[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Next Issue
Volume 14, November-2
Previous Issue
Volume 14, October-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 14, Issue 21 (November-1 2022) – 301 articles

Cover Story (view full-size image): Complementary and alternative medicine (CAM) therapies include a wide range of procedures and products that are often used by cancer patients to directly combat cancer and protect normal cells from the toxic effects of conventional therapies. Most often, their use is self-prescribed, based on a collection of scattered information from websites and advice from relatives or friends, possibly leading to unexpected toxicities and a reduction in the efficacy of cancer therapy. Despite claims of promising and potential benefits made by prescribers, many CAMs lack clear scientific evidence of their safety and efficacy. Given the widespread use of CAM, in this review we focused on the most important known data on the interaction risk between anticancer drugs and CAMs in accordance with the meaning of integrative medicine, providing a practical guide for caregivers and patients. 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:
28 pages, 2331 KiB  
Review
Ewing Sarcoma Meets Epigenetics, Immunology and Nanomedicine: Moving Forward into Novel Therapeutic Strategies
by Sara Sánchez-Molina, Elisabet Figuerola-Bou, Víctor Sánchez-Margalet, Luis de la Cruz-Merino, Jaume Mora, Enrique de Álava Casado, Daniel José García-Domínguez and Lourdes Hontecillas-Prieto
Cancers 2022, 14(21), 5473; https://doi.org/10.3390/cancers14215473 - 7 Nov 2022
Cited by 5 | Viewed by 4846
Abstract
Ewing Sarcoma (EWS) is an aggressive bone and soft tissue tumor that mainly affects children, adolescents, and young adults. The standard therapy, including chemotherapy, surgery, and radiotherapy, has substantially improved the survival of EWS patients with localized disease. Unfortunately, this multimodal treatment remains [...] Read more.
Ewing Sarcoma (EWS) is an aggressive bone and soft tissue tumor that mainly affects children, adolescents, and young adults. The standard therapy, including chemotherapy, surgery, and radiotherapy, has substantially improved the survival of EWS patients with localized disease. Unfortunately, this multimodal treatment remains elusive in clinics for those patients with recurrent or metastatic disease who have an unfavorable prognosis. Consistently, there is an urgent need to find new strategies for patients that fail to respond to standard therapies. In this regard, in the last decade, treatments targeting epigenetic dependencies in tumor cells and the immune system have emerged into the clinical scenario. Additionally, recent advances in nanomedicine provide novel delivery drug systems, which may address challenges such as side effects and toxicity. Therefore, therapeutic strategies stemming from epigenetics, immunology, and nanomedicine yield promising alternatives for treating these patients. In this review, we highlight the most relevant EWS preclinical and clinical studies in epigenetics, immunotherapy, and nanotherapy conducted in the last five years. Full article
Show Figures

Figure 1

Figure 1
<p>Immune therapies currently explored in EWS. (<b>A</b>) Immune checkpoint inhibitors block the interaction of immune checkpoint molecules (e.g., PD-1 or CTLA-4) with its inhibitory ligands to stimulate the immune response. (<b>B</b>) Adoptive cell therapy involves the infusion of modified autologous T-cells or allogenic NK cells. T-cells can be genetically modified to express a chimeric antigen receptor (CAR) specific of a tumor-associated antigen (e.g., EGFR) that can be recognized by major histocompatibility complex (MHC)-independent mechanisms. In contrast, T-cells isolated from tumors can be stimulated with an oncolytic peptide (e.g., LTX-315) and reinfusioned back to mediate an antitumoral MHC-dependent response. Transfer of NK cells from healthy donors is based on the innate ability of NK cells to kill tumor cells through various mechanisms such as granzyme B release. (<b>C</b>) Antibody-based therapies involve the use of specific antibodies targeting tumor-associated antigens (e.g., GD2). (<b>D</b>) Cancer vaccines stimulate the immune system response of the host through various mechanisms. The VIGIL vaccine in EWS is based on the tumor cells engineered to express GM-CSF and a bifunctional shRNA that prevents immunosuppression by TGFβ1-2 release. Reinfusion of these tumor cells, thus, promotes antigen-presentation and the adaptive immune response.</p>
Full article ">Figure 2
<p>Summarizing the Nanoparticles and nanomaterials used in EWS studies.</p>
Full article ">
22 pages, 735 KiB  
Review
Histone Deacetylase Functions in Gastric Cancer: Therapeutic Target?
by Amandine Badie, Christian Gaiddon and Georg Mellitzer
Cancers 2022, 14(21), 5472; https://doi.org/10.3390/cancers14215472 - 7 Nov 2022
Cited by 10 | Viewed by 2439
Abstract
Gastric cancer (GC) is one of the most aggressive cancers. Therapeutic treatments are based on surgery combined with chemotherapy using a combination of platinum-based agents. However, at metastatic stages of the disease, survival is extremely low due to late diagnosis and resistance mechanisms [...] Read more.
Gastric cancer (GC) is one of the most aggressive cancers. Therapeutic treatments are based on surgery combined with chemotherapy using a combination of platinum-based agents. However, at metastatic stages of the disease, survival is extremely low due to late diagnosis and resistance mechanisms to chemotherapies. The development of new classifications has not yet identified new prognostic markers for clinical use. The studies of epigenetic processes highlighted the implication of histone acetylation status, regulated by histone acetyltransferases (HATs) and by histone deacetylases (HDACs), in cancer development. In this way, inhibitors of HDACs (HDACis) have been developed and some of them have already been clinically approved to treat T-cell lymphoma and multiple myeloma. In this review, we summarize the regulations and functions of eighteen HDACs in GC, describing their known targets, involved cellular processes, associated clinicopathological features, and impact on survival of patients. Additionally, we resume the in vitro, pre-clinical, and clinical trials of four HDACis approved by Food and Drug Administration (FDA) in cancers in the context of GC. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Role of HDAC1 in different signaling pathways involved in GC progression. HDAC1 promotes GC cell proliferation via lncRNA HRCEG repression, lncRNAs BC01600 and AF116637 upregulation, or MORC2/p21 pathway. HDAC1 favors metastatic abilities of GC cells through miR-34a repression inducing CD44 expression. HDAC1 promotes HIF-1α activity leading to glycolysis in GC cells. HDAC1 inhibits CRADD transcription and consequently caspase-2-dependent apoptosis. HDAC1 is a resistance factor to chemotherapies in GC such as anthracycline, via repression of CITED2, and doxorubicin, inhibiting its fixation on DNA. Figure has been generated using Biorender (<a href="http://Biorender.com" target="_blank">Biorender.com</a>).</p>
Full article ">
13 pages, 775 KiB  
Review
Aptamers, a New Therapeutic Opportunity for the Treatment of Multiple Myeloma
by Ane Amundarain, Fernando Pastor, Felipe Prósper and Xabier Agirre
Cancers 2022, 14(21), 5471; https://doi.org/10.3390/cancers14215471 - 7 Nov 2022
Cited by 2 | Viewed by 2631
Abstract
Multiple Myeloma (MM) remains an incurable disease due to high relapse rates and fast development of drug resistances. The introduction of monoclonal antibodies (mAb) has caused a paradigm shift in MM treatment, paving the way for targeted approaches with increased efficacy and reduced [...] Read more.
Multiple Myeloma (MM) remains an incurable disease due to high relapse rates and fast development of drug resistances. The introduction of monoclonal antibodies (mAb) has caused a paradigm shift in MM treatment, paving the way for targeted approaches with increased efficacy and reduced toxicities. Nevertheless, antibody-based therapies face several difficulties such as high immunogenicity, high production costs and limited conjugation capacity, which we believe could be overcome by the introduction of nucleic acid aptamers. Similar to antibodies, aptamers can bind to their targets with great affinity and specificity. However, their chemical nature reduces their immunogenicity and production costs, while it enables their conjugation to a wide variety of cargoes for their use as delivery agents. In this review, we summarize several aptamers that have been tested against MM specific targets with promising results, establishing the rationale for the further development of aptamer-based strategies against MM. In this direction, we believe that the study of novel plasma cell surface markers, the development of intracellular aptamers and further research on aptamers as building blocks for complex nanomedicines will lead to the generation of next-generation targeted approaches that will undoubtedly contribute to improve the management and life quality of MM patients. Full article
(This article belongs to the Special Issue Aptamers and Cancer)
Show Figures

Figure 1

Figure 1
<p>Overview of purified protein-based SELEX. The initial RNA/ssDNA aptamer pool is generated from a commercial ssDNA oligonucleotide library consisting of a central random core sequence flanked by fixed 5′ and 3′ primer binding regions to allow library amplification. To obtain RNA/ssDNA aptamers for the first selection cycle, the library is amplified by PCR and subjected to in vitro transcription or strand separation, respectively. A purified protein-based SELEX approach has four key steps: (<b>1</b>) the aptamer library is initially incubated with the purified target protein and (<b>2</b>) bound sequences are isolated from the unbound ones via different partitioning strategies. The bound sequences are then (<b>3</b>) recovered and (<b>4</b>) re-amplified to obtain the new aptamer library for the next selection cycle. For RNA aptamers, recovered RNA aptamers must be retrotranscribed for subsequent PCR amplification, and the amplification product is then converted into RNA again via in vitro transcription. For ssDNA, recovered aptamers are directly amplified by PCR and ssDNA aptamers are obtained by strand separation. After n selection cycles, the aptamer pool is sequenced to identify the enriched aptamer sequences. Image made in ©BioRender.</p>
Full article ">Figure 2
<p>Current aptamers for precision medicine in MM. (<b>1</b>) Aptamer against AXII, (<b>2</b>) BCMA targeted aptamer, (<b>3</b>) aptamer against C-MET, (<b>4</b>) conjugated CD38-doxorubicin aptamer and (<b>5</b>) NOX-A12 RNA aptamer for CXCL12. All the aptamers except the NOX-A12 spiegelmer are directed towards receptors expressed in the MM plasma cell membrane. Receptor–ligand interactions are depicted with black arrows, while the mechanism of action of aptamers is depicted with red arrows for antagonist aptamers, and blue arrows for cargo delivery aptamers. BM: bone marrow; Doxo: doxorubicin; OBL: osteoblast; sAXII: soluble annexin A2. Image made in ©BioRender.</p>
Full article ">
12 pages, 947 KiB  
Systematic Review
The Effects of Patient-Reported Outcome Screening on the Survival of People with Cancer: A Systematic Review and Meta-Analysis
by Caterina Caminiti, Giuseppe Maglietta, Francesca Diodati, Matteo Puntoni, Barbara Marcomini, Silvia Lazzarelli, Carmine Pinto and Francesco Perrone
Cancers 2022, 14(21), 5470; https://doi.org/10.3390/cancers14215470 - 7 Nov 2022
Cited by 22 | Viewed by 2327
Abstract
This study examined the effects of the routine assessment of patient-reported outcomes (PROs) on the overall survival of adult patients with cancer. We included clinical trials and observational studies with a control group that compared PRO monitoring interventions in cancer clinical practice to [...] Read more.
This study examined the effects of the routine assessment of patient-reported outcomes (PROs) on the overall survival of adult patients with cancer. We included clinical trials and observational studies with a control group that compared PRO monitoring interventions in cancer clinical practice to usual care. The Cochrane risk-of-bias tools were used. In total, six studies were included in the systematic review: two randomized trials, one population-based retrospectively matched cohort study, two pre–post with historical control studies and one non-randomized controlled trial. Half were multicenter, two were conducted in Europe, three were conducted in the USA and was conducted in Canada. Two studies considered any type of cancer, two were restricted to lung cancer and two were restricted to advanced forms of cancer. PRO screening was electronic in four of the six studies. The meta-analysis included all six studies (intervention = 130.094; control = 129.903). The pooled mortality outcome at 1 year was RR = 0.77 (95%CI 0.76–0.78) as determined by the common effect model and RR = 0.82 (95%CI 0.60–1.12; p = 0.16) as determined by the random-effects model. Heterogeneity was statistically significant (I2 = 73%; p < 0.01). The overall risk of bias was rated as moderate in five studies and serious in one study. This meta-analysis seemed to indicate the survival benefits of PRO screening. As routine PRO monitoring is often challenging, more robust evidence regarding the effects of PROs on mortality would support systematic applications. Full article
(This article belongs to the Special Issue Quality of Life and Side Effects Management in Cancer Treatment)
Show Figures

Figure 1

Figure 1
<p>A PRISMA 2020 flow diagram of the process of identifying studies (both included and excluded).</p>
Full article ">Figure 2
<p>A forest plot of the survival pooled effect and a summary of the risk of bias (traffic light plot and summary bar plot) [<a href="#B32-cancers-14-05470" class="html-bibr">32</a>,<a href="#B33-cancers-14-05470" class="html-bibr">33</a>,<a href="#B34-cancers-14-05470" class="html-bibr">34</a>,<a href="#B35-cancers-14-05470" class="html-bibr">35</a>,<a href="#B36-cancers-14-05470" class="html-bibr">36</a>,<a href="#B37-cancers-14-05470" class="html-bibr">37</a>].</p>
Full article ">
9 pages, 1057 KiB  
Article
Reinduction of Hedgehog Inhibitors after Checkpoint Inhibition in Advanced Basal Cell Carcinoma: A Series of 12 Patients
by Viola K. DeTemple, Jessica C. Hassel, Michael M. Sachse, Imke Grimmelmann, Ulrike Leiter, Christoffer Gebhardt, Julia Eckardt, Claudia Pföhler, Yenny Angela, Hanna Hübbe and Ralf Gutzmer
Cancers 2022, 14(21), 5469; https://doi.org/10.3390/cancers14215469 - 7 Nov 2022
Cited by 3 | Viewed by 1997
Abstract
For patients with advanced basal cell carcinoma (aBCC) first-line treatment with hedgehog inhibitors (HHIs) and second-line treatment with PD1 inhibitors (PD1i) is available, offering combination and sequencing options. Here, we focus on the efficacy and safety of HHI reinduction after PD1i failure. Retrospective [...] Read more.
For patients with advanced basal cell carcinoma (aBCC) first-line treatment with hedgehog inhibitors (HHIs) and second-line treatment with PD1 inhibitors (PD1i) is available, offering combination and sequencing options. Here, we focus on the efficacy and safety of HHI reinduction after PD1i failure. Retrospective data analysis was performed with 12 patients with aBCC (locally advanced (n = 8)/metastatic (n = 4)). These patients (male:female 6:6, median age 68 years) initially received HHIs, leading to complete/partial response (66%) or stable disease (33%). Median treatment duration was 20.8 (2–64.5) months until discontinuation due to progression (n = 8), adverse events (n = 3), or patient request (n = 1). Subsequent PD1 inhibition (pembrolizumab 42%, cemiplimab 58%) yielded a partial response (8%), stable disease (33%), or progression (59%). Median treatment duration was 4.1 (0.8–16.3) months until discontinuation due to progression (n = 9), adverse events (n = 1), patient request (n = 1), or missing drug approval (n = 1). HHI reinduction resulted in complete/partial response (33%), stable disease (50%), or progression (17%). Median treatment duration was 3.6 (1–29) months. Response duration in the four responding patients was 2–29+ months. Thus, a subgroup of patients with aBCC responded to reinduction of HHI following PD1i failure. Therefore, this sequential treatment represents a feasible treatment option. Full article
(This article belongs to the Special Issue Skin Cancer: Epidemiology, Prevention and Quality of Life)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Course of treatment across all 12 patients depicted as swimmers plot.</p>
Full article ">Figure 2
<p>Exemplary photodocumentation of one patient during the course of treatment. Photodocumentation for patient 7 with locally advanced basal cell carcinoma along the treatment sequence HHI–PD1i–HHI. (<b>a</b>) Starting point before systemic treatment with locally advanced basal cell carcinoma on the left flank. (<b>b</b>) Initial partial response to vismodegib within 16 months of treatment, followed by (<b>c</b>) progressive disease under HHI after another 7 months. (<b>d</b>) Further progressive disease under cemiplimab within 6 months after only short term partial response. (<b>e</b>) Tumor stabilization was reached within 4 months under HHI reinduction with sonidegib.</p>
Full article ">
26 pages, 2070 KiB  
Review
Wnt/β-Catenin Signaling as a Driver of Stemness and Metabolic Reprogramming in Hepatocellular Carcinoma
by Rainbow Wing Hei Leung and Terence Kin Wah Lee
Cancers 2022, 14(21), 5468; https://doi.org/10.3390/cancers14215468 - 7 Nov 2022
Cited by 21 | Viewed by 5019
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer death worldwide due to its high rates of tumor recurrence and metastasis. Aberrant Wnt/β-catenin signaling has been shown to play a significant role in HCC development, progression and clinical impact on tumor behavior. Accumulating [...] Read more.
Hepatocellular carcinoma (HCC) is a major cause of cancer death worldwide due to its high rates of tumor recurrence and metastasis. Aberrant Wnt/β-catenin signaling has been shown to play a significant role in HCC development, progression and clinical impact on tumor behavior. Accumulating evidence has revealed the critical involvement of Wnt/β-catenin signaling in driving cancer stemness and metabolic reprogramming, which are regarded as emerging cancer hallmarks. In this review, we summarize the regulatory mechanism of Wnt/β-catenin signaling and its role in HCC. Furthermore, we provide an update on the regulatory roles of Wnt/β-catenin signaling in metabolic reprogramming, cancer stemness and drug resistance in HCC. We also provide an update on preclinical and clinical studies targeting Wnt/β-catenin signaling alone or in combination with current therapies for effective cancer therapy. This review provides insights into the current opportunities and challenges of targeting this signaling pathway in HCC. Full article
(This article belongs to the Special Issue Wnt Pathway Targets in Cancer)
Show Figures

