CancersPub Date : 2025-05-29DOI: 10.3390/cancers17111812
Valentina Andrade-Perez, Noël J-M Raynal
{"title":"Interplay Between the Epigenome, the Microenvironment, and the Immune System in Neuroblastoma.","authors":"Valentina Andrade-Perez, Noël J-M Raynal","doi":"10.3390/cancers17111812","DOIUrl":"10.3390/cancers17111812","url":null,"abstract":"<p><p>Neuroblastoma (NB) is the most prevalent extracranial childhood tumor and the third leading cause of death from cancer in children. Despite having a high overall survival rate for low- and intermediate-risk patients, survival rates for high-risk cases remain unsatisfactory. The current standard treatment for high-risk NB involves surgery, chemotherapy, radiotherapy, autologous stem cell transplantation, immunotherapy with anti-ganglioside GD2, and differentiation therapy with isotretinoin. Besides not being enough to achieve a high survival rate in high-risk patients, these treatments are associated with significant side effects. With next-generation sequencing technologies, a better understanding of the genetic and epigenetic landscapes of NB has been achieved. This has led to the study of novel treatments to improve the overall survival rate of high-risk NB and reduce the toxicity of conventional treatments. Current research is focusing on the development of targeted drugs for genetic and epigenetic alterations, and protein degraders. Moreover, immunotherapy to enhance anticancer immune responses and by using cell-engineering techniques with chimeric antigen receptor (CAR) T and NK cells are being explored to target NB cells. Here, we review promising novel treatment strategies for NB, which target genetics, epigenetics, the tumor microenvironment, and the immune landscape, highlighting preclinical studies and ongoing clinical trials.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153772/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282540","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-29DOI: 10.3390/cancers17111817
Nicole Lisa Li-Ann Goh, Nur Jannah Abdul Rahim, Rituparna Bhatt, Si En Ong, Khai Yee Lim, Anastasia Gandini, Zhen Zeng, Snehlata Kumari, Brian Gabrielli
{"title":"Checkpoint Kinase 1 Inhibitor Combined with Low Dose Hydroxyurea Promotes ATM-Activated NF-κB-Dependent Pro-Inflammatory Chemokine Expression in Melanomas.","authors":"Nicole Lisa Li-Ann Goh, Nur Jannah Abdul Rahim, Rituparna Bhatt, Si En Ong, Khai Yee Lim, Anastasia Gandini, Zhen Zeng, Snehlata Kumari, Brian Gabrielli","doi":"10.3390/cancers17111817","DOIUrl":"10.3390/cancers17111817","url":null,"abstract":"<p><strong>Background/objectives: </strong>Melanoma has a rising incidence worldwide. Current treatments are effective, although the development of resistance is common. A novel anti-cancer treatment using checkpoint kinase 1 inhibitor (CHK1i), SRA737, in combination with low-dose hydroxyurea (LDHU), has been demonstrated to effectively kill tumour cells and promote an anti-tumour immune response through the treatment-induced release of pro-inflammatory chemokines and cytokines. These chemokines/cytokines modify the tumour microenvironment from an immunosuppressive to an inflamed state to recruit anti-tumour immune cells.</p><p><strong>Methods: </strong>A panel of human melanoma cell lines was assessed using a panel of chemokines and cytokine expression, and the mechanism of their regulation was investigated.</p><p><strong>Results: </strong>We demonstrate that SRA737 + LDHU upregulates pro-inflammatory chemokines in human melanoma cells in response to SRA737 + LDHU through the ATM-NF-κB signalling pathway. The increased chemokine expression corresponded to the increase in secretion of pro-inflammatory chemokines from tumour cells following SRA737 + LDHU treatment. However, inhibiting NF-κB and ATM did not affect SRA737 + LDHU-induced cell killing. Increased expression of non-NF-κB target genes with SRA737 + LDHU suggests that other transcriptional pathways are also activated and may contribute to the increasing cytokine/chemokine gene expression in response to treatment.</p><p><strong>Conclusions: </strong>SRA737 + LDHU upregulates pro-inflammatory chemokine expression through an ATM-NF-κB-dependent mechanism.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153532/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282497","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-29DOI: 10.3390/cancers17111816
