in [...] Read more.
Even if its completed form is complex, cancer originates from one or two events that happened to a single cell. A simplified model can play a role in understanding how cancer initiates at the beginning. The pathophysiology of leukemia has been studied in the most detailed manner among all human cancers. In this review, based on milestone papers and the latest research developments in hematology, acute promyelocytic leukemia (APL), chronic myeloid leukemia (CML), and acute myeloid leukemia (AML) with RUNX1-RUNX1T1 are selected to consider minimal requirements for cancer initiation. A one-hit model can be applied to the initiation of APL and CML whereas a two-hit model is more suitable to the initiation of AML with RUNX1-RUNX1T1 and other AMLs. Even in cancer cells with multiple genetic abnormalities, there must be a few mutant genes critical for the mutant clone to survive and proliferate. Such genes should be identified and characterized in each case in order to develop individualized target therapy. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) A model of a normal bone marrow showing the myeloid maturation pathway. HPSC indicated by grey circles are located close to a stroma cell in niche. (<b>B</b>) A model of APL. Red cells represent the mutant clone.</p>
Full article ">Figure 2
<p>A model of chronic phase CML. Red cells represent the mutant clone.</p>
Full article ">Figure 3
<p>A model of the pre-leukemia stage of AML. Red cells represent the mutant clone.</p>
Full article ">Figure 4
<p>A model of the overt stage of AML. Red cells represent the mutant clone.</p>
Full article ">Figure 5
<p>(<b>A</b>) A scheme of a normal colon crypt. Stem cells at the bottom of the crypt give rise to progeny of epithelial cells which proliferate and mature. Terminally differentiated cells are pushed away into the bowel lumen. (<b>B</b>) Red mutant cells proliferate rapidly to form a polyp but can be pushed out in the meantime.</p>
Full article ">
19 pages, 4792 KiB  
Article
Curcumin-Dichloroacetate Hybrid Molecule as an Antitumor Oral Drug against Multidrug-Resistant Advanced Bladder Cancers
by Kunj Bihari Gupta, Truett L. Taylor, Siva S. Panda, Muthusamy Thangaraju and Bal. L. Lokeshwar
Cancers 2024, 16(17), 3108; https://doi.org/10.3390/cancers16173108 - 8 Sep 2024
Viewed by 1501
Abstract
Tumor cells produce excessive reactive oxygen species (ROS) but cannot detoxify ROS if they are due to an external agent. An agent that produces toxic levels of ROS, specifically in tumor cells, could be an effective anticancer drug. CMC-2 is a molecular hybrid [...] Read more.
Tumor cells produce excessive reactive oxygen species (ROS) but cannot detoxify ROS if they are due to an external agent. An agent that produces toxic levels of ROS, specifically in tumor cells, could be an effective anticancer drug. CMC-2 is a molecular hybrid of the bioactive polyphenol curcumin conjugated to dichloroacetate (DCA) via a glycine bridge. The CMC-2 was tested for its cytotoxic antitumor activities and killed both naïve and multidrug-resistant (MDR) bladder cancer (BCa) cells with equal potency (<1.0 µM); CMC-2 was about 10–15 folds more potent than curcumin or DCA. Growth of human BCa xenograft in mice was reduced by >50% by oral gavage of 50 mg/kg of CMC-2 without recognizable systemic toxicity. Doses that used curcumin or DCA showed minimum antitumor effects. In vitro, the toxicity of CMC-2 in both naïve and MDR cells depended on increased intracellular ROS in tumor cells but not in normal cells at comparable doses. Increased ROS caused the permeabilization of mitochondria and induced apoptosis. Further, adding N-Acetyl cysteine (NAC), a hydroxyl radical scavenger, abolished excessive ROS production and CMC-2’s cytotoxicity. The lack of systemic toxicity, equal potency against chemotherapy -naïve and resistant tumors, and oral bioavailability establish the potential of CMC-2 as a potent drug against bladder cancers. Full article
(This article belongs to the Special Issue Feature Paper in Section “Cancer Therapy” in 2024)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of CMC-2.</p>
Full article ">Figure 2
<p>Cur, DCA, and CMC-2 are cytotoxic to BCa cells but were relatively nontoxic to non-transformed bladder epithelial cells, UROtsa: (<b>A</b>). Viability of tumorigenic, chemo-sensitive (HT-1376, 253J, T24, and 5637), chemo-resistant (5637 GemR, 5637 CisR, and 5637 G+C R) and non-tumorigenic (UROtsa) human BCa cell lines, treated with drugs or drug-diluent (control). (<b>B</b>). The viability of chemo-resistant human BCa, 5637 CisR, 5637 GemR, and 5637 G+C R cells as percent of untreated control. The MTT reduction assay was performed after 72 h of respective drug treatment. Results are expressed as mean of percent cell viability ± standard deviation (SD) (<span class="html-italic">n</span> = 4). Except for the non-tumorigenic UROtsa cells, CMC-2 was significantly more cytotoxic than Cur or DCA (<span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">Figure 3
<p>Clonal survival of human BCa cells exposed to DCA, Cur, and CMC-2: (<b>A</b>) Colonies formed by surviving cells after treatment with several doses of CMC-2, Cur, and DCA for three days that were then allowed to form colonies of &gt;16 cells in the absence of drugs for next 4–7 days. (<b>B</b>–<b>D</b>). Graphs represent the mean of number of surviving colonies ± standard deviation (<span class="html-italic">n</span> = 3) for each cell type: (<b>B</b>) HT-1376 cells, (<b>C</b>) 5637 naïve, and (<b>D</b>) 5637 G+C R. Paired <span class="html-italic">t</span>-tests were used to determine the significance of difference between control and the exposed cells. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.005.</p>
Full article ">Figure 4
<p>Estimating oxidative stress: reactive oxygen species (ROS) produced in (<b>A</b>) HT-1376 and (<b>B</b>) 5637 naïve cells after treatment with DCA, Cur, and CMC-2 for various periods, as estimated using a DCFDA probe. The relative fluorescence unit (RFU) was converted to the percent ROS value. Data represented here are means of ROS produced ± standard deviation (<span class="html-italic">n</span> = 4) after subtracting the ROS values generated in the untreated cells. Cells treated with H<sub>2</sub>O<sub>2</sub> (0.2 µM) served as a positive control. ROS levels were significantly higher (<span class="html-italic">p</span> ≤ 0.05) in CMC-2-treated cells at the dose level of 10 µM of DCA, Cur, and CMC-2 for all treatment periods in both cell lines.</p>
Full article ">Figure 5
<p>Estimating mitochondrial ROS (mt-ROS): the mt-ROS produced in the (<b>A</b>) HT-1376 and (<b>B</b>). 5637 naïve cells after the treatment of several doses of DCA, Cur, and CMC-2 exposed for several periods, as estimated using MitoSOX. The relative fluorescence unit (RFU) was converted to percent mt-ROS. Data represented here are means of mt-ROS produced ± standard deviation (<span class="html-italic">n</span> = 4) after subtracting the ROS values generated in the untreated cells. Cells treated with H<sub>2</sub>O<sub>2</sub> (0.2 µM) served as a positive control. mt-ROS levels were significantly higher (<span class="html-italic">p</span> ≤ 0.05) in CMC-2-treated cells at the dose level of 10 µM of DCA, Cur, and CMC-2 for all treatment periods in both cell lines.</p>
Full article ">Figure 6
<p>NAC abolishes ROS generation in CMC-2-treated cells: the level of ROS produced in (<b>A</b>) HT-1376, (<b>B</b>) 5637 naïve cells, after treatment with several doses of CMC-2 with and without N-acetyl-L-cysteine (NAC, 1 mM), was estimated using a probe DCFDA for 30 min, and intracellular fluorescence was measured. The relative fluorescence unit (RFU) was converted to the percent ROS value. Data represented here are means of ROS produced ± standard deviation (<span class="html-italic">n</span> = 4) after subtracting the ROS values generated in the untreated cells. Cells treated with H<sub>2</sub>O<sub>2</sub> (0.2 µM) served as a positive control.</p>
Full article ">Figure 7
<p>NAC abolishes CMC-2-induced cytotoxicity. MTT assay-based cellular viability was estimated in HT-1376, T24, 5637 naïve, and 5637 G+C R cells after treatment with several concentrations of CMC-2, NAC alone, and in combination. The percent cell viability was calculated relative to the untreated control cells, and results are expressed as the mean of percent cell viability ± standard deviation (<span class="html-italic">n</span> = 4) for each treatment. Paired <span class="html-italic">t</span>-tests were used to determine the significance of the difference in the cellular viability between CMC-2 and the combination treatment group. *** <span class="html-italic">p</span> ≤ 0.005.</p>
Full article ">Figure 8
<p>CMC-2 reduces mitochondrial transmembrane potential (ψM). Levels of ψM in human BC HT-1376 and 5637 naïve cells were estimated by JC-1 aggregates using flow cytometry after a 24 h treatment with DCA, Cur, and CMC-2. (<b>A</b>). Distribution of cells based on red and green fluorescence channels. (<b>B</b>). Bar graph demonstrating the red/green ratio of JC-1 fluorescence. The red/green ratio was significantly decreased in cells treated with all three drugs (*** <span class="html-italic">p</span> ≤ 0.001).</p>
Full article ">Figure 9
<p>CMC-2-induced cytotoxicity reveals cell cycle arrest in the G1 phase. Cell cycle phase resolution was analyzed by flow cytometry. HT-1376 and 5637 naïve cells were treated with DCA, Cur, and CMC-2 for 24 h, and PI-stained nuclei suspensions were analyzed by flow cytometry to resolve individual cell cycle phase fractions based on the DNA content. (<b>A</b>). Cell cycle phase distribution of each treatment in both cells. (<b>B</b>). Bar graphs represent cell cycle phases upon drug treatment in both cell lines. Flow cytometry data were collected and analyzed using the NovoCyte Quanteon and NovoExpress software. The experiment was performed three times; data shown are mean ± SD, and pairwise <span class="html-italic">t</span>-tests were used to determine significance * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01. (<b>C</b>). Immunoblots of Cyclin E1 in total cell lysates treated with DCA, Cur, and CMC-2 in both cell types. See <a href="#app1-cancers-16-03108" class="html-app">Figure S2</a>: uncropped blots. β-actin is shown as a loading control in all lanes. All experiments were performed thrice, and a representative immunoblot image is displayed.</p>
Full article ">Figure 10
<p>Estimating the markers for apoptotic cell death by measuring the level of Annexin V in the outer leaflet of the plasma membrane. (<b>A</b>) Distribution of bladder cancer cells labeled with Annexin V-FITC (bound to externalized phosphatidyl serine) and PI (DNA) after 24 h of treatment of DCA (10 μM), Cur (10 μM), CMC-2 (1 μM) and Staurosporine (Sta, 0.2 μM; a positive inducer of apoptosis). The single-cell suspensions were analyzed by flow cytometry to determine the intensity of (green) and (red) fluorescence of individual cells. Quadrant analysis shows the percentage of live cells undergoing early- (low green bars) or late-stage (high green bars) apoptosis. (<b>B</b>). Quantification of the relative distribution of cells in each quadrant. (<b>C</b>,<b>D</b>) Immunoblot analysis of protein markers related to apoptotic cell death: cleaved and full-length PARP, Bax, Bcl-2, Bcl-XL, AIF (apoptosis-inducing factor), p-AKT (Ser-473), and AKT. β-actin and GAPDH were used as a loading control. See <a href="#app1-cancers-16-03108" class="html-app">Figure S2</a>: uncropped blots. Three separate preparations of cell lysates from independent experiments were analyzed by immunoblotting, and the best representative image of one set of cell lysates is shown here (*** <span class="html-italic">p</span> ≤ 0.001, when compared to untreated control).</p>
Full article ">Figure 11
<p>Antitumor activity of DCA, Cur, and CMC-2 in HT-1376 xenografted mice. (<b>A</b>) Growth curve of tumor volume of each group. (<b>B</b>) Terminal tumor images from formalin-fixed whole tumors of each group. (<b>C</b>) Body weight of each tumor-bearing animal throughout this study. (<b>D</b>) H&amp;E-stained tumor sections of each group to demonstrate the histological details of tumor cells and stroma. Paired <span class="html-italic">t</span>-tests were used to determine significance. Data are represented as mean ± SD, *** <span class="html-italic">p</span> ≤ 0.005.</p>
Full article ">
13 pages, 1686 KiB  
Systematic Review
Impact of Indocyanine Green Dose on Sentinel Lymph Node Mapping in Cervical Cancer: A Systematic Review
by Joel Laufer, Santiago Scasso and Andrea Papadia
Cancers 2024, 16(17), 3107; https://doi.org/10.3390/cancers16173107 - 8 Sep 2024
Viewed by 1084
Abstract
Over the past decade, SLN mapping has become increasingly important in cervical cancer surgery. ICG is the most commonly used tracer due to its high bilateral detection rates, ease of use, and safety. However, there is no consensus on the optimal ICG dose, [...] Read more.
Over the past decade, SLN mapping has become increasingly important in cervical cancer surgery. ICG is the most commonly used tracer due to its high bilateral detection rates, ease of use, and safety. However, there is no consensus on the optimal ICG dose, leading to variability in outcomes. This systematic review aims to evaluate the impact of different ICG doses on SLN detection in early-stage cervical cancer, identifying the most effective and safe dose for clinical practice. A comprehensive search was conducted in MEDLINE/PubMed up to May 2024. Studies included assessed SLN mapping using ICG in stage IA2-IIA/IIB cervical cancer. Exclusions were applied to studies not reporting ICG dose or using multiple tracers without dose-specific results. Twelve studies were included, with ICG concentrations ranging from 0.25 mg/mL to 25 mg/mL and injection volumes from 1 to 10 mL. Overall SLN detection rates ranged from 88% to 100%, while bilateral detection rates varied between 74.1% and 98.5%. The most consistent results were obtained with an ICG concentration of 1.25 mg/mL and a 4 mL injection volume. In conclusion, an ICG concentration of 1.25 mg/mL with a 4 mL injection volume is recommended for effective SLN mapping in cervical cancer, achieving high detection rates with minimal variability. Standardizing this dose in clinical practice is suggested to improve reproducibility and outcomes. Full article
(This article belongs to the Special Issue Cervical Cancer: Screening and Treatment in 2024-2025)
Show Figures