Figure 1

Figure 1
<p>Regulation of Wnt/β-catenin signaling in HCC. Wnt/β-catenin signaling in HCC is regulated by (<b>A</b>) DNA methylation, (<b>B</b>) histone modification and (<b>C</b>) non-coding RNAs.</p>
Full article ">Figure 2
<p>The role of Wnt/β-catenin signaling on cancer metabolism. Wnt/β-catenin plays crucial role in (<b>A</b>) aerobic glycolysis, (<b>B</b>) glutaminolysis and (<b>C</b>) fatty acid metabolism of HCC cells.</p>
Full article ">Figure 3
<p>The role of Wnt/β-catenin signaling on cancer stemness and drug resistance. Intrinsic and extrinsic regulation of Wnt/β-catenin signaling lead to tumor growth, cancer stemness, EMT and drug resistance of HCC cells.</p>
Full article ">
18 pages, 804 KiB  
Review
Post-Neoadjuvant Treatment Strategies for Patients with Early Breast Cancer
by Elisa Agostinetto, Flavia Jacobs, Véronique Debien, Alex De Caluwé, Catalin-Florin Pop, Xavier Catteau, Philippe Aftimos, Evandro de Azambuja and Laurence Buisseret
Cancers 2022, 14(21), 5467; https://doi.org/10.3390/cancers14215467 - 7 Nov 2022
Cited by 8 | Viewed by 6074
Abstract
Pre-surgical treatments in patients with early breast cancer allows a direct estimation of treatment efficacy, by comparing the tumor and the treatment. Patients who achieve a pathological complete response at surgery have a better prognosis, with lower risk of disease recurrence and death. [...] Read more.
Pre-surgical treatments in patients with early breast cancer allows a direct estimation of treatment efficacy, by comparing the tumor and the treatment. Patients who achieve a pathological complete response at surgery have a better prognosis, with lower risk of disease recurrence and death. Hence, clinical research efforts have been focusing on high-risk patients with residual disease at surgery, who may be “salvaged” through additional treatments administered in the post-neoadjuvant setting. In the present review, we aim to illustrate the development and advantages of the post-neoadjuvant setting, and to discuss the available strategies for patients with early breast cancer, either approved or under investigation. This review was written after literature search on main scientific databases (e.g., PubMed) and conference proceedings from major oncology conferences up to 1 August 2022. T-DM1 and capecitabine are currently approved as post-neoadjuvant treatments for patients with HER2-positive and triple-negative breast cancer, respectively, with residual disease at surgery. More recently, other treatment strategies have been approved for patients with high-risk early breast cancer, including the immune checkpoint inhibitor pembrolizumab, the PARP inhibitor olaparib and the CDK 4/6 inhibitor abemaciclib. Novel agents and treatment combinations are currently under investigation as promising post-neoadjuvant treatment strategies. Full article
(This article belongs to the Special Issue Post-neoadjuvant Strategies in Breast Cancer)
Show Figures

Figure 1

Figure 1
<p>Simplified representation of post-neoadjuvant treatment strategy for breast cancer patients. Patients eligible for preoperative treatment receive neoadjuvant chemotherapy. If at the time of surgery the tumor is no longer detectable in the specimen (i.e., a pathological complete response (pCR) is achieved), these patients could be potentially candidate to de-escalation treatment strategies, as pCR is associated with lower risk of disease recurrence. The potential de-escalation strategy can apply to surgery, radiotherapy, or systemic adjuvant therapy. Of note, the management of a patient with a pCR is multifactorial, and requires multidisciplinary discussion. Indeed, caution should be paid to avoid the removal of too many treatment components. In case of invasive residual disease, patients may be considered for additional post-neoadjuvant treatments, for instance with chemotherapy (i.e., capecitabine in triple-negative breast cancer) or antibody drug conjugates (i.e., T-DM1 in HER2-positive breast cancer). The evaluation of residual disease can be done using various biomarkers for risk assessment (e.g., Ki67, TILs, RCB, gene expression, and genetic alterations). Residual disease can be classified according to the Residual Cancer Burden index, that combines pathologic measurements of primary tumor (size and cellularity) and nodal metastases (number and size), and classifies the specimen in one of four classes (RCB 0, i.e., pCR, RCB I, RCB II, RCB III). A higher RCB index (i.e., RCB III) indicates a larger amount of residual disease, and it is associated with a higher risk of recurrence.Abbreviations: CDKi 4/6: cyclin-dependent kinase inhibitor; ctDNA: circulating tumor DNA; HER2, human epidermal growth factor receptor 2; HR+, hormone receptor-positive breast cancer; LVI: lymphovascular invasion; NAC: neoadjuvant chemotherapy; NGS: next-generation sequencing; PARPi: poly ADP-ribose polymerase inhibitor; pCR: pathologic complete response; RCB: residual cancer burden; RD: residual disease; TDM1: trastuzumab-emtansine; TILS: tumors-infiltrating lymphocytes.</p>
Full article ">Figure 2
<p>Proportion of pathological complete response (pCR) after neoadjuvant therapy according to breast cancer subtypes. HR-positive/HER2-negative (HR+/HER2−) is the most prevalent subtype in breast cancer, occurring in approximately 70% of patients, followed by HER2−positive (HER2+) and TNBC subtypes (approximately 20% and 10%, respectively). Subtype-specific pCR rate are 8.3% in HR+/HER2−, 18.7% in HER2+/HR+, 38.9% in HER2+/HR− and 31.1% in TNBC (original figure based on literature data, i.e., Houssami et al., Meta-analysis of the association of breast cancer subtype and pathologic complete response to neoadjuvant chemotherapy, Eur J Cancer 2012. doi: 10.1016/j.ejca.2012.05.023) [<a href="#B9-cancers-14-05467" class="html-bibr">9</a>]. Abbreviations: HR: hormonal receptors; TNBC: triple-negative breast cancer; pCR: pathological complete response.</p>
Full article ">
16 pages, 1185 KiB  
Systematic Review
Speech and Language Errors during Awake Brain Surgery and Postoperative Language Outcome in Glioma Patients: A Systematic Review
by Ellen Collée, Arnaud Vincent, Clemens Dirven and Djaina Satoer
Cancers 2022, 14(21), 5466; https://doi.org/10.3390/cancers14215466 - 7 Nov 2022
Cited by 9 | Viewed by 3093
Abstract
Awake craniotomy with direct electrical stimulation (DES) is the standard treatment for patients with gliomas in eloquent areas. Even though language is monitored carefully during surgery, many patients suffer from postoperative aphasia, with negative effects on their quality of life. Some perioperative factors [...] Read more.
Awake craniotomy with direct electrical stimulation (DES) is the standard treatment for patients with gliomas in eloquent areas. Even though language is monitored carefully during surgery, many patients suffer from postoperative aphasia, with negative effects on their quality of life. Some perioperative factors are reported to influence postoperative language outcome. However, the influence of different intraoperative speech and language errors on language outcome is not clear. Therefore, we investigate this relation. A systematic search was performed in which 81 studies were included, reporting speech and language errors during awake craniotomy with DES and postoperative language outcomes in adult glioma patients up until 6 July 2020. The frequencies of intraoperative errors and language status were calculated. Binary logistic regressions were performed. Preoperative language deficits were a significant predictor for postoperative acute (OR = 3.42, p < 0.001) and short-term (OR = 1.95, p = 0.007) language deficits. Intraoperative anomia (OR = 2.09, p = 0.015) and intraoperative production errors (e.g., dysarthria or stuttering; OR = 2.06, p = 0.016) were significant predictors for postoperative acute language deficits. Postoperatively, the language deficits that occurred most often were production deficits and spontaneous speech deficits. To conclude, during surgery, intraoperative anomia and production errors should carry particular weight during decision-making concerning the optimal onco-functional balance for a given patient, and spontaneous speech should be monitored. Further prognostic research could facilitate intraoperative decision-making, leading to fewer or less severe postoperative language deficits and improvement of quality of life. Full article
(This article belongs to the Special Issue Advances of Brain Mapping in Cancer Research)
Show Figures

Figure 1

Figure 1
<p>PRISMA flowchart of total records identified through searching of databases.</p>
Full article ">Figure 2
<p>Language status in the preoperative (T0) and postoperative phases (T1–T4).</p>
Full article ">Figure 3
<p>Significant preoperative and intraoperative predictors for postoperative language deficits at T1 (<span class="html-italic">n</span> = 589) and T3 (<span class="html-italic">n</span> = 456). T0 = preoperatively and postoperatively, T1 = 1–10 days, T3 = ≥3–8 months, OR = odds ratio and CI = confidence interval. This figure describes the binary logistic regression (T1 and T3) with the predictors of preoperative language status, intraoperative speech and language error categories, where the dependent variable is postoperative language outcome and reference categories are no preoperative language deficits and intraoperative speech arrest. * = <span class="html-italic">p</span> &lt; 0.05. ** = <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
18 pages, 1941 KiB  
Article
The Glasgow Prognostic Score Predicts Survival Outcomes in Neuroendocrine Neoplasms of the Gastro–Entero–Pancreatic (GEP-NEN) System
by Niklas Gebauer, Maria Ziehm, Judith Gebauer, Armin Riecke, Sebastian Meyhöfer, Birte Kulemann, Nikolas von Bubnoff, Konrad Steinestel, Arthur Bauer and Hanno M. Witte
Cancers 2022, 14(21), 5465; https://doi.org/10.3390/cancers14215465 - 7 Nov 2022
Cited by 1 | Viewed by 1854
Abstract
Background: Across a variety of solid tumors, prognostic implications of nutritional and inflammation-based risk scores have been identified as a complementary resource of risk stratification. Methods: In this retrospective study, we performed a comparative analysis of several established risk scores and ratios, such [...] Read more.
Background: Across a variety of solid tumors, prognostic implications of nutritional and inflammation-based risk scores have been identified as a complementary resource of risk stratification. Methods: In this retrospective study, we performed a comparative analysis of several established risk scores and ratios, such as the Glasgow Prognostic Score (GPS), in neuroendocrine neoplasms of the gastro–entero–pancreatic (GEP-NEN) system with respect to their prognostic capabilities. Clinicopathological and treatment-related data for 102 GEP-NEN patients administered to the participating institutions between 2011 and 2021 were collected. Scores/ratios significantly associated with overall or progression-free survival (OS, PFS) upon univariate analysis were subsequently included in a Cox-proportional hazard model for the multivariate analysis. Results: The median age was 62 years (range 18–95 years) and the median follow-up period spanned 51 months. Pancreatic or intestinal localization at the initial diagnosis were present in 41 (40.2%) and 44 (43.1%) cases, respectively. In 17 patients (16.7%), the primary manifestation could not be ascertained (NNUP; neuroendocrine neoplasms of unknown primary). Histological grading (HG) revealed 24/102 (23.5%) NET/NEC (poorly differentiated; high grade G3) and 78/102 (76.5%) NET (highly or moderately differentiated; low–high grade G1–G2). In total, 53/102 (51.9%) patients presented with metastatic disease (UICC IV), 11/102 (10.7%) patients presented with multifocal disease, and 56/102 (54.9%) patients underwent a primary surgical or endoscopic approach, whereas 28 (27.5%) patients received systemic cytoreductive treatment. The univariate analysis revealed the GPS and PI (prognostic index), as well as UICC-stage IV, HG, and the Charlson comorbidity index (CCI) to predict both the PFS and OS in GEP-NEN patients. However, the calculation of the survival did not separate GPS subgroups at lower risk (GPS 0 versus GPS 1). Upon the subsequent multivariate analysis, GPS was the only independent predictor of both OS (p < 0.0001; HR = 3.459, 95% CI = 1.263–6.322) and PFS (p < 0.003; HR = 2.119, 95% CI = 0.944–4.265). Conclusion: In line with previous results for other entities, the present study revealed the GPS at baseline to be the only independent predictor of survival across all stages of GEP-NEN, and thus supports its clinical utility for risk stratification in this group of patients. Full article
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)
Show Figures