Franchesca König, Chanel Davidoff, Katarzyna Ibanez, Sinchun Hwang, Ilan Goldstein, Darren R Feldman, Boglarka Gyurkocza, Sergio A Giralt, Ioannis Politikos, Doris M Ponce, Michael Scordo, Grigory Syrkin, Christian M Custodio, Gunjan L Shah
{"title":"Post-Transplant Pain and Paralysis: Neurologic Amyotrophy as an Atypical Cause of Shoulder Dysfunction Following Hematopoietic Stem Cell Transplant.","authors":"Franchesca König, Chanel Davidoff, Katarzyna Ibanez, Sinchun Hwang, Ilan Goldstein, Darren R Feldman, Boglarka Gyurkocza, Sergio A Giralt, Ioannis Politikos, Doris M Ponce, Michael Scordo, Grigory Syrkin, Christian M Custodio, Gunjan L Shah","doi":"10.3390/cancers17111816","DOIUrl":"10.3390/cancers17111816","url":null,"abstract":"<p><strong>Background: </strong>Neuralgic amyotrophy (NA), also known as Parsonage-Turner syndrome, brachial neuritis, and idiopathic brachial plexopathy, is a rare and potentially debilitating peripheral nerve disorder characterized by acute-onset shoulder pain followed by progressive motor deficits. It is often under-recognized, with an estimated incidence of 1 to 3 per 100,000 annually, though some studies suggest the actual prevalence may be significantly higher. The condition typically progresses through three phases, an acute painful phase, a phase of weakness, and a recovery phase, with sensory disturbances common in addition to motor weakness. The exact pathogenesis of NA remains unclear, though it is thought to involve a combination of genetic, environmental, and immunological factors. While neurologic complications following hematopoietic stem cell transplantation (HSCT), such as neuropathies and myopathies, have been documented, NA remains exceedingly rare in this context, with only a few reported cases. The pathophysiology in HSCT patients is hypothesized to involve immune dysregulation, graft-versus-host disease (GvHD), infection, and the effects of immunosuppressive therapy. Diagnosis is primarily clinical, supported by electrodiagnostic studies and MRI, though no laboratory markers exist. The management of NA is largely supportive and multimodal, focusing on pain control and rehabilitation.</p><p><strong>Objectives: </strong>The objective of this study was to describe the characteristics, clinical course, and outcomes of patients admitted for HSCT who were subsequently diagnosed with NA.</p><p><strong>Study design: </strong>This retrospective case series from a single institution examined nine (N = 9) patients who developed acute shoulder pain following HSCT. We collected data on demographics, transplant details, clinical features, MRI findings, and electrodiagnostic studies, summarized using descriptive statistics. The diagnosis of neurologic amyotrophy was based on clinical presentation and corroborated by imaging and electrodiagnostic results. Long-term follow-up was assessed to evaluate symptom recovery.</p><p><strong>Results: </strong>Between August 2020 and July 2022, nine patients (44% male, median age 60) were diagnosed with NA following autologous (n = 4) or allogeneic (n = 5) HSCT. The onset of severe shoulder pain occurred at a median of 9 days post-transplant (range 1-21 days), with the majority of patients experiencing unilateral pain, predominantly affecting the right shoulder (55%). Neurologic weakness developed on average 5.1 days after pain onset, and sensory deficits were observed in all but one patient. MRI findings revealed muscle edema, atrophy, and enhancement in six patients, while electromyography confirmed NA in five. Due to the small sample size, statistical analyses, including <i>p</i>-values, confidence intervals, and trend comparisons, were not performed, and thus no conclusions can be drawn regarding a","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153850/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282586","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-29DOI: 10.3390/cancers17111820
