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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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

Article Types

Countries / Regions

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

Search Results (42,827)

Search Parameters:
Keywords = oncology

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 497 KiB  
Review
Artificial Intelligence in Oncology
by Krzysztof Jeziorski and Robert Olszewski
Appl. Sci. 2025, 15(1), 269; https://doi.org/10.3390/app15010269 (registering DOI) - 30 Dec 2024
Abstract
The aim of the article is to highlight the key role of artificial intelligence in modern oncology. The search for scientific publications was carried out through the following web search engines: PubMed, PMC, Web of Science, Scopus, Embase and Ebsco. Artificial intelligence plays [...] Read more.
The aim of the article is to highlight the key role of artificial intelligence in modern oncology. The search for scientific publications was carried out through the following web search engines: PubMed, PMC, Web of Science, Scopus, Embase and Ebsco. Artificial intelligence plays a special role in oncology and is considered to be the future of oncology. The largest application of artificial intelligence in oncology is in diagnostics (more than 80%), particularly in radiology and pathology. This can help oncologists not only detect cancer at an early stage but also forecast the possible development of the disease by using predictive models. Artificial intelligence plays a special role in clinical trials. AI makes it possible to accelerate the discovery and development of new drugs, even if not necessarily successfully. This is done by detecting new molecules. Artificial intelligence enables patient recruitment by combining diverse demographic and medical patient data to match the requirements of a given research protocol. This can be done by reducing population heterogeneity, or by prognostic and predictive enrichment. The effectiveness of artificial intelligence in oncology depends on the continuous learning of the system based on large amounts of new data but the development of artificial intelligence also requires the resolution of some ethical and legal issues. Full article
(This article belongs to the Section Biomedical Engineering)
Show Figures

Figure 1

Figure 1
<p>The relationship between artificial intelligence, machine learning and deep learning according to Shimizu and Nakayama [<a href="#B7-applsci-15-00269" class="html-bibr">7</a>].</p>
Full article ">
13 pages, 2106 KiB  
Article
The Efficacy of FOLFIRI Plus Ramucirumab in Recurrent Colorectal Cancer Refractory to Adjuvant Chemotherapy with Oxaliplatin/Fluoropyrimidine—Including Biomarker Analyses
by Naotoshi Sugimoto, Shingo Noura, Takeshi Kato, Shinichi Yoshioka, Taishi Hata, Atsushi Naito, Mitsuyoshi Tei, Hiroshi Tamagawa, Takamichi Komori, Yoshihito Ide, Takayuki Fukuzaki, Katsuki Danno, Genta Sawada, Yoshinori Kagawa, Toshio Shimokawa, Norikatsu Miyoshi, Takayuki Ogino, Mamoru Uemura, Hirofumi Yamamoto, Kohei Murata, Yuichiro Doki and Hidetoshi Eguchiadd Show full author list remove Hide full author list
Cancers 2025, 17(1), 91; https://doi.org/10.3390/cancers17010091 (registering DOI) - 30 Dec 2024
Abstract
Background: FOLFIRI (5-FU + leucovorin + irinotecan) plus ramucirumab is one of the standards in second-line metastatic colorectal cancer (CRC) patients progressing after treatment with oxaliplatin/fluoropyrimidine with bevacizumab, but there is no evidence on its efficacy without prior bevacizumab. Moreover, VEGF-D has not [...] Read more.
Background: FOLFIRI (5-FU + leucovorin + irinotecan) plus ramucirumab is one of the standards in second-line metastatic colorectal cancer (CRC) patients progressing after treatment with oxaliplatin/fluoropyrimidine with bevacizumab, but there is no evidence on its efficacy without prior bevacizumab. Moreover, VEGF-D has not been confirmed as a predictive biomarker for ramucirumab’s efficacy, either. Methods: The RAINCLOUD study was a multicenter, single-arm, phase II trial conducted in Japan. Patients with recurrent CRC pretreated with fluoropyrimidine and oxaliplatin without bevacizumab were analyzed. The primary endpoint was progression-free survival (PFS). The secondary endpoints measured were overall survival (OS), overall response rate (ORR), and safety. Results: A total of 48 patients were enrolled from 15 sites between September 2017 and September 2020. Their median age was 63.5 years (25~77), 20.1% had a right-sided tumor, and 68.8% had RAS-mutant cancer. The median PFS was 8.9 months (90% CI: 6.3–11.8), so the primary endpoint was met. Their median OS and ORR were 22.3 months (95% CI: 17.4-NA) and 41.7% (95% CI: 4.9–7.6), respectively. An incidence of grade 3/4 adverse events that reached over 5% applied to neutropenia (44%), leucopenia (10%), and hypertension (8%). In the biomarker analysis, the serum VEGF-D levels post-treatment were higher than those pre-treatment, but the PFS in those with high VEGF-D levels trended towards being worse than that in those with low VEGF-D (7.6M/5.6M (p = 0.095; HR: 0.56)). Instead, those with low TSP-2 had a better PFS than those with high TSP-2 (7.5M/4.3M (p = 0.022; HR: 0.45)). Conclusions: Our data suggested that FOLFIRI plus ramucirumab was effective and tolerable for CRC refractory to fluoropyrimidine and oxaliplatin without anti-angiogenesis. Serum VEGF-D levels may not be predictive but TSP-2 may be a potential prognostic biomarker for ramucirumab’s efficacy. Full article
(This article belongs to the Section Cancer Drug Development)
Show Figures