Figure 1

Figure 1
<p>Stacked bar plots demonstrate the distribution of the relevant clinicopathological insights for each GPS subgroup ((<b>A</b>) age; (<b>B</b>) sex; (<b>C</b>) B symptoms; (<b>D</b>) ECOG performance status; (<b>E</b>) UICC stage; (<b>F</b>) histological grading). (<b>G</b>) The distribution of the GPS in relation to the primary tumor sites. Significant correlations are marked with * (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Visualization of Pearson’s correlation with respect to the clinical characteristics and inflammation-/nutritional-status-based risk scores/ratios. High degrees of correlation are colored in red and low degrees of correlation are colored in blue.</p>
Full article ">Figure 3
<p>Progression-free (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>,<b>I</b>,<b>K</b>) and overall (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>,<b>J</b>,<b>L</b>) survival according to the Glasgow Prognostic Score (GPS) (log-rank GPS 0 vs. GPS 1 vs. GPS 2; (<b>A</b>,<b>B</b>)), CRP/albumin ratio (CAR) (log-rank test; (<b>C</b>,<b>D</b>)) and primary tumor sites (log-rank test; (<b>K</b>,<b>L</b>)) in GEP-NEN patients. (<b>E</b>–<b>J</b>) The Kaplan–Meier analysis (PFS and OS) according to the histological grading among the GPS subtypes.</p>
Full article ">
19 pages, 3728 KiB  
Systematic Review
Anastomotic Leak in Ovarian Cancer Cytoreduction Surgery: A Systematic Review and Meta-Analysis
by Massimiliano Fornasiero, Georgios Geropoulos, Konstantinos S. Kechagias, Kyriakos Psarras, Konstantinos Katsikas Triantafyllidis, Panagiotis Giannos, Georgios Koimtzis, Nikoletta A. Petrou, James Lucocq, Christos Kontovounisios and Dimitrios Giannis
Cancers 2022, 14(21), 5464; https://doi.org/10.3390/cancers14215464 - 7 Nov 2022
Cited by 2 | Viewed by 2640
Abstract
Introduction: Anastomotic leaks (AL) following ovarian cytoreduction surgery could be detrimental, leading to significant delays in commencing adjuvant chemotherapy, prolonged hospital stays and increased morbidity. The aim of this study was to investigate risk factors associated with anastomotic leaks after ovarian cytoreduction surgery. [...] Read more.
Introduction: Anastomotic leaks (AL) following ovarian cytoreduction surgery could be detrimental, leading to significant delays in commencing adjuvant chemotherapy, prolonged hospital stays and increased morbidity. The aim of this study was to investigate risk factors associated with anastomotic leaks after ovarian cytoreduction surgery. Material and methods: The MEDLINE (via PubMed), Cochrane Library, EMBASE and Scopus bibliographical databases were searched. Original clinical studies investigating risk factors for AL in ovarian cytoreduction surgery were included. Results: Eighteen studies with non-overlapping populations reporting on patients undergoing cytoreduction surgery for ovarian cancer (n = 4622, including 344 cases complicated by AL) were included in our analysis. Patients undergoing ovarian cytoreduction surgery complicated by AL had a significantly higher rate of 30-day mortality but no difference in 60-day mortality. Multiple bowel resections were associated with an increased risk of postoperative AL, while no association was observed with body mass index (BMI), American Society of Anesthesiologists (ASA) score, age, smoking, operative approach (primary versus interval cytoreductive, stapled versus hand-sewn anastomoses and formation of diverting stoma), neoadjuvant chemotherapy and use of hyperthermic intraperitoneal chemotherapy (HIPEC). Discussion: Multiple bowel resections were the only clinical risk factor associated with increased risk for AL after bowel surgery in the ovarian cancer population. The increased 30-day mortality rate in patients undergoing ovarian cytoreduction complicated by AL highlights the need to minimize the number of bowel resections in this population. Further studies are required to clarify any association between neoadjuvant chemotherapy and decreased AL rates. Full article
(This article belongs to the Special Issue Systematic Reviews and Meta-Analyses of Genitourinary Cancers)
Show Figures

Figure 1

Figure 1
<p>PRISMA flowchart.</p>
Full article ">Figure 2
<p>Basic patient characteristics.</p>
Full article ">Figure 3
<p>ASA scores.</p>
Full article ">Figure 4
<p>Chemotherapy administration.</p>
Full article ">Figure 5
<p>Intraoperative outcomes.</p>
Full article ">Figure 6
<p>Stoma formation and primary cytoreduction risk factors.</p>
Full article ">Figure 7
<p>Mortality rates.</p>
Full article ">
14 pages, 1640 KiB  
Article
Enhanced Recovery after Uterine Corpus Cancer Surgery: A 10 Year Retrospective Cohort Study of Robotic Surgery in an NHS Cancer Centre
by Christina Uwins, Radwa Hablase, Hasanthi Assalaarachchi, Anil Tailor, Alexandra Stewart, Jayanta Chatterjee, Patricia Ellis, Simon S. Skene, Agnieszka Michael and Simon Butler-Manuel
Cancers 2022, 14(21), 5463; https://doi.org/10.3390/cancers14215463 - 7 Nov 2022
Cited by 4 | Viewed by 2690
Abstract
Royal Surrey NHS Foundation Trust introduced robotic surgery for uterine corpus cancer in 2010 to support increased access to minimally invasive surgery, a central element of an enhanced recovery after surgery (ERAS) pathway. More than 1750 gynaecological oncology robotic procedures have now been [...] Read more.
Royal Surrey NHS Foundation Trust introduced robotic surgery for uterine corpus cancer in 2010 to support increased access to minimally invasive surgery, a central element of an enhanced recovery after surgery (ERAS) pathway. More than 1750 gynaecological oncology robotic procedures have now been performed at Royal Surrey NHS Foundation Trust. A retrospective cohort study was performed of patients undergoing surgery for uterine corpus cancer between the 1 January 2010 and the 31 December 2019 to evaluate its success. Data was extracted from the dedicated gynaecological oncology database and a detailed notes review performed. During this time; 952 patients received primary surgery for uterine corpus cancer; robotic: n = 734; open: n = 164; other minimally invasive surgery: n = 54. The introduction of the Da VinciTM robot to Royal Surrey NHS Foundation Trust was associated with an increase in the minimally invasive surgery rate. Prior to the introduction of robotic surgery in 2008 the minimally invasive surgery (MIS) rate was 33% for women with uterine corpus cancer undergoing full surgical staging. In 2019, 10 years after the start of the robotic surgery program 91.3% of women with uterine corpus cancer received robotic surgery. Overall the MIS rate increased from 33% in 2008 to 92.9% in 2019. Robotic surgery is associated with a low 30-day mortality (0.1%), low return to theatre (0.5%), a low use of blood transfusion and intensive care (1.8% & 7.2% respectively), low conversion to open surgery (0.5%) and a reduction in median length of stay from 6 days (in 2008) to 1 day, regardless of age/BMI. Robotic survival is consistent with published data. Introduction of the robotic program for the treatment of uterine cancer increased productivity and was associated with a highly predicable patient pathway of care, for high-risk patients, with reduced demands on health services. Future health care commissioning should further expand access to robotic surgery nationally for women with uterine corpus cancer. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Implementation of Robotic Surgery for Corpus Cancer at Royal Surrey NHS Foundation Trust.</p>
Full article ">Figure 2
<p>Robotic Cohort—Association between BMI/Age and Median EBL/Length of stay (LOS).</p>
Full article ">Figure 3
<p>Robotic Cohort—Overall, Disease Specific Survival &amp; Relapse Free Survival by FIGO Stage (2009).</p>
Full article ">Figure 4
<p>Growth in the use of Robotic Surgery at Royal Surrey NHS Foundation Trust, Guildford.</p>
Full article ">
13 pages, 281 KiB  
Article
Impact of Lymph Node Dissection on Postoperative Complications of Total Thyroidectomy in Patients with Thyroid Carcinoma
by Gregory Baud, Arnaud Jannin, Camille Marciniak, Benjamin Chevalier, Christine Do Cao, Emmanuelle Leteurtre, Amandine Beron, Georges Lion, Samuel Boury, Sebastien Aubert, Brigitte Bouchindhomme, Marie-Christine Vantyghem, Robert Caiazzo and François Pattou
Cancers 2022, 14(21), 5462; https://doi.org/10.3390/cancers14215462 - 7 Nov 2022
Cited by 17 | Viewed by 2447
Abstract
Background: Lymph node dissection (LND) in primary treatment of differentiated thyroid carcinoma is controversial. The aim of our retrospective study was to analyse the risk factors of post-thyroidectomy complications and to assess the morbidity of lymph node dissection, especially in the central neck [...] Read more.
Background: Lymph node dissection (LND) in primary treatment of differentiated thyroid carcinoma is controversial. The aim of our retrospective study was to analyse the risk factors of post-thyroidectomy complications and to assess the morbidity of lymph node dissection, especially in the central neck compartment, since prophylactic central lymph node dissection has not been proven to bring an overall survival benefit. Methods: We performed a retrospective analysis of postoperative complications from 1547 consecutive patients with differentiated thyroid carcinoma in an academic department of endocrine surgery over a period of 10 years. Results: A total of 535 patients underwent lymph node dissection, whereas the other 1012 did not. The rate of postoperative hypoparathyroidism was higher in patients with LND (17.6% vs. 11.4%, p = 0.001). No significant difference in the rate of permanent hypoparathyroidism (2.4% vs. 1.3%, p = 0.096) was observed between these two groups. A multivariate analysis was performed. Female gender, ipsilateral and bilateral central LND (CLND), parathyroid autotransplantation, and the presence of the parathyroid gland on the resected thyroid were associated with transient hypoparathyroidism. Bilateral CLND and the presence of the parathyroid gland on specimen were associated with permanent hypoparathyroidism. The rate of transient recurrent laryngeal nerve (RLN) injury (15.3% vs. 5.4%, p < 0.001) and permanent RLN injury (6.5% vs. 0.9%, p < 0.001) were higher in the LND group. In multivariate analysis, ipsilateral and bilateral lateral LND (LLND) were the main predictive factors of transient and permanent RLN injury. Bilateral RLN injury (2.6% vs. 0.4%, p < 0.001), chyle leakage (2.4% vs. 0%, p < 0.001), other nerve injuries (2.2% vs. 0%, p < 0.001), and abscess (2.4% vs. 0.5%, p = 0.001) were higher in the patients with LND. Conclusions: The surgical technique and the extent of lymph node dissection during surgery for thyroid carcinoma increase postoperative morbidity. A wider knowledge of lymph-node-dissection-related complications associated with thyroid surgery could help surgeons to carefully evaluate the surgical and medical therapeutic options. Full article
(This article belongs to the Special Issue Management and Treatment of Endocrine Tumors)
14 pages, 1759 KiB  
Article
A Risk Model for Patients with PSA-Only Recurrence (Biochemical Recurrence) Based on PSA and PSMA PET/CT: An Individual Patient Data Meta-Analysis
by Rie von Eyben, Daniel S. Kapp, Manuela Andrea Hoffmann, Cigdem Soydal, Christian Uprimny, Irene Virgolini, Murat Tuncel, Mathieu Gauthé and Finn E. von Eyben
Cancers 2022, 14(21), 5461; https://doi.org/10.3390/cancers14215461 - 7 Nov 2022
Cited by 4 | Viewed by 2258 | Correction
Abstract
An individual patient meta-analysis followed 1216 patients with PSA-only recurrence (biochemical recurrence, BCR) restaged with [68Ga]Ga-PSMA-11 PET/CT before the salvage treatment for median 3.5 years and analyzed the overall survival (OS). A new risk model included a good risk group with [...] Read more.
An individual patient meta-analysis followed 1216 patients with PSA-only recurrence (biochemical recurrence, BCR) restaged with [68Ga]Ga-PSMA-11 PET/CT before the salvage treatment for median 3.5 years and analyzed the overall survival (OS). A new risk model included a good risk group with a prescan PSA < 0.5 ng/mL (26%), an intermediate risk group with a prescan PSA > 0.5 ng/mL and a PSMA PET/CT with 1 to 5 positive sites (65%), and a poor risk group with a prescan PSA > 0.5 ng/mL and a PSA PET/CT with > 5 positive sites (9%) (p < 0.0001, log rank test). The poor risk group had a five-year OS > 60%. Adding a BCR risk score by the European Association of Urology did not significantly improve the prediction of OS (p = 0.64). In conclusion, the restaging PSMA PET/CT markedly predicted the 5-year OS. The new risk model for patients with PSA-only relapse requires a restaging PSMA PET/CT for patients with a prescan PSA > 0.5 ng/mL and has a potential use in new trials aiming to improve the outcome for patients with PSA-only recurrence who have polysites prostate cancer detected on PSMA PET/CT. Full article
(This article belongs to the Special Issue Prostate-Specific Membrane Antigen (PSMA))
Show Figures

Figure 1

Figure 1
<p>The PRISMA chart diagram shows how the meta-analysis selected and included the cohorts.</p>
Full article ">Figure 2
<p>The five cohorts had grossly similar relation between the prescan PSA and the number of positive sites on the restaging PSMA PET/CT. The cohorts are named by the cities for the study centers.</p>
Full article ">Figure 3
<p>The Forest plot of the patients with PSA-only relapse in the five cohorts. (<b>A</b>) a third of the patients had a negative restaging PSMA PET/CT, (<b>B</b>) more than half of the patients had one to five positive sites, (<b>C</b>) a tenth of the patients had more than five positive sites.</p>
Full article ">Figure 3 Cont.
<p>The Forest plot of the patients with PSA-only relapse in the five cohorts. (<b>A</b>) a third of the patients had a negative restaging PSMA PET/CT, (<b>B</b>) more than half of the patients had one to five positive sites, (<b>C</b>) a tenth of the patients had more than five positive sites.</p>
Full article ">Figure 4
<p>A new risk model shows a significant difference in overall survival between the good-, intermediate-, and poor risk groups (<span class="html-italic">p</span> &lt; 0.0001). The figure compares the overall survival for two patient groups by the number of positive findings on restaging PSMA PET/CT: 1–5 sites vs. &gt; 5 sites.</p>
Full article ">Figure 5
<p>The EAU BCR risk classification significantly separates the patients with PSA relapse into two groups with different overall survival (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 6
<p>Design for a phase III trial of patients with PSA-only recurrence and high risk according to the new risk model. The trial compares an aggressive multimodality salvage treatment with salvage treatment with established treatments. Abbreviations: ARI second generation androgen receptor inhibitor, MDT metastasis directed therapy, OS overall survival, PFS progression-free survival, SaRT Salvage radiation therapy.</p>
Full article ">
22 pages, 1413 KiB  
Review
The Monocyte, a Maestro in the Tumor Microenvironment (TME) of Breast Cancer
by Hoda T. Amer, Ulrike Stein and Hend M. El Tayebi
Cancers 2022, 14(21), 5460; https://doi.org/10.3390/cancers14215460 - 7 Nov 2022
Cited by 21 | Viewed by 4707
Abstract
Breast cancer (BC) is well-known for being a leading cause of death worldwide. It is classified molecularly into luminal A, luminal B HER2−, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These subtypes differ in their prognosis; thus, understanding the tumor microenvironment [...] Read more.
Breast cancer (BC) is well-known for being a leading cause of death worldwide. It is classified molecularly into luminal A, luminal B HER2−, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These subtypes differ in their prognosis; thus, understanding the tumor microenvironment (TME) makes new treatment strategies possible. The TME contains populations that exhibit anti-tumorigenic actions such as tumor-associated eosinophils. Moreover, it contains pro-tumorigenic populations such as tumor-associated neutrophils (TANs), or monocyte-derived populations. The monocyte-derived populations are tumor-associated macrophages (TAMs) and MDSCs. Thus, a monocyte can be considered a maestro within the TME. Moreover, the expansion of monocytes in the TME depends on many factors such as the BC stage, the presence of macrophage colony-stimulating factor (M-CSF), and the presence of some chemoattractants. After expansion, monocytes can differentiate into pro-inflammatory populations such as M1 macrophages or anti-inflammatory populations such as M2 macrophages according to the nature of cytokines present in the TME. Differentiation to TAMs depends on various factors such as the BC subtype, the presence of anti-inflammatory cytokines, and epigenetic factors. Furthermore, TAMs and MDSCs not only have a role in tumor progression but also are key players in metastasis. Thus, understanding the monocytes further can introduce new target therapies. Full article
Show Figures