Whi-An Kwon, Jae Young Joung
{"title":"T-Cell Engager Therapy in Prostate Cancer: Molecular Insights into a New Frontier in Immunotherapy.","authors":"Whi-An Kwon, Jae Young Joung","doi":"10.3390/cancers17111820","DOIUrl":"10.3390/cancers17111820","url":null,"abstract":"<p><p>Advanced prostate cancer (PCa) remains lethal despite standard therapies, and immune checkpoint inhibitors offer limited benefit in its \"immune-cold\" microenvironment. T-cell engagers (TCEs)-bispecific antibodies linking CD3 on T-cells to tumor-associated antigens (TAAs)-provide potent, MHC-independent cytotoxicity, overcoming a key resistance mechanism. While early PSMA-targeted TCEs established proof-of-concept, recent data, notably for six transmembrane epithelial antigen of the prostate 1 (STEAP1)-targeting agents like Xaluritamig, demonstrate more substantial objective responses, highlighting progress through improved target selection and molecular design. This review synthesizes the evolving landscape of TCEs targeting PSMA, STEAP1, and DLL3 in PCa. We critically evaluate emerging clinical evidence, arguing that realizing the significant therapeutic potential of TCEs requires overcoming key challenges, including cytokine release syndrome (CRS), limited response durability, and antigen escape. We contend that future success hinges on sophisticated engineering strategies (e.g., affinity tuning, masking, multispecific constructs) and rationally designed combination therapies tailored to disease-specific hurdles. Strategies for toxicity mitigation, the crucial role of biomarker-driven patient selection, and potential integration with existing treatments are also discussed. Accumulating evidence supports TCEs becoming a new therapeutic pillar for advanced PCa, but achieving this demands sustained innovation focused on optimizing efficacy and safety. This review critically connects molecular engineering advancements with clinical realities and future imperatives.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153726/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282607","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-29DOI: 10.3390/cancers17111814
Maen Abdelrahim, Abdullah Esmail, Richard D Kim, Sukeshi Patel Arora, Junaid Arshad, Ioannis A Kournoutas, Conor D O'Donnell, Todor I Totev, Amie Tan, Fan Mu, Shravanthi M Seshasayee, Sairy Hernandez, Nguyen H Tran
{"title":"Real-World Experiences Using Atezolizumab + Bevacizumab for the Treatment of Unresectable Hepatocellular Carcinoma: A Multicenter Study.","authors":"Maen Abdelrahim, Abdullah Esmail, Richard D Kim, Sukeshi Patel Arora, Junaid Arshad, Ioannis A Kournoutas, Conor D O'Donnell, Todor I Totev, Amie Tan, Fan Mu, Shravanthi M Seshasayee, Sairy Hernandez, Nguyen H Tran","doi":"10.3390/cancers17111814","DOIUrl":"10.3390/cancers17111814","url":null,"abstract":"<p><p><b>Objective</b>: This study aimed to evaluate the characteristics, clinical outcomes, and resource use of patients with unresectable hepatocellular carcinoma (uHCC) treated with first-line (1L) atezolizumab plus bevacizumab (A+B) at five United States (US) institutions: the Mayo Clinic, Houston Methodist, Moffitt Cancer Center, Mays Cancer Center, and University of Arizona. <b>Methods</b>: Treating oncologists extracted data from medical charts of patients with uHCC who were treated with A+B after 1 January 2019. Real-world progression-free survival (rwPFS) and overall survival (OS) were assessed using the Kaplan-Meier method for the overall cohort and for a \"trial-like\" subgroup with characteristics similar to those in the IMbrave150 trial (Eastern Cooperative Oncology Group Performance Status [ECOG PS] 0-1, Child-Pugh [CP] class A, albumin-bilirubin grade 1-2). <b>Results</b>: Of the 300 patients in the overall cohort (median age of 68 years; 12% ECOG PS ≥ 2; 73% CP A; 26% CP B; median follow-up of 8.7 months), the median rwPFS was 6.8 (95% confidence interval [CI]: 5.8, 8.4) months, and