Figure 1

Figure 1
<p>Overall quality assessment of the studies included in the systematic review, according to the QUADAS-2 tool.</p>
Full article ">Figure 2
<p>PRISMA Flow chart.</p>
Full article ">Figure 3
<p>ICG concentration used in different studies [<a href="#B17-cancers-16-03107" class="html-bibr">17</a>,<a href="#B18-cancers-16-03107" class="html-bibr">18</a>,<a href="#B19-cancers-16-03107" class="html-bibr">19</a>,<a href="#B20-cancers-16-03107" class="html-bibr">20</a>,<a href="#B21-cancers-16-03107" class="html-bibr">21</a>,<a href="#B22-cancers-16-03107" class="html-bibr">22</a>,<a href="#B23-cancers-16-03107" class="html-bibr">23</a>,<a href="#B24-cancers-16-03107" class="html-bibr">24</a>,<a href="#B25-cancers-16-03107" class="html-bibr">25</a>,<a href="#B26-cancers-16-03107" class="html-bibr">26</a>,<a href="#B27-cancers-16-03107" class="html-bibr">27</a>,<a href="#B28-cancers-16-03107" class="html-bibr">28</a>].</p>
Full article ">Figure 4
<p>ICG volume used in different studies [<a href="#B17-cancers-16-03107" class="html-bibr">17</a>,<a href="#B18-cancers-16-03107" class="html-bibr">18</a>,<a href="#B19-cancers-16-03107" class="html-bibr">19</a>,<a href="#B20-cancers-16-03107" class="html-bibr">20</a>,<a href="#B21-cancers-16-03107" class="html-bibr">21</a>,<a href="#B22-cancers-16-03107" class="html-bibr">22</a>,<a href="#B23-cancers-16-03107" class="html-bibr">23</a>,<a href="#B24-cancers-16-03107" class="html-bibr">24</a>,<a href="#B25-cancers-16-03107" class="html-bibr">25</a>,<a href="#B26-cancers-16-03107" class="html-bibr">26</a>,<a href="#B27-cancers-16-03107" class="html-bibr">27</a>,<a href="#B28-cancers-16-03107" class="html-bibr">28</a>].</p>
Full article ">
13 pages, 1365 KiB  
Commentary
Amitotic Cell Division, Malignancy, and Resistance to Anticancer Agents: A Tribute to Drs. Walen and Rajaraman
by Razmik Mirzayans and David Murray
Cancers 2024, 16(17), 3106; https://doi.org/10.3390/cancers16173106 - 8 Sep 2024
Viewed by 2159
Abstract
Cell division is crucial for the survival of living organisms. Human cells undergo three types of cell division: mitosis, meiosis, and amitosis. The former two types occur in somatic cells and germ cells, respectively. Amitosis involves nuclear budding and occurs in cells that [...] Read more.
Cell division is crucial for the survival of living organisms. Human cells undergo three types of cell division: mitosis, meiosis, and amitosis. The former two types occur in somatic cells and germ cells, respectively. Amitosis involves nuclear budding and occurs in cells that exhibit abnormal nuclear morphology (e.g., polyploidy) with increased cell size. In the early 2000s, Kirsten Walen and Rengaswami Rajaraman and his associates independently reported that polyploid human cells are capable of producing progeny via amitotic cell division, and that a subset of emerging daughter cells proliferate rapidly, exhibit stem cell-like properties, and can contribute to tumorigenesis. Polyploid cells that arise in solid tumors/tumor-derived cell lines are referred to as polyploid giant cancer cells (PGCCs) and are known to contribute to therapy resistance and disease recurrence following anticancer treatment. This commentary provides an update on some of these intriguing discoveries as a tribute to Drs. Walen and Rajaraman. Full article
(This article belongs to the Special Issue The Role of Chromosomal Instability in Cancer)
Show Figures

Figure 1

Figure 1
<p>Timeline of findings related to the DNA damage response that led to the somatic mutation theory of cancer. Reproduced from Huang and Zhou [<a href="#B15-cancers-16-03106" class="html-bibr">15</a>].</p>
Full article ">Figure 2
<p>Fate of giant cancer cells based on discoveries reported by Walen, Rajaraman, Erenpreisa and their colleagues in the early 2000s (reviewed in [<a href="#B42-cancers-16-03106" class="html-bibr">42</a>]). These authors revealed the following sequence of events: “Cancer cells exposed to DNA-damaging agents may fail to activate early cell cycle checkpoints and thus replicate their genome on damaged templates. While some cells may die, a great number may retain viability and acquire diverse types of nuclear abnormalities including micronuclei, multiple nuclei and a highly enlarged nucleus. A proportion of multinucleated/polyploid giant cells may undergo neotic cell division, which is characterized by karyokinesis via nuclear budding followed by asymmetric cytokinesis, resulting in the generation of small mononuclear karyoplasts (also called Raju cells). These karyoplasts may resume mitotic division, and may eventually produce highly metastatic and therapy resistant descendants” [<a href="#B42-cancers-16-03106" class="html-bibr">42</a>]. Reproduced from our 2008 review [<a href="#B42-cancers-16-03106" class="html-bibr">42</a>].</p>
Full article ">Figure 3
<p>The role of PGCCs in cancer remission (dormancy) and relapse. At the macroscopic level, (e.g., observed in live animals [<a href="#B82-cancers-16-03106" class="html-bibr">82</a>,<a href="#B83-cancers-16-03106" class="html-bibr">83</a>]), the initial cancer remission post-therapy is followed by a dormant state (also known as “senescence-like dormancy” [<a href="#B84-cancers-16-03106" class="html-bibr">84</a>] and “minimal residual disease” [<a href="#B85-cancers-16-03106" class="html-bibr">85</a>]), and eventual tumor relapse in most patients. At the microscopic level, in response to ionizing radiation [<a href="#B78-cancers-16-03106" class="html-bibr">78</a>] or chemotherapeutic drugs [<a href="#B82-cancers-16-03106" class="html-bibr">82</a>,<a href="#B83-cancers-16-03106" class="html-bibr">83</a>] (i, green circles), the majority of bulk (para-diploid) cancer cells undergo polyploidization and/or cell fusion, generating PGCCs (ii, maroon circles). During the subsequent transitional state (iii, blue circles), a proportion of PGCCs undergo depolyploidization and generate high frequencies of rapidly proliferating small-sized progeny cells that repopulate the tumor (iv, pink circles).</p>
Full article ">Figure 4
<p>Fluorescence images showing the nuclear morphology of 2800T (Li-Fraumeni syndrome) dermal fibroblasts at different passage numbers. P, polyploid/multinucleated giant cells. Scale bars, 50 µm. Reproduced from Mirzayans et al. [<a href="#B96-cancers-16-03106" class="html-bibr">96</a>] with permission.</p>
Full article ">Figure A1
<p>The development and fate of multinucleated giant cells following exposure to ionizing radiation, as reported by Puck and Marcus over 60 years ago [<a href="#B45-cancers-16-03106" class="html-bibr">45</a>]. (For details, see [<a href="#B46-cancers-16-03106" class="html-bibr">46</a>]).</p>
Full article ">
13 pages, 256 KiB  
Article
Brain Metastases as Inaugural Sign of Non-Small Cell Lung Carcinoma: Case Series and Review of Literature
by Alexandra Pușcașu, Fabien Moinard-Butot, Simon Nannini, Cathie Fischbach, Roland Schott and Laura Bender
Cancers 2024, 16(17), 3105; https://doi.org/10.3390/cancers16173105 - 8 Sep 2024
Viewed by 1187
Abstract
In the era of immune checkpoint inhibitors (ICI), managing non-oncogene driven non-small cell lung cancer (NSCLC) with brain metastases (BM) is challenging, especially when brain involvement is the initial sign. Patients with newly diagnosed brain metastatic NSCLC without epidermal growth factor receptor (EFGR) [...] Read more.
In the era of immune checkpoint inhibitors (ICI), managing non-oncogene driven non-small cell lung cancer (NSCLC) with brain metastases (BM) is challenging, especially when brain involvement is the initial sign. Patients with newly diagnosed brain metastatic NSCLC without epidermal growth factor receptor (EFGR) nor anaplastic lymphoma kinase (ALK) alterations were retrospectively included. Twenty-five patients were analyzed; 15 (60%) had symptomatic BM as the first sign (group 1), while 10 (40%) had BM discovered during complementary examinations (group 2). Fourteen patients (56%) had concomitant extracerebral metastases, primarily in group 2. Eight (32%) had oligometastatic disease, with seven in group 1. Over half received chemotherapy and pembrolizumab as first-line treatment. BM surgical resection occurred in twelve (80%) patients in group 1 and one in group 2. Median cerebral progression-free survival was 10 months: 12 in group 1 and 5 in group 2. Median overall survival was 25 months: not reached in group 1 and 6 months in group 2. This case series highlights survival outcomes for patients with inaugural BM, a demographic underrepresented in pivotal trials. Oligometastatic disease and symptomatic BM as initial signs seem associated with better prognosis due to increased use of multimodal local approaches. Combining local approaches with first-line ICI+/− chemotherapy appears to improve survival in brain metastatic NSCLC. A literature review was conducted to explore key questions regarding upfront ICI alone or in combination with systemic drugs or local approaches in brain metastatic NSCLC. Full article
(This article belongs to the Section Cancer Metastasis)
20 pages, 2673 KiB  
Article
Immune Cell Molecular Pharmacodynamics of Lanreotide in Relation to Treatment Response in Patients with Gastroenteropancreatic Neuroendocrine Tumors
by Sabah Alaklabi, Orla Maguire, Harsha Pattnaik, Yali Zhang, Jacky Chow, Jianmin Wang, Hans Minderman and Renuka Iyer
Cancers 2024, 16(17), 3104; https://doi.org/10.3390/cancers16173104 - 7 Sep 2024
Viewed by 1256
Abstract
The CLARINET trial led to the approval of lanreotide for the treatment of patients with gastroenteropancreatic neuroendocrine tumors (NETs). It is hypothesized that lanreotide regulates proliferation, hormone synthesis, and other cellular functions via binding to somatostatin receptors (SSTR1–5) present in NETs. However, our [...] Read more.
The CLARINET trial led to the approval of lanreotide for the treatment of patients with gastroenteropancreatic neuroendocrine tumors (NETs). It is hypothesized that lanreotide regulates proliferation, hormone synthesis, and other cellular functions via binding to somatostatin receptors (SSTR1–5) present in NETs. However, our knowledge of how lanreotide affects the immune system is limited. In vitro studies have investigated functional immune response parameters with lanreotide treatment in healthy donor T cell subsets, encompassing the breadth of SSTR expression, apoptosis induction, cytokine production, and activity of transcription factor signaling pathways. In our study, we characterized in vitro immune mechanisms in healthy donor T cells in response to lanreotide. We also studied the in vivo effects by looking at differential gene expression pre- and post-lanreotide therapy in patients with NET. Immune-focused gene and protein expression profiling was performed on peripheral blood samples from 17 NET patients and correlated with clinical response. In vivo, lanreotide therapy showed reduced effects on wnt, T cell receptor (TCR), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) signaling in CD8+ T cells in responders compared to non-responders. Compared to non-responders, responders showed reduced effects on cytokine and chemokine signaling but greater effects on ubiquitination and proteasome degradation genes. Our results suggest significant lanreotide pharmacodynamic effects on immune function in vivo, which correlate with responses in NET patients. This is not evident from experimental in vitro settings. Full article
(This article belongs to the Special Issue Updates in Neuroendocrine Neoplasms)
Show Figures