Figure 1

Figure 1
<p>TAMs in the breast cancer TME. 1. The effect of TAMs on T cells: 1.a. TAMs can suppress T-cell proliferation via the programmed death-ligand 1 (PDL-1) [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 1.b. TGF-beta affects T-cell function by upregulation of PDL-1 on TAMs [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 1.c. Tregs are induced by IL-10, TGF-B, and PDGF-2, thus suppressing T cells [<a href="#B45-cancers-14-05460" class="html-bibr">45</a>]. 1.d. Treg recruitment happens through CCL7/8/22 [<a href="#B45-cancers-14-05460" class="html-bibr">45</a>]. 1.e. Increased activity of arginase enzyme and iNOS result in the increased level of NO and RNOS (ONOO<sup>−</sup>) leading to nitrosylation and thus impairing T-cell self-stimulation by IL-2 in addition to nitration of TCR signaling complex altering T-cell function [<a href="#B23-cancers-14-05460" class="html-bibr">23</a>]. 2. The effect of TAMs on NK cells: 2.a. The inhibitory effect of TAMs can be due to the expression of PDL-1 (which is highly expressed on TAMs) or 2.b. it can be due to TGF-B secretion [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 2.c. TAMs also suppress NK IFN-Y production [<a href="#B42-cancers-14-05460" class="html-bibr">42</a>]. 3. TAMs induce angiogenesis by releasing VEGF, PDGF, and IL-8 [<a href="#B23-cancers-14-05460" class="html-bibr">23</a>]. 4. Interaction between TAMs and cancer cells: 4.a. It was observed that VCAM1+ tumor cells have increased survival in a leukocyte-rich environment due to the adhesion of leukocyte receptors on BC cells (VCAM1) to TAM α 4 integrin [<a href="#B21-cancers-14-05460" class="html-bibr">21</a>]. 4.b. TGF-B was shown to upregulate PDL-1 on cancer cells, thus inducing an inhibitory effect on immune cells [<a href="#B23-cancers-14-05460" class="html-bibr">23</a>]. 4.c. There is a paracrine loop between cancer cells and TAMs. TAMs secrete epidermal growth factor (EGF) that binds to EGFRs on the cancer cells [<a href="#B21-cancers-14-05460" class="html-bibr">21</a>]. 4.d. TAMs express M-CSFR, which is a monocyte colony-stimulating factor receptor also known as colony-stimulating factor 1 receptor (CSF-1R or cFMS). The M-CSFR binds to the M-CSF (CSF-1) that is produced by cancer cells [<a href="#B41-cancers-14-05460" class="html-bibr">41</a>]. The binding of cancer cells with TAMs allows the co-migration of two different cell types, thus enhancing invasion, motility, and intravasation [<a href="#B21-cancers-14-05460" class="html-bibr">21</a>]. 5. CD163 and C206 are considered the commonly used self-markers for TAMs. 6. MCP-1 is a monocyte recruiter that is produced by TAMs; monocytes respond to TME and differentiate into TAMs [<a href="#B46-cancers-14-05460" class="html-bibr">46</a>]. 7.a. TAMs coordinate in the extracellular proteolysis through the secretion of tissue-remodeling cysteine cathepsin proteases that contribute to ECM and collagen degradation [<a href="#B47-cancers-14-05460" class="html-bibr">47</a>]. 7.b. Moreover, MMPs contribute to collagen degradation. Types I, III, IV, and VI are the major collagens that play an important role in tumors [<a href="#B47-cancers-14-05460" class="html-bibr">47</a>]. Myeloid cells remodel ECM by degrading the collagen through matrix metalloproteinases (MMPs) [<a href="#B34-cancers-14-05460" class="html-bibr">34</a>].</p>
Full article ">Figure 2
<p>IL-10: key inducers and key anti-inflammatory mechanisms. a. Inducing agents increase the release of IL-10 from different immune cells [<a href="#B25-cancers-14-05460" class="html-bibr">25</a>,<a href="#B54-cancers-14-05460" class="html-bibr">54</a>,<a href="#B55-cancers-14-05460" class="html-bibr">55</a>] b. IL-10 acts as an inhibitor for a number of pro-inflammatory cytokines released by macrophages/monocytes. c. IL-10 induces a small subset of genes in human monocytes [<a href="#B56-cancers-14-05460" class="html-bibr">56</a>]. d. IL-10 is an inhibitor for NF-kB (nuclear localization of nuclear factor kB), a transcriptional factor responsible for the expression of inflammatory genes; however, other transcriptional factors such as NF-IL6, AP-1, and AP-2 were not affected by IL-10 [<a href="#B57-cancers-14-05460" class="html-bibr">57</a>].</p>
Full article ">Figure 3
<p>Role of TAMs and MDSCs in metastasis: 1. Tumor growth and invasion. 1.a. TAMs produce MMPs (MMP1/2/3/7/9) and cathepsin. 1.b. Chemokines such as acidic and rich in cysteine (SPARC), chemokine (C-C motif) ligand 18 (CCL18), αvβ5 integrins, phosphatidylinositol transfer protein 3 (PITPNM3), epidermal growth factor (EGF), EGF receptor (EGFR), colony-stimulating factor 1 (CSF-1), and CSF-1 receptor (CSF-1R) allow the interaction between tumor cells, thus facilitating the invasion step [<a href="#B72-cancers-14-05460" class="html-bibr">72</a>].1.c. MDSCs facilitate the invasion step by producing MMPs and TGF-beta [<a href="#B73-cancers-14-05460" class="html-bibr">73</a>]. 2. Intravasation. 2.a. TAMs and MDSCs release VEGF-A [<a href="#B72-cancers-14-05460" class="html-bibr">72</a>,<a href="#B73-cancers-14-05460" class="html-bibr">73</a>]. 3. Extravasation. 3.a. Metastasis-associated macrophages (MAMs) accumulate at the metastatic site and also release VEGF-A. 3.b. The CCL2-CCR2 signaling pathway activates the CCL3-CCR1 signaling pathway in MAMs, leading to their accumulation in the metastatic site and thus attracting more tumor cells and prolonging the process of seeding. 4. Seeding [<a href="#B72-cancers-14-05460" class="html-bibr">72</a>]. 5. The metastatic niche in the lungs is induced by MDSCs through the production of CXCL-17 which recruits more MDSCs in the lungs, leading to an increase in the levels of platelet-derived growth factor-beta (PDGF-beta) that will induce angiogenesis, thus creating favorable conditions for metastatic cells [<a href="#B74-cancers-14-05460" class="html-bibr">74</a>].</p>
Full article ">
13 pages, 1123 KiB  
Article
Preoperative Serum Markers and Risk Classification in Intrahepatic Cholangiocarcinoma: A Multicenter Retrospective Study
by Masaki Kaibori, Kengo Yoshii, Hisashi Kosaka, Masato Ota, Koji Komeda, Masaki Ueno, Daisuke Hokutou, Hiroya Iida, Kosuke Matsui and Mitsugu Sekimoto
Cancers 2022, 14(21), 5459; https://doi.org/10.3390/cancers14215459 - 7 Nov 2022
Cited by 4 | Viewed by 2163
Abstract
Accurate risk stratification selects patients who are expected to benefit most from surgery. This retrospective study enrolled 225 Japanese patients with intrahepatic cholangiocellular carcinoma (ICC) who underwent hepatectomy between January 2009 and December 2020 and identified preoperative blood test biomarkers to formulate a [...] Read more.
Accurate risk stratification selects patients who are expected to benefit most from surgery. This retrospective study enrolled 225 Japanese patients with intrahepatic cholangiocellular carcinoma (ICC) who underwent hepatectomy between January 2009 and December 2020 and identified preoperative blood test biomarkers to formulate a classification system that predicted prognosis. The optimal cut-off values of blood test parameters were determined by ROC curve analysis, with Cox univariate and multivariate analyses identifying prognostic factors. Risk classifications were established using classification and regression tree (CART) analysis. CART analysis revealed decision trees for recurrence-free survival (RFS) and overall survival (OS) and created three risk classifications based on machine learning of preoperative serum markers. Five-year rates differed significantly (p < 0.001) between groups: 60.4% (low-risk), 22.8% (moderate-risk), and 4.1% (high-risk) for RFS and 69.2% (low-risk), 32.3% (moderate-risk), and 9.2% (high-risk) for OS. No difference in OS was observed between patients in the low-risk group with or without postoperative adjuvant chemotherapy, although OS improved in the moderate group and was prolonged significantly in the high-risk group receiving chemotherapy. Stratification of patients with ICC who underwent hepatectomy into three risk groups for RFS and OS identified preoperative prognostic factors that predicted prognosis and were easy to understand and apply clinically. Full article
(This article belongs to the Collection Treatment of Hepatocellular Carcinoma and Cholangiocarcinoma)
Show Figures

Figure 1

Figure 1
<p>The decision tree model based on CA19-9, CAR, and CRP for predicting recurrence-free survival. CA19-9: carbohydrate antigen 19-9; CRP: C-reactive protein; CAR: CRP/albumin ratio.</p>
Full article ">Figure 2
<p>Survival outcomes. (<b>A</b>) Recurrence-free survival among three groups stratified according to the three risk classifications. (<b>B</b>) Overall survival among three groups stratified according to the three risk classifications. (<b>C</b>) Recurrence-free survival among three groups stratified according to the 8th edition of the AJCC staging system. (<b>D</b>) Overall survival among three groups stratified according to the 8th edition of the AJCC staging system. CI: confidence interval; HR: hepatic resection; AIC: Akaike information criterion.</p>
Full article ">Figure 3
<p>Comparison of survival outcomes after hepatic resection between patients with or without adjuvant chemotherapy. (<b>A</b>) Recurrence-free survival in the low-risk group. (<b>B</b>) Overall survival in the low-risk group. (<b>C</b>) Recurrence-free survival in the moderate-risk group. (<b>D</b>) Overall survival in the moderate-risk group. (<b>E</b>) Recurrence-free survival in the high-risk group. (<b>F</b>) Overall survival in the high-risk group. CI: confidence interval; HR: hepatic resection.</p>
Full article ">
18 pages, 4588 KiB  
Review
Intravascular NK/T-Cell Lymphoma: What We Know about This Diagnostically Challenging, Aggressive Disease
by Magda Zanelli, Paola Parente, Francesca Sanguedolce, Maurizio Zizzo, Andrea Palicelli, Alessandra Bisagni, Illuminato Carosi, Domenico Trombetta, Luca Mastracci, Linda Ricci, Saverio Pancetti, Giovanni Martino, Giuseppe Broggi, Rosario Caltabiano, Alberto Cavazza and Stefano Ascani
Cancers 2022, 14(21), 5458; https://doi.org/10.3390/cancers14215458 - 6 Nov 2022
Cited by 10 | Viewed by 2320
Abstract
Intravascular lymphoma is a form of lymphoid malignancy characterized by neoplastic cells growing almost exclusively within the lumina of small- to medium-sized blood vessels. Most cases are of B-cell origin with rare cases of natural killer or T-cell lineage. Extranodal sites are affected, [...] Read more.
Intravascular lymphoma is a form of lymphoid malignancy characterized by neoplastic cells growing almost exclusively within the lumina of small- to medium-sized blood vessels. Most cases are of B-cell origin with rare cases of natural killer or T-cell lineage. Extranodal sites are affected, mainly the skin and central nervous system, although any organ may be involved. Intravascular NK/T-cell lymphoma deserves special attention because of its clinicopathologic features and the need for adequate immunophenotyping combined with clonality test for a proper diagnosis. Moreover, intravascular NK/T-cell lymphoma is strongly linked to Epstein–Barr virus (EBV), which is considered to play a role in tumorigenesis and to be responsible for the aggressive behavior of the disease. In this paper, we review the current knowledge on this rare lymphoma and, in particular, the most recent advances about its molecular landscape. The main distinguishing features with other EBV-related entities, such as extranodal NK/T-cell lymphoma, EBV-positive primary nodal T/NK-cell lymphoma, and aggressive NK-cell leukemia, are discussed to help pathologists obtain the correct diagnosis and consequently develop an adequate and prompt therapy response. Full article
(This article belongs to the Special Issue Advances in NK/T-cell Lymphoma, Epidemiology, Biology and Therapy)
Show Figures

Figure 1

Figure 1
<p>High-power view showing kidney parenchyma with large, atypical cells growing within vascular spaces (see within red circle; hematoxylin and eosin, 200× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 2
<p>High-power view showing central nervous system parenchyma with atypical cells within a vascular space (hematoxylin and eosin, 400× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 3
<p>Medium-power view showing bone marrow with CD3-positive atypical cells within sinusoidal spaces (CD3 immunostaining, 100× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 4
<p>High-power view showing lung parenchyma with perforin-positive atypical cells within vascular spaces (perforin immunostaining, 200× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 5
<p>High-power view showing lung parenchyma with EBER-positive atypical cells within vascular spaces (in situ hybridization for EBV-encoded RNA, 400× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 6
<p>High-power view showing a diffuse and polymorphic proliferation of atypical, medium- to large-sized lymphoid cells (hematoxylin and eosin, 200× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 7
<p>High-power view showing a diffuse proliferation of CD56-positive atypical cells (CD56 immunostaining, 200× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 8
<p>High-power view showing CD30-positive atypical cells (CD30 immunostaining, 400× magnification, previously unpublished, original image from S.A.).</p>
Full article ">Figure 9
<p>Medium-power view showing a diffuse proliferation of EBER-positive cells (in situ hybridization for EBV-encoded RNA, 100× magnification, previously unpublished, original image from S.A.).</p>
Full article ">
18 pages, 1880 KiB  
Article
An Effective Method for Lung Cancer Diagnosis from CT Scan Using Deep Learning-Based Support Vector Network
by Imran Shafi, Sadia Din, Asim Khan, Isabel De La Torre Díez, Ramón del Jesús Palí Casanova, Kilian Tutusaus Pifarre and Imran Ashraf
Cancers 2022, 14(21), 5457; https://doi.org/10.3390/cancers14215457 - 6 Nov 2022
Cited by 57 | Viewed by 11922
Abstract
The diagnosis of early-stage lung cancer is challenging due to its asymptomatic nature, especially given the repeated radiation exposure and high cost of computed tomography(CT). Examining the lung CT images to detect pulmonary nodules, especially the cell lung cancer lesions, is also tedious [...] Read more.
The diagnosis of early-stage lung cancer is challenging due to its asymptomatic nature, especially given the repeated radiation exposure and high cost of computed tomography(CT). Examining the lung CT images to detect pulmonary nodules, especially the cell lung cancer lesions, is also tedious and prone to errors even by a specialist. This study proposes a cancer diagnostic model based on a deep learning-enabled support vector machine (SVM). The proposed computer-aided design (CAD) model identifies the physiological and pathological changes in the soft tissues of the cross-section in lung cancer lesions. The model is first trained to recognize lung cancer by measuring and comparing the selected profile values in CT images obtained from patients and control patients at their diagnosis. Then, the model is tested and validated using the CT scans of both patients and control patients that are not shown in the training phase. The study investigates 888 annotated CT scans from the publicly available LIDC/IDRI database. The proposed deep learning-assisted SVM-based model yields 94% accuracy for pulmonary nodule detection representing early-stage lung cancer. It is found superior to other existing methods including complex deep learning, simple machine learning, and the hybrid techniques used on lung CT images for nodule detection. Experimental results demonstrate that the proposed approach can greatly assist radiologists in detecting early lung cancer and facilitating the timely management of patients. Full article
Show Figures