Figure 1

Figure 1
<p>CONSORT flowchart showing the disposition of the enrolled patients at the time of the data design.</p>
Full article ">Figure 2
<p>Kaplan–Meier survival analysis of (<b>a</b>) PFS, progression-free survival, and (<b>b</b>) OS, overall survival; CI, confidence interval.</p>
Full article ">Figure 3
<p>Dynamics of angiogenesis factors. PlGF: placental growth factor; VEGF-A: vascular endothelial growth factor A; VEGF-D: vascular endothelial growth factor D; TSP-2: Thrombospondin-2.</p>
Full article ">Figure 4
<p>Progression-free survival according to angiogenesis factors pre-treatment (high = red; low = green).</p>
Full article ">Figure 5
<p>Overall survival according to angiogenesis factors pre-treatment (high = red; low = green).</p>
Full article ">
14 pages, 415 KiB  
Article
Debriefing Methodologies in Nursing Simulation: An Exploratory Study of the Italian Settings
by Sonia Lomuscio, Emanuele Capogna, Stefano Sironi, Marco Sguanci, Sara Morales Palomares, Giovanni Cangelosi, Gaetano Ferrara, Stefano Mancin, Antonio Amodeo, Anne Destrebecq, Mauro Parozzi and Susy Dal Bello
Nurs. Rep. 2025, 15(1), 7; https://doi.org/10.3390/nursrep15010007 (registering DOI) - 30 Dec 2024
Abstract
Background: As part of simulation-based learning, it is well known that debriefing plays a crucial role; ineffective debriefing can lead to a reiteration of errors in decision-making and a poor understanding of one’s limitations, compromising the learner’s psychological safety and making future simulated [...] Read more.
Background: As part of simulation-based learning, it is well known that debriefing plays a crucial role; ineffective debriefing can lead to a reiteration of errors in decision-making and a poor understanding of one’s limitations, compromising the learner’s psychological safety and making future simulated learning experiences less effective. In Italy, although simulation has been used in nursing education for more than 20 years, there is a general lack of data regarding the elements of debriefing. Methods: An exploratory, cross-sectional, multicenter nationwide study was conducted to identify current debriefing practices in Italian simulation-based nursing education. A non-probability sample of all directors of the Italian Bachelor school of Nursing and the directors of simulation centers on Italian national territory was surveyed with an online questionnaire. Results: Fifty-four nursing degree programs and 11 simulation centers participated in the survey. Significant differences were found between debriefing practices used by simulation centers and those used by the Bachelor School of Nursing. Specifically, differences concerned the training of debriefers, the knowledge of a debriefing framework, the use of different rooms for debriefing and the time spent on this activity. Conclusions: There is an emerging need for a harmonization process in Italian nursing education debriefing practices that would align the current reality with the best practices of the literature. Full article
22 pages, 1059 KiB  
Article
Factors Influencing the Duration of Maintenance Therapy in Metastatic Colorectal Cancer
by Théo Fourrier, Caroline Truntzer, Morgane Peroz, Valentin Derangère, Julie Vincent, Leila Bengrine-Lefèvre, Audrey Hennequin, Rémi Palmier, David Orry, Thomas Rabel and François Ghiringhelli
Cancers 2025, 17(1), 88; https://doi.org/10.3390/cancers17010088 (registering DOI) - 30 Dec 2024
Abstract
Background/Objectives: Metastatic colorectal cancer (mCRC) is mainly treated with 5-Fluoro-Uracil (5-FU), Oxaliplatin and Irinotecan chemotherapies and anti-Epidermal Growth Factor Receptor (EGFR) or anti-Vascular Endothelial Growth Factor (VEGF) targeted therapies. Due to chemotherapy-related toxicity, patients receive induction treatment to achieve tumour response followed by [...] Read more.
Background/Objectives: Metastatic colorectal cancer (mCRC) is mainly treated with 5-Fluoro-Uracil (5-FU), Oxaliplatin and Irinotecan chemotherapies and anti-Epidermal Growth Factor Receptor (EGFR) or anti-Vascular Endothelial Growth Factor (VEGF) targeted therapies. Due to chemotherapy-related toxicity, patients receive induction treatment to achieve tumour response followed by maintenance therapy with less cytotoxic molecules or a chemotherapy-free interval to reduce chemotherapy-related toxicity. In this study, the aim was to determine the patient, cancer and treatment factors that influence the duration of maintenance therapy (DMT). Methods: We collected retrospective data on a cohort of 133 patients treated at the Centre Georges François Leclerc (CGFL) cancer centre in Dijon between March 2014 and June 2022. Patients had unresectable or potentially resectable diseases. They received first-line induction treatment with chemotherapy and/or targeted therapy and maintenance treatment, defined as the interruption of at least one chemotherapy agent. Results: In the multivariate analysis, age (HR: 1.02, 95% CI 1.00–1.04, p = 0.031), N2 nodal status (HR: 1.78, 95% CI 1.09–2.89, p = 0.021) and the presence of peritoneal metastases (HR:2.05, 95% CI 1.25–3.36, p = 0.004), as well as baseline carcino-embryonic antigen (CEA) level (HR:1.10, 95% CI 1.00–1.20, p = 0.052), were significantly associated to poor DMT. Local treatment of liver metastases also significantly reduced the DMT (HR:0.49, 95% CI 0.28–0.86, p = 0.013). In our cohort, induction triplet chemotherapy significantly increased the CEA delta (70% vs. 44%, p = 0.047) compared to doublet chemotherapy and led to a higher rate of liver surgery (40% vs. 21%, p =0.014) and a trend for a higher rate of local treatment of metastases (62% vs. 45%, p = 0.059). Conclusions: Duration of maintenance therapy is determined by the initial patient and colorectal cancer characteristics. However, it is significantly increased by local treatment of liver metastases. By reducing the tumour burden, a triplet induction chemotherapy regimen increases the rate of liver metastase resection. Full article
(This article belongs to the Special Issue Contemporary Treatment of Colorectal Cancer)
16 pages, 1164 KiB  
Review
Effect of Gut Dysbiosis on Onset of GI Cancer
by Seema Kumari, Mundla Srilatha and Ganji Purnachandra Nagaraju
Cancers 2025, 17(1), 90; https://doi.org/10.3390/cancers17010090 (registering DOI) - 30 Dec 2024
Abstract
Dysbiosis in the gut microbiota plays a significant role in GI cancer development by influencing immune function and disrupting metabolic functions. Dysbiosis can drive carcinogenesis through pathways like immune dysregulation and the release of carcinogenic metabolites, and altered metabolism, genetic instability, and pro-inflammatory [...] Read more.
Dysbiosis in the gut microbiota plays a significant role in GI cancer development by influencing immune function and disrupting metabolic functions. Dysbiosis can drive carcinogenesis through pathways like immune dysregulation and the release of carcinogenic metabolites, and altered metabolism, genetic instability, and pro-inflammatory signalling, contributing to GI cancer initiation and progression. Helicobacter pylori infection and genotoxins released from dysbiosis, lifestyle and dietary habits are other factors that contribute to GI cancer development. Emerging diagnostic and therapeutic approaches show promise in colorectal cancer treatment, including the multitarget faecal immunochemical test (mtFIT), standard FIT, and faecal microbiota transplantation (FMT) combined with PD-1 inhibitors. We used search engine databases like PubMed, Scopus, and Web of Science. This review discusses the role of dysbiosis in GI cancer onset and explores strategies such as FMT, probiotics, and prebiotics to enhance the immune response and improve cancer therapy outcomes. Full article
23 pages, 557 KiB  
Article
Reproducing the NIRS-QST Clinical Dose Calculations for Carbon Ion Radiotherapy Using Microdosimetric Probability Density Distributions
by Alessio Parisi, Keith M. Furutani, Shannon Hartzell and Chris J. Beltran
Radiation 2025, 5(1), 2; https://doi.org/10.3390/radiation5010002 (registering DOI) - 30 Dec 2024
Abstract
Ion radiotherapy requires accurate relative biological effectiveness (RBE) calculations to account for the markedly different biological effects of ions compared to photons. Microdosimetric RBE models rely on descriptions of the energy deposition at the microscopic scale, either through radial dose distributions (RDDs) or [...] Read more.
Ion radiotherapy requires accurate relative biological effectiveness (RBE) calculations to account for the markedly different biological effects of ions compared to photons. Microdosimetric RBE models rely on descriptions of the energy deposition at the microscopic scale, either through radial dose distributions (RDDs) or microdosimetric probability density distributions. While RDD approaches focus on the theoretical description of the energy deposition around the ion track, microdosimetric distributions offer the advantage of being experimentally measurable, which is crucial for quality assurance programs. As the results of microdosimetric RBE models depend on whether RDD or microdosimetric distributions are used, the model parameters are not interchangeable between these approaches. This study presents and validates a method to reproduce the published reference biological and clinical dose calculations at NIRS-QST for only carbon ion radiotherapy by using the modified microdosimetric kinetic model (MKM) alongside microdosimetric distributions instead of the reference RDD approach. To achieve this, Monte Carlo simulations were performed to estimate the variation of the radiation quality within and outside the field of pristine and spread-out Bragg peaks. By appropriately optimizing the modified MKM parameters for microdosimetric distributions assessed within water spheres, we successfully reproduced the results of calculations using the reference NIRS-QST RDD, generally within 2%. Full article