Figure 1

Figure 1
<p>SSTR expression and in vitro effects of lanreotide on healthy human T cell function and survival. (<b>A</b>) Expression of SSTR receptors 1–5 on Tc (<b>left</b> graph), Th (<b>middle</b> graph), and Treg (<b>right</b> graph) cell populations showing SSTR2 to be the most abundantly expressed SSTR in all T cell populations studied. (<b>B</b>) Effects of in vitro lanreotide exposure on T cell function and survival. Function measured as IFNg (<b>left</b> graph) and IL-2 (<b>middle</b> graph) production, and survival measured by apoptosis assay (<b>right</b> graph). (<b>C</b>) Effects of in vitro lanreotide exposure on transcription factor signaling measured by activity of NFAT (<b>left</b> graph), NFkB (<b>middle</b> graph) or ERK1/2 (<b>right</b> graph). N = 10 for all experiments.</p>
Full article ">Figure 2
<p>Proteomic and transcriptional analysis of in vivo lanreotide treatment in T cell subsets from NET patients. Paired samples obtained before and after treatment were analyzed for each patient. Volcano plots show a linear mixed model (LMM) analysis of Tc (<b>left</b> graphs), Th (<b>middle</b> graphs), and Treg (<b>right</b> graphs) cell populations fitted to test three different effects. The ‘Patient Effect’ (<b>upper</b> graphs) estimated if any gene was significantly different between Responder and Non-responder pre-treatment, where non-responders served as reference. The ‘Treatment effect’ (<b>middle</b> graphs) estimated if any genes were significantly changed between pre-treatment and post-treatment (regardless of response). For this analysis, the pre-treatment samples served as a reference. The ‘Interaction Effect’ estimated whether any genes were significantly changed before and after treatment in responders versus non-responders, where non-responders served as reference (lower graphs). Plots show the statistical significance (<span class="html-italic">p</span> value) versus the magnitude of change (fold change) for each of the 9 categories.</p>
Full article ">Figure 3
<p>Expression of genes of note from transcriptional analysis of in vivo lanreotide treatment in T cell subsets from NET patients. (<b>A</b>) Expression of TXNIP in non-responders (red bar) and responders (teal bar) in Tc (<b>left</b> graph), Th (<b>middle</b> graph), and Treg cells (<b>right</b> graph). (<b>B</b>) Most significantly altered genes between non-responders and responders pre-treatment shown for Th (<b>left</b> graph), Treg (<b>middle</b> graph), and Tc cells (<b>right</b> graph). (<b>C</b>) Expression of TXNIP in Tc cells pre- and post-treatment in non-responders and responders indicates upregulation with lanreotide treatment in responders only. (<b>D</b>) Interaction effect, i.e., significant change in gene expression between non-responders and responders following treatment shown for CCRL2 in TC cells (<b>right</b> graph).</p>
Full article ">Figure A1
<p>Gating strategy used in sorting T cell sub-populations. Isolating T cells for sorting followed a hierarchical gating strategy where, from left to right, upper to lower, singlets were selected based on forward scatter area against height. A lymphoid population was then selected based on forward scatter area against side scatter area. (Cells). Viable T cells were selected as CD3 positive and negative for LiveDead viability dye. CD4 negative, CD8 positive TC cells were selected. CD8 negative, CD4 positive were further gated as either CD25 negative, CD127 positive TH cells or CD127 dim/negative, CD25 positive Treg cells.</p>
Full article ">Figure A2
<p>T cell proportions in NET patients. The overall T cell percentages were assessed for all NET patient samples that had been sorted for NanoString analysis. The % CD3+ cells (<b>left</b> graph) is derived from the ‘cells’ population in the gating strategy. CD8+ and CD4+ cells are both derived from the CD3+ population (<b>middle</b> graphs). The Treg population is derived from the CD4+ cells (<b>right</b> graph).</p>
Full article ">
23 pages, 5206 KiB  
Article
Fibroblast Growth Factor 2 (FGF2) Activates Vascular Endothelial Growth Factor (VEGF) Signaling in Gastrointestinal Stromal Tumors (GIST): An Autocrine Mechanism Contributing to Imatinib Mesylate (IM) Resistance
by Sergei Boichuk, Pavel Dunaev, Aigul Galembikova and Elena Valeeva
Cancers 2024, 16(17), 3103; https://doi.org/10.3390/cancers16173103 - 7 Sep 2024
Cited by 2 | Viewed by 1346
Abstract
We showed previously that the autocrine activation of the FGFR-mediated pathway in GIST lacking secondary KIT mutations was a result of the inhibition of KIT signaling. We show here that the FGF2/FGFR pathway regulates VEGF-A/VEGFR signaling in IM-resistant GIST cells. Indeed, recombinant FGF2 [...] Read more.
We showed previously that the autocrine activation of the FGFR-mediated pathway in GIST lacking secondary KIT mutations was a result of the inhibition of KIT signaling. We show here that the FGF2/FGFR pathway regulates VEGF-A/VEGFR signaling in IM-resistant GIST cells. Indeed, recombinant FGF2 increased the production of VEGF-A by IM-naive and resistant GIST cells. VEGF-A production was also increased in KIT-inhibited GIST, whereas the neutralization of FGF2 by anti-FGF2 mAb attenuated VEGFR signaling. Of note, BGJ 398, pan FGFR inhibitor, effectively and time-dependently inhibited VEGFR signaling in IM-resistant GIST T-1R cells, thereby revealing the regulatory role of the FGFR pathway in VEGFR signaling for this particular GIST cell line. This also resulted in significant synergy between BGJ 398 and VEGFR inhibitors (i.e., sunitinib and regorafenib) by enhancing their pro-apoptotic and anti-proliferative activities. The high potency of the combined use of VEGFR and FGFR inhibitors in IM-resistant GISTs was revealed by the impressive synergy scores observed for regorafenib or sunitinib and BGJ 398. Moreover, FGFR1/2 and VEGFR1/2 were co-localized in IM-resistant GIST T-1R cells, and the direct interaction between the aforementioned RTKs was confirmed by co-immunoprecipitation. In contrast, IM-resistant GIST 430 cells expressed lower basal levels of FGF2 and VEGF-A. Despite the increased expression VEGFR1 and FGFR1/2 in GIST 430 cells, these RTKs were not co-localized and co-immunoprecipitated. Moreover, no synergy between FGFR and VEGFR inhibitors was observed for the IM-resistant GIST 430 cell line. Collectively, the dual targeting of FGFR and VEGFR pathways in IM-resistant GISTs is not limited to the synergistic anti-angiogenic treatment effects. The dual inhibition of FGFR and VEGFR pathways in IM-resistant GISTs potentiates the proapoptotic and anti-proliferative activities of the corresponding RTKi. Mechanistically, the FGF2-induced activation of the FGFR pathway turns on VEGFR signaling via the overproduction of VEGF-A, induces the interaction between FGFR1/2 and VEGFR1, and thereby renders cancer cells highly sensitive to the dual inhibition of the aforementioned RTKs. Thus, our data uncovers the novel mechanism of the cross-talk between the aforementioned RTKs in IM-resistant GISTs lacking secondary KIT mutations and suggests that the dual blockade of FGFR and VEGFR signaling might be an effective treatment strategy for patients with GIST-acquired IM resistance via KIT-independent mechanisms. Full article
Show Figures

Figure 1

Figure 1
<p>FGFR, VEGFR, and KIT signaling pathways in IM-naive (GIST T-1) and IM-resistant (GIST T-1R, GIST 430) cells. (<b>A</b>) Expression of FGF/FGFR signaling proteins in GIST cells; (<b>B</b>) expression of the VEGF/FGFR signaling proteins in GIST cells; (<b>C</b>) changes in the relative expression level of VEGF-A and VEGFR1, 2, and 3 in GIST T-1 vs. T-1R and GIST 430 cells, as determined by quantitative RT-PCR. For internal control, the amplification of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used; data are presented as median ± SD. Significant differences with <span class="html-italic">p</span> &lt; 0.05 (*), <span class="html-italic">p</span> &lt; 0.01 (**) from <span class="html-italic">n</span> ≥ 3 using unpaired Student’s <span class="html-italic">t</span>-test. (<b>D</b>) Expression of the KIT signaling proteins in GIST cells. Actin stain was used as a loading control for all experiments shown in (<b>A</b>,<b>B</b>,<b>D</b>). (<b>E</b>) Concentration of VEGF-A (pg/mL) in supernatants of IM-naive (GIST T-1) and resistant (GIST T-1R and 430) cells measured by ELISA, as described in <a href="#sec4-cancers-16-03103" class="html-sec">Section 4</a>. Data are presented as median ± SD. Significant differences with <span class="html-italic">p</span> &lt; 0.05 (*), <span class="html-italic">p</span> &lt; 0.01 (**) from <span class="html-italic">n</span> ≥ 3 using unpaired Student’s <span class="html-italic">t</span>-test. The original Western blot figures can be found in <a href="#app1-cancers-16-03103" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) The exogenous FGF2 activates VEGFR signaling in GIST T-1 cells via up-regulation of VEGF-A. (<b>A</b>) FGF-2 stimulates, whereas anti-FGF2 neutralizing Abs abrogates VEGFR signaling GIST T-1 cells. Cells were treated with FGF2 (100 ng/mL), IM (0.02 µM) alone or in the presence of anti-FGF2 neutralizing Abs (20 µg/mL) for 72 h. Expression of VEGF-A, total and phosphorylated forms of VEGFR, KIT, MAPK, AKT, and STAT1 was assessed by immunoblot analysis. Actin stain was used as a loading control for each sample. (<b>B</b>) Concentration of VEGF-A (pg/mL, measured by ELISA) in supernatants of IM-naive (GIST T-1) cells treated with DMSO (control), FGF-2 (100 ng/mL), IM (0.02 µM) alone or in the presence of anti-FGF-2 Abs (20 µg/mL) for 72 h. Data are presented as median ± SD. Significant differences with <span class="html-italic">p</span> &lt; 0.05 (*), <span class="html-italic">p</span> &lt; 0.01 (**) from <span class="html-italic">n</span> ≥ 3 using unpaired Student’s <span class="html-italic">t</span>-test. (<b>C</b>) Changes in the relative expression level of mRNA VEGF-A and VEGFR1, 2, and 3 in GIST T-1 after treatment with FGF-2 (100 ng/mL), as determined by quantitative RT-PCR. The amplification of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used for internal control. Data are presented as median ± SD. Significant differences with <span class="html-italic">p</span> &lt; 0.05 (*) from <span class="html-italic">n</span> ≥ 3 using unpaired Student’s <span class="html-italic">t</span>-test. The original Western blot figures can be found in <a href="#app1-cancers-16-03103" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 3
<p>Inhibition of FGFR signaling attenuates activation of FGFR and VEGFR pathways in GIST T-1R cells. GIST T-1R cells were treated with BGJ 398 (2 µM) or regorafenib (REGO) (1 µM) for 24 h and subjected for WB analysis to examine expression of total and phosphorylated forms of VEGFR, FGFR, and FRS-2. Actin staining was used to show the comparable amounts of protein loaded into each sample. The original Western blot figures can be found in <a href="#app1-cancers-16-03103" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 4
<p>(<b>A</b>–<b>D</b>). Crosstalk between FGFR1/2 and VEGFR1/2 in IM-resistant GISTs. GIST T-1 R (<b>A</b>) and GIST 430 (<b>B</b>) were subjected to the double immunofluorescence staining for FGFR1 or 2 and VEGFR1 or 2. To outline the nucleus, the images were also merged with DAPI staining. (<b>C</b>) Percentages of cells with co-localized RTKs from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05; (<b>D</b>) co-immunoprecipitation of FGFR1 or 2 with VEGFR1 in IM-resistant GISTs. FGFR1 or -2 expression in GIST cell lysates immunoprecipitated by anti-VGFR1 Abs to demonstrate endogenous complex formation. The original Western blot figures can be found in <a href="#app1-cancers-16-03103" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 5
<p>(<b>A</b>,<b>B</b>) Pro-apoptotic and anti-proliferative activities of sunitinib (SU) (<b>A</b>) or regorafenib (REGO) (<b>B</b>) used alone or in combination with BGJ 398 in GIST T-1R cells. Cells were treated for 72 h and subjected to WB analysis to examine expression of apoptotic markers—cleaved forms of PARP and caspase-3 and downstream signaling molecules of FGFR and VEGFR pathways: total and phosphorylated forms of AKT, MAPK, and STAT-1. Actin staining was used to show the comparable amounts of protein loaded into each sample. The original Western blot figures can be found in <a href="#app1-cancers-16-03103" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 6
<p>(<b>A</b>–<b>D</b>) Anti-proliferative activity BGJ 398 used in combination with sunitinib (SU) or regorafenib (REGO) in GIST-T1R cells. Cells were treated with RTKis (BGJ398 1 µM, SU 0.5µM, REGO 0.5µM) for 72 h (<b>A</b>) Changes in growth kinetics of GIST-T1R cells treated with DMSO (control), SU or BGJ 398 alone and in combination; (<b>B</b>) changes in growth kinetics of GIST-T1R cells treated with DMSO (control), REGO, or BGJ 398 alone and in combination; (<b>C</b>) (<b>Upper</b>) panel—representative images of crystal violet staining of GIST-T1R cells treated with SU or BGJ 398 alone or in combination. (<b>Lower</b>) panel—quantification of crystal violet staining of GIST cells, as shown in the upper panel. *: <span class="html-italic">p</span> ≤ 0.001; (<b>D</b>) (<b>Upper</b>) panel—representative images of crystal violet staining of GIST-T1R cells treated with REGO or BGJ 398 alone or in combination. (<b>Lower</b>) panel—quantification of crystal violet staining of GIST cells, as shown in the upper panel. *: <span class="html-italic">p</span> ≤ 0.001. The culture dishes for (<b>C</b>) and (<b>D</b>) were stained with crystal violet and photographed. The cells treated with DMSO were used as a control. Quantification of crystal violet staining of GIST cells is described in <a href="#sec4-cancers-16-03103" class="html-sec">Section 4</a>.</p>
Full article ">Figure 7
<p>Assessment of the synergy between BGJ 398 and sunitinib (SU) (<b>A</b>) or regorafenib (REGO) (<b>B</b>) observed for IM-resistant GIST T-1R cells (ZIP model).</p>
Full article ">Figure 8
<p>Inhibition of KIT signaling activates FGFR and VEGFR signaling via overproduction of FGF-2 and VEGF-A in IM-resistant GIST-T1 cells.</p>
Full article ">
11 pages, 1254 KiB  
Article
HLA-A01 and HLA-B27 Supertypes, but Not HLA Homozygocity, Correlate with Clinical Outcome among Patients with Non-Small Cell Lung Cancer Treated with Pembrolizumab in Combination with Chemotherapy
by Afaf Abed, Anna Reid, Ngie Law, Michael Millward and Elin S. Gray
Cancers 2024, 16(17), 3102; https://doi.org/10.3390/cancers16173102 - 7 Sep 2024
Viewed by 1138
Abstract
Introduction: Maximal heterozygosity on the human leukocyte antigen (HLA) loci has been found to be associated with improved survival and development of immune-related adverse events (irAEs) among NSCLC patients treated with immunotherapy. Here, we investigated the effect of germline HLA-I/-II on clinical outcomes [...] Read more.
Introduction: Maximal heterozygosity on the human leukocyte antigen (HLA) loci has been found to be associated with improved survival and development of immune-related adverse events (irAEs) among NSCLC patients treated with immunotherapy. Here, we investigated the effect of germline HLA-I/-II on clinical outcomes among NSCLC patients treated with first-line pembrolizumab in combination with chemotherapy. Method: We prospectively recruited patients with NSCLC who were commencing first-line pembrolizumab in combination with chemotherapy. DNA from white blood cells was used for high-resolution HLA-I/II typing. Results: Of the 65 patients recruited, 53 complied with the inclusion criteria. We did not find an association between HLA-I/-II homozygosity and clinical outcome among the studied population. However, the presence of HLA-A01 was associated with unfavourable PFS (HR = 2.32, 95%CI 1.13–4.77, p = 0.022) and worsening OS (HR = 2.86, 95%CI 1.06–7.70, p = 0.038). The presence of HLA-B27 was associated with improved PFS (HR = 0.35, 95%CI 0.18–0.71, p = 0.004) and trends toward improving OS. None of the HLA-I supertypes were associated with the development or worsening of irAEs. Conclusions: The absence of association between genomic HLA-I/-II homozygosity and clinical outcome among patients with advanced NSCLC treated with pembrolizumab in combination with chemotherapy might reflect a diminished role for HLA molecules among patients with low or no PD-L1. HLA-A01 and HLA-B27 might have a role in predicting clinical outcomes among this cohort of patients. Further studies are needed to explore biomarkers for this group of patients. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of HLA-I/II homozygosity on PFS and OS among patients with advanced NSCLC treated with pembrolizumab in combination with chemotherapy. (<b>A</b>,<b>B</b>) Comparison of patients with homozygosity at one or more HLA-I loci on PFS and OS, respectively. (<b>C</b>,<b>D</b>) Comparison of patients with homozygosity at one or more HLA-II loci on PFS and OS, respectively. Kaplan–Meier curves were compared using log-rank analysis. HLA, human leukocyte antigen; NSCLC, non-small cell lung cancer; OS, overall survival; PFS, progression-free survival.</p>
Full article ">Figure 2
<p>Association of HLA-I supertypes with (<b>A</b>) PFS and (<b>B</b>) OS. Forest plot indicates hazard ratio (HR) with 95% confidence interval (CI). The number of patients with each supertype is indicated with (N), with % representing frequency in relation to the full cohort of 53 patients with NSCLC treated with pembrochemotherapy. HLA-B58 is not represented in the forest plot, as its frequency was only in two patients.</p>
Full article ">Figure 3
<p>The correlation between HLA-I supertypes and the development of any irAE. N: number of patients who developed irAEs with a specific supertype, with % representing the frequency in relation to the full number of patients that developed irAEs (16 patients); the RR: relative risks, associated CI: confidence intervals and <span class="html-italic">p</span>-values of a two-sided Fisher’s exact test are represented; irAE: immune-related adverse event.</p>
Full article ">
12 pages, 9453 KiB  
Article
The Local Rhombus-Shaped Flap—An Easy and Reliable Technique for Oncoplastic Breast Cancer Surgery and Defect Closure in Breast and Axilla
by Hisham Fansa and Sora Linder
Cancers 2024, 16(17), 3101; https://doi.org/10.3390/cancers16173101 - 6 Sep 2024
Viewed by 1038
Abstract
Primarily, breast-conserving therapy is an oncological intervention, but eventually it is judged by its cosmetic result. Remaining cavities from tumor resection can promote seromas, delay healing and cause lasting discomfort. Additionally, volume loss, dislocation of nipple/areola and fat necrosis lead to (cosmetically) unfavorable [...] Read more.
Primarily, breast-conserving therapy is an oncological intervention, but eventually it is judged by its cosmetic result. Remaining cavities from tumor resection can promote seromas, delay healing and cause lasting discomfort. Additionally, volume loss, dislocation of nipple/areola and fat necrosis lead to (cosmetically) unfavorable results, aggravated by radiotherapy. Oncoplastic surgery can reduce these sequelae. A local flap that has rarely been used in breast cancer surgery is the Limberg rhombic flap. The tumor defect is planned as a rhombus. The sides of the rhombus are of equal length and ideally have an angle of 60° and 120°. The flap that closes the defect is planned as an extension of equal length of the short diagonal. The second incision of the flap is placed according to the defect angle of 60°, running parallel to the defect at the same length. This creates a second rhombus. The flap is transposed into the defect, and the donor area is primarily closed. It is axially perfused and safe with a 1:1 length-to-width ratio. Compared to local perforator flaps, defect closure is easily managed without microsurgical skills. In the breast, the flap can be used in volume replacement and volume displacement techniques as an all-layer flap to cover defects, or it can be deepithelialized and buried. In the axilla, it can cover full-thickness defects when skin is involved. The advantages of the rhombic flap are its safety and simplicity to add volume and close defects, thus reducing the complexity of oncoplastic surgery. Full article
(This article belongs to the Special Issue Trends in Mastectomy and Breast Reconstruction for Cancer)
Show Figures