Figure 1

Figure 1
<p>CT scans based on maximum intensity projection.</p>
Full article ">Figure 2
<p>Flow of the proposed approach.</p>
Full article ">Figure 3
<p>Flow diagram of the proposed system.</p>
Full article ">Figure 4
<p>Architecture of proposed hybrid model (CNN + SVM).</p>
Full article ">Figure 5
<p>Proposed CNN for classification of nodules (Cancerous-1, Non-Cancerous-0).</p>
Full article ">Figure 6
<p>Wide neural net for classification of lung nodules.</p>
Full article ">Figure 7
<p>Proposed hybrid model using CNN with NB.</p>
Full article ">Figure 8
<p>Proposed hybrid model combining CNN and DT.</p>
Full article ">Figure 9
<p>Hybrid model CNN + RF.</p>
Full article ">Figure 10
<p>Comparison of all applied techniques on LUNA16 dataset.</p>
Full article ">Figure 11
<p>Comparison with other existing CAD systems.</p>
Full article ">
22 pages, 1365 KiB  
Review
Targeted Therapy and Immunotherapy for Heterogeneous Breast Cancer
by Xiaolu Sun, Kuai Liu, Shuli Lu, Weina He and Zixiu Du
Cancers 2022, 14(21), 5456; https://doi.org/10.3390/cancers14215456 - 6 Nov 2022
Cited by 16 | Viewed by 3935
Abstract
Breast cancer (BC) is the most common malignancy in women worldwide, and it is a molecularly diverse disease. Heterogeneity can be observed in a wide range of cell types with varying morphologies and behaviors. Molecular classifications are broadly used in clinical diagnosis, including [...] Read more.
Breast cancer (BC) is the most common malignancy in women worldwide, and it is a molecularly diverse disease. Heterogeneity can be observed in a wide range of cell types with varying morphologies and behaviors. Molecular classifications are broadly used in clinical diagnosis, including estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), and breast cancer gene (BRCA) mutations, as indicators of tumor heterogeneity. Treatment strategies differ according to the molecular subtype. Besides the traditional treatments, such as hormone (endocrine) therapy, radiotherapy, and chemotherapy, innovative approaches have accelerated BC treatments, which contain targeted therapies and immunotherapy. Among them, monoclonal antibodies, small-molecule inhibitors and antibody–drug conjugates, and targeted delivery systems are promising armamentarium for breast cancer, while checkpoint inhibitors, CAR T cell therapy, cancer vaccines, and tumor-microenvironment-targeted therapy provide a more comprehensive understanding of breast cancer and could assist in developing new therapeutic strategies. Full article
(This article belongs to the Special Issue Nanobiomaterials for Cancer Early Detection and Therapy)
Show Figures

Figure 1

Figure 1
<p>Molecular classification of breast cancer and corresponding treatments.</p>
Full article ">Figure 2
<p>Nanoparticles with varying characteristics in targeting drug delivery systems.</p>
Full article ">Figure 3
<p>The tumor microenvironment of breast cancer.</p>
Full article ">
10 pages, 19804 KiB  
Article
Recurrence of Oral Leukoplakia after CO2 Laser Resection: A Prospective Longitudinal Study
by Adela Rodriguez-Lujan, Pia López-Jornet and Eduardo Pons-Fuster López
Cancers 2022, 14(21), 5455; https://doi.org/10.3390/cancers14215455 - 6 Nov 2022
Cited by 4 | Viewed by 4965
Abstract
Aim: The aim of this study is to assess the efficacy of CO2 laser treatment in oral leukoplakia and to analyse the recurrence rate of oral leukoplakia lesions at 18-month follow-up. Materials and methods: A prospective clinical study regarding CO2 laser [...] Read more.
Aim: The aim of this study is to assess the efficacy of CO2 laser treatment in oral leukoplakia and to analyse the recurrence rate of oral leukoplakia lesions at 18-month follow-up. Materials and methods: A prospective clinical study regarding CO2 laser treatment for oral leukoplakia was conducted, in which 39 patients with a total of 53 oral leukoplakias were included. Follow-up was performed at 18 months post-surgery and the following variables were studied: sex, age, associated risk factors, clinical classification, size, location and presence of epithelial dysplasia, recurrence, and rate of malignant transformation after resection. Results: In the analysis of the final results 18 months after baseline, a treatment success rate of 43.75% was observed. Oral leukoplakia recurred in 54.17% of cases, and 2.08% of leukoplakias progressed to cancer. Among all the studied variables (age, tobacco use, size, location, clinical type or histology), no significant differences were found with regard to recurrence. Conclusion: The use of CO2 laser therapy to treat leukoplakia lesions is sufficient to remove such lesions. However, parameters that can assess recurrence need to be sought. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>). Oral leukoplakia Before CO<sub>2</sub>. (<b>B</b>) Immediately after treatment.</p>
Full article ">Figure 2
<p>(<b>A</b>) Lesion before CO<sub>2</sub>. (<b>B</b>) Immediately after carbon dioxide surgery.</p>
Full article ">Figure 3
<p>(<b>A</b>) before removal with CO<sub>2</sub> laser on lingual margin and (<b>B</b>) 3 months after treatment.</p>
Full article ">Figure 4
<p>Survival curve developed by the Kaplan-Meier method. Patient disease-free survival at 18 months was 23.3 ± 0.06, with an estimated 10.2 ± 0.8 months (95% CI = 8.63–11.8) on average until recurrence of the lesion.</p>
Full article ">
15 pages, 7120 KiB  
Article
Discovery of Mitochondrial Complex I Inhibitors as Anticancer and Radiosensitizer Drugs Based on Compensatory Stimulation of Lactate Release
by Junjie Lan, Octavia Cadassou, Cyril Corbet, Olivier Riant and Olivier Feron
Cancers 2022, 14(21), 5454; https://doi.org/10.3390/cancers14215454 - 6 Nov 2022
Cited by 7 | Viewed by 2313
Abstract
Cancer cells may stimulate glycolytic flux when O2 becomes insufficient. Increase in L-lactate release therefore appears as an escape mechanism to drugs targeting mitochondrial respiration but also represents a response that may be exploited to screen for compounds blocking either mitochondrial carriers [...] Read more.
Cancer cells may stimulate glycolytic flux when O2 becomes insufficient. Increase in L-lactate release therefore appears as an escape mechanism to drugs targeting mitochondrial respiration but also represents a response that may be exploited to screen for compounds blocking either mitochondrial carriers of oxidizable substrates or the electron transport chain. Here, we developed a screening procedure based on the capacity of cancer cells to release L-lactate to gain insights on the development of mitochondrial complex I inhibitors. For this purpose, we synthesized derivatives of carboxyamidotriazole, a compound previously described as a potential OXPHOS inhibitor. Two series of derivatives were generated by cycloaddition between benzylazide and either cyanoacetamides or alkynes. A primary assay measuring L-lactate release as a compensatory mechanism upon OXPHOS inhibition led us to identify 15 hits among 28 derivatives. A secondary assay measuring O2 consumption in permeabilized cancer cells confirmed that 12 compounds among the hits exhibited reversible complex I inhibitory activity. Anticancer effects of a short list of 5 compounds identified to induce more L-lactate release than reference compound were then evaluated on cancer cells and tumor-mimicking 3D spheroids. Human and mouse cancer cell monolayers exhibiting high level of respiration in basal conditions were up to 3-fold more sensitive than less oxidative cancer cells. 3D tumor spheroids further revealed potency differences between selected compounds in terms of cytotoxicity but also radiosensitizing activity resulting from local reoxygenation. In conclusion, this study documents the feasibility to efficiently screen in 96-well plate format for mitochondrial complex I inhibitors based on the capacity of drug candidates to induce L-lactate release. Full article
(This article belongs to the Special Issue The Role of Lactate Isomers in Cancer)
Show Figures