13 pages, 461 KiB  
Review
Self-Expandable Metal Stents for Obstructing Colon Cancer and Extracolonic Cancer: A Review of Latest Evidence
by Pedro Marílio Cardoso and Eduardo Rodrigues-Pinto
Cancers 2025, 17(1), 87; https://doi.org/10.3390/cancers17010087 (registering DOI) - 30 Dec 2024
Abstract
Colorectal cancer (CRC) is a leading cause of cancer mortality, with many patients presenting with malignant colorectal obstruction (MCO). Self-expandable metal stents (SEMSs) have emerged as a minimally invasive key intervention, both as a bridge to surgery (BTS) in curative setting sand for [...] Read more.
Colorectal cancer (CRC) is a leading cause of cancer mortality, with many patients presenting with malignant colorectal obstruction (MCO). Self-expandable metal stents (SEMSs) have emerged as a minimally invasive key intervention, both as a bridge to surgery (BTS) in curative setting sand for palliation in advanced disease. This review aims to provide an evidence-based analysis of SEMS indications, contraindications, and efficacy across curative and palliative contexts, with focus on long-term outcomes. Based on data from recent trials and guidelines, we examine SEMS placement outcomes, focusing on specific scenarios, including BTS for left-sided MCO, chemotherapy (with angiogenic agents) safety during stent therapy, the optimal timing between SEMS placement and surgery, and oncological outcomes. We also discuss the use of SEMSs in challenging contexts such as proximal colon obstruction and extracolonic obstruction, and the relevant technical considerations. Findings indicate that using a SEMS in the BTS setting reduces emergency surgery needs, minimizes complications, and decreases stoma formation. Long-term oncologic outcomes, particularly recurrence, are still debated, but recent evidence shows that SEMS placement is safe, without worsening long term outcomes. Palliative SEMS placement shows high efficacy in symptom relief with manageable adverse events. Success depends on patient selection and technical expertise, with multidisciplinary approaches essential for optimal outcomes. Full article
(This article belongs to the Special Issue Endoscopic Advances in Gastrointestinal Oncology)
11 pages, 255 KiB  
Article
RadiOnCOVID: Multicentric Analysis of the Impact of COVID-19 on Patients Undergoing Radiation Therapy in Italy
by Andrea Emanuele Guerini, Giulia Marvaso, Sandro Tonoli, Giulia Corrao, Maria Ausilia Teriaca, Matteo Sepulcri, Melissa Scricciolo, Alessandro Gava, Sabrina Montrone, Niccolò Giaj-Levra, Barbara Noris Chiorda, Giovanna Mantello, Francesco Fiorica, Simona Borghesi, Liliana Belgioia, Angela Caroli, Alba Fiorentino, Radioncovid Study Group, Barbara Alicja Jereczek-Fossa, Stefano Maria Magrini and Michela Buglioneadd Show full author list remove Hide full author list
Cancers 2025, 17(1), 86; https://doi.org/10.3390/cancers17010086 (registering DOI) - 30 Dec 2024
Abstract
Methods and materials: Patients with ongoing or planned anticancer treatment at 19 Italian Radiation Oncology centers were included in the study retrospectively from 3 February 2020 to 31 December 2020 and prospectively from 1 January 2021 to 31 May 2021. Anonymized data were [...] Read more.
Methods and materials: Patients with ongoing or planned anticancer treatment at 19 Italian Radiation Oncology centers were included in the study retrospectively from 3 February 2020 to 31 December 2020 and prospectively from 1 January 2021 to 31 May 2021. Anonymized data were processed through a specific website and database. Antineoplastic treatment characteristics and timing and outcomes of COVID-19 and its impact on radiotherapy or systemic therapy were described. Results: The retrieved cohort included 41,039 patients that received treatment or were planned for therapy in the study period. Overall, 123 patients had a confirmed COVID-19 diagnosis during antineoplastic treatment (group A) and 99 patients before treatment start (group B). The incidence of COVID-19 across the whole cohort in the index period was 0.54% (groups A + B) and 0.30% considering only group A. A total of 60 patients developed severe COVID-19, and a total of 45 patients died as a consequence of the infection (incidence of 0.15% and 0.11%, respectively). Nonetheless, mortality among COVID-19 patients was high, with an attributable death rate after confirmed infection of 20.27%. Among the 123 patients in group A, 37.4% required temporary treatment suspension, 32.5% definitive suspension and 37 patients continued treatment while positive. As for the 99 patients in group B, 53.5% experienced temporary delay, 20.2% experienced definitive treatment suspension and 26.3% had no delay. Conclusions: Most of the patients with a COVID-19 diagnosis in our cohort recovered and completed their treatment; nonetheless, the attributable death rate after confirmed infection was 20.27%, and mortality was high among cancer patients with severe COVID-19 presentation. The global incidence of death due to COVID-19 or severe COVID-19 was low and decreased over time. Radiation oncology activity could be safely continued during the COVID-19 pandemic with the adoption of adequate preventive measures. Full article
(This article belongs to the Section Infectious Agents and Cancer)
10 pages, 2008 KiB  
Article
High Incidence of Isolated Tumor Cells in Sentinel Node Biopsies of Thin Melanomas: A Potential Factor in the Paradoxical Prognosis of Stage IIIA Cutaneous Melanoma?
by Andrea Ronchi, Giuseppe D’Abbronzo, Emma Carraturo, Giuseppe Argenziano, Gabriella Brancaccio, Camila Scharf, Elvira Moscarella, Teresa Troiani, Francesco Iovino, Salvatore Tolone, Mario Faenza, Gerardo Cazzato and Renato Franco
Diagnostics 2025, 15(1), 69; https://doi.org/10.3390/diagnostics15010069 (registering DOI) - 30 Dec 2024
Abstract
Background/Objectives: This study aims to evaluate whether the presence of isolated tumor cells (ITCs) correlates with specific stages of cutaneous melanoma, potentially shedding light on their prognostic significance and the paradoxical survival outcomes in stage IIIA. Methods: This study analyzed cases [...] Read more.
Background/Objectives: This study aims to evaluate whether the presence of isolated tumor cells (ITCs) correlates with specific stages of cutaneous melanoma, potentially shedding light on their prognostic significance and the paradoxical survival outcomes in stage IIIA. Methods: This study analyzed cases of sentinel lymph node biopsies for cutaneous melanoma between 2021 and 2023. It included patients with CM diagnoses, available histological slides, and clinical information about the neoplasia stage. The correlation between the primary tumor stage and the presence of isolated tumor cells was statistically analyzed. Results: This study analyzed 462 sentinel lymph node biopsies, revealing 77.1% negative cases and 22.9% positive cases. Isolated tumor cells were observed in 24 cases (5.2%), most commonly in the early stages (e.g., pT1b and pT2a). Statistical analysis confirmed a significant correlation between ITC presence and early-stage neoplasms (p = 0.014). Conclusions: Although ITCs prompt upstaging, their prognostic impact appears limited, especially in thin melanomas, where survival aligns more closely with stage IB than stage IIIA. This aligns with findings from breast cancer studies where ITCs are not equated to metastases in staging due to their minimal impact on prognosis. Current melanoma staging practices could benefit from differentiating ITCs from larger metastatic deposits to better reflect the actual metastatic burden and guide treatment decisions. Full article
(This article belongs to the Special Issue Latest Advances in Diagnosis and Management of Skin Cancer)
13 pages, 1928 KiB  
Article
Non-Optic Glioma-like Lesions in Adult Neurofibromatosis Type 1 Patients
by Walter Taal, Bart Zick, Bart J. Emmer and Martin J. van den Bent
Diagnostics 2025, 15(1), 67; https://doi.org/10.3390/diagnostics15010067 (registering DOI) - 30 Dec 2024
Abstract
Purpose: Physicians face clinical dilemmas in the diagnosis of non-optic intraparenchymal lesions on MRI brain scans of patients with neurofibromatosis type 1. As the incidence and evolution of these lesions into adulthood remain unclear, we conducted a retrospective study on this topic. Methods: [...] Read more.
Purpose: Physicians face clinical dilemmas in the diagnosis of non-optic intraparenchymal lesions on MRI brain scans of patients with neurofibromatosis type 1. As the incidence and evolution of these lesions into adulthood remain unclear, we conducted a retrospective study on this topic. Methods: All adult neurofibromatosis type 1 patients who had at least one MRI brain scan in our center were selected for this study. Brain lesions with contrast enhancement after gadolinium administration and/or mass effect were named “glioma-like lesions”. Results: In our cohort of 396 adult neurofibromatosis type 1 patients, 182 had at least one MRI scan of the brain. A total of 48 glioma-like lesions were found in 38/182 patients. The majority of glioma-like lesions remained stable, decreased in size or even disappeared during a median follow-up time of 8.5 years. Twelve glioma-like lesions in 11/182 patients were resected or biopsied, and histology showed gliomas of astrocytic origin (WHO grade 1–4). Conclusions: It was concluded from these data that asymptomatic glioma-like lesions on MRI brain scans in neurofibromatosis type 1 patients, either with contrast enhancement and/or mass effect, had an indolent nature. Mildly symptomatic or asymptomatic patients can therefore be followed without invasive diagnostic and therapeutic procedures. Full article
(This article belongs to the Special Issue Pathology and Diagnosis of Neurological Disorders)
Show Figures