Figure 1

Figure 1
<p>Unfavorable cosmetic result 2 years after externally conducted breast-conserving therapy on the left breast and adjuvant radiation therapy. (<b>a</b>) The nipple–areola complex (NAC) is dislocated from the midline. (<b>b</b>) Volume loss and scarring further impair the cosmetic result. A primary local flap for volume replacement or oncoplastic reduction mastopexy would have been a better choice.</p>
Full article ">Figure 2
<p>Principle of the rhombic flap according to Limberg. (<b>a</b>) The defect (red) is shaped like a rhombus. The sides of the rhombus are of equal length and have an ideal angle of 60° and 120°. The flap (purple) has the same measurements as the defect. The flap that closes the defect is planned as an extension of the short diagonal (halves the two 120° angles, here A to C = A′ to B′). The second incision of the flap, B′ to C′, runs parallel to the defect, again with the same length. The base of the flap is not incised. The flap is transposed into the defect so that A′ is sutured to A, B′ to B and C′ to C. (<b>b</b>) The shape of the final scar after the defect and the donor site of the flap are closed.</p>
Full article ">Figure 3
<p>(<b>a</b>) The exact planning of the position (axis) of the rhombic defect usually has some play. Additionally, the definitive flap can be harvested 90° around the defect; thus, four options exist to close the defect, here named option 1 to 4. The best flap would be the one that adds the same amount of volume and whose donor site is best concealed, or whose donor site skin is most elastic to facilitate primary closure. (<b>b</b>) A larger defect (red) can be divided into two rhombi (dotted line). Each of the two smaller rhombi is closed by a separate Limberg flap (purple flap 1 and yellow flap 2). Even larger defects can be divided into 3 or 4 rhomboids. Caution: The primary closure of 4 Limberg flaps that are not buried results in a swastika as a scar at the end.</p>
Full article ">Figure 4
<p>Patient with bilateral invasive breast cancer; recurrence in the upper lateral side of the left breast. (<b>a</b>) The all-layered tumor resection was planned in a rhombic shape. The flap was pedicled dorsally and inferiorly with the donor site in an area of excess skin. Alternative pedicles would have been placed dorsally and cranially, or cranially and anteriorly. However, by placing it dorsally and inferiorly, the area of skin excess was used best with direct closure of the donor site. (<b>b</b>) Result after 4 months. No dislocation of the NAC, no visible volume loss of the breast.</p>
Full article ">Figure 5
<p>Patient with axillary recurrence on the left axilla 9 years after initial invasive breast cancer diagnosis: (<b>a</b>) Involvement of the skin is clearly visible. A resection with primary closure will result in a contracture and limitation of movement of the arm. (<b>b</b>) The resection was planned in a rhombus shape. The flap was pedicled inferiorly. There is no need for “complicated” flaps. Larger resections and defect coverage can also be performed with a rhombus-shaped flap. (<b>c</b>) Resected axillary recurrence with skin. (<b>d</b>) 3 months after surgery with free margins: primary healing of the flap. No contracture in the axilla, and no impaired range of movement. The patient then received radiotherapy and adjuvant immunotherapy.</p>
Full article ">Figure 6
<p>53-year-old patient with invasive breast cancer on the right breast. (<b>a</b>) Preoperative view with firm breast. (<b>b</b>) Wire marking and delineation of tumor borders. Tumor size: 3.5 × 4 cm. (<b>c</b>) The tumor is resected via a direct incision in this case. No skin resection is necessary. The flap is planned as a rhombus. The pedicle is placed cranially and dorsally. (<b>d</b>) Resected specimen; tumor weight is 85 g. (<b>e</b>) After tumor resection, the flap is incised in all layers. White star indicates the parenchymal defect after tumor resection. (<b>f</b>) Volume and NAC position are evaluated in the seating position with the flap temporarily fitted. Marking of the buried area of the flap. (<b>g</b>) Deepithelialization of the inferior, buried part of the flap. Fixation of the flap with Vicryl 3-0. (<b>h</b>) Immediately after wound closure, using Monocryl 4-0 intracutaneously. Drains are not necessary. (<b>i</b>,<b>j</b>) illustrate two weeks after surgery. Volume appears symmetrical, and NAC is in the right position, despite the large resection. (<b>k</b>,<b>l</b>) show results four months after surgery and eight weeks after radiation therapy. The ultrasound (<b>k</b>) shows no dead space/cavity or necrosis. The flap has healed well. The white arrows show the anterior deepithelialized buried part of the flap. The frontal view shows a symmetrical result and no NAC dislocation after radiation therapy.</p>
Full article ">Figure 6 Cont.
<p>53-year-old patient with invasive breast cancer on the right breast. (<b>a</b>) Preoperative view with firm breast. (<b>b</b>) Wire marking and delineation of tumor borders. Tumor size: 3.5 × 4 cm. (<b>c</b>) The tumor is resected via a direct incision in this case. No skin resection is necessary. The flap is planned as a rhombus. The pedicle is placed cranially and dorsally. (<b>d</b>) Resected specimen; tumor weight is 85 g. (<b>e</b>) After tumor resection, the flap is incised in all layers. White star indicates the parenchymal defect after tumor resection. (<b>f</b>) Volume and NAC position are evaluated in the seating position with the flap temporarily fitted. Marking of the buried area of the flap. (<b>g</b>) Deepithelialization of the inferior, buried part of the flap. Fixation of the flap with Vicryl 3-0. (<b>h</b>) Immediately after wound closure, using Monocryl 4-0 intracutaneously. Drains are not necessary. (<b>i</b>,<b>j</b>) illustrate two weeks after surgery. Volume appears symmetrical, and NAC is in the right position, despite the large resection. (<b>k</b>,<b>l</b>) show results four months after surgery and eight weeks after radiation therapy. The ultrasound (<b>k</b>) shows no dead space/cavity or necrosis. The flap has healed well. The white arrows show the anterior deepithelialized buried part of the flap. The frontal view shows a symmetrical result and no NAC dislocation after radiation therapy.</p>
Full article ">
26 pages, 2978 KiB  
Review
Scoping Review: Methods and Applications of Spatial Transcriptomics in Tumor Research
by Kacper Maciejewski and Patrycja Czerwinska
Cancers 2024, 16(17), 3100; https://doi.org/10.3390/cancers16173100 - 6 Sep 2024
Viewed by 3037
Abstract
Spatial transcriptomics (ST) examines gene expression within its spatial context on tissue, linking morphology and function. Advances in ST resolution and throughput have led to an increase in scientific interest, notably in cancer research. This scoping study reviews the challenges and practical applications [...] Read more.
Spatial transcriptomics (ST) examines gene expression within its spatial context on tissue, linking morphology and function. Advances in ST resolution and throughput have led to an increase in scientific interest, notably in cancer research. This scoping study reviews the challenges and practical applications of ST, summarizing current methods, trends, and data analysis techniques for ST in neoplasm research. We analyzed 41 articles published by the end of 2023 alongside public data repositories. The findings indicate cancer biology is an important focus of ST research, with a rising number of studies each year. Visium (10x Genomics, Pleasanton, CA, USA) is the leading ST platform, and SCTransform from Seurat R library is the preferred method for data normalization and integration. Many studies incorporate additional data types like single-cell sequencing and immunohistochemistry. Common ST applications include discovering the composition and function of tumor tissues in the context of their heterogeneity, characterizing the tumor microenvironment, or identifying interactions between cells, including spatial patterns of expression and co-occurrence. However, nearly half of the studies lacked comprehensive data processing protocols, hindering their reproducibility. By recommending greater transparency in sharing analysis methods and adapting single-cell analysis techniques with caution, this review aims to improve the reproducibility and reliability of future studies in cancer research. Full article
Show Figures