Figure 1

Figure 1
<p>Main synthetic paths to generate carboxyamidotriazole derivatives. The key fragments benzyl azide, <span class="html-italic">N</span>-substituted-2-cyanoacetamides and alkynes were prepared by conventional synthetic methods. The 5-amino-4-carboxyamido-1,2,3-triazole derivatives were assembled by a 1,3-dipolar cycloaddition between 2-cyanoacetamide or <span class="html-italic">N</span>-substituted 2-cyanoacetamides with the corresponding benzyl azides, using sodium ethoxide as a base, as previously reported [<a href="#B31-cancers-14-05454" class="html-bibr">31</a>,<a href="#B32-cancers-14-05454" class="html-bibr">32</a>]. Another series of <span class="html-italic">N</span>-substituted 1,2,3-triazoles, lacking the amino group on the 5-position of the heterocycle, were also prepared by Huisgen-Click cycloaddition reaction between a suitable benzyl azide and an alkyne, using the standard copper acetate/sodium ascorbate system as a catalyst [<a href="#B33-cancers-14-05454" class="html-bibr">33</a>].</p>
Full article ">Figure 2
<p>Screening procedure to identify anticancer activity of <span class="html-italic">bona fide</span> mitochondrial complex I inhibitors. The 3-step procedure includes the evaluation of (1) the stimulation of glycolysis upon OXPHOS inhibition (based on the increase in extracellular L-lactate release), (2) complex I inhibition (based on the measurement of real-time oxygen consumption rate (OCR) in permeabilized cancer cells exposed to a NADH-generating system and (3) the capacity to impact the growth of 3D tumor spheroids recapitulating tumor metabolic heterogeneity (through the determination of growth inhibitory effects, re-oxygenation and radio-sensitization).</p>
Full article ">Figure 3
<p>Compound-induced L-lactate release (Assay #1). Culture medium was collected after 24 h exposure to the indicated compounds (10 µM) and the amount of extracellular L-lactate release was determined in a semi-automated manner; dose-dependent effects of complex I inhibitor IACS are shown on the right (shaded bars). Bar graph depicts the extent of L-lactate secretion above the basal release as determined in the absence of any compound (n = 3, ** <span class="html-italic">p</span> &lt; 0.01 for increased L-lactate release vs. reference compound <b>aa</b>).</p>
Full article ">Figure 4
<p>Mitochondrial complex I activity inhibition (Assay #2). (<b>A</b>–<b>D</b>)<b>.</b> Graphs depict changes in O<sub>2</sub> consumption rate (OCR) measured with the Seahorse technology on permeabilized CT26 cancer cells upon successive addition of pyruvate/malate, the indicated inhibitor and succinate. OXPHOS I inhibitor IACS was used as a control in the experiments testing reference compound <b>aa</b> (<b>A</b>), and compound <b>aa</b> (10µM) was used as reference for testing <b>au, ah</b> and <b>at</b> derivatives (<b>B</b>–<b>D</b>) (n = 6 per compound). (<b>E</b>,<b>F</b>)<b>.</b> Growth inhibitory effects of cultured CT26 (<b>E</b>) and 4T1 (<b>B</b>) cancer cells exposed to 100 nM IACS or 1 µM <b>aa</b>, <b>ag</b>, <b>aj</b> and <b>au</b> compounds for 72 h; data represent the reduced extent (expressed as %) of cell viability (determined with Presto Blue) when compared with vehicle conditions (n = 3).</p>
Full article ">Figure 5
<p>Inhibition of 3D tumor spheroid growth (Assay #3). Graphs depict (<b>A</b>) the direct growth inhibitory effects of the indicated compounds and (<b>B</b>) their capacity to exert radiosensitizing effects as determined using 3D tumor CT26 spheroids (n = 6 per condition). ** <span class="html-italic">p</span> &lt; 0.01 for spheroid sizes at day 10 significantly smaller than spheroids treated with <b>aa</b> (<b>A</b>) or <b>aa</b> + irradiation (<b>B</b>) and <sup>§§</sup> <span class="html-italic">p</span> &lt; 0.01 for spheroid sizes at day 10 significantly smaller than corresponding spheroids at day 3 (<b>B</b>). Drug exposure without (<b>A</b>) or with irradiation (<b>B</b>) was associated with a significant reduction in spheroid sizes vs. untreated spheroids (not shown).</p>
Full article ">Figure 6
<p>Radiosensitizing effects of selected complex I inhibitors. (<b>A</b>) Extent of surviving cancer cells post-exposure to radiotherapy and the indicated compounds (n = 3, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. vehicle condition). (<b>B</b>) Representative pictures of pimonidazole immunostaining revealing a reduction in the hypoxic fraction of 3D tumor CT26 spheroids induced by the indicated compound; this experiment was repeated twice with similar results.</p>
Full article ">
19 pages, 3903 KiB  
Article
CAXII Is a Surrogate Marker for Luminal Breast Tumors Regulated by ER and GATA3
by Lucas Porras, Faustine Gorse, Ndeye Khady Thiombane, Louis Gaboury and Sylvie Mader
Cancers 2022, 14(21), 5453; https://doi.org/10.3390/cancers14215453 - 6 Nov 2022
Cited by 1 | Viewed by 2412
Abstract
Estrogen receptor alpha (ERα) expression in ~2/3 breast tumors selects patients for hormonal therapies. Tumors negative for ERα but positive for the progesterone receptor (PR, encoded by PGR) have also been candidates for ER-targeting therapies, as PR expression may reflect undetected ER [...] Read more.
Estrogen receptor alpha (ERα) expression in ~2/3 breast tumors selects patients for hormonal therapies. Tumors negative for ERα but positive for the progesterone receptor (PR, encoded by PGR) have also been candidates for ER-targeting therapies, as PR expression may reflect undetected ER activity. Conversely, PR status in ER+ tumors predicts a worse therapeutic response. Our analysis of breast tumor transcriptome datasets, however, revealed that in tumors with lower PGR expression, the clinical PR status does not correlate accurately with the expression of ESR1 or of ER target genes, including PGR itself. We identified carbonic anhydrase 12 (CA12) as an estrogen target gene better correlated with ESR1 than PGR, reflecting CA12 regulation by both ERα and the luminal factor and upstream ESR1 regulator GATA3. Immunostaining supported strong positive correlations at the protein level with ERα and GATA3 in a cohort of 118 tumors. Most ER+PR tumors expressed CAXII at levels similar to those of ER+PR+ tumors, consistent with observations in tumor transcriptome datasets and with active estrogenic signaling in some ER+PR breast cancer cell lines. The few ERPR+ tumors did not express CAXII or the other luminal markers FOXA1 and GATA3. Overall, CAXII is a luminal marker that can help interpret ER status in single ER/PR positive tumors. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>PR status imperfectly reflects <span class="html-italic">ESR1</span> expression, ER status and activity. Pair-wise scatterplots for the expression of <span class="html-italic">ESR1</span> vs <span class="html-italic">PGR</span> (<b>A</b>,<b>B</b>,<b>E</b>), <span class="html-italic">TFF1</span> (<b>C</b>), <span class="html-italic">GREB1</span> (<b>D</b>) and <span class="html-italic">CA12</span> (<b>F</b>) generated using MiSTIC [<a href="#B55-cancers-14-05453" class="html-bibr">55</a>]. Boxes identify 90% of <span class="html-italic">ESR1</span><sup>low</sup> tumors, and the portion of <span class="html-italic">ESR1</span><sup>high</sup> tumors identified using the same threshold of positivity for different ER target genes. Percentages of total tumors are shown. Enrichment of <span class="html-italic">ESR1</span><sup>high</sup><span class="html-italic">PGR</span><sup>low</sup> tumors (highlighted in green) in the CIT luminal B tumor subtype is shown in the inset of panel (<b>E</b>).</p>
Full article ">Figure 2
<p><span class="html-italic">CA12</span> mRNA levels are highly correlated with those of <span class="html-italic">ESR1</span>, <span class="html-italic">FOXA1</span> and <span class="html-italic">GATA3</span> in breast tumors. (<b>A</b>) Minimum spanning tree representing a cluster of genes expressed preferentially in luminal and molecular apocrine breast tumors, identified by gene correlation analysis from a TCGA breast cancer transcriptomic dataset comprising 754 tumors using MiSTIC software [<a href="#B55-cancers-14-05453" class="html-bibr">55</a>]. Genes encoding transcription factors (<span class="html-italic">ESR1</span>, <span class="html-italic">FOXA1</span>, <span class="html-italic">GATA3, SPDEF</span>, <span class="html-italic">XBP1</span> and <span class="html-italic">AR</span>) are highlighted in orange, whereas <span class="html-italic">CA12</span> is indicated in red. (<b>B</b>) Pair-wise correlation scatterplots for <span class="html-italic">CA12</span> and <span class="html-italic">ESR1</span> (top), <span class="html-italic">GATA3</span> (middle) or <span class="html-italic">FOXA1</span> (bottom) expression levels in the same cohort, with CIT tumor subtypes highlighted by different colors. Luminal A tumors appear in dark blue, luminal B in green, luminal C in light blue, molecular apocrine in magenta and basal-like in red. Normal-like or non-classified tumors are shown in black. Pearson correlation coefficients are indicated at the bottom of each pair-wise correlation scatterplot.</p>
Full article ">Figure 3
<p><span class="html-italic">ESR1</span><sup>high</sup> cell lines express high levels of <span class="html-italic">CA12</span>, but variable levels of <span class="html-italic">PGR</span>. Pair-wise scatterplots for the expression of <span class="html-italic">ESR1</span> vs those of <span class="html-italic">CA12</span> (<b>A</b>) or <span class="html-italic">PGR</span> (<b>B</b>). Values are Log2(Rpkm+1). RNA levels and cell line subtyping are from [<a href="#B61-cancers-14-05453" class="html-bibr">61</a>]. Selected cell lines are labeled.</p>
Full article ">Figure 4
<p>ERα and GATA3 positively regulate <span class="html-italic">CA12</span> expression in luminal breast cancer cell lines. (<b>A</b>) Schematic <span class="html-italic">CA12</span> gene organization with exons (grey boxes) and putative enhancers (red boxes). Predicted binding motifs for ERα and GATA3 are highlighted in bold in sequences for enhancers 1 and 2. (<b>B</b>) MCF-7, T-47D and ZR-75-1 cell lines were cultured in hormone-depleted medium for five days and then treated with E2 (25 nM). <span class="html-italic">CA12</span> mRNA levels were measured by RT-qPCR at 4, 8 and 16 h. <span class="html-italic">RPLP0</span> and <span class="html-italic">YWHAZ</span> were used as housekeeping genes for normalization. Values are presented as ratios over the expression in vehicle-treated samples for each time point. Asterisks indicate statistically significant regulations (Average of <span class="html-italic">n</span> = 3; *, <span class="html-italic">p</span> ≤ 0.05; **, <span class="html-italic">p</span> ≤ 0.01; ****, <span class="html-italic">p</span> ≤ 0.0001; Student’s unpaired <span class="html-italic">t</span>-test). (<b>C</b>) CAXII protein levels from MCF-7, T-47D and ZR-75-1 cells were analyzed at 0, 4, 8, 16 and 24 h after treatment with E2 by Western blotting (<span class="html-italic">n</span> = 3, a representative blot is shown). β-actin or Lamin B1 housekeeping proteins were used as loading controls. (<b>D</b>) MCF-7 cells were cultured in hormone-depleted medium for three days and then treated with E2 (25 nM) for 1 h before fixation and collection. Binding of ERα, FOXA1 and GATA3 to <span class="html-italic">CA12</span> enhancers was examined by ChIP-qPCR. Relative immunoprecipitation levels (ratios to IgG in vehicle-treated cells) are shown. The assay was performed twice with similar results. Asterisks indicate statistically significant binding compared to IgG in vehicle-treated cells for each enhancer from one experiment performed in triplicates (*, <span class="html-italic">p</span> ≤ 0.05; one-way ANOVA test, Dunnett’s multiple comparisons test). (<b>E</b>) MCF-7 cells were cultured in hormone-depleted medium for three days and transfected with a SMARTpool of siRNAs targeting <span class="html-italic">CA12</span> and two different siRNAs targeting <span class="html-italic">ESR1</span>, <span class="html-italic">FOXA1</span> or <span class="html-italic">GATA3</span>. Cells were then collected two days after transfection. Protein levels of CAXII, ERα, FOXA1 and GATA3 were analyzed by Western blotting (<span class="html-italic">n</span> = 2). β-actin was used as a loading control. (<b>F</b>,<b>G</b>) T-47D and ZR-75-1 cells were cultured in hormone-depleted medium for three days and transfected with two different siRNAs targeting <span class="html-italic">GATA3</span>. Cells were then collected two days after transfection. Protein levels of CAXII and GATA3 were analyzed by Western blotting in T-47D (<b>F</b>) and ZR-75-1 (<b>G</b>) cells; CAXII levels were quantified for both cell lines using ImageJ (average of <span class="html-italic">n</span> = 3 for quantification, one representative blot shown; *, <span class="html-italic">p</span> ≤ 0.05; **, <span class="html-italic">p</span> ≤ 0.01; Student’s unpaired <span class="html-italic">t</span>-test). Lamin B1 was used as a loading control.</p>
Full article ">Figure 5
<p>CAXII expression is increased in tumoral epithelial cells with loss of polarity. CAXII staining (in brown) of normal adjacent mammary tissue (<b>A</b>), columnar cell lesion (<b>B</b>), in situ breast ductal carcinoma (<b>C</b>) and invasive breast ductal carcinoma (<b>D</b>) at 20× and 40× magnification. The apical (A) and basal (B) poles of a luminal cell are labeled.</p>
Full article ">Figure 6
<p>Protein expression levels of CAXII and of the luminal transcription factors ERα, FOXA1 and GATA3 are correlated in breast tumors. Correlations between CAXII and ERα (<b>A</b>), GATA3 (<b>B</b>) or FOXA1 (<b>C</b>) expression scores are shown for all tumors in the TMAs. Scores were computed by QuPath using membrane (CAXII) and nuclear signal intensities (ERα, GATA3, FOXA1). ER<sup>+</sup>PR<sup>+</sup> tumors are highlighted in dark blue circles, ER<sup>+</sup>PR<sup>−</sup> tumors in light blue circles, ER<sup>−</sup>PR<sup>+</sup> tumors in yellow diamonds, ER<sup>−</sup>HER2<sup>+</sup> tumors in green squares and ER<sup>−</sup>HER2<sup>−</sup> tumors in red circles. Representative co-staining for CAXII (brown) and each of these luminal transcription factors (green) is also shown.</p>
Full article ">Figure 7
<p>CAXII expression discriminates ER<sup>+</sup> from ER<sup>−</sup> breast tumors. (<b>A</b>) Boxplot representing CAXII score distribution according to ER, PR and HER2 status (7 arrays, <span class="html-italic">n</span> = 118; ns, <span class="html-italic">p</span> &gt; 0.05; ****, <span class="html-italic">p</span> ≤ 0.0001; Dunn’s multiple comparisons post-hoc test performed after Kruskall-Wallis test (<span class="html-italic">p</span> &lt; 0.0001)). ER<sup>−</sup>PR<sup>+</sup> tumors are highlighted in yellow in the ER<sup>−</sup>HER2<sup>−</sup> group. (<b>B</b>) Representative cores for the four observed phenotypes of CAXII and ER status after co-staining with CAXII (brown) and ERα (green). Scores were computed by QuPath using membrane (CAXII) and nuclear (ERα) signal intensities.</p>
Full article ">
16 pages, 1397 KiB  
Article
Oncological Outcomes of Distal Ureterectomy for High-Risk Urothelial Carcinoma: A Multicenter Study by The French Bladder Cancer Committee
by Alexandra Masson-Lecomte, Victoire Vaillant, Mathieu Roumiguié, Stéphan Lévy, Benjamin Pradère, Michaël Peyromaure, Igor Duquesne, Alexandre De La Taille, Cédric Lebâcle, Adrien Panis, Olivier Traxer, Priscilla Leon, Maud Hulin, Evanguelos Xylinas, François Audenet, Thomas Seisen, Yohann Loriot, Yves Allory, Morgan Rouprêt and Yann Neuzillet
Cancers 2022, 14(21), 5452; https://doi.org/10.3390/cancers14215452 - 6 Nov 2022
Cited by 14 | Viewed by 2751
Abstract
Upper urinary tract urothelial carcinoma (UTUC) is an uncommon disease and its gold-standard treatment is radical nephroureterectomy (RNU). Distal ureterectomy (DU) might be an alternative for tumors of the distal ureter but its indications remain unclear. Here, we aimed to evaluate the oncological [...] Read more.
Upper urinary tract urothelial carcinoma (UTUC) is an uncommon disease and its gold-standard treatment is radical nephroureterectomy (RNU). Distal ureterectomy (DU) might be an alternative for tumors of the distal ureter but its indications remain unclear. Here, we aimed to evaluate the oncological outcomes of DU for UTUC of the pelvic ureter. We performed a multicenter retrospective analysis of patients with UTUC who underwent DU. The primary endpoint was 5-year cancer-specific survival (CSS), followed by overall survival (OS), intravesical recurrence-free (IVR) and homolateral urinary tract recurrence-free (HUR) survivals as secondary endpoints. Univariate and multivariate Cox regressions were performed to assess factors associated with outcomes. 155 patients were included, 91% of which were high-risk. 5-year CSS was 84.4%, OS was 71.9%, IVR-free survival was 43.6% and HUR-free survival was 74.4%. Multifocality, high grade and tumor size were the most significant predictors of survival endpoints. Of note, neither hydronephrosis nor pre-operative diagnostic ureteroscopy/JJ stent were associated with any of the endpoints. Perioperative morbidity was minimal. In conclusion, DU stands as a possible alternative to RNU for UTUC of the pelvic ureter. Close monitoring is mandatory due to the high risk of recurrence in the remaining urinary tract. Full article
(This article belongs to the Special Issue Advance and New Insights in Bladder Cancer)
Show Figures

Figure 1

Figure 1
<p>Flow chart for patients’ selection.</p>
Full article ">Figure 2
<p>Kaplan–Meier modelling of overall survival (5-year OS: 71.9%, lower—upper 95%CI 0.632–0.819).</p>
Full article ">Figure 3
<p>Kaplan–Meier modelling of cancer-specific survival (5-year CSS: 84.4%, lower—upper 95% CI 0.777–0.918).</p>
Full article ">Figure 4
<p>Kaplan–Meier modelling of intra vesical recurrence (5-year IV free survival: 43.6%, lower—upper 95% CI 0.349–0.545).</p>
Full article ">Figure 5
<p>Kaplan–Meier modelling of homolateral upper tact recurrence (5-year HUR free survival: 74.4%, lower—upper 95% CI 0.658–0.840).</p>
Full article ">
20 pages, 1034 KiB  
Article
A Retrospective, Single-Institution Experience of Bullous Pemphigoid as an Adverse Effect of Immune Checkpoint Inhibitors
by Walid Shalata, Sarah Weissmann, Sapir Itzhaki Gabay, Kim Sheva, Omar Abu Saleh, Ashraf Abu Jama, Alexander Yakobson and Keren Rouvinov
Cancers 2022, 14(21), 5451; https://doi.org/10.3390/cancers14215451 - 5 Nov 2022
Cited by 17 | Viewed by 2965
Abstract
Immune checkpoint inhibitors are a class of cancer treatment drugs that stimulate the immune system’s ability to fight tumor cells. These drugs are monoclonal antibodies targeting im-mune-inhibiting proteins on cancer cells, such as CTLA-4 and PD-1/PD-L1. Immune checkpoint inhibitors cause many immune-related adverse [...] Read more.
Immune checkpoint inhibitors are a class of cancer treatment drugs that stimulate the immune system’s ability to fight tumor cells. These drugs are monoclonal antibodies targeting im-mune-inhibiting proteins on cancer cells, such as CTLA-4 and PD-1/PD-L1. Immune checkpoint inhibitors cause many immune-related adverse events. Cutaneous toxicities are of the most common adverse effects and occur with a range of severity. Bullous Pemphigoid is a rare adverse event with a high impact on quality of life that may occur after immune checkpoint inhibitor treatment. In this article, we investigate current research on immune checkpoint inhibitors, cutaneous adverse events, and common presentations and treatments, with a specific focus on Bullous Pemphigoid, its characteristics, onset timing, and treatment. Significant findings include a negative skew in the onset of presentation. Furthermore, we describe exclusive cases. Full article
Show Figures

Figure 1

Figure 1
<p>Flow diagram of the single-center, retrospective, observational study of advanced or metastatic brain pineoblastoma, renal cell carcinoma, and urothelial carcinoma.</p>
Full article ">Figure 2
<p>Median weeks until presentation of Bullous Pemphigoid adverse event by treatment type.</p>
Full article ">
13 pages, 623 KiB  
Review
Long Non-Coding RNA and microRNA Interplay in Colorectal Cancer and Their Effect on the Tumor Microenvironment
by Marie Rajtmajerová, Andriy Trailin, Václav Liška, Kari Hemminki and Filip Ambrozkiewicz
Cancers 2022, 14(21), 5450; https://doi.org/10.3390/cancers14215450 - 5 Nov 2022
Cited by 9 | Viewed by 3093
Abstract
As the current staging and grading systems are not sufficient to stratify patients for therapy and predict the outcome of the disease, there is an urgent need to understand cancer in its complexity. The mutual relationship between tumour and immune or stromal cells [...] Read more.
As the current staging and grading systems are not sufficient to stratify patients for therapy and predict the outcome of the disease, there is an urgent need to understand cancer in its complexity. The mutual relationship between tumour and immune or stromal cells leads to rapid evolution and subsequent genetic and epigenetic changes. Immunoscore has been introduced as a diagnostic tool for colorectal cancer (CRC) only recently, emphasising the role of the specific tumor microenvironment in patient’s prognosis and overall outcome. Despite the fact that non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), cannot be translated into proteins, they significantly affect cell’s transcriptome and translatome. miRNA binding to mRNA efficiently blocks its translation and leads to mRNA destruction. On the other hand, miRNAs can be bound by lncRNAs or circular RNAs (circRNAs), which prevents them from interfering with translation. In this way, ncRNAs create a multi-step network that regulates the cell’s translatome. ncRNAs are also shed by the cell as exogenous RNAs and they are also found in exosomes, suggesting their role in intercellular communication. Hence, these mechanisms affect the tumor microenvironment as much as protein signal molecules. In this review, we provide an insight into the current knowledge of the microenvironment, lncRNAs’, and miRNAs’ interplay. Understanding mechanisms that underlie the evolution of a tissue as complex as a tumour is crucial for the future success in therapy. Full article
Show Figures

Figure 1

Figure 1
<p>lncRNA–miRNA interaction. Similarly to mRNAs, miRNAs undergo a series of processing steps in the nucleus as well as in the cytoplasm. Some miRNAs can also be produced from their lncRNA precursors. Fully matured mRNAs as well as all non-coding RNAs (such as miRNAs and lncRNAs) are localised in the cytoplasm where lncRNAs and mRNAs compete for miRNAs. This binding can either block or trigger the final translational process and is therefore an important post-transcriptional regulatory step in gene expression.</p>
Full article ">
18 pages, 3411 KiB  
Article
CRISPR/Cas9 Edited RAS & MEK Mutant Cells Acquire BRAF and MEK Inhibitor Resistance with MEK1 Q56P Restoring Sensitivity to MEK/BRAF Inhibitor Combo and KRAS G13D Gaining Sensitivity to Immunotherapy
by Elizabeth Turner, Luping Chen, John G. Foulke, Zhizhan Gu and Fang Tian
Cancers 2022, 14(21), 5449; https://doi.org/10.3390/cancers14215449 - 5 Nov 2022
Cited by 5 | Viewed by 3675
Abstract
BRAF V600E mutation drives uncontrolled cell growth in most melanomas. While BRAF V600E tumors are initially responsive to BRAF inhibitors, prolonged treatment results in inhibitor resistance and tumor regrowth. Clinical data have linked the NRAS Q61K, KRAS G13D and MEK1 Q56P mutations [...] Read more.
BRAF V600E mutation drives uncontrolled cell growth in most melanomas. While BRAF V600E tumors are initially responsive to BRAF inhibitors, prolonged treatment results in inhibitor resistance and tumor regrowth. Clinical data have linked the NRAS Q61K, KRAS G13D and MEK1 Q56P mutations to the BRAF inhibitor resistance. However, development of novel therapeutics is hindered by the lack of relevant isogeneic cell models. We employed CRISPR/Cas9 genome engineering to introduce NRAS Q61K, KRAS G13D and MEK1 Q56P mutations into the A375 melanoma cell line with endogenously high expression of BRAF V600E. The resulting isogenic cell lines are resistant to BRAF inhibitors. The A375 MEK1 Q56P isogenic cells are additionally resistant to MEK inhibitors as single agent, but interestingly, these cells become sensitive to MEK/BRAF inhibitor combo. Our results suggest that resistance in the NRAS and MEK isogenic lines is driven by constitutive MEK/ERK signaling, while the resistance in the KRAS isogenic line is driven by EGFR overexpression. Interestingly, the KRAS G13D isogenic line displays elevated PD-L1 expression suggesting the KRAS G13D mutation could be a potential indication for immunotherapy. Overall, these three novel isogenic cell models with endogenous level RAS and MEK1 point mutations provide direct bio-functional evidence demonstrating that acquiring a drug-resistant gene drives tumor cell survival and may simultaneously introduce new indications for combo therapy or immunotherapy in the clinic. Full article
(This article belongs to the Special Issue Targeting the (Un)Usual Suspects in Cancer)
Show Figures