Figure 1

Figure 1
<p>The evolution of asymptomatic glioma-like lesions in NF1 patients. A total of 48 asymptomatic glioma-like lesions, defined as MRI lesions exhibiting clear mass effect and/or contrast enhancement, were found in 38 out of 182 adult NF1 patients, with at least 1 MRI scan of the brain after the age of 18 years old. The size of every lesion is measured on axial T2-weighted MRI brain images, and the product of the largest diameter and the perpendicular diameter is plotted against the patient’s age. Lines with the same color represent lesions within the same patient. Solid lines present enhancing lesions. Dotted lines represent non-enhancing lesions, but the open balls in these dotted lines represent temporary contrast enhancement of the same lesion. The 3 R’s mean that the lesions have been resected because of fast radiological progression (see <a href="#diagnostics-15-00067-t004" class="html-table">Table 4</a>; patient 1–3).</p>
Full article ">Figure 2
<p><b>A non-enhancing space-occupying glioma-like lesion (GLL).</b> (<b>a</b>) A GLL in the left cerebellar peduncle in an 8-year-old neurofibromatosis type 1 patient. The MRI was performed because of a mild ataxia of the left extremities. The MRI shows a space-occupying lesion with high signal on the T2-weighted images (the yellow arrowheads point to the diffuse hyperintense lesion, while the blue arrowhead points to compressions of the 4th ventricle from the left side). The lesion in the cerebellar peduncle was regarded as a low-grade glioma. The lesion was followed up frequently and no treatment was given. The patient’s symptoms improved within years, as the lesion and its space-occupying appearance were also disappearing. (<b>b</b>) MRI of the same patient at the age of 22 years old shows a significant decrease in the T2 lesion and its mass effect.</p>
Full article ">Figure 3
<p>An enhancing glioma-like lesion. (<b>a</b>) The brain MRI shows a lesion in in the left frontal cerebral lobe of a 26-year-old neurofibromatosis type 1 patient. The MRI scan was performed because of screening purposes and the patient did not have neurological signs or symptoms. The lesion shows some mass effect and clear contrast enhancement. This lesion was characterized as a low-grade glioma (most probably a pilocytic astrocytoma). (<b>b</b>) The brain MRI at the age of 30 years shows a reduction in size of the enhancing part of the lesion, without any intervention.</p>
Full article ">Figure 4
<p><b>Diagnostic flowchart for glioma-like lesions.</b> Focal areas of signal intensity (FASIs) appear hyperintense on T2-weighted MRI and do not exhibit space-occupying characteristics or enhancement after intravenous contrast administration. FASIs are typically asymptomatic and tend to decrease in number and size with advancing age. Consequently, there is no requirement for repeat brain MRI in cases of FASIs.</p>
Full article ">
37 pages, 12904 KiB  
Review
Targeting Invasion: The Role of MMP-2 and MMP-9 Inhibition in Colorectal Cancer Therapy
by Alireza Shoari, Arghavan Ashja Ardalan, Alexandra M. Dimesa and Mathew A. Coban
Biomolecules 2025, 15(1), 35; https://doi.org/10.3390/biom15010035 (registering DOI) - 30 Dec 2024
Abstract
Colorectal cancer (CRC) remains one of the most prevalent and lethal cancers worldwide, prompting ongoing research into innovative therapeutic strategies. This review aims to systematically evaluate the role of gelatinases, specifically MMP-2 and MMP-9, as therapeutic targets in CRC, providing a critical analysis [...] Read more.
Colorectal cancer (CRC) remains one of the most prevalent and lethal cancers worldwide, prompting ongoing research into innovative therapeutic strategies. This review aims to systematically evaluate the role of gelatinases, specifically MMP-2 and MMP-9, as therapeutic targets in CRC, providing a critical analysis of their potential to improve patient outcomes. Gelatinases, specifically MMP-2 and MMP-9, play critical roles in the processes of tumor growth, invasion, and metastasis. Their expression and activity are significantly elevated in CRC, correlating with poor prognosis and lower survival rates. This review provides a comprehensive overview of the pathophysiological roles of gelatinases in CRC, highlighting their contribution to tumor microenvironment modulation, angiogenesis, and the metastatic cascade. We also critically evaluate recent advancements in the development of gelatinase inhibitors, including small molecule inhibitors, natural compounds, and novel therapeutic approaches like gene silencing techniques. Challenges such as nonspecificity, adverse side effects, and resistance mechanisms are discussed. We explore the potential of gelatinase inhibition in combination therapies, particularly with conventional chemotherapy and emerging targeted treatments, to enhance therapeutic efficacy and overcome resistance. The novelty of this review lies in its integration of recent findings on diverse inhibition strategies with insights into their clinical relevance, offering a roadmap for future research. By addressing the limitations of current approaches and proposing novel strategies, this review underscores the potential of gelatinase inhibitors in CRC prevention and therapy, inspiring further exploration in this promising area of oncological treatment. Full article
Show Figures