Figure 1

Figure 1
<p>Current interests of spatial transcriptomics in cancer research. (<b>A</b>) The number of unique spatial transcriptomics (ST) samples related to cancer research deposited at the Gene Expression Omnibus (GEO) repository. (<b>B</b>) The number of spatial transcriptomics (ST) publications related to tumor research per year indexed in PubMed. Data up to 31 December 2023. See the individual queries in <a href="#app1-cancers-16-03100" class="html-app">Table S1</a>.</p>
Full article ">Figure 2
<p>Standard ST analysis workflow of sequencing-based methods. For array-based platforms, mRNAs are labeled with arrays of spatially barcoded probes with a sequence indicating location before sequencing. In the microdissection-based approach, cells or regions of interest can be directly microdissected and their locations recorded before their transcriptomes are sequenced. Later on, sequencing results undergo standardized pre-processing to obtain count matrices including expression values of detected genes per each spot/cell. Next, typical data analysis incorporates quality control with filtering of low-quality spatial regions, expression normalization, feature selection, possible data integration with other samples or data types, tissue segmentation with image analysis, and data annotation. The final steps include data exploratory methods and interpretation, which are highly study-specific; thus, analysis workflows may differ per article. <span class="html-italic">ROI</span>—region of interest. <span class="html-italic">UMI</span>—unique molecular identifier. Based on Williams et al. [<a href="#B10-cancers-16-03100" class="html-bibr">10</a>] and Fang et al. [<a href="#B18-cancers-16-03100" class="html-bibr">18</a>].</p>
Full article ">Figure 3
<p>Scoping analysis workflow, study selection criteria, and number of studies at each phase. We thoroughly analyzed 41 articles, which consisted of 38 neoplasm biology-based articles (<span class="html-italic">biology-oriented</span>) and 3 articles focused entirely on machine learning (ML) applications in the biomedical domain (<span class="html-italic">ML-oriented</span>). <span class="html-italic">Not tumor-based</span>—articles did not use tumor tissues. <span class="html-italic">data analysis tool</span>—articles focused entirely on presenting new data analysis methods and did not directly derive any new biological insights. <span class="html-italic">methodology</span>—articles were related to library preparation or sequencing protocols (e.g., methods comparison).</p>
Full article ">Figure 4
<p>Overview of studies taken into account for the scoping analysis. (<b>A</b>) Proportions of articles’ publication years. (<b>B</b>) Proportions of data availability approaches of analyzed studies. Only studies that collected and processed their own ST samples are included. (<b>C</b>) Proportions of the availability of code developed by analyzed studies. Only studies that used programming language-based solutions (e.g., R/Python libraries) for data analysis are included. (<b>D</b>) The number of organs whose sample tissues were collected from for every analyzed study. Two studies utilized samples from multiple anatomical regions. <span class="html-italic">Y</span> (yes)—available, authors state the repository and access number where the developed data/code can be found. <span class="html-italic">N</span> (no)—unavailable, authors do not mention at all that developed data/code is accessible in any way; <span class="html-italic">UR</span> (upon request)—authors declare that the data/code is available upon reasonable request to appropriate corresponding authors.</p>
Full article ">Figure 5
<p><b>Trends in sample origin and their development platform.</b> (<b>A</b>) Proportions of sample sources for each analyzed study. The tag <span class="html-italic">own</span> represents that authors collected and processed samples fully on their own. Other data sources are publicly available repositories. One study was classified as both <span class="html-italic">own</span> and <span class="html-italic">GEO</span> because original data findings were later confirmed using publicly available data from GEO. (<b>B</b>) Proportions of spatial transcriptomics sequencing-based platforms employed by studies that collected and processed their own samples.</p>
Full article ">Figure 6
<p><b>Trends in standards of data quality-control reporting and expression normalization methods.</b> (<b>A</b>) Proportions of reported data quality-control protocols. Studies analyzing data from the publicly available 10x Genomics repository are not included. <span class="html-italic">Y</span> (yes)—quality control (e.g., filtering, removal of specific spots, and sequencing quality) was reported completely and exhaustively with all necessary details. <span class="html-italic">ND</span> (no details)—quality control was reported as an employed data processing step but without any explicit information on how it was performed. <span class="html-italic">N</span> (no)—authors did not report any quality-control measures in their study. (<b>B</b>) The number of times each normalization protocol was mentioned in the analyzed articles that used the Visium (10x Genomics) platform for their spatial transcriptomics data development. One article was classified thrice. <span class="html-italic">ND</span> (no details)—authors declared that data were normalized but did not provide any specific algorithm or protocol.</p>
Full article ">Figure 7
<p>Overview of reported software used for ST data analysis, which was mentioned in (<b>A</b>) at least two Visium-based analyzed studies and (<b>B</b>) at least one GeoMx-based analyzed study.</p>
Full article ">Figure 8
<p>Insights of ST data analysis trends. (<b>A</b>) Functions/methods used for ST data processing and analysis (e.g., transforming, reshaping, clustering, and integrating) that were used in at least two articles. (<b>B</b>) Data dimensionality reduction methods reported in the analyzed articles whenever mentioned. (<b>C</b>) Other data sources used alongside spatial transcriptomics in analyzed articles. The tag <span class="html-italic">cell culture</span> represents the group of standard methods and analyses used while performing cell cultures. <span class="html-italic">Mass spectrometry</span> also includes mass cytometry protocols. Any animal assays were omitted in this analysis.</p>
Full article ">Figure 9
<p>The analysis of current ST application trends. (<b>A</b>) Article keywords that were associated with at least two articles. (<b>B</b>) Number of times each category of analysis was performed on ST data in at least two articles. Categories were assigned to each article based on a scoping analysis of all articles. <span class="html-italic">Tumor heterogeneity</span>—here, an overloaded umbrella term for distinct analyses of tumor landscape composition, tumor microenvironment, and cell–cell interactions, categorized together only for bias reduction of current study. <span class="html-italic">DEA</span>—differential expression analysis (e.g., identification of differentially expressed genes between tissue types, highly variable genes, and spatially variable genes). <span class="html-italic">GSEA</span>—gene set enrichment analysis (e.g., tumor hallmarks, pathway enrichment, and signature assessments). <span class="html-italic">cell-type deconvolution</span>—predictions of specific cell types present in each spot (e.g., immune infiltration based on annotated single-cell data). <span class="html-italic">GO</span>—gene ontology analysis. <span class="html-italic">KEGG</span>—pathway analysis based on the Kyoto Encyclopedia of Genes and Genomes database. <span class="html-italic">expression distribution</span>—simple plotting of spatial expression distribution of genes of interest (this category does not overlap with <span class="html-italic">tumor heterogeneity</span>). <span class="html-italic">cell trajectory analysis</span>—pseudotime analysis (e.g., spot-wise relative distances). <span class="html-italic">GSVA</span>—gene set variation analysis. <span class="html-italic">ssGSEA</span>—single-sample gene set enrichment analysis. <span class="html-italic">WGCNA</span>—weighted correlation network analysis (e.g., for gene co-expression). <span class="html-italic">predictive model</span>—ST data were fed into a machine-learning model to create spatially resolved-bsed predictions.</p>
Full article ">
4 pages, 177 KiB  
Editorial
Fecundity and Nutrient Deficiency Following Obesity Treatment: Implications for Young-Onset Cancer Risk in Offspring
by Savio George Barreto, Chris Moy, Stephen J. Pandol and Lilian Kow
Cancers 2024, 16(17), 3099; https://doi.org/10.3390/cancers16173099 - 6 Sep 2024
Viewed by 829
Abstract
Young-onset adult cancers have been an emerging problem over the last three decades in Australia [...] Full article
(This article belongs to the Section Cancer Epidemiology and Prevention)
18 pages, 1058 KiB  
Review
Developmental Therapeutics in Metastatic Prostate Cancer: New Targets and New Strategies
by Jingsong Zhang and Juskaran S. Chadha
Cancers 2024, 16(17), 3098; https://doi.org/10.3390/cancers16173098 - 6 Sep 2024
Viewed by 2027
Abstract
There is an unmet need to develop new treatments for metastatic prostate cancer. With the development of targeted radioligand therapies, bispecific T cell engagers, antibody–drug conjugates and chimeric antigen receptor T cell (CAR T) therapies, tumor-associated cell surface antigens have emerged as new [...] Read more.
There is an unmet need to develop new treatments for metastatic prostate cancer. With the development of targeted radioligand therapies, bispecific T cell engagers, antibody–drug conjugates and chimeric antigen receptor T cell (CAR T) therapies, tumor-associated cell surface antigens have emerged as new therapeutic targets in metastatic prostate cancer. Ongoing and completed clinical trials targeting prostate-specific membrane antigen (PSMA), six transmembrane epithelial antigens of the prostate 1 (STEAP1), kallikrein-related peptidase 2 (KLK2), prostate stem cell antigen (PSCA), and delta-like protein 3 (DLL3) in metastatic prostate cancer were reviewed. Strategies for sequential or combinational therapy were discussed. Full article
(This article belongs to the Special Issue New Insights into Urologic Oncology)
Show Figures

Figure 1

Figure 1
<p>Clinical course of an mCRPC patient who was enrolled in the anti-PSMAxCD28 BiTE study followed by Lutetium Lu177 vivpivotide tetraxetan (Lu177). (<b>a</b>) Coronal CT images of baseline (<b>left</b>), post cycle 1 (<b>middle</b>), and post cycle 2 (<b>right</b>) liver metastases (yellow arrow) before and during treatment with anti-PSMAxCD28/REGN5678 and Cemiplimab per NCT05125016 protocol. (<b>b</b>) Changes in PSA values (y-axis) over time. R: REGN5678; C: Cemiplimab; C1PD: post cycle 1 progressive disease in the liver; C2PR: post cycle 2 partial response in liver.</p>
Full article ">Figure 2
<p>Schema of the phase I γδ enriched autologous CAR T trial targeting PSCA in mCRPC (NCT06193486). Zoledronic acid will be given prior to day-14 leukapheresis. The lymphodepletion regimen includes cyclophosphamide 500 mg/m<sup>2</sup> and fludarabine (30 mg/m<sup>2</sup>) administered over 3 days (days-5, -4, -3). Fresh CAR T product is infused at day 0, which will be followed by a 28-day observation period for dose-limiting toxicity (DLT).</p>
Full article ">
12 pages, 1254 KiB  
Article
Deep Learning Model for Predicting Lung Adenocarcinoma Recurrence from Whole Slide Images
by Ziyu Su, Usman Afzaal, Shuo Niu, Margarita Munoz de Toro, Fei Xing, Jimmy Ruiz, Metin N. Gurcan, Wencheng Li and M. Khalid Khan Niazi
Cancers 2024, 16(17), 3097; https://doi.org/10.3390/cancers16173097 - 6 Sep 2024
Viewed by 1230
Abstract
Lung cancer is the leading cause of cancer-related death in the United States. Lung adenocarcinoma (LUAD) is one of the most common subtypes of lung cancer that can be treated with resection. While resection can be curative, there is a significant risk of [...] Read more.
Lung cancer is the leading cause of cancer-related death in the United States. Lung adenocarcinoma (LUAD) is one of the most common subtypes of lung cancer that can be treated with resection. While resection can be curative, there is a significant risk of recurrence, which necessitates close monitoring and additional treatment planning. Traditionally, microscopic evaluation of tumor grading in resected specimens is a standard pathologic practice that informs subsequent therapy and patient management. However, this approach is labor-intensive and subject to inter-observer variability. To address the challenge of accurately predicting recurrence, we propose a deep learning-based model to predict the 5-year recurrence of LUAD in patients following surgical resection. In our model, we introduce an innovative dual-attention architecture that significantly enhances computational efficiency. Our model demonstrates excellent performance in recurrent risk stratification, achieving a hazard ratio of 2.29 (95% CI: 1.69–3.09, p < 0.005), which outperforms several existing deep learning methods. This study contributes to ongoing efforts to use deep learning models for automatically learning histologic patterns from whole slide images (WSIs) and predicting LUAD recurrence risk, thereby improving the accuracy and efficiency of treatment decision making. Full article
(This article belongs to the Section Cancer Informatics and Big Data)
Show Figures

Figure 1

Figure 1
<p>Overview of our tumor bulk segmentation pipeline, including tumor patch prediction, tumor bulk mask refinement, and tumor bulk extraction.</p>
Full article ">Figure 2
<p>Overview of the DAMIL architecture. The tumor bulk region from a WSI is extracted and cropped into patches during the preprocessing step. All patches are further encoded into vectors by a feature extractor model. The encoded vectors go through a cross-attention module followed by a gated-attention module. The final representation is used for RC/NRC prediction.</p>
Full article ">Figure 3
<p>Kaplan–Meier plots for recurrence-free probability for 5-year follow-up.</p>
Full article ">
11 pages, 968 KiB  
Review
Decision Variables for the Use of Radioactive Iodine in Patients with Thyroid Cancer at Intermediate Risk of Recurrence
by Samantha K. Newman, Armando Patrizio and Laura Boucai
Cancers 2024, 16(17), 3096; https://doi.org/10.3390/cancers16173096 - 6 Sep 2024
Cited by 1 | Viewed by 1268
Abstract
The use of radioactive iodine (RAI) after total thyroidectomy for patients at the American Thyroid Association (ATA) who are at intermediate risk of recurrence is controversial. This is due to the lack of prospective randomized trials proving a benefit to recurrence or survival [...] Read more.
The use of radioactive iodine (RAI) after total thyroidectomy for patients at the American Thyroid Association (ATA) who are at intermediate risk of recurrence is controversial. This is due to the lack of prospective randomized trials proving a benefit to recurrence or survival of RAI therapy in this group. In the absence of such evidence, clinicians struggle to recommend for or against this therapeutic approach which frequently results in overtreatment. This review describes key elements in the decision-making process that help clinicians more comprehensively evaluate the need for RAI therapy in patients with thyroid cancer at intermediate risk of recurrence. A clear definition of the purpose of RAI therapy should be conveyed to patients. In this sense, adjuvant RAI therapy intends to decrease recurrence, and ablation therapy is used to facilitate surveillance. Better stratification of the intermediate risk category into a low–intermediate subgroup and an intermediate–high-risk subgroup results in less heterogeneity and a more precise prediction of recurrence risk. The evaluation of post-operative thyroglobulin levels may prevent the overtreatment of low–intermediate-risk patients when their thyroglobulin level is <2.5 ng/mL. the integration of tumor genomics (when available) alongside pathologic features can enhance the ability of the clinician to predict iodine concentration in thyroid cancer cells. Finally, a detailed consideration of the adverse effects of RAI, patients’ comorbidities, and patient preferences will result in a patient-centered personalized approach. Systematic examination of these variables will ultimately provide a framework for making more educated decisions on the use of RAI in patients at intermediate risk of recurrence that will prevent overtreatment and minimize harm. Full article
(This article belongs to the Special Issue Feature Paper in Section 'Cancer Epidemiology and Prevention' in 2024)
Show Figures

Figure 1

Figure 1
<p>Key variables in the decision-making process for RAI use in patients with thyroid cancer at intermediate risk of recurrence.</p>
Full article ">
11 pages, 1337 KiB  
Article
Real-Life Outcomes of Adjuvant Targeted Therapy and Anti-PD1 Agents in Stage III/IV Resected Melanoma
by Gabriele Roccuzzo, Paolo Fava, Chiara Astrua, Matteo Giovanni Brizio, Giovanni Cavaliere, Eleonora Bongiovanni, Umberto Santaniello, Giulia Carpentieri, Luca Cangiolosi, Camilla Brondino, Valentina Pala, Simone Ribero and Pietro Quaglino
Cancers 2024, 16(17), 3095; https://doi.org/10.3390/cancers16173095 - 6 Sep 2024
Cited by 3 | Viewed by 1075
Abstract
This study was carried out at the Dermatologic Clinic of the University of Turin, Italy, to assess the effectiveness and safety of adjuvant therapy in patients who received either targeted therapy (TT: dabrafenib + trametinib) or immunotherapy (IT: nivolumab or pembrolizumab) for up [...] Read more.
This study was carried out at the Dermatologic Clinic of the University of Turin, Italy, to assess the effectiveness and safety of adjuvant therapy in patients who received either targeted therapy (TT: dabrafenib + trametinib) or immunotherapy (IT: nivolumab or pembrolizumab) for up to 12 months. A total of 163 patients participated, including 147 with stage III and 19 with stage IV with no evidence of disease. The primary outcomes were relapse-free survival (RFS), distant metastasis-free survival (DMFS), and overall survival (OS). At 48 months, both TT and IT approaches yielded comparable outcomes in terms of RFS (55.6–55.4%, p = 0.532), DMFS (58.2–59.8%, p = 0.761), and OS (62.4–69.5%, p = 0.889). Whilst temporary therapy suspension was more common among TT-treated patients compared to IT-treated individuals, therapy discontinuation due to adverse events occurred at comparable rates in both groups. Predictors of relapse included mitoses, lymphovascular invasion, ulceration, and positive sentinel lymph nodes. Overall, the proportion of BRAF-mutated patients receiving IT stood at 7.4%, lower than what was observed in clinical trials. Full article
(This article belongs to the Special Issue Advances in Skin Cancer: Diagnosis, Treatment and Prognosis)
Show Figures