Figure 1

Figure 1
<p>CRISPR/Cas9 engineering of isogenic A375 models of drug-resistant melanoma. (<b>A</b>) Schematic diagram of the CRISPR/Cas9 editing strategy used for the introduction of point mutations associated with BRAF and MEK inhibitor resistance into A375 melanoma cells. Two guide RNAs in complex with Cas9 (sg1 and sg2, scissors) were used to create double-stranded breaks in the intronic regions (black lines) to either side of the target exon (black box). A donor plasmid containing a copy of the target exon with the desired point mutation (star) and flanking intronic sequences was used as a repair template. This strategy ensures that any indels resulting from imperfect sequence repair at the Cas9 cut sites (red lines) are spliced out during mRNA processing and do not affect the resulting cellular protein; (<b>B</b>) Sanger sequencing of genomic DNA from the resulting KRAS G13D heterozygous (left), NRAS Q61K heterozygous (middle), and MEK1 Q56P homozygous (right) A375 isogenic lines.</p>
Full article ">Figure 2
<p>Isogenic melanoma models are resistant to BRAF inhibitors but not to BRAF non-specific chemotherapeutics in 2D tissue culture. (<b>A</b>) Dabrafenib resistance of A375 melanoma models in 2D tissue culture; (<b>B</b>) Vemurafenib resistance of A375 melanoma models in 2D tissue culture; (<b>C</b>) No resistance to the BRAF non-specific chemotherapeutic doxorubicin in 2D tissue culture; (<b>D</b>) Immunoblot demonstrating BRAF inhibitor resistance in EGFR pathway signaling in NRAS Q61K and KRAS G13D A375 melanoma models. Cells were treated with 1.0 μM of the indicated drug for 90 min prior to harvesting protein; (<b>E</b>) Dose-response curve for the MEK inhibitor trametinib in the MEK1 Q56P melanoma model in 2D tissue culture; (<b>F</b>) The same curve as in (<b>E</b>) for the MEK inhibitor binimetinib.</p>
Full article ">Figure 3
<p>Effect of tissue culture format on EGFR pathway signaling in drug-resistant melanoma models. (<b>A</b>) A375 WT, NRAS Q61K, KRAS G13D, and MEK1 Q56P cells growing in a 2D monolayer (top) and as 3D spheroids (bottom). For spheroid formation, 500 cells from each line were seeded in each well of a 96-well ultra-low attachment spheroid microplate and grown for five days before imaging; (<b>B</b>) Immunoblots tracking EGFR pathway signaling in engineered A375 melanoma model cells in 2D and 3D tissue culture. Protein was harvested from each cell line growing in 2D culture, and cellular protein from 3D spheroids was collected from spheroids seeded at 500 cells per 96-well and grown for seven days. Total protein (20 μg) was loaded in each lane and samples were blotted for total EGFR, total MEK1/2, phospho-MEK1/2 (Ser217/221), total ERK1/2 (p44/42 MAPK), phospho-ERK1/2 (Thr202/Tyr204), AKT, phospho-AKT (Ser473), and GAPDH.</p>
Full article ">Figure 4
<p>A375 Ras mutant melanoma models are resistant to BRAF inhibitors in 3D tissue culture and A375 MEK1 melanoma model is resistant to both MEK and BRAF inhibitors in 3D tissue culture. (<b>A</b>) BRAF inhibitor resistance in A375 RAS mutant melanoma models in 3D tissue culture. For each indicated cell type, 500 cells were seeded in each well of a ULA spheroid microplate and grown for three days in the absence of drug. After 72 h the indicated BRAF-specific inhibitor or the BRAF-nonspecific chemotherapeutic agent doxorubicin was added (Db = dabrafenib 25 nM, Vb = vemurafenib 50 nM, Dx = doxorubicin 100 nM) and the spheroids were grown for an additional three days. Spheroids were then stained with 2 μM Calcein AM green and NucBlue live-cell nuclear marker for 90 min and then imaged; (<b>B</b>) Average spheroid size of RAS mutant melanoma models following drug treatment relative to the un-drugged condition. Statistical analysis was performed using two-way ANOVA with multiple comparisons, each condition represents at least <span class="html-italic">n</span> = 3 spheroids; (<b>C</b>) MEK and BRAF inhibitor resistance in A375 MEKQ56P spheroid melanoma model. Drugged spheroids were handled as in (<b>A</b>) and treated with 25 nM dabrafenib, 50 nM vemurafenib, and 50 nM of the MEK inhibitors trametinib (Tb) and binimetinib (Bb), respectively, or 100 nM doxorubicin; (<b>D</b>) Average numbers of nuclei per spheroid were calculated relative to the un-drugged condition. Results from at least <span class="html-italic">n</span> = 3 spheroids for each condition were averaged and plotted, statistical analysis was performed using two-way ANOVA with multiple comparisons. ****, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>MEK1 Q56P melanoma model is sensitive to combination BRAF/MEK inhibitor treatment in both 2D and 3D tissue culture. (<b>A</b>) Dose-response curves for A375 WT cells in 2D tissue culture with dabrafenib (grey line), trametinib (black line), or a combination of dabrafenib and trametinib (red line, molarity indicates total drug concentration); (<b>B</b>) Dose-response curves for NRAS Q61K cells in 2D tissue culture with dabrafenib, trametinib, or a combination of dabrafenib and trametinib; (<b>C</b>) Dose-response curves for MEK1 Q56P cells in 2D tissue culture with dabrafenib, trametinib, or combination; (<b>D</b>) Dose-response curves for MEK1 Q56P cells in 2D tissue culture with dabrafenib, trametinib, or combination. Lower survival with combination indicates synergistic drug killing in this line; (<b>E</b>) Immunoblot demonstrating synergistic inhibition of the MEK/ERK signaling pathway in MEK1 Q56P melanoma model cells in 2D tissue culture. Cells were treated with either 1.0 μM of the indicated inhibitor compound or with 0.5 μM each of each indicated drug for 90 min prior to harvesting protein (Db = dabrafenib 1.0 μM, Vb = vemurafenib 1.0 μM, Tb = trametinib 1.0 μM, Bb = binimetinib 1.0 μM, Db/Tb = dabrafenib 0.5 μM + trametinib 0.5 μM); (<b>F</b>) Model of synergistic inhibition of the RAS/RAF/MEK/ERK pathway by combination MEK and BRAF inhibitor treatment. Orange star indicates the primary BRAF V600E mutation which drives cell proliferation in the absence of BRAF inhibitor. Treatment with BRAF inhibitor results in secondary MEK1 Q65P mutation (black outlined orange star). In the presence of these two mutations, upstream pathway inhibition with BRAF inhibitor in combination with downstream pathway inhibition with MEK inhibitor (red arrows) leads to less cell survival and proliferation than is observed when each drug is used alone; (<b>G</b>) Susceptibility of MEK1 Q56P cells grown in 3D tissue culture to dabrafenib, trametinib, and combination drug treatment. For each indicated cell type, 500 cells were seeded in each well of a ULA spheroid microplate and grown for three days in the absence of drug. The spheroids were then treated with 12 nM dabrafenib, 4 nM trametinib, a combination of 6 nM dabrafenib and 2 nM trametinib, or vehicle control for an additional three days. Spheroids were then stained with 2 μM Calcein AM green and NucBlue live-cell nuclear marker for 90 min and then imaged; (<b>H</b>) Spheroid sizes were calculated relative to the un-drugged condition. Results from at least <span class="html-italic">n</span> = 3 spheroids for each condition were averaged and plotted, statistical analysis was performed using two-way ANOVA with multiple comparisons. ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>PD-L1 is constitutively expressed in KRAS G13D, but not in A375 WT, NRAS Q61K, or MEK1 Q56P melanoma models. (<b>A</b>) Flow cytometry analysis of cell surface PD-L1 expression in A375 WT, NRAS Q61K, KRAS G13D, and MEK1 Q56P melanoma models grown in 2D tissue culture. Cells were treated overnight with 200 ng/µL interferon gamma (red), or mock treated (blue). The following day the cells were stained with either anti-PD-L1 or isotype control (grey); (<b>B</b>) PD-L1 immunoblot of total cellular protein from A375 WT, NRAS Q61K, and KRAS G13D cells grown in either 2D or 3D tissue culture; (<b>C</b>) Indirect immunofluorescence staining of PD-L1 in A375 WT, NRAS Q61K and KRAS G13D melanoma model lines.</p>
Full article ">
15 pages, 1168 KiB  
Review
Prostate Cancer Stem Cells: The Role of CD133
by Jianhui Yang, Omar Aljitawi and Peter Van Veldhuizen
Cancers 2022, 14(21), 5448; https://doi.org/10.3390/cancers14215448 - 5 Nov 2022
Cited by 17 | Viewed by 2859
Abstract
Prostate cancer stem cells (PCSCs), possessing self-renewal properties and resistance to anticancer treatment, are possibly the leading cause of distant metastasis and treatment failure in prostate cancer (PC). CD133 is one of the most well-known and valuable cell surface markers of cancer stem [...] Read more.
Prostate cancer stem cells (PCSCs), possessing self-renewal properties and resistance to anticancer treatment, are possibly the leading cause of distant metastasis and treatment failure in prostate cancer (PC). CD133 is one of the most well-known and valuable cell surface markers of cancer stem cells (CSCs) in many cancers, including PC. In this article, we focus on reviewing the role of CD133 in PCSC. Any other main stem cell biomarkers in PCSC reported from key publications, as well as about vital research progress of CD133 in CSCs of different cancers, will be selectively reviewed to help us inform the main topic. Full article
Show Figures

Figure 1

Figure 1
<p>PC cell lines can be de-differentiated to PCSCs or stem-like PC cells by chemotherapy, radiotherapy, serum starvation, sphere culture, and genomic manipulation.</p>
Full article ">Figure 2
<p>Summary of some known upstream and downstream molecular events related to CD133. The upstream events result in the upregulation of CD 133, and downstream events after CD133 induction are directly or indirectly related to increased stemness properties of PC cells, mainly via CD133/PI3K/AKT/Wnt/β-Catenin signaling axis.</p>
Full article ">Figure 3
<p>The proposed main mechanism of the role of CD133 in treatment resistance. The phosphorylated tyrosine 828 of CD133 mediates direct interaction with p85 of phosphoinositide 3-kinase (PI3K), followed by phosphorylated AK and translocation of NF-κB/p65. The latter enhances the transcription of Bcl-2, some members of the Inhibitors of Apoptosis Proteins (IAPs) family and multidrug resistance 1 (MDR-1).</p>
Full article ">
23 pages, 2358 KiB  
Article
Testicular Neoplasms: Primary Tumour Size Is Closely Interrelated with Histology, Clinical Staging, and Tumour Marker Expression Rates—A Comprehensive Statistical Analysis
by Klaus-Peter Dieckmann, Hendrik Isbarn, Francesca Grobelny, Cansu Dumlupinar, Julia Utschig, Christian Wülfing, Uwe Pichlmeier and Gazanfer Belge
Cancers 2022, 14(21), 5447; https://doi.org/10.3390/cancers14215447 - 5 Nov 2022
Cited by 12 | Viewed by 2359
Abstract
The role of primary tumour size (TS) in the clinical course of testicular tumours is incompletely understood. We retrospectively evaluated 641 consecutive patients with testicular neoplasms with regard to TS, histology, clinical stage (CS), serum tumour marker (STM) expression and patient age using [...] Read more.
The role of primary tumour size (TS) in the clinical course of testicular tumours is incompletely understood. We retrospectively evaluated 641 consecutive patients with testicular neoplasms with regard to TS, histology, clinical stage (CS), serum tumour marker (STM) expression and patient age using descriptive statistical methods. TS ≤ 10 mm was encountered in 13.6% of cases. Median TS of 10 mm, 30 mm, 35 mm, and 53 mm were found in benign tumours, seminomas, nonseminomas, and other malignant tumours, respectively. In cases with TS ≤ 10 mm, 50.6% had benign tumours. Upon receiver operating characteristics analysis, TS of > 16 mm revealed 81.5% sensitivity and 81.0% specificity for detecting malignancy. In subcentimeter germ cell tumours (GCTs), 97.7% of cases had CS1, and CS1 frequency dropped with increasing TS. Expression rates of all STMs significantly increased with TS. MicroRNA-371a-3p (M371) serum levels had higher expression rates than classical STMs, with a rate of 44.1% in subcentimeter GCTs. In all, TS is a biologically relevant factor owing to its significant associations with CS, STM expression rates and histology. Importantly, 50% of subcentimeter testicular neoplasms are of benign nature, and M371 outperforms the classical markers even in subcentimeter tumours. Full article
(This article belongs to the Section Cancer Biomarkers)
Show Figures