the median OS was 14.4 (95% CI: 12.3, 18.2) months. In the trial-like subgroup (<i>n</i> = 194), the median rwPFS was 8.8 (95% CI: 7.6, 12.1) months and the median OS was 19.5 (95% CI: 14.6, 24.7) months. A significantly lower proportion of patients with CP A compared with CP B (39.7% vs. 73.4%) experienced hospitalization within one year of A+B initiation, whereas hospitalizations due to treatment-related adverse events were similar. <b>Conclusions</b>: This study provides insights into the real-world effectiveness of 1L A+B in a diverse US patient cohort, with results from trial-like patients supporting the reproducible efficacy of A+B in clinical practice.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153728/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282593","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-29DOI: 10.3390/cancers17111813
Linhan Ye, Zongyao Chen, Jingcheng Zhang, Qiaolin Li
{"title":"Modeling the Transitional Phase of Epithelial Cells Reveals Prognostic and Therapeutic Targets in Pancreatic Ductal Adenocarcinoma.","authors":"Linhan Ye, Zongyao Chen, Jingcheng Zhang, Qiaolin Li","doi":"10.3390/cancers17111813","DOIUrl":"10.3390/cancers17111813","url":null,"abstract":"<p><p><b>Background:</b> Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor prognosis, and its progression is driven by epithelial plasticity and tumor microenvironment remodeling. Finding biomarkers that are responsible for the turning point from the early stage to the aggressive phase would facilitate clinical management. <b>Method:</b> In this study, we employed single-cell RNA sequencing to characterize a distinct subpopulation of highly proliferative epithelial cells undergoing a transitional phase during PDAC progression. By linking to cell cycle dysregulation, epithelial differentiation, and clinical staging, we constructed a gene-based risk score model using Lasso Cox regression. The expression of selected genes within the model was further validated using qPCR. <b>Results:</b> The model demonstrated robust predictive power for patient prognosis, TNM staging, and chemotherapy sensitivity. Further analysis of the tumor microenvironment revealed intensified crosstalk between a specific fibroblast subpopulation and transitional epithelial cells, mediated largely by collagen signaling. This stromal-epithelial interaction was found to contribute to the fibrotic barrier characteristic of PDAC. Additionally, immune profiling uncovered altered infiltration patterns, particularly involving natural killer (NK) cells in high-risk patients, suggesting mechanisms of immune tolerance and inhibition. <b>Conclusions:</b> These findings offer potential avenues for early detection, risk stratification, and targeted therapeutic strategies in PDAC.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153703/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282565","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-29DOI: 10.3390/cancers17111815
Stefania Rizzo, Luca Bellesi, Ebticem Ben Khalifa, Stefano Presilla, Andrea D'Ermo, Francesco Magoga, Matteo Merli, Ermidio Rezzonico, Oriana D'Ecclesiis, Filippo Del Grande
{"title":"Radiation Dose Reduction in Cancer Imaging with New-Model CT Scanners: A Quality of Care Evaluation.","authors":"Stefania Rizzo, Luca Bellesi, Ebticem Ben Khalifa, Stefano Presilla, Andrea D'Ermo, Francesco Magoga, Matteo Merli, Ermidio Rezzonico, Oriana D'Ecclesiis, Filippo Del Grande","doi":"10.3390/cancers17111815","DOIUrl":"10.3390/cancers17111815","url":null,"abstract":"<p><strong>Background/objectives: </strong>The primary aim of this study was to evaluate whether the replacement of roughly one-decade-old computed tomography (CT) scanners with new-model CT scanners were associated with an additional reduction in the radiation dose delivered to oncological patients, in a radiological setting where the optimization of protocols had already reached very low radiation doses. An exploratory secondary objective was to evaluate the potential differences in the objective image quality between the CT scans obtained before and after the installation of the new-generation CT scanners.