Figure 1

Figure 1
<p>Relapse-free survival at 48 months according to stage (<b>a</b>) and therapy (<b>b</b>).</p>
Full article ">Figure 2
<p>Distant metastasis-free survival at 48 months according to stage (<b>a</b>) and therapy (<b>b</b>).</p>
Full article ">Figure 3
<p>Overall survival at 48 months according to stage (<b>a</b>) and therapy (<b>b</b>).</p>
Full article ">
16 pages, 538 KiB  
Review
Transdermal Fentanyl in Patients with Cachexia—A Scoping Review
by Andrea Carlini, Emanuela Scarpi, Carla Bettini, Andrea Ardizzoni, Costanza Maria Donati, Laura Fabbri, Francesca Ghetti, Francesca Martini, Marianna Ricci, Elisabetta Sansoni, Maria Valentina Tenti, Alessio Giuseppe Morganti, Eduardo Bruera, Marco Cesare Maltoni and Romina Rossi
Cancers 2024, 16(17), 3094; https://doi.org/10.3390/cancers16173094 - 5 Sep 2024
Viewed by 1385
Abstract
Cachectic patients frequently require transdermal fentanyl (TDF) for pain management, but data on its efficacy and safety are scarce and inconsistent. This scoping review aims to analyze the evidence concerning TDF administration in patients with cachexia irrespective of the underlying pathology. The primary [...] Read more.
Cachectic patients frequently require transdermal fentanyl (TDF) for pain management, but data on its efficacy and safety are scarce and inconsistent. This scoping review aims to analyze the evidence concerning TDF administration in patients with cachexia irrespective of the underlying pathology. The primary objective is to assess the analgesic efficacy and tolerability of TDF in cachectic patients. The secondary objective is to identify cachexia characteristics that may influence fentanyl pharmacokinetics (PK). A comprehensive search of PubMed, Embase, and Web of Science databases was conducted up to March 2024. The review included observational and clinical studies on cachectic patients with moderate to severe pain treated with TDF patches at any dosage or frequency. Phase 1 trials, animal studies, case reports, preclinical studies and conference abstracts were excluded. Nine studies were included: four studies reported that cachexia negatively impacted TDF efficacy, increasing required doses and lowering plasma concentrations; three studies found minimal or no impact of cachexia on TDF efficacy and PK; two studies suggested that cachexia might improve TDF outcomes. Study quality ranged from moderate to high, according to the National Institutes of Health (NIH) Quality Assessment Tool. The current evidence is insufficient to provide any definitive recommendations for TDF prescribing in cachectic patients. Full article
(This article belongs to the Section Cancer Survivorship and Quality of Life)
Show Figures

Figure 1

Figure 1
<p>Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews (PRISMA-ScR) flow diagram.</p>
Full article ">
16 pages, 3309 KiB  
Article
A Comprehensive Analysis of Skin Cancer Concerns and Protective Practices in Manitoba, Canada, Highlights Lack of Skin Cancer Awareness and Predominance of High-Risk Sun Exposure Behaviors
by François Lagacé, Santina Conte, Lorena A. Mija, Amina Moustaqim-Barrette, Farhan Mahmood, Jonathan LeBeau, Alyson McKenna, Mahan Maazi, Johnny Hanna, Alexandra Sarah Victoria Kelly, Elham Rahme, Travis J. Hrubeniuk, Sandra Peláez and Ivan V. Litvinov
Cancers 2024, 16(17), 3093; https://doi.org/10.3390/cancers16173093 - 5 Sep 2024
Cited by 2 | Viewed by 1421
Abstract
The rapidly increasing skin cancer rates in Canada are alarming, with current data estimating that 1/3 of Canadians will be affected in their lifetime. Thus, deeper understanding of high-risk sun exposure behaviors is needed to help counter this trend. Only limited action has [...] Read more.
The rapidly increasing skin cancer rates in Canada are alarming, with current data estimating that 1/3 of Canadians will be affected in their lifetime. Thus, deeper understanding of high-risk sun exposure behaviors is needed to help counter this trend. Only limited action has been taken by federal/provincial governments to reduce skin cancer incidence. A cross-sectional survey study was conducted in Manitoba, with frequency counts, means, and percentages used to encapsulate responses. Age- and gender-adjusted odds ratios were calculated using logistic regression analyses. Our study identified worrying inadequacies in sun protective behaviors and attitudes, with the threat of such high-risk behaviors amplified by a lack of skin cancer awareness. Alarming elements were noted in participants’ sun exposure history (>65% reported a history of sunburns, >50% previously used a tanning bed, and >75% recently tanned for pleasure), beliefs and attitudes (>50% believe that they look better/healthier with a tan, and >40% believe that having a base tan is protective against further sun damage), and sun protection efforts (sun protective clothing was used <60% of the time, sunscreen was used by <50%, and there was a lack of knowledge about sunscreen characteristics in ~30% of respondents), in addition to significant differences being established between demographic subgroups (based on gender, age, skin phototype, income, and education attained). This study provides worrisome insight onto the grim landscape of sun protective behaviors and attitudes in Manitoba, which will inevitably translate into higher skin cancer rates and should serve as a call to action to promote targeted public health messaging in this jurisdiction and beyond. Full article
(This article belongs to the Special Issue Skin Cancer: Risk Factors and Prevention)
Show Figures

Figure 1

Figure 1
<p>Comparison of sun exposure, melanoma risk factors, sun protection habits, and level of worry for melanoma between women (<span class="html-italic">n</span> = 2369) and men (<span class="html-italic">n</span> = 964), depicted as a bar graph (<b>a</b>) and corresponding forest plot (<b>b</b>), between Fitzpatrick skin phototypes I-III (<span class="html-italic">n</span> = 2650) and phototypes IV-VI (<span class="html-italic">n</span> = 684) as a bar graph (<b>c</b>) and corresponding forest plot (<b>d</b>), and between ages 18 and 49 years (<span class="html-italic">n</span> = 1120) vs. individuals aged ≥50 years (<span class="html-italic">n</span> = 2227) as a bar graph (<b>e</b>) and corresponding forest plot (<b>f</b>). Odds ratios (OR) are adjusted for age and gender where appropriate.</p>
Full article ">Figure 2
<p>Comparison of sun exposure, melanoma risk factors, sun protection habits, and level of worry for melanoma between those that have completed a university degree (<span class="html-italic">n</span> = 1833) and those that have not completed a university degree (<span class="html-italic">n</span> = 1459) (<b>a</b>) and corresponding forest plot (<b>b</b>), and between individuals with an annual income ≥CAD 50,000 (<span class="html-italic">n</span> = 2430) and individuals with an annual income &lt;CAD 50,000 (<span class="html-italic">n</span> = 338) as a bar graph (<b>c</b>) and corresponding forest plot (<b>d</b>). Odds ratios (OR) are adjusted for age and gender where appropriate.</p>
Full article ">
17 pages, 1874 KiB  
Article
Apoptosis of Pancreatic Cancer Cells after Co-Treatment with Eugenol and Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand
by Hyun Hee Kim, Suk-Young Lee and Dae-Hee Lee
Cancers 2024, 16(17), 3092; https://doi.org/10.3390/cancers16173092 - 5 Sep 2024
Viewed by 1091
Abstract
Pancreatic cancer is a refractory cancer with limited treatment options. Various cancer types are resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Eugenol, the main component of clove oil, exhibits anticancer, anti-inflammatory, and antioxidant effects. However, no studies have reported that eugenol increases [...] Read more.
Pancreatic cancer is a refractory cancer with limited treatment options. Various cancer types are resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Eugenol, the main component of clove oil, exhibits anticancer, anti-inflammatory, and antioxidant effects. However, no studies have reported that eugenol increases TRAIL sensitivity by upregulating death receptor (DR) expression. Here, we aimed to investigate eugenol as a potent TRAIL sensitizer. Increased apoptosis and inhibition of cell proliferation was observed in pancreatic cancer cells treated with eugenol and TRAIL compared with those treated with eugenol alone. Eugenol upregulated the expression of DR5, inhibited the FLICE-inhibitory protein (FLIP), an anti-apoptotic protein, and increased p53, a tumor suppressor protein. In addition, eugenol induced the generation of reactive oxygen species (ROS) and caused endoplasmic reticulum (ER) stress. C/EBP-homologous protein (CHOP) knockdown using siRNA decreased the expression of DR5 and reduced the combined effects of eugenol and TRAIL. These results demonstrate that eugenol enhances TRAIL-induced apoptosis by upregulating DR5 through the ROS-mediated ER stress–CHOP pathway, which enhances ER stress by inducing p53 and downregulating FLIP expression. This suggests that eugenol has the potential to treat pancreatic cancer by increasing cell sensitivity to TRAIL. Full article
(This article belongs to the Section Cancer Pathophysiology)
Show Figures