Figure 1

Figure 1
<p>Tumour sizes observed in the four histologic groups. Box and whisker plots showing the distribution of tumour sizes stratified by histologic subtypes of testicular neoplasms. The boxes display the first quartile, median and third quartile. The whiskers are defined as the largest or lowest observed value that falls within 1.5 times the interquartile range measured from Q3 or Q1, respectively. Area of box relates to sample size. □ outliers; + denotes arithmetic mean; SE seminoma; NS nonseminoma; BT benign tumours; OM other malignancies.</p>
Full article ">Figure 2
<p>Typical subcentimeter testicular neoplasm. Surgical specimen of a 6 mm sized benign Leydig cell tumour excised by testis sparing surgery.</p>
Full article ">Figure 3
<p>Proportions of histologic subgroups in testicular tumours sized &gt;10 mm and ≤10 mm.</p>
Full article ">Figure 4
<p>ROC analysis for predicting histology of a testicular neoplasm by its size.</p>
Full article ">Figure 5
<p>Probability curve for prediction of malignant histology of a testicular neoplasm. The logistic regression curve indicates the probability of a given tumour size to predict malignancy. Shadowed area represents 95% confidence intervals. Neoplasms with a size of ≤8 mm involve a 50% probability of malignancy, while tumour sizes of ≥25 mm, ≥33 mm, and ≥39 mm involve probabilities of 95%, 99%, and 100%, respectively.</p>
Full article ">Figure 6
<p>Tumour sizes in clinical stages in germ cell tumours. Box and whisker plots showing the distribution of tumour sizes stratified by clinical stages of testicular neoplasms. The boxes display the first quartile, median and third quartile. The whiskers are defined as the largest or lowest observed value that falls within 1.5 times the interquartile range measured from Q3 or Q1, respectively. Area of box denotes sample size. □ outliers; + denotes arithmetic mean; CS clinical stage.</p>
Full article ">Figure 7
<p>Expression rates of M371 and AFP and/or bHCG in germ cell tumours in relation to size of primary tumour in seminoma (<b>A</b>) and in nonseminoma (<b>B</b>). Blue columns denote expression rates of AFP and/or bHCG in five categories of tumour size, red columns indicate expression rates of M371. Overall, the expression rates of all tumour markers were higher in nonseminoma than in seminoma. All markers showed a significant trend towards lower expression rates with decreasing tumour size. M371 had higher expression rates than the other markers even in the smallest tumour size category. Error bars represent 95% CIs.</p>
Full article ">
18 pages, 5727 KiB  
Article
Water-Soluble Truncated Fatty Acid–Porphyrin Conjugates Provide Photo-Sensitizer Activity for Photodynamic Therapy in Malignant Mesothelioma
by Sam Bonsall, Simeon Hubbard, Uthaman Jithin, Joseph Anslow, Dylan Todd, Callum Rowding, Tom Filarowski, Greg Duly, Ryan Wilson, Jack Porter, Simon Turega and Sarah Haywood-Small
Cancers 2022, 14(21), 5446; https://doi.org/10.3390/cancers14215446 - 5 Nov 2022
Cited by 2 | Viewed by 2955
Abstract
Clinical trials evaluating intrapleural photodynamic therapy (PDT) are ongoing for mesothelioma. Several issues still hinder the development of PDT, such as those related to the inherent properties of photosensitizers. Herein, we report the synthesis, photophysical, and photobiological properties of three porphyrin-based photosensitizers conjugated [...] Read more.
Clinical trials evaluating intrapleural photodynamic therapy (PDT) are ongoing for mesothelioma. Several issues still hinder the development of PDT, such as those related to the inherent properties of photosensitizers. Herein, we report the synthesis, photophysical, and photobiological properties of three porphyrin-based photosensitizers conjugated to truncated fatty acids (C5SHU to C7SHU). Our photosensitizers exhibited excellent water solubility and high PDT efficiency in mesothelioma. As expected, absorption spectroscopy confirmed an increased aggregation as a consequence of extending the fatty acid chain length. In vitro PDT activity was studied using human mesothelioma cell lines (biphasic MSTO-211H cells and epithelioid NCI-H28 cells) alongside a non-malignant mesothelial cell line (MET-5A). The PDT effect of these photosensitizers was initially assessed using the colorimetric WST-8 cell viability assay and the mode of cell death was determined via flow cytometry of Annexin V-FITC/PI-stained cells. Photosensitizers appeared to selectively localize within the non-nuclear compartments of cells before exhibiting high phototoxicity. Both apoptosis and necrosis were induced at 24 and 48 h. As our pentanoic acid-derivatized porphyrin (C5SHU) induced the largest anti-tumor effect in this study, we put this forward as an anti-tumor drug candidate in PDT and photo-imaging diagnosis in mesothelioma. Full article
(This article belongs to the Special Issue Recent Research on Mesothelioma)
Show Figures

Figure 1

Figure 1
<p>Absorption spectra normalized of porphyrins C5SHU, C6SHU, and C7SHU spectra (<b>a</b>), (<b>b</b>), and (<b>c</b>), respectively, in different solvents made up from the 6 mM NaOH stock solutions. Solid black lines represent the spectra of the porphyrin in DMSO, black dotted line represent the spectra of the porphyrin in water and the black dashed line represents the spectra of the porphyrin in 1 M HCL.</p>
Full article ">Figure 2
<p>Example of C7SHU cellular localization. MSTO-211H cells were treated with 20 μg/mL PS C7SHU at a concentration of 10 μg/mL for 24 h. Images present are presented as a bright field, a Texas red channel and a composite image. Scale bar: 150 µm.</p>
Full article ">Figure 3
<p>Example of cellular localization. The MSTO-211H, NCI-H28, and Met-5a cell lines were treated with PS C5SHU, C6SHU and C7SHU at concentrations of 15 or 20 μg/mL dependent on EC<sub>50</sub> for 24 h. Images present are composite images of the bright field image and the Texas red channel. Scale bar: 150 µm.</p>
Full article ">Figure 4
<p>PS-induced cytotoxicity on MSTO-211H cells as determined by the WST-8 assay 24 and 48 h after PDT. Cells received C5SHU, C6SHU, and C7SHU treatments in dark and light conditions. Results are expressed as the mean ± SD of three independent experiments performed in triplicate. The varying significance of light treatments relative to the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001) and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 5
<p>PS-induced cytotoxicity on NCI-H28 cells as determined by the WST-8 assay 24 and 48 h after PDT. Cells received C5SHU, C6SHU, and C7SHU treatments in dark and light conditions. Results are expressed as the mean ± SD of three independent experiments performed in triplicate. The varying significance of light treatments relative to the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001), and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 6
<p>PS-induced cytotoxicity on Met-5a cells as determined by the WST-8 assay 24 and 48 h after PDT treatment. Cells received C5SHU, C6SHU and C7SHU treatments in dark and light conditions. Results are expressed as the mean ± SD of three independent experiments performed in triplicate. The varying significance of light treatments relative to the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001) and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 7
<p>Cell death analysis of C5SHU-treated MSTO-211H, NCI-H28, and Met-5a cells as determined by the Annexin V-FITC/PI assay 24 h after treatment. Results are expressed as the mean ± SD of three independent experiments. The varying significance of viable populations relative to that of the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 8
<p>Cell death analysis of C5SHU-treated MSTO-211H, NCI-H28, and Met-5a cells as determined by the Annexin V-FITC/PI assay 48 h after treatment. Results are expressed as the mean ± SD of three independent experiments. The varying significance of viable populations relative to that of the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001), and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 9
<p>Cell death analysis of C6SHU-treated MSTO-211H, NCI-H28, and Met-5a cells as determined by the Annexin V-FITC/PI assay 24 h after treatment. Results are expressed as the mean ± SD of three independent experiments. The varying significance of viable populations relative to that of the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001), and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 10
<p>Cell death analysis of C6SHU-treated MSTO-211H, NCI-H28, and Met-5a cells as determined by the Annexin V-FITC/PI assay 48 h after treatment. Results are expressed as the mean ± SD of three independent experiments. The varying significance of viable populations relative to that of the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001), and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 11
<p>Cell death analysis of C7SHU-treated MSTO-211H, NCI-H28, and Met-5a cells as determined by the Annexin V-FITC/PI assay 24 h after treatment. Results are expressed as the mean ± SD of three independent experiments. The varying significance of viable populations relative to that of the untreated control are indicated by * (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Figure 12
<p>Cell death analysis of C7SHU-treated MSTO-211H, NCI-H28, and Met-5a cells as determined by the Annexin V-FITC/PI assay 48 h after treatment. Results are expressed as the mean ± SD of three independent experiments. The varying significance of viable populations relative to that of the untreated control are indicated by ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001), and **** (<span class="html-italic">p</span> ≤ 0.0001).</p>
Full article ">Scheme 1
<p>Synthesis of porphyrin PS C5SHU, C6SHU, and C7SHU from porphyrin 1 through ether bond formation followed by the hydrolysis of ester intermediates 2, 3, and 4.</p>
Full article ">
14 pages, 820 KiB  
Article
Reliable Flow-Cytometric Approach for Minimal Residual Disease Monitoring in Patients with B-Cell Precursor Acute Lymphoblastic Leukemia after CD19-Targeted Therapy
by Ekaterina Mikhailova, Olga Illarionova, Alexander Komkov, Elena Zerkalenkova, Ilgar Mamedov, Larisa Shelikhova, Yulia Olshanskaya, Natalia Miakova, Galina Novichkova, Alexander Karachunskiy, Michael Maschan and Alexander Popov
Cancers 2022, 14(21), 5445; https://doi.org/10.3390/cancers14215445 - 5 Nov 2022
Cited by 10 | Viewed by 2961
Abstract
We aimed to develop an antibody panel and data analysis algorithm for multicolor flow cytometry (MFC), which is a reliable method for minimal residual disease (MRD) detection in patients with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) treated with CD19-directed therapy. The development of [...] Read more.
We aimed to develop an antibody panel and data analysis algorithm for multicolor flow cytometry (MFC), which is a reliable method for minimal residual disease (MRD) detection in patients with B-cell precursor acute lymphoblastic leukemia (BCP-ALL) treated with CD19-directed therapy. The development of the approach, which was adapted for the case of possible CD19 loss, was based on the additional B-lineage marker expression data obtained from a study of primary BCP-ALL patients, an analysis of the immunophenotypic changes that occur during blinatumomab or CAR-T therapy, and an analysis of very early CD19-negative normal BCPs. We have developed a single-tube 11-color panel for MFC-MRD detection. CD22- and iCD79a-based primary B-lineage gating (preferably consecutive) was recommended. Based on patterns of antigen expression changes and the relative expansion of normal CD19-negative BCPs, guidelines for MFC data analysis and interpretation were established. The suggested approach was tested in comparison with the molecular techniques: IG/TR gene rearrangement detection by next-generation sequencing (NGS) and RQ-PCR for fusion-gene transcripts (FGTs). Qualitative concordance rates of 82.8% and 89.8% were obtained for NGS-MRD and FGT-MRD results, respectively. We have developed a sensitive and reliable approach that allows MFC-MRD monitoring after CD19-directed treatment, even in the case of possible CD19 loss. Full article
(This article belongs to the Special Issue 2nd Edition: Minimal Residual Disease of Cancers)
Show Figures

Figure 1

Figure 1
<p>Analysis algorithm of BCP-ALL MRD detection after CD19-directed therapy.</p>
Full article ">
22 pages, 5079 KiB  
Review
Classic and New Markers in Diagnostics and Classification of Breast Cancer
by Roman Beňačka, Daniela Szabóová, Zuzana Guľašová, Zdenka Hertelyová and Jozef Radoňák
Cancers 2022, 14(21), 5444; https://doi.org/10.3390/cancers14215444 - 5 Nov 2022
Cited by 44 | Viewed by 9950
Abstract
Breast cancer remains the most frequently diagnosed form of female’s cancer, and in recent years it has become the most common cause of cancer death in women worldwide. Like many other tumours, breast cancer is a histologically and biologically heterogeneous disease. In recent [...] Read more.
Breast cancer remains the most frequently diagnosed form of female’s cancer, and in recent years it has become the most common cause of cancer death in women worldwide. Like many other tumours, breast cancer is a histologically and biologically heterogeneous disease. In recent years, considerable progress has been made in diagnosis, subtyping, and complex treatment of breast cancer with the aim of providing best suited tumour-specific personalized therapy. Traditional methods for breast cancer diagnosis include mammography, MRI, biopsy and histological analysis of tumour tissue in order to determine classical markers such as estrogen and progesterone receptors (ER, PR), cytokeratins (CK5/6, CK14, C19), proliferation index (Ki67) and human epidermal growth factor type 2 receptor (HER2). In recent years, these methods have been supplemented by modern molecular methodologies such as next-generation sequencing, microRNA, in situ hybridization, and RT-qPCR to identify novel molecular biomarkers. MicroRNAs (miR-10b, miR-125b, miR145, miR-21, miR-155, mir-30, let-7, miR-25-3p), altered DNA methylation and mutations of specific genes (p16, BRCA1, RASSF1A, APC, GSTP1), circular RNA (hsa_circ_0072309, hsa_circRNA_0001785), circulating DNA and tumour cells, altered levels of specific proteins (apolipoprotein C-I), lipids, gene polymorphisms or nanoparticle enhanced imaging, all these are promising diagnostic and prognostic tools to disclose any specific features from the multifaceted nature of breast cancer to prepare best suited individualized therapy. Full article
Show Figures

Figure 1

Figure 1
<p>BC. Left: main histopathological cancer types (red) together with other pathological findings. Middle: scheme of terminal ductal-lobular unit illustrating location of basal-myoepithelial cells (milk ejection) and luminal cells (milk production). Right: main targets of BC metastasis (frequency). Arrows indicate metastatic spread: violet-local, blue—via lymph, red—through blood. Data combined [<a href="#B19-cancers-14-05444" class="html-bibr">19</a>,<a href="#B21-cancers-14-05444" class="html-bibr">21</a>,<a href="#B26-cancers-14-05444" class="html-bibr">26</a>,<a href="#B38-cancers-14-05444" class="html-bibr">38</a>]. Certain items on right credited to Servier Medical Arts (CC-BY-3.0 licence; <a href="https://creativecommons.org/licenses/by/3.0/" target="_blank">https://creativecommons.org/licenses/by/3.0/</a>, accessed on 12 October 2022).</p>
Full article ">Figure 2
<p>Molecular subtypes of BC and preferred places they metastasize. Upper panel: molecular subtypes of BC. TN, triple negative tumour. ER and PR, oestrogen and progesterone receptor; EGF, epidermal growth factor receptor; HER2+, HER2+/neu/EGF-like growth factor receptor; Ki67, proliferation related antigen; p53, product of <span class="html-italic">TP53</span> tumour suppressor gene. Lower panel: most common organ and tissue sites of metastases of different types of tumours. Individual types are arranged in the same order as often they metastasize (in bold the rate exceeds 40%). Data combined [<a href="#B6-cancers-14-05444" class="html-bibr">6</a>,<a href="#B9-cancers-14-05444" class="html-bibr">9</a>,<a href="#B19-cancers-14-05444" class="html-bibr">19</a>,<a href="#B20-cancers-14-05444" class="html-bibr">20</a>,<a href="#B25-cancers-14-05444" class="html-bibr">25</a>,<a href="#B45-cancers-14-05444" class="html-bibr">45</a>]. Some picture items credited to Servier Medical Arts (CC-BY-3.0 licence; <a href="https://creativecommons.org/licenses/by/3.0/" target="_blank">https://creativecommons.org/licenses/by/3.0/</a>, accessed on 12 October 2022).</p>
Full article ">Figure 3
<p>BC candidates arranged according to functional categories. Data were collected from several sources [<a href="#B9-cancers-14-05444" class="html-bibr">9</a>,<a href="#B12-cancers-14-05444" class="html-bibr">12</a>,<a href="#B13-cancers-14-05444" class="html-bibr">13</a>,<a href="#B20-cancers-14-05444" class="html-bibr">20</a>,<a href="#B23-cancers-14-05444" class="html-bibr">23</a>,<a href="#B24-cancers-14-05444" class="html-bibr">24</a>,<a href="#B25-cancers-14-05444" class="html-bibr">25</a>,<a href="#B52-cancers-14-05444" class="html-bibr">52</a>,<a href="#B53-cancers-14-05444" class="html-bibr">53</a>].</p>
Full article ">Figure 4
<p>Algorithm of immunohistochemical and molecular genetic analysis of a tumour sample Combined [<a href="#B60-cancers-14-05444" class="html-bibr">60</a>].</p>
Full article ">Figure 5
<p>Multigene BC tests. Venn diagram is presenting numbers of genes/proteins evaluated by commercially available multigene tests. Abbreviations: ER(+) and ER(−) oestrogen receptor positive or negative cancers, respectively; PR(+) and PR(−), progesterone receptor positive or negative cancers; HER2(+) overexpression of HER2 oncoprotein; N(−) not present in lymph nodes, N(+) present in 1 to 3 lymph nodes [<a href="#B21-cancers-14-05444" class="html-bibr">21</a>].</p>
Full article ">
Previous Issue
Back to TopTop