Figure 1

Figure 1
<p>The multidomain structure of MMP-9. The following domains are shown: the signal peptide (purple), the propeptide (green), the catalytic domain (yellow), the three fibronectin repeats (blue), the metal binding site (orange), the OG domain (brown), and the PEX domain (red).</p>
Full article ">Figure 2
<p>The image illustrates the transition from benign growths to malignant cell invasion in colorectal cancer, highlighting the abnormal cell growth and the presence of matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9, that facilitate cancer cell invasion. (Illustration created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 3
<p>The image illustrates the comprehensive approaches for gelatinase inhibition in colorectal cancer therapy, encompassing natural sources, small chemical molecules, proteins, peptides, and microRNA-based strategies. (Illustration created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 4
<p>Schematic representation of the underlying process by which CPT inhibits CT26 colon cancer. CPT suppresses the production of MMP-2 and MMP-9 and enhances the production of TIMP-1 and TIMP-2, resulting in the prevention of tumor invasion by CPT. The CPT suppresses the expression of inflammatory factors, hence reducing inflammation and inhibiting tumor inflammation. In addition, the PI3K/Akt/mTOR pathway is crucial for tumor growth. CPT effectively hinders the phosphorylation of PI3K, Akt, and mTOR, hence suppressing tumor growth. These combined effects contribute to the anti-tumor impact of CPT. Adapted with permission from [<a href="#B63-biomolecules-15-00035" class="html-bibr">63</a>], Elsevier, 2018.</p>
Full article ">Figure 5
<p>Structure of the human MMP-9-specific pyrrole-imidazole (PI) polyamide. Adapted with permission from [<a href="#B115-biomolecules-15-00035" class="html-bibr">115</a>], Wiley, 2010.</p>
Full article ">Figure 6
<p>(<b>A</b>) 3D binding mode of 8 (yellow sticks), (<b>B</b>) 2D binding mode of 8, (<b>C</b>) 3D binding mode of 16 (magenta sticks), (<b>D</b>) 2D binding mode of 16 in the catalytic domain of MMP-9 (PDB ID: 1GKC). Adapted with permission from [<a href="#B126-biomolecules-15-00035" class="html-bibr">126</a>], Elsevier, 2021.</p>
Full article ">Figure 7
<p>miR-124 specifically targets PD-L1 and STAT3. The STAT3 and PD-L1 pathways have a significant impact on the progression of colorectal cancer and the immunological response of T cells. Thus, by modifying downstream effectors like MMP-9, it is possible to limit CRC carcinogenesis through the regulation of PD-L1 and STAT3 expression and activity. Adapted with permission from [<a href="#B149-biomolecules-15-00035" class="html-bibr">149</a>], Wiley, 2010.</p>
Full article ">
16 pages, 231 KiB  
Article
Patient–Nurse Communication in an Oncology Hospital Setting: A Qualitative Study
by Laura Iacorossi, Giovanna D’Antonio, Maria Condoleo, Lara Guariglia, Fabrizio Petrone, Simona Molinaro and Anita Caruso
Healthcare 2025, 13(1), 50; https://doi.org/10.3390/healthcare13010050 (registering DOI) - 30 Dec 2024
Abstract
Background: Communication is an important aspect in making patients competent to define, process, and manage their disease condition as well as to intercept and satisfy psychosocial needs. Communication between patient and nurse is central to the learning and orientating process since the nurse [...] Read more.
Background: Communication is an important aspect in making patients competent to define, process, and manage their disease condition as well as to intercept and satisfy psychosocial needs. Communication between patient and nurse is central to the learning and orientating process since the nurse has the greatest frequency and continuity of relationship with patients and their families. This study aims to investigate the quality of communication between patient and nurse and the factors that promote or hinder effective communication from the oncology patient’s perspective within an inpatient hospital setting. Methods: A descriptive qualitative study was conducted with one-to-one semi-structured interviews analyzed using the Framework Analysis methodology. The population consisted of oncologic patients admitted to the Medical Oncology Units of the Regina Elena National Cancer Institute in Rome. Data were analyzed using Ritchie and Spencer’s Framework Analysis. Results: The sample comprised 20 patients, with an average age of 61.35, admitted to the Medical Oncology Units of the Regina Elena National Cancer Institute in Rome. Three themes emerged: positive communication as an element of care, factors fostering communication, and factors hindering communication. Conclusions: The sample interviewed deems the quality of communication satisfactory. Familiar communication style, direct language, and caring are factors fostering communication. In contrast, lack of communication between medical and nursing staff, shortage of staff, and lack of time are considered communication barriers. Advanced communication competencies in nurses are crucial for effectively addressing the emotional and psychosocial needs of cancer patients, fostering a more empathetic and supportive care environment. Full article
(This article belongs to the Special Issue Nursing Competencies: New Advances in Nursing Care)
12 pages, 787 KiB  
Article
Implementation of a Hepatitis B Screening Program in Patients Receiving Systemic Anti-Cancer Therapy
by Jennifer Leigh, Ranjeeta Mallick, Stephanie Brule, Lisa Rambout, Jennifer Newton, Dominick Bossé, Curtis Cooper and Joanna Gotfrit
Curr. Oncol. 2025, 32(1), 20; https://doi.org/10.3390/curroncol32010020 (registering DOI) - 30 Dec 2024
Abstract
Cancer patients receiving non-endocrine therapies are at risk of hepatitis B virus (HBV) reactivation (HBVr). Guidelines recommend HBV screening prior to treatment. The Ottawa Hospital Cancer Center implemented a screening pilot for all patients receiving FOLFOX-based regimens between January and April 2023. We [...] Read more.
Cancer patients receiving non-endocrine therapies are at risk of hepatitis B virus (HBV) reactivation (HBVr). Guidelines recommend HBV screening prior to treatment. The Ottawa Hospital Cancer Center implemented a screening pilot for all patients receiving FOLFOX-based regimens between January and April 2023. We assessed the pilot from a quality improvement perspective. Charts were retrospectively reviewed, and patient and disease characteristics were collected. The primary endpoint was to identify the proportion of patients who underwent HBV screening prior to treatment start. Univariate analyses assessed the association between baseline characteristics and failure to screen. Quality metrics were also reviewed. There were 32/42 patients (76.2%) who completed screening, and 5 (11.9%) had a positive screen. The majority of eligible patients (59.5%) completed screening prior to the first treatment as intended. Four of five patients who tested positive were referred to Infectious Diseases. Of those, one received antivirals for chronic HBV. There were no treatment delays due to pending screening and no HBV reactivation. Receipt of prior systemic therapy was significantly associated with failure to screen (55 vs. 95%, OR 17.1 (95% CI 1.92–153), p = 0.011). The results of this pilot highlight the importance of building HBV screening into standardized treatment plans and engaging all team members to ensure high levels of screening. Prior systemic therapy receipt was associated with failure to screen, and thus, programs should include education on the necessity of screening as recommended by medical guidelines. Full article
(This article belongs to the Special Issue Gastrointestinal Cancers in Eastern Canada)
Show Figures

Figure 1

Figure 1
<p>HBV Screening Process Overview. Outline of the HBV screening program, including steps taken, treatment plan orders, communications distributed, and best practice advisories received by various members of the healthcare team.</p>
Full article ">
18 pages, 2615 KiB  
Article
The Vimentin-Targeting Drug ALD-R491 Partially Reverts the Epithelial-to-Mesenchymal Transition and Vimentin Interactome of Lung Cancer Cells
by Marieke Rosier, Anja Krstulović, Hyejeong Rosemary Kim, Nihardeep Kaur, Erhumuoghene Mary Enakireru, Deebie Symmes, Katalin Dobra, Ruihuan Chen, Caroline A. Evans and Annica K. B. Gad
Cancers 2025, 17(1), 81; https://doi.org/10.3390/cancers17010081 (registering DOI) - 30 Dec 2024
Abstract
Background: The epithelial-to-mesenchymal transition (EMT) is a common feature in early cancer invasion. Increased vimentin is a canonical marker of the EMT; however, the role of vimentin in EMT remains unknown. Methods: To clarify this, we induced EMT in lung cancer cells with [...] Read more.
Background: The epithelial-to-mesenchymal transition (EMT) is a common feature in early cancer invasion. Increased vimentin is a canonical marker of the EMT; however, the role of vimentin in EMT remains unknown. Methods: To clarify this, we induced EMT in lung cancer cells with TGF-β1, followed by treatment with the vimentin-targeting drug ALD-R491, live-cell imaging, and quantitative proteomics. Results: We identified 838 proteins in the intermediate filament fraction of cells. TGF-β1 treatment increased the proportion of vimentin in this fraction and the levels of 24 proteins. Variants of fibronectin showed the most pronounced increase (137-fold), followed by regulators of the cytoskeleton, cell motility, and division, such as the mRNA-splicing protein SON. TGF-β1 increased cell spreading and cell migration speed, and changed a positive correlation between cell migration speed and persistence to negative. ALD-R491 reversed these mesenchymal phenotypes to epithelial and the binding of RNA-binding proteins, including SON. Conclusions: These findings present many new interactors of intermediate filaments, describe how EMT and vimentin filament dynamics influence the intermediate filament interactome, and present ALD-R491 as a possible EMT-inhibitor. The observations support the hypothesis that the dynamic turnover of vimentin filaments and their interacting proteins govern mesenchymal cell migration, EMT, cell invasion, and cancer metastasis. Full article
(This article belongs to the Special Issue Extracellular Matrix Proteins in Cancer)
Show Figures