Figure 1

Figure 1
<p>Cell proliferation inhibition assay of eugenol and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on human pancreatic cancer cell lines. (<b>a</b>) Chemical structure of eugenol. Graphs showing the proliferation rate of various cell lines after exposure to (<b>b</b>) eugenol and (<b>c</b>) TRAIL. (<b>d</b>) In the crystal violet assay and (<b>e</b>) colony formation assay, PANC-1 cells were treated with eugenol and stained with crystal violet. Each result is presented as the mean of three independent experiments; asterisks indicate significant differences at * <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.</p>
Full article ">Figure 2
<p>Effect of combined eugenol and TRAIL treatment on cell proliferation of PANC-1 cells. (<b>a</b>) HPDE cells and (<b>b</b>) PANC-1 cells were treated with eugenol and TRAIL; cell viability was determined. (<b>c</b>) In the crystal violet assay and (<b>d</b>) colony formation assay, PANC-1 cells were treated with eugenol and TRAIL, stained with crystal violet. (<b>e</b>) PANC-1 cells were treated with eugenol and TRAIL, and the migration of cells was assessed using a wound healing assay. Each result is presented as the mean of three independent experiments; asterisks indicate significant differences at * <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.</p>
Full article ">Figure 3
<p>Effect of co-treatment with eugenol and TRAIL on TRAIL-induced apoptosis of PANC-1 cells. Dual fluorescence scatter plot and graphs showing the apoptosis rate of PANC-1 cells after exposure to (<b>a</b>) eugenol and (<b>b</b>) TRAIL. (<b>c</b>) PANC-1 cells treated with both eugenol and TRAIL for 24 h. Flow cytometry was performed using double staining with Annexin V and PI. (<b>d</b>) Effect of combined treatment with eugenol and TRAIL on levels of apoptosis-associated proteins. (<b>e</b>) ELISA-based luminescence assays were used to measure caspase 3/7 activities following treatment with eugenol and TRAIL. * <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. The original Western blot figures can be found in <a href="#app1-cancers-16-03092" class="html-app">Figure S1</a>.</p>
Full article ">Figure 4
<p>Eugenol induces DR5 expression in PANC-1 cells. (<b>a</b>) The protein levels of pro-apoptotic and anti-apoptotic proteins were examined using Western blot analysis. Death receptor expression levels after eugenol treatment in (<b>b</b>) HPDE cells and (<b>c</b>) PANC-1 cells. Flow cytometry results showing (<b>d</b>) DR5 and (<b>e</b>) DR4 expression on the cell surface. (<b>f</b>) Western blotting results showing DR5 expression from various cell lines. *** <span class="html-italic">p</span> &lt; 0.001; ns = not significant. The original Western blot figures can be found in <a href="#app1-cancers-16-03092" class="html-app">Figure S1</a>.</p>
Full article ">Figure 5
<p>Eugenol induces ER stress in PANC-1 cells. (<b>a</b>) PANC-1 cells exposed to varying concentrations of eugenol for 24 h. The expression levels of ER stress-related proteins were analyzed using Western blotting; (<b>b</b>) PANC-1 cells treated with 400 μM eugenol and stained with DCFH-DA. (<b>c</b>) ROS production levels from the DCFH-DA analysis following 12 h treatment with 400 μM of eugenol in HPDE and PANC-1 cells. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; ns = not significant. The original Western blot figures can be found in <a href="#app1-cancers-16-03092" class="html-app">Figure S1</a>.</p>
Full article ">Figure 6
<p>Eugenol-induced upregulation of DR5 and CHOP mediated by ROS. (<b>a</b>) PANC-1 cells pretreated with 10 mM NAC for 1 h, treated with 400 μM eugenol for 12 h, and stained with DCFH-DA. The fluorescence intensity of DCF-DA expressed in the cells was measured using flow cytometry. (<b>b</b>) PANC-1 cells pretreated with NAC for 1 h, treated with eugenol for 12 h, and stained with DCFH-DA and DHE. The fluorescence intensity of DCF-DA in the cells was determined using a fluorescence microscope at a magnification of 10×. (<b>c</b>) PANC-1 cells pretreated with NAC for 1 h and subsequently treated with eugenol for 24 h. DR5 and CHOP expression were analyzed by Western blotting. PANC-1 cells pretreated with NAC and subsequently treated with eugenol or TRAIL, followed by analysis for (<b>d</b>) DR5 and CHOP expression and (<b>e</b>) PARP and cleaved-PARP expression using Western blotting. (<b>f</b>) The dual fluorescence scatter plot and graphs illustrate the apoptosis rate of PANC-1 cells following exposure to eugenol, TRAIL, and NAC. PANC-1 cells pretreated with NAC and subsequently treated with eugenol or TRAIL were analyzed for apoptosis using flow cytometry following Annexin V and PI staining. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001. The original Western blot figures can be found in <a href="#app1-cancers-16-03092" class="html-app">Figure S1</a>.</p>
Full article ">Figure 7
<p>Induction of DR5 by eugenol mediated through CHOP activation. (<b>a</b>) The expression levels of DR5 and CHOP were analyzed by Western blotting in PANC-1 cells treated with eugenol over a time course. (<b>b</b>) PANC-1 cells were transfected with CHOP siRNA for 6 h and then treated with eugenol or TRAIL, followed by the determination of protein expression levels of PARP, DR5, and CHOP using Western blotting. (<b>c</b>) After transfection with CHOP siRNA, PANC-1 cells were treated with eugenol or TRAIL and subsequently stained with Annexin V and PI, followed by apoptosis analysis using flow cytometry. (<b>d</b>) Diagram of the mechanisms of action of eugenol. Pathway activation, signal propagation, or expression promotion is indicated by the symbol “⟶”, whereas pathway inactivation, signaling suppression, or expression downregulation is represented by the symbol “⊣”. ** <span class="html-italic">p</span> &lt; 0.01. The original Western blot figures can be found in <a href="#app1-cancers-16-03092" class="html-app">Figure S1</a>.</p>
Full article ">Figure 7 Cont.
<p>Induction of DR5 by eugenol mediated through CHOP activation. (<b>a</b>) The expression levels of DR5 and CHOP were analyzed by Western blotting in PANC-1 cells treated with eugenol over a time course. (<b>b</b>) PANC-1 cells were transfected with CHOP siRNA for 6 h and then treated with eugenol or TRAIL, followed by the determination of protein expression levels of PARP, DR5, and CHOP using Western blotting. (<b>c</b>) After transfection with CHOP siRNA, PANC-1 cells were treated with eugenol or TRAIL and subsequently stained with Annexin V and PI, followed by apoptosis analysis using flow cytometry. (<b>d</b>) Diagram of the mechanisms of action of eugenol. Pathway activation, signal propagation, or expression promotion is indicated by the symbol “⟶”, whereas pathway inactivation, signaling suppression, or expression downregulation is represented by the symbol “⊣”. ** <span class="html-italic">p</span> &lt; 0.01. The original Western blot figures can be found in <a href="#app1-cancers-16-03092" class="html-app">Figure S1</a>.</p>
Full article ">
12 pages, 2767 KiB  
Article
Initial Experience of Single-Port Robotic Lobectomy for Large-Sized Non-Small Cell Lung Cancer: A Single-Center Retrospective Study
by Jun Hee Lee, Byung Mo Gu, Hwan Seok Yong, Soon Young Hwang and Hyun Koo Kim
Cancers 2024, 16(17), 3091; https://doi.org/10.3390/cancers16173091 - 5 Sep 2024
Viewed by 977
Abstract
Single-port robotic-assisted thoracic surgery (SP-RATS) lobectomy using the da Vinci Xi system has been performed by several pioneers. However, due to the severe collisions and the steep learning curve, this approach is not yet widely used. This study aimed to evaluate the feasibility [...] Read more.
Single-port robotic-assisted thoracic surgery (SP-RATS) lobectomy using the da Vinci Xi system has been performed by several pioneers. However, due to the severe collisions and the steep learning curve, this approach is not yet widely used. This study aimed to evaluate the feasibility of SP-RATS lobectomy for large-sized non-small cell lung cancer (NSCLC). As we believe that for large-sized tumors it is reasonable to make a slightly larger incision, we performed SP-RATS lobectomy for large-sized NSCLC (greater than 5 cm) through a single incision (6–8 cm). Eleven patients underwent SP-RATS lobectomy using the da Vinci Xi system at our institution from April 2022 to May 2024. The median tumor size on computed tomography and on pathology was 6.6 cm [interquartile range (IQR), 6.1–7.5 cm] and 6 cm [IQR, 5.1–7.1], respectively. The median total operative time was 198 min [IQR, 159–260 min], and the median postoperative length of stay was 4 days [IQR, 4–10 days], with no major postoperative complications (≥grade III on the Clavien–Dindo classification). Our approach may combine the benefits of single-port surgery with those of robotic surgery and is safe, feasible, and may promote better outcomes in patients with large-sized NSCLC. Full article
(This article belongs to the Special Issue State of the Art: Cardiothoracic Tumors)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Port mapping for single-port robotic-assisted thoracic surgery for large-sized tumors. (<b>A</b>) The anatomy for an incision site. The red line indicates an incision site. The blue dotted line indicates the posterior axillary line. A 6–8 cm utility incision is made at the 7th–8th ICS. (<b>B</b>) Layout of robotic arm positions. During the procedure, an 8 mm port is used on both arms (arm 1 and arm 3) to reduce collisions (<a href="#cancers-16-03091-f002" class="html-fig">Figure 2</a>A). When using the robotic stapler, the 8 mm port on either arm 1 or arm 3, where the stapler will be inserted, is replaced with a 12 mm port (<a href="#cancers-16-03091-f002" class="html-fig">Figure 2</a>B). After stapling, we replaced the 12 mm port with an 8 mm port to reduce any collisions between instruments.</p>
Full article ">Figure 2
<p>Port layout for stapling. (<b>A</b>) Usually, two 8 mm ports are placed at the left and right edges of the incision. (<b>B</b>) During stapling, an 8 mm port at the position where the stapler will be inserted is replaced with a 12 mm port.</p>
Full article ">
20 pages, 4232 KiB  
Article
Molecular Insights into the Anticancer Activity of Withaferin-A: The Inhibition of Survivin Signaling
by Renu Wadhwa, Jia Wang, Seyad Shefrin, Huayue Zhang, Durai Sundar and Sunil C. Kaul
Cancers 2024, 16(17), 3090; https://doi.org/10.3390/cancers16173090 - 5 Sep 2024
Cited by 1 | Viewed by 1536
Abstract
Survivin, a member of the IAP family, functions as a homodimer and inhibits caspases, the key enzymes involved in apoptosis. Several Survivin inhibitors, including YM-155, Debio1143, EM1421, LQZ-7I, and TL32711, have emerged as potential anticancer drugs awaiting validation in clinical trials. Due to [...] Read more.
Survivin, a member of the IAP family, functions as a homodimer and inhibits caspases, the key enzymes involved in apoptosis. Several Survivin inhibitors, including YM-155, Debio1143, EM1421, LQZ-7I, and TL32711, have emerged as potential anticancer drugs awaiting validation in clinical trials. Due to the high cost and adverse side effects of synthetic drugs, natural compounds with similar activity have also been in demand. In this study, we conducted molecular docking assays to evaluate the ability of Wi-A and Wi-N to block Survivin dimerization. We found that Wi-A, but not Wi-N, can bind to and prevent the homodimerization of Survivin, similar to YM-155. Therefore, we prepared a Wi-A-rich extract from Ashwagandha leaves (Wi-AREAL). Experimental analyses of human cervical carcinoma cells (HeLa and ME-180) treated with Wi-AREAL (0.05–0.1%) included assessments of viability, apoptosis, cell cycle, migration, invasion, and the expression levels (mRNA and protein) of molecular markers associated with these phenotypes. We found that Wi-AREAL led to growth arrest mediated by the upregulation of p21WAF1 and the downregulation of several proteins (CDK1, Cyclin B, pRb) involved in cell cycle progression. Furthermore, Wi-AREAL treatment activated apoptosis signaling, as evidenced by reduced PARP-1 and Bcl-2 levels, increased procaspase-3, and elevated Cytochrome C. Additionally, treating cells with a nontoxic low concentration (0.01%) of Wi-AREAL inhibited migration and invasion, as well as EMT (epithelial–mesenchymal transition) signaling. By combining computational and experimental approaches, we demonstrate the potential of Wi-A and Wi-AREAL as natural inhibitors of Survivin, which may be helpful in cancer treatment. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

Figure 1
<p>Computational analysis reveals Survivin inhibition by Withaferin-A and YM-155. (<b>A</b>) Visualization of the interaction between homodimers of Survivin involving amino acid residues Gly99, Leu98, Leu96, and Glu94. (<b>B</b>) Visualization of the interaction between YM-155 and the Survivin homodimer-forming region; the interaction of YM-155 with Thr5 and Leu96 is seen. (<b>C</b>) Visualization of the interaction between Wi-A and the Survivin homodimer-forming region; the interaction of Wi-A with Thr5 is seen. (<b>D</b>) Root means square deviation plot of the simulations for YM-155 and Wi-A interactions with Survivin. (<b>E</b>,<b>F</b>) Interaction fraction diagrams of Wi-A (<b>E</b>) and YM-155 (<b>F</b>) over the simulation were obtained using Schrodinger simulation analysis. (<b>G</b>,<b>H</b>) Hydrogen bond occupancy plot of Wi-A (<b>G</b>) and YM-155 (<b>H</b>) with Survivin obtained using VMD analysis.</p>
Full article ">Figure 2
<p>Effect of Wi-AREAL extract on the viability of cervical cancer cells. A dose-dependent decrease in viability (<b>A</b>) and increase in apoptotic cells (<b>B</b>) was observed in the short term (24–48 h treatment). (<b>C</b>) Long-term effects (10–15 days) showed a dose-dependent colony number and size reduction. Quantitation from three independent experiments is shown below (mean ± SD, n = 3), * <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 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 3
<p>The effect of YM-155 and Wi-AREAL on Survivin protein and mRNA levels in HeLa cells. Dose-dependent effect of YM-155 on cell viability (<b>A</b>). Cells treated with YM-155 and Wi-AREAL showed a decrease in Survivin protein (<b>B</b>–<b>D</b>) as determined by Western blotting (<b>B</b>,<b>C</b>) and immunostaining (<b>D</b>). Quantitation from three independent experiments is shown below (mean ± SD, n = 3), * <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 (Student’s <span class="html-italic">t</span>-test). (<b>E</b>) Cells treated with YM-155 and Wi-AREAL showed a decrease in Survivin mRNA (mean ± SD, n = 3), *** <span class="html-italic">p</span> &lt; 0.001 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 4
<p>Cell cycle analysis of control and Wi-AREAL-treated HeLa and ME-180 cells. The treated cells exhibited G2-M arrest (<b>A</b>,<b>B</b>), increased p21WAF1, and decreased CDK1, cyclinB1, and pRb (<b>C</b>,<b>D</b>) levels in both cell lines. Quantitation from three independent experiments is shown on the right (mean ± SD, n = 3), * <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 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 5
<p>Flow cytometry analyses of control and Wi-AREAL-treated cells. The treated cells showed a dose-dependent increase in apoptotic cells (<b>A</b>,<b>B</b>) and Cytochrome C (<b>C</b>,<b>D</b>) and a decrease in PARP-1, Bcl2, and procaspase (<b>C</b>,<b>D</b>) in both cell lines. Quantitation from three independent experiments is shown on the right (mean ± SD, n = 3), * <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 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p>Effect of a low dose of Wi-AREAL on cell migration and invasion characteristics. ME-180 cells treated with Wi-AREAL showed a decrease in cell migration (<b>A</b>) and invasion (<b>B</b>) comparable to that in cells treated with either Wi-A or YM-155. Quantitation from three independent experiments is shown on the right (mean ± SD, n = 3), *** <span class="html-italic">p</span> &lt; 0.001 (Student’s <span class="html-italic">t</span>-test). The treated cells showed decreased hnRNP-K, mortalin, CARF proteins (<b>C</b>), and mRNA (<b>D</b>) (mean ± SD, n = 3), * <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 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 7
<p>Effect of a low dose of Wi-AREAL on EMT (epithelial–mesenchymal transition). The treated cells showed reduced N-cadherin, Vimentin, Wnt-1, β-catenin, and TCF-4 at the protein (<b>A</b>,<b>B</b>) and mRNA (<b>C</b>) levels. (mean ± SD, n = 3), * <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 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 8
<p>The schematic model illustrates the impact of Wi-REAL on cancer cell characteristics through the inhibition of Survivin signaling.</p>
Full article ">
18 pages, 763 KiB  
Review
Future AI Will Most Likely Predict Antibody-Drug Conjugate Response in Oncology: A Review and Expert Opinion
by Navid Sobhani, Alberto D’Angelo, Matteo Pittacolo, Giuseppina Mondani and Daniele Generali
Cancers 2024, 16(17), 3089; https://doi.org/10.3390/cancers16173089 - 5 Sep 2024
Cited by 2 | Viewed by 2233
Abstract
The medical research field has been tremendously galvanized to improve the prediction of therapy efficacy by the revolution in artificial intelligence (AI). An earnest desire to find better ways to predict the effectiveness of therapy with the use of AI has propelled the [...] Read more.
The medical research field has been tremendously galvanized to improve the prediction of therapy efficacy by the revolution in artificial intelligence (AI). An earnest desire to find better ways to predict the effectiveness of therapy with the use of AI has propelled the evolution of new models in which it can become more applicable in clinical settings such as breast cancer detection. However, in some instances, the U.S. Food and Drug Administration was obliged to back some previously approved inaccurate models for AI-based prognostic models because they eventually produce inaccurate prognoses for specific patients who might be at risk of heart failure. In light of instances in which the medical research community has often evolved some unrealistic expectations regarding the advances in AI and its potential use for medical purposes, implementing standard procedures for AI-based cancer models is critical. Specifically, models would have to meet some general parameters for standardization, transparency of their logistic modules, and avoidance of algorithm biases. In this review, we summarize the current knowledge about AI-based prognostic methods and describe how they may be used in the future for predicting antibody-drug conjugate efficacy in cancer patients. We also summarize the findings of recent late-phase clinical trials using these conjugates for cancer therapy. Full article
Show Figures