Figure 1

Figure 1
<p>TGF-β1-induced cell and cytoskeletal EMT phenotypes in A549 lung cancer cells, with and without ALD-R491 treatment. A549 cells treated without or with TGF-β1, and subsequent ALD-R491 treatment (R491), with regards to (<b>A</b>) vimentin, F-actin, or nuclei, as indicated, with merged images showing vimentin (red), F-actin (green), and nuclei (blue). Scale bars 10 (<b>left</b>) and 50 µm (<b>right</b>). (<b>B</b>) Protein levels of EMT markers (<b>left</b>), as quantified (<b>right</b>) for control (dark grey), treatment with TGF-β1 (white), followed by ALD-R491 (light grey). Bar plots show mean, error bars standard deviation (SD), black dots the values of each biological repeat, <span class="html-italic">p</span> ≤ 0.05 (*). Representative cells are shown.</p>
Full article ">Figure 2
<p><b>ALD-R491 reverses EMT-dependent cell spreading and migration phenotypes to normal.</b> A549 lung cancer cell treated without (dark grey) or with TGF-β1 (white) and ALD-R491 (R491) (light grey), as indicated, shown in (<b>A</b>) cell monolayers, scale bar 100 µm, (<b>B</b>) cell aspect ratio, circularity, spreading area, and (<b>C</b>) cell migration speed, persistence and correlation between speed and persistence for control (dark grey), treatment with TGF-β1 (white), followed by ALD-R491 (light grey). Bar plots show mean, error bars standard deviation (SD), <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 3
<p><b>Treatment with ALD-R491 and nuclear division during mitosis.</b> A549 lung cancer cells treated without (<b>top left</b>) and with TGF-β1 (<b>top right</b>), followed by ALD-R491 (R491) (<b>lower left</b>). Bright field images (BF), DNA (Hoechst) and merged images with BF and DNA (blue), are shown, as indicated. White arrows indicating dividing cells. Scalebar, 20 µm, with quantifications shown (<b>lower right</b>), with A549 treated without (dark grey) and TGF- β1 (white), followed by ALD-R491 (light grey), with regard to the proportion of nuclear division of all nuclei (left) and deformed, not-dividing proportion of dividing nuclei (right). Bar plots show mean, error bars, standard error of the mean (SEM), <span class="html-italic">p</span> ≤ 0.01 (**).</p>
Full article ">Figure 4
<p><b>TGF-β1 increases vimentin and reduces keratin in the intermediate filament fraction of the cytoskeleton</b>. The proportion of (<b>left</b>) keratin (dark grey), vimentin (white), and lamins (light grey) in the intermediate filament fraction, or of the (<b>right</b>) lamin A (dark grey), lamin B1 (white), and B2 (light grey) of all lamins in A549 lung cancer cell treated without (control) or with TGF-β1 and subsequent ALD-R491, as indicated. Bar plots show mean, error bars standard deviation (SD), <span class="html-italic">p</span> ≤ 0.05 (*), <span class="html-italic">p</span> ≤ 0.01 (**).</p>
Full article ">Figure 5
<p>The EMT increases binding to the intermediate filaments of proteins that regulate the cell–extracellular matrix adhesion, cytoskeleton, cell shape, and cell motility, while ALD-R491 reduces binding of RNA metabolism and function. The changes of the vimentin interactome upon EMT (<b>left</b>) and by additional ALD-R491 treatment (<b>right</b>), shown as percentages, with biological function (<b>top</b>), cellular compartment (<b>middle</b>), and molecular function (<b>lower</b>), as indicated.</p>
Full article ">
25 pages, 8739 KiB  
Article
DNAJB1-PKAc Kinase Is Expressed in Young Patients with Pediatric Liver Cancers and Enhances Carcinogenic Pathways
by Yasmeen Fleifil, Ruhi Gulati, Katherine Jennings, Alexander Miethke, Alexander Bondoc, Gregory Tiao, James I. Geller, Rebekah Karns, Lubov Timchenko and Nikolai Timchenko
Cancers 2025, 17(1), 83; https://doi.org/10.3390/cancers17010083 (registering DOI) - 30 Dec 2024
Abstract
Background and Aims: Hepatoblastoma (HBL) and fibrolamellar hepatocellular carcinoma (FLC) are the most common liver malignancies in children and young adults. FLC oncogenesis is associated with the generation of the fusion kinase, DNAJB1-PKAc (J-PKAc). J-PKAc has been found in 90% of FLC patients’ [...] Read more.
Background and Aims: Hepatoblastoma (HBL) and fibrolamellar hepatocellular carcinoma (FLC) are the most common liver malignancies in children and young adults. FLC oncogenesis is associated with the generation of the fusion kinase, DNAJB1-PKAc (J-PKAc). J-PKAc has been found in 90% of FLC patients’ tumors but not in other liver cancers. Since previous studies of J-PKAc were performed with adolescent patients, we asked if young children may express J-PKAc and if there are consequences of such expression. Methods: The biobank of the pediatric HBL/HCN-NOS specimens was examined by QRT-PCR, Western blots, RNA-Seq, and immunostaining with fusion-specific antibodies. Results: J-PKAc is expressed in 70% of the HBL/HCN-NOS patients. RNA-Seq analysis revealed that HBL tumors that do not have cells expressing J-PKAc show elevated expression of the membrane attack complex (MAC), which eliminates cells expressing J-PKAc. The fusion-positive HBL/HCN-NOS samples have several signaling pathways that are different from fusion-negative HBLs. Upregulated pathways included genes involved in the G1 to S transition and in liver cancer. Downregulated pathways included over 60 tumor suppressors, the CYP family, and the SLC family. The repression of these genes involves J-PKAc-β-catenin-TCF4-mediated elevation of the HDAC1-Sp5 pathway. The identified upregulated and downregulated pathways are direct targets of the fusion kinase. The J-PKAc kinase is also detected in livers of 1-year-old children with biliary atresia (BA). Conclusions: J-PKAc is expressed in both HBL tumor and BA liver samples, contributing to the development of HBL and creating a transcriptome profiling consistent with the potential development of liver cancer in young patients. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