Figure 1

Figure 1
<p>Clinically tested ADCs. This bar graph shows the 277 ADCs that have undergone clinical trials along with their trial status (completed, active/recruiting, not yet recruiting, suspended, and unknown). Additionally, to the right of the main Total bar, the active/recruiting ADCs are broken down into additional columns to highlight their highest developmental stage (phases 1–4 [P1–P4]).</p>
Full article ">Figure 2
<p>Artificial intelligence assisted antibody-drug conjugate selection for the treatment of cancer.</p>
Full article ">
15 pages, 4099 KiB  
Article
Exposed Phosphatidylserine as a Biomarker for Clear Identification of Breast Cancer Brain Metastases in Mouse Models
by Lulu Wang, Alan H. Zhao, Chad A. Arledge, Fei Xing, Michael D. Chan, Rolf A. Brekken, Amyn A. Habib and Dawen Zhao
Cancers 2024, 16(17), 3088; https://doi.org/10.3390/cancers16173088 - 5 Sep 2024
Viewed by 1148
Abstract
Brain metastasis is the most common intracranial malignancy in adults. The prognosis is extremely poor, partly because most patients have more than one brain lesion, and the currently available therapies are nonspecific or inaccessible to those occult metastases due to an impermeable blood–tumor [...] Read more.
Brain metastasis is the most common intracranial malignancy in adults. The prognosis is extremely poor, partly because most patients have more than one brain lesion, and the currently available therapies are nonspecific or inaccessible to those occult metastases due to an impermeable blood–tumor barrier (BTB). Phosphatidylserine (PS) is externalized on the surface of viable endothelial cells (ECs) in tumor blood vessels. In this study, we have applied a PS-targeting antibody to assess brain metastases in mouse models. Fluorescence microscopic imaging revealed that extensive PS exposure was found exclusively on vascular ECs of brain metastases. The highly sensitive and specific binding of the PS antibody enables individual metastases, even micrometastases containing an intact BTB, to be clearly delineated. Furthermore, the conjugation of the PS antibody with a fluorescence dye, IRDye 800CW, or a radioisotope, 125I, allowed the clear visualization of individual brain metastases by optical imaging and autoradiography, respectively. In conclusion, we demonstrated a novel strategy for targeting brain metastases based on our finding that abundant PS exposure occurs on blood vessels of brain metastases but not on normal brain, which may be useful for the development of imaging and targeted therapeutics for brain metastases. Full article
(This article belongs to the Special Issue Brain Metastases: From Mechanisms to Treatment)
Show Figures

Figure 1

Figure 1
<p>Longitudinal MRI monitoring of the intracranial distribution of brain metastases and permeability of the BTB in the 231-Br model. (<b>a</b>) Three consecutive coronal sections of high-resolution MRI were obtained from a representative mouse brain bearing 231-Br metastases. At week 3, T<sub>2</sub>-w images revealed multiple hyperintense metastases across the brain (yellow arrowheads), only two of which were enhanced on T<sub>1</sub>-w post-contrast images (green arrows), indicating a leaky BTB. At week 4, several new lesions appeared on T<sub>2</sub>-w images (yellow arrowheads), while those lesions seen on the prior scan became larger. An increased number of the contrast-enhanced lesions were seen at this time (green arrows). (<b>b</b>) A total of 464 231-Br brain metastases were identified by MRI, including non-permeable (<span class="html-italic">n</span> = 304) and permeable (<span class="html-italic">n</span> = 160) metastases based on T<sub>1</sub>-w post-contrast images. A plot of permeability versus size indicated that larger metastases tend to be leaky. However, there was no significant difference in tumor size between the permeable and non-permeable metastases (<span class="html-italic">p</span> = 0.1).</p>
Full article ">Figure 2
<p>PS exposed exclusively in vascular endothelial cells of brain metastases. (<b>a</b>) Multiple brain lesions were detected on three consecutive coronal sections of T<sub>2</sub>-w images (arrow) in a 231-Br mouse, while a few lesions were enhanced on T<sub>1</sub>-w post-contrast images (arrow). Immediately after MRI, the mouse was injected i.v. with 1N11 (150 µg). Four hours later, the mouse was perfused, and the brain was dissected. (<b>b</b>) Corresponding H&amp;E sections depicted more microscopic lesions that were invisible by MRI. (<b>c</b>–<b>f</b>) Diffuse brain metastases were labeled on one of the H&amp;E sections (arrow; (<b>c</b>)). Immunofluorescence staining on a consecutive section showed that 1N11 (red; (<b>d</b>)) localized to every tumor lesion, even microscopic lesions identified by H&amp;E staining (arrow; bar = 1 mm; (<b>c</b>)). The binding of 1N11 to tumor vessels (CD31, blue; (<b>e</b>)) was confirmed by the magenta color in the merged images (<b>f</b>). (<b>g</b>–<b>j</b>) A region containing positive 1N11 (outlined in (<b>d</b>)) was selected and magnified (bar = 100 µm). The merged image showed that 1N11 (red, (<b>h</b>)) co-localized with almost every CD31-positive tumor vessel (blue, (<b>g</b>)) to give a magenta color (<b>i</b>). Vessels in nearby normal brain (star, (<b>i</b>)) were not stained by 1N11. The tumor regions were distinguished from normal brain by the presence of GFP in the tumor (<b>j</b>).</p>
Full article ">Figure 3
<p>Staining with 1N11 was antigen-specific. 231-Br mice were injected i.v. with either 1N11 or the control antibody, Aurexis (150 µg). Four hours later, the mice were perfused, and the brains were dissected. (<b>a</b>–<b>c</b>) A brain region containing a tumor lesion was stained with anti-CD31 for blood vessels (<b>a</b>) and co-stained for 1N11 (<b>b</b>). The merged image showed that 1N11 co-localized with CD31-positive tumor vessels. Vessels in nearby normal brain (containing no GFP) were not stained by 1N11 (<b>c</b>). (<b>d</b>–<b>f</b>) In contrast, the control antibody, Aurexis, showed no staining of blood vessels of brain metastases (bar = 100 µm). (<b>g</b>) For the group of 231-Br mice, the percentage of PS-positive vessels in brain metastases was 93 ± 5%, while it was only 2 ± 2% for the control antibody, Aurexis. * <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>TNF-α-induced PS externalization in vascular endothelial cells and the overexpression of TNF-α was detected in brain metastases. (<b>a</b>) A group of 231-Br mice receiving i.v. 1N11 followed by i.v. pimonidazole was used to study tumor hypoxia. A representative region containing positive 1N11 staining (red) was co-stained for pimonidazole (blue). There was essentially no positive pimonidazole staining in the brain metastasis (GFP). (<b>b</b>) HUVECs were treated with/without TNF-α (20 ng/mL) for 24 h before fixation, then immunocytostained with 1N11 (red), cytoskeleton (phalloidin, green), and nuclear (DAPI, blue). The merged image showed numerous cell surface-exposed PS in the TNF-α-treated cells, while there was no PS exposure in the control cells. (<b>c</b>) Anti-TNF-α staining revealed marked expression of TNF-α in the 231-Br brain metastasis but not in normal brains.</p>
Full article ">Figure 5
<p>PS-targeted NIR imaging of brain metastases. NIR dye, IRDye-800CW-labeled 1N11 F(ab’)<sub>2</sub>, was injected into the mice bearing 231-Br (4 wks post-tumor cell injection) or 4T1 (2 wks post-tumor cell injection) brain metastases. (<b>a</b>) Twenty-four hours later, ex vivo NIR imaging of the whole brain detected distinct 800CW signals from multiple regions of a 231-Br brain, while a clear contrast of a single brain lesion was seen in a 4T1 brain. (<b>b</b>) A series of unstained coronal brain cryosections (10 µm, bottom row) was obtained after the brain in (<b>a</b>) was imaged. The NIR imaging revealed a clear tumor contrast on the series of brain sections, which correlated well with H&amp;E staining (middle row) of tumors and T1-contrast-enhanced MR images (top row). (<b>c</b>) Quantification of the light intensity in the tumor versus the contralateral normal brain obtained a ratio of 5.7 ± 0.8 with 800CW-1N11, which was significantly higher than that of the control antibody, 800CW-Aurexis (1.5 ± 0.4; * <span class="html-italic">p</span> &lt; 0.05). (<b>d</b>) Tumor vascular endothelial cells were immunostained with anti-CD31 (green) in the cryosections adjacent to those used in (<b>b</b>). 800CW-1N11 signals (red) from the same field were detected with an NIR filter set (<b>e</b>). (<b>f</b>) The merged image revealed extensive 800CW-1N11 bound to vascular endothelial cells, while some were detected in the extravascular space (DAPI, blue).</p>
Full article ">Figure 6
<p>Autoradiography imaging of I-125-labeled 1N11 in targeting brain metastases. (<b>a</b>) T<sub>2</sub>-w MR images of 4 consecutive 1 mm thick coronal slices covering 4 mm brain tissues post-bregma were acquired from a normal control mouse brain (<b>top</b>) and a 231-Br brain (<b>bottom</b>), respectively. Multiple tumors in the 231-Br brain were identified (arrow). An image stack across the 4 slices was created for each animal. For the 231-Br brain, the tumor lesions (red) from each slice were projected on the stacked image to correlate with the autoradiography study. (<b>b</b>) After the MRI, each mouse was given i.v. 60 µCi <sup>125</sup>I-1N11. Forty-eight hours later, the mice were perfused, and mouse brains were dissected. Using a mouse brain matrix, 4 mm thick brain tissues post-bregma, correlating with the MRI, were cut from each mouse brain and laid with the cutting face on the autoradiograph film. After 12 h of incubation, autoradiograph images showed multiple hot spots on the tumor brain, while a clean background signal was observed on the normal brain. There was a general spatial correlation between tumor lesions on the MRI and hot spots on the autoradiograph. (<b>c</b>) Significantly higher uptake of <sup>125</sup>I-1N11 was observed in individual brain metastases as compared to normal brain tissues (a ratio of 3.6 ± 0.8; <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
10 pages, 1172 KiB  
Review
Clinical and Histologic Variants of CD8+ Cutaneous T-Cell Lymphomas
by Madisen A. Swallow, Goran Micevic, Amanda Zhou, Kacie R. Carlson, Francine M. Foss and Michael Girardi
Cancers 2024, 16(17), 3087; https://doi.org/10.3390/cancers16173087 - 5 Sep 2024
Viewed by 1598
Abstract
Although the vast majority of CTCL subtypes are of the CD4+ T-helper cell differentiation phenotype, there is a spectrum of CD8+ variants that manifest wide-ranging clinical, histologic, and phenotypic features that inform the classification of the disease. CD8, like CD4, and cytotoxic molecules [...] Read more.
Although the vast majority of CTCL subtypes are of the CD4+ T-helper cell differentiation phenotype, there is a spectrum of CD8+ variants that manifest wide-ranging clinical, histologic, and phenotypic features that inform the classification of the disease. CD8, like CD4, and cytotoxic molecules (including TIA and granzyme) are readily detectable via IHC staining of tissue and, when expressed on the phenotypically abnormal T-cell population, can help distinguish specific CTCL subtypes. Nonetheless, given that the histopathologic differential for CD8+ lymphoproliferative disorders and lymphomas may range from very indolent lymphomatoid papulosis (LyP) to aggressive entities like CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma (AECTCL), CD8 and/or cytotoxic molecule expression alone is insufficient for diagnosis and is not in itself an indicator of prognosis. We present a review of CTCL subtypes that can demonstrate CD8 positivity: CD8+ mycosis fungoides (MF), LyP type D, subcutaneous panniculitis-like T-cell lymphoma (SPTCL), primary cutaneous gamma/delta T-cell lymphoma (PCGDTL), CD8+ AECTCL, and acral CD8+ T-cell lymphoproliferative disorder (acral CD8+ TCLPD). These diseases may have different clinical manifestations and distinctive treatment algorithms. Due to the rare nature of these diseases, it is imperative to integrate clinical, histologic, and immunohistochemical findings to determine an accurate diagnosis and an appropriate treatment plan. Full article
(This article belongs to the Special Issue Cutaneous Lymphoma)
Show Figures

Figure 1

Figure 1
<p>The cutaneous T-cell lymphoma entities divided into typically CD4-positive and CD8-positive lymphomas.</p>
Full article ">Figure 2
<p>When constructing a diagnostic decision tree for CD8+ T-cell lymphoma, it is crucial to consider the key distinguishing features, particularly immunohistochemistry (IHC) differences and clinical decision points. The IHC differences, shown in the diamonds, include cytotoxic markers such as granzyme and TIA, as well as CD30 positivity. Clinical decision points, shown in rectangles, are also essential components of the decision-making process. Together, these elements help in accurately differentiating and diagnosing CD8+ T-cell lymphoma.</p>
Full article ">
15 pages, 638 KiB  
Review
The Surgical Renaissance: Advancements in Video-Assisted Thoracoscopic Surgery and Robotic-Assisted Thoracic Surgery and Their Impact on Patient Outcomes
by Jennifer M. Pan, Ammara A. Watkins, Cameron T. Stock, Susan D. Moffatt-Bruce and Elliot L. Servais
Cancers 2024, 16(17), 3086; https://doi.org/10.3390/cancers16173086 - 5 Sep 2024
Viewed by 1159
Abstract
Minimally invasive thoracic surgery has advanced the treatment of lung cancer since its introduction in the 1990s. Video-assisted thoracoscopic surgery (VATS) and robotic-assisted thoracic surgery (RATS) offer the advantage of smaller incisions without compromising patient outcomes. These techniques have been shown to be [...] Read more.
Minimally invasive thoracic surgery has advanced the treatment of lung cancer since its introduction in the 1990s. Video-assisted thoracoscopic surgery (VATS) and robotic-assisted thoracic surgery (RATS) offer the advantage of smaller incisions without compromising patient outcomes. These techniques have been shown to be safe and effective in standard pulmonary resections (lobectomy and sub-lobar resection) and in complex pulmonary resections (sleeve resection and pneumonectomy). Furthermore, several studies show these techniques enhance patient outcomes from early recovery to improved quality of life (QoL) and excellent oncologic results. The rise of RATS has yielded further operative benefits compared to thoracoscopic surgery. The wristed instruments, neutralization of tremor, dexterity, and magnification allow for more precise and delicate dissection of tissues and vessels. This review summarizes of the advancements in minimally invasive thoracic surgery and the positive impact on patient outcomes. Full article
(This article belongs to the Special Issue Advancements in Lung Cancer Surgical Treatment and Prognosis)
Show Figures

Figure 1

Figure 1
<p>Distribution of cases and modalities over time. Modalities were thoracotomy (dark blue bars), VATS (grey bars), and RATS (blue bars). ([2022] Servais et al. Reproduced with permission from the authors [<a href="#B9-cancers-16-03086" class="html-bibr">9</a>]).</p>
Full article ">
Previous Issue
Back to TopTop