Figure 1
<p>Identification of the J-PKAc kinase in 1–3-year-old children with hepatoblastoma and with HCN-NOS. (<b>A</b>) The identification of J-PKAc in a HBL patient (HBL72). The top image is a Western blot to PKAc. FLC: protein extract from an FLC patient (control sample); B: background (adjacent to the tumor) region of the liver; T: tumor. The bottom image shows the detection of the J-PKAc fusion mRNA in the tumor of HBL72. (<b>B</b>) Identification of J-PKAc in pediatric HBL, HCC, and HCN-NOS specimens in a large biobank at CCHMC. The top panel is a Western blot with HBL specimens using antibodies that recognize both fusion and native PKAc. Specimens with high levels of J-PKAc are shown in red. The bottom left image shows QRT-PCR analysis of the J-PKAc fusion transcript in two J-PKAc fusion-positive samples. The bottom right image shows the detection of the fusion kinase in HCN-NOS77. (<b>C</b>) Generation of the J-PKAc-fusion-specific antibody. Top: Peptide sequence that was used for the generation of a fusion-specific antibody. Bottom: Western blots of background and tumor sections of FLC51 and FLC lung metastasis with the fusion antibody. The membrane was re-probed with antibodies that recognize both J-PKAc and native PKAc. (<b>D</b>) Immunohistochemistry of tumor sections of three FLC patients with the fusion-specific antibody. Magnification = 10×. Scale bars = 50 μm. Inserts show individual cells under 40×. (<b>E</b>) Expression of the fusion J-PKAc in HBLs/HCN-NOSs was examined by Western blot with Fus-Abs. The membrane was re-probed with Abs to cdc2 (cdk1) and to β-actin as a loading control. (<b>F</b>) Immunohistochemistry of tumor sections of samples HBL114 and HCN-NOS108. Images of individual cells are shown in boxes. Magnification = 10×. Scale bars = 50 μm.</p>
Full article ">Figure 2
<p>RNA-Seq analyses of J-PKAc-positive HBLs/HCN-NOSs and J-PKAc-negative HBLs. (<b>A</b>) Pathways that are upregulated in fusion-negative HBLs (<span class="html-italic">n</span> = 3) compared to background tissue (<span class="html-italic">n</span> = 3). (<b>B</b>) Pathways that are downregulated in fusion-positive (<span class="html-italic">n</span> = 5) HBLs compared to background tissue (<span class="html-italic">n</span> = 3). (<b>C</b>) Nuclear and membrane immunostaining in fusion-positive HBL114 and HCN-NOS108. (<b>D</b>) Examination of the integrity of the fusion-positive membranes in the patients with FLC by immunostaining with Fus-Abs. Black arrows point to cells with membrane and nuclear Fus-staining. Examples of the destruction of fusion-positive membranes in tumors of samples FLC110 and FLCH01 are shown by open red arrows. (<b>E</b>) A hypothesis for the process of distraction of fusion-expressing cells in the fusion-negative HBL patients.</p>
Full article ">Figure 2 Cont.
<p>RNA-Seq analyses of J-PKAc-positive HBLs/HCN-NOSs and J-PKAc-negative HBLs. (<b>A</b>) Pathways that are upregulated in fusion-negative HBLs (<span class="html-italic">n</span> = 3) compared to background tissue (<span class="html-italic">n</span> = 3). (<b>B</b>) Pathways that are downregulated in fusion-positive (<span class="html-italic">n</span> = 5) HBLs compared to background tissue (<span class="html-italic">n</span> = 3). (<b>C</b>) Nuclear and membrane immunostaining in fusion-positive HBL114 and HCN-NOS108. (<b>D</b>) Examination of the integrity of the fusion-positive membranes in the patients with FLC by immunostaining with Fus-Abs. Black arrows point to cells with membrane and nuclear Fus-staining. Examples of the destruction of fusion-positive membranes in tumors of samples FLC110 and FLCH01 are shown by open red arrows. (<b>E</b>) A hypothesis for the process of distraction of fusion-expressing cells in the fusion-negative HBL patients.</p>
Full article ">Figure 3
<p>The cell line flc110, derived from the Fibrolamellar HLC110 patient, contains cells with disintegrated fusion-positive membranes. (<b>A</b>) Examination of the J-PKAc kinase in tumors of the FLC110 patient. Cytoplasm and nuclear extracts from background (adjacent, non-tumor) and tumor sections were examined by a Western blot assay with antibodies to PKAc, Abs to the N-terminus of DNAJB1, and Fus-Abs. The upper image shows the integrity of the proteins determined by Coomassie staining. (<b>B</b>) Detection of the fusion J-PKAc mRNA by QRT-PCR. (<b>C</b>) Generation of the FLC110-derived cell line. Images of cells exiting the tumor (exit) and images of cells after 1st and 2nd passages are shown. (<b>D</b>) Western blots of WCEs from HepG2 and flc110 cells with fusion-specific Abs and with Abs to PKAc. (<b>E</b>) Immunostaining of flc110 cells with fusion-specific Abs. (<b>F</b>) Fusion/DAPI/merged images of flc110 cells stained with Fus-Abs. Cells with membrane and nuclear immunostaining are shown. Arrows show fusion-positive cells with destructed nuclei and cells with cell damage.</p>
Full article ">Figure 4
<p>RNA-Seq-based comparison of pathways that are different in J-PKAc-positive HBLs/HCN-NOSs compared to J-PKAc-negative HBLs. (<b>A</b>) List of compared HBL/HCN-NOS samples. (<b>B</b>) A heatmap that shows the differences in pathways between fusion-positive HBLs/HCN-NOSs and fusion-negative HBLs. (<b>C</b>) Pathways that are upregulated in fusion-positive (<span class="html-italic">n</span> = 5) HBLs. (<b>D</b>) Pathways that are downregulated in fusion-positive HBLs (<span class="html-italic">n</span> = 5) compared to fusion-negative HBLs (<span class="html-italic">n</span> = 3). (<b>E</b>) Levels of mRNAs coding for markers of proliferation cdk1 and cdk4 as averages of duplicate measurements in fusion-positive (<span class="html-italic">n</span> = 7) vs. fusion-negative samples (<span class="html-italic">n</span> = 8). Paired <span class="html-italic">t</span>-tests were performed (*: <span class="html-italic">p</span> &lt; 0.05); differences are not significant in unlabeled plots. (<b>F</b>) List of tumor suppressors and levels of their fold-reduction in fusion-positive HBLs/HCN-NOSs compared to fusion-negative HBLs. QRT-PCR shows a reduction of the tumor suppressor histidine-rich glycoprotein (HRG) in a large biobank of HBL/HCN-NOS samples (<span class="html-italic">n</span> = 40) and confirmation of the reduction in HRG in fusion-positive HBLs/HCN-NOSs (<span class="html-italic">n</span> = 5) compared to fusion-negative HBLs (<span class="html-italic">n</span> = 3) as averages of duplicate measurements. The bottom QRT-PCR graph shows a reduction in HRG in the FLC patients (<span class="html-italic">n</span> = 5) as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed (****: <span class="html-italic">p</span> &lt; 0.0001); differences are not significant in unlabeled plots.</p>
Full article ">Figure 5
<p>J-PKAc-positive HBLs are characterized by high levels of fibrosis. (<b>A</b>) QRT-PCR showed that levels of AKAP12 and AKAP13 mRNAs are increased in fusion-positive (<span class="html-italic">n</span> = 7) vs. fusion-negative HBLs/HCCs (<span class="html-italic">n</span> = 5). Levels of Col2A1 mRNAs are increased in fusion-positive (<span class="html-italic">n</span> = 5) vs. fusion-negative HBLs/HCCs (<span class="html-italic">n</span> = 3), and levels of Col1A1 mRNAs are increased in fusion-positive (<span class="html-italic">n</span> = 8) compared to fusion-negative HBLs/HCCs (<span class="html-italic">n</span> = 10). Paired <span class="html-italic">t</span>-tests were performed (*: <span class="html-italic">p</span> &lt; 0.05); differences are not significant in unlabeled plots. (<b>B</b>) H&amp;E staining of tumors and backgrounds of J-PKAc-negative and J-PKAc-positive HBLs, HCCs, and HCN-NOSs. Arrows show lamellar structures (for FLC) and lamellar-like structures (for HBLs). (<b>C</b>) Sirius Red staining of fusion-negative and fusion-positive HBLs. FLC110 and FLC6; staining of the tumor sections of two FLC patients. Arrows show Sirius Red-positive lamellar-like structures.</p>
Full article ">Figure 6
<p>Components of the complement cascade/MAC and expression of the genes of bile acid/drug metabolisms that are reduced in J-PKAc-positive HBLs. (<b>A</b>) Upper left: RNA-Seq-based list and fold reduction in mRNAs coding for the complement cascade and membrane attack complex. Middle: QRT-PCR shows the levels of corresponding mRNAs in fusion-negative (<span class="html-italic">n</span> = 3) and fusion-positive (<span class="html-italic">n</span> = 5) HBLs as averages of duplicate measurements. Right: Levels of FCN3 in a large biobank (<span class="html-italic">n</span> = 42) were determined by QRT-PCR as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed (****: <span class="html-italic">p</span> &lt; 0.0001); differences are not significant in unlabeled plots. (<b>B</b>) QRT-PCR shows levels of mRNAs coding for complement cascade/MAC in Huh6 cells transfected with the fusion kinase (<span class="html-italic">n</span> = 3) compared to untransfected cells (<span class="html-italic">n</span> = 3) as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed (*: <span class="html-italic">p</span> &lt; 0.05, ***: <span class="html-italic">p</span> &lt; 0.001); differences are not significant in unlabeled plots. (<b>C</b>) Levels of complement cascade/MAC in the patients with FLC (<span class="html-italic">n</span> = 5) as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed; differences are not significant in unlabeled plots. (<b>D</b>,<b>E</b>) The list of members of CYP and SLC families that are downregulated in fusion-positive HBLs relative to fusion-negative HBLs. The right part shows QRT-PCR for CYP2B6 and SLC members in fusion-positive (<span class="html-italic">n</span> = 5) and fusion-negative HBLs (<span class="html-italic">n</span> = 3) and CYP3A4 in fusion-positive (<span class="html-italic">n</span> = 7) and fusion-negative (<span class="html-italic">n</span> = 9) HBLs as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed; differences are not significant in unlabeled plots.</p>
Full article ">Figure 7
<p>Levels of mRNAs in patients with fibrolamellar HCC which were differentially expressed in fusion-positive HBLs. (<b>A</b>) A Western blot of protein extracts from 5 FLC patients with Fus-Abs shows expression of J-PKAc in tumor sections of the livers. (<b>B</b>) Levels of CYP and SLC family mRNAs are reduced in the FLC patients. Left: Red color shows mRNAs that are downregulated in fusion-positive HBLs compared to fusion-negative HBLs. Right: QRT-PCR analysis is shown as averages of duplicate measurements in the FLC patients (<span class="html-italic">n</span> = 5). Paired <span class="html-italic">t</span>-tests were performed; differences are not significant in unlabeled plots. (<b>C</b>) Levels of Sp5 (<span class="html-italic">n</span> = 6) and HDAC1 (<span class="html-italic">n</span> = 8) are increased in the tumors of the FLC samples, shown as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed (**: <span class="html-italic">p</span> &lt; 0.01); differences are not significant in unlabeled plots. (<b>D</b>) Markers of liver proliferation, cdk1 and cdk4, and the marker of fibrosis, Col1A1, are increased in the FLC samples (<span class="html-italic">n</span> = 5), shown as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed; differences are not significant in unlabeled plots. (<b>E</b>) The ph-S675-β-catenin pathway is enhanced in patients with J-PKAc. Left: a diagram showing the J-PKAc-β-catenin pathway described in our previous paper [<a href="#B5-cancers-17-00083" class="html-bibr">5</a>]. Western: levels of the fusion J-PKAc kinase and ph-S675-β-catenin were examined by Western blot. Bar graphs show ratios of ph-S675-β-catenin to β-actin. (<b>F</b>) Levels of J-PKAc-β-catenin-CEGR/ALCD pathway-dependent stem cell markers and cancer-associated mRNAs in J-PKAc-negative (<span class="html-italic">n</span> = 5) and J-PKAc-positive HBLs/HCN-NOSs (<span class="html-italic">n</span> = 8), shown as averages of duplicate measurements. Unpaired <span class="html-italic">t</span>-tests were performed; differences are not significant in unlabeled plots.</p>
Full article ">Figure 8
<p>The J-PKAc fusion kinase directly regulates pathways that are differentially expressed in J-PKAc-positive HBLs. (<b>A</b>,<b>B</b>) Ectopic expression of J-PKAc in HepG2 (<b>A</b>) and Huh6 (<b>B</b>) cells enhanced phosphorylation of b-catenin at Ser675. Upper images show Western blots with proteins from cells transfected with lipofectamine (Lip) and DNAJB1-PKAc plasmid using Abs to PKAc (which recognize the native and fusion kinases) and with Abs to ph-S675-β-catenin. The bar graphs below show levels of ph-S675-β-catenin calculated as a ratio to the loading control β-actin in duplicate. Unpaired <span class="html-italic">t</span>-tests were performed (*: <span class="html-italic">p</span> &lt; 0.05); differences are not significant in unlabeled plots. (<b>C</b>) A summary of the elevation in mRNAs corresponding to J-PKAc-β-catenin-dependent genes in HepG2 (<span class="html-italic">n</span> = 3) and in Huh6 cells (<span class="html-italic">n</span> = 3) transfected with a DNAJB1-PKAc-expressing plasmid compared to untransfected HepG2 (<span class="html-italic">n</span> = 3) and Huh6 cells (<span class="html-italic">n</span> = 3), shown as averages of duplicate measurements. (<b>D</b>,<b>E</b>) Images of HepG2 and Huh6 cells transfected with DNAJB1-PKAc plasmid in triplicate and stained with the fusion-specific antibody (green), HDAC1 (red), and DAPI. Three fields of images for each type of cell are shown. The bottom images show high magnifications of the merges of the positive cells. Arrows show cells with co-localization of the J-PKAc and HDAC1. (<b>F</b>) Left: levels of mRNAs of HDAC1-Sp5-dependent CYP and SLC genes in cells transfected by DNAJB1-PKAc. Right: levels of mRNA coding for the tumor suppressor HRG in transfected cells. Both are shown as averages of duplicate measurements for transfected (<span class="html-italic">n</span> = 3) compared to untransfected cells (<span class="html-italic">n</span> = 3). Paired <span class="html-italic">t</span>-tests were performed (*: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01); differences are not significant in unlabeled plots.</p>
Full article ">Figure 9
<p>The expression of the J-PKAc fusion kinase in a patient-derived HBL cell line determines the formation of elongated lamellar-like structures. (<b>A</b>) Expression of the fusion kinase in the livers of the HBL patients, the tumors of which were used for the generation of cell lines. The position of the fusion kinase is shown by a red arrow. (<b>B</b>) Generation of hbl111 and hbl114 cell lines. The left part shows a “cell-free-exit” strategy; the right part shows Western blots of whole-cell extracts isolated from generated cell lines with Abs to the C-terminal part of the PKAc and with Fus-Abs. (<b>C</b>) Immunostaining of the hbl114 cell line with fusion-specific Abs at 3–5 days after plating. (<b>D</b>) Typical images of tumor clusters and elongated lamellar-like structures in cell lines hbl111, flc110, and hbl114 at 12 days after plating at low density. (<b>E</b>) Immunostaining of the flc110 and hbl114 cells with Fus-Abs at 12 days after plating at low density. (<b>F</b>) Elimination of the fusion kinase by inhibition of the β-catenin-CEGR/ALCD pathway prevents the formation of elongated structures in the hbl114 cell line. The upper image shows Western blots of proteins isolated from DMSO- and PRI-724-treated hbl114 cells with Abs to PKAc and with Fus-Abs. The bottom part shows images of cells forming elongated structures and cells after treatments with PRI-724.</p>
Full article ">Figure 10
<p>Examination of cancer pathways in chemo-resistant patient HBL116. (<b>A</b>) QRT-PCR analysis of cancer pathways in a tumor of patient HBL116. (<b>B</b>) β-III-tubulin staining of background and tumor sections. (<b>C</b>) Examination of J-PKAc in the liver of patient HBL116 by immunostaining.</p>
Full article ">Figure 10 Cont.
<p>Examination of cancer pathways in chemo-resistant patient HBL116. (<b>A</b>) QRT-PCR analysis of cancer pathways in a tumor of patient HBL116. (<b>B</b>) β-III-tubulin staining of background and tumor sections. (<b>C</b>) Examination of J-PKAc in the liver of patient HBL116 by immunostaining.</p>
Full article ">Figure 11
<p>The fusion J-PKAc contributes to the chemo-resistance of the hbl116 cell line derived from chemo-resistant patient HBL116. (<b>A</b>) Expression of the fusion J-PKAc in patient HBL116. A Western blot was performed with cytoplasmic and nuclear extracts from background and tumor specimens of the liver of patient HBL116. Antibodies to PKAc and Fus-Abs were used. Red arrows show the fusion J-PKAc kinase, black arrows show native PKAc. (<b>B</b>) Generation of the hbl116 cell line using the “cell free exit” protocol. Images of the exit of cells and hbl116 cells that form elongated clusters are shown. (<b>C</b>) hbl116 cells express the fusion J-PKAc protein. A Western blot was performed with Abs to PKAc and Fus-Abs. (<b>D</b>) Typical images of the hbl116 cells treated with cisplatin, PRI-724, and with a combination of cisplatin and PRI-724. (<b>E</b>) Expression of J-PKAc-dependent genes in treated hbl116 cells. A Western blot was performed with Abs, as shown on the right. (<b>F</b>) Examination of the β-catenin-TCF4-p300 pathway. Western blot and Co-IP studies are shown. (<b>G</b>) A summary of the studies showing the role of J-PKAc in the resistance of hbl116 cells to cisplatin.</p>
Full article ">Figure 12
<p>A portion of the patients with biliary atresia expresses J-PKAc, which correlates with an elevation in J-PKAc-ph-S675-β-catenin-dependent genes and fibrolamellar HCC-specific genes and with an elevation in mRNAs coding for cell cycle proteins. (<b>A</b>) A table showing pathological characteristics of six BA patients, the livers of whom were investigated. Age and levels of ALT/AST in the fusion-positive patients are shown in red. (<b>B</b>) Western blots of protein extracts from 6 BA patients with antibodies to PKAc, to the fusion kinase, to J-PKAc-β-catenin-TCF4 axis, and to downstream targets and proliferation markers. Coomassie staining on the top shows the integrity of proteins. Bar graphs show levels of proteins as ratios to β-actin determined by ImageJ software. (<b>C</b>) Expression of FLC-specific mRNAs in all available fusion-positive BA samples (<span class="html-italic">n</span> = 3) compared to the fusion-negative samples (<span class="html-italic">n</span> = 3), shown as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed; differences are not significant in unlabeled plots. (<b>D</b>) Expression of markers of proliferation in all available fusion-positive BA samples (<span class="html-italic">n</span> = 3) compared to the fusion-negative BA samples (<span class="html-italic">n</span> = 3), shown as averages of duplicate measurements. Paired <span class="html-italic">t</span>-tests were performed; differences are not significant in unlabeled plots. (<b>E</b>) Expression of the fusion kinase in recently collected specimens from patients with HBL was detected by Western blot with antibodies to PKAc (upper) and with Fus-Abs (bottom). The membrane was re-probed with an antibody to β-actin. (<b>F</b>) A hypothesis for the elimination of fusion-expressing cells with age and for the development of FLC in the patients who failed to eliminate the fusion-expressing cells. (<b>G</b>) A summary showing J-PKAc-dependent pathways that are increased in fusion-positive HBLs/HCN-NOSs.</p>
Full article ">
Back to TopTop