</p><p><strong>Methods: </strong>Chest and abdominal CT examinations conducted for oncologic purposes were retrospectively selected from two time periods-prior to scanner replacement (2022) and following an upgrade (2024)-after five CT systems in our radiology department were replaced. We extracted and compared the CT dose index (CTDI) and dose length product (DLP) for each CT phase. For the objective image quality evaluation, we calculated the signal-to-noise ratio (SNR) and the contrast-to-noise ratio (CNR) at the center of the liver and the aorta. An appropriate statistical analysis was performed and a <i>p</i>-value < 0.05 was considered significant.</p><p><strong>Results: </strong>We included 14,601 CT acquisitions, of which 9013 (61.7%) were performed before and 5588 (38.3%) after the replacement of the CT scanners. There were significantly lower values for the CTDI and DLP with the new CT scanners compared to the old ones. The CTDI with the new CT scanners was significantly lower in all phases (<i>p</i>-value = 0.002 for unenhanced phase, and <i>p</i> < 0.001 for arterial, portal venous, and delayed phases). The DLP using the new CT scanners was significantly lower in the arterial, portal venous, and delayed phases (<i>p</i> < 0.001), and it was not significantly different in the unenhanced phase (<i>p</i> = 0.36). There was no significant difference in the SNR at the liver level (<i>p</i> = 0.72) or at the aorta level (<i>p</i> = 0.51). There was no significant difference in the CNR at the liver level (<i>p</i> = 0.24), whereas the CNR was higher with the new CT scanners at the aorta level (<i>p</i> = 0.03).</p><p><strong>Conclusions: </strong>The transition to new-model CT scanners resulted in a significant reduction in the radiation dose delivered by chest and abdomen CT scans, without compromising the objective image quality.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153870/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-28DOI: 10.3390/cancers17111797
James Yu, Jose M Laborde, Robin Park, Moazzam Shahzad, Youngchul Kim, Jaekyung Cheon, Iman Imanirad, Richard D Kim, Tiago Biachi de Castria, Nicole L Nardella, Mokenge Malafa, Jason W Denbo, Jason B Fleming, Sarah E Hoffe, Jessica M Frakes, Andrew J Sinnamon, Jose M Pimiento, Pamela J Hodul, Dae Won Kim
{"title":"Association of Pathologic Response and Adjuvant Chemotherapy with Survival in Resected Pancreatic Ductal Adenocarcinoma Following Neoadjuvant Therapy.","authors":"James Yu, Jose M Laborde, Robin Park, Moazzam Shahzad, Youngchul Kim, Jaekyung Cheon, Iman Imanirad, Richard D Kim, Tiago Biachi de Castria, Nicole L Nardella, Mokenge Malafa, Jason W Denbo, Jason B Fleming, Sarah E Hoffe, Jessica M Frakes, Andrew J Sinnamon, Jose M Pimiento, Pamela J Hodul, Dae Won Kim","doi":"10.3390/cancers17111797","DOIUrl":"10.3390/cancers17111797","url":null,"abstract":"<p><p><b>Background:</b> In patients with curatively resected pancreatic adenocarcinoma who have undergone neoadjuvant chemotherapy (NACT), evidence supporting the benefit of additional adjuvant chemotherapy (ACT) remains limited. We aim to identify favorable factors contributing to survival benefits in resected pancreatic adenocarcinoma after NACT. <b>Methods</b>: This is a retrospective cohort study of pancreatic adenocarcinoma patients who underwent NACT followed by curative surgical resection between 2008 and 2023 at a single academic institution. Univariate and multivariable analyses were conducted to identify factors contributing to disease-free survival (DFS) and overall survival (OS). <b>Results</b>: A total of 230 patients with a median age of 68 years (IQR, 62-72 years) were included. All patients underwent curative surgical resection. Of these, 42% received neoadjuvant modified (m) FOLFIRINOX (96/230), 15% received gemcitabine plus nab-paclitaxel (GEM-NAB) (34/230), and 43% received gemcitabine, docetaxel, and capecitabine (GTX) (100/230). In univariate analysis, lower College of American Pathologists (CAP) tumor regression grade (TRG) (0-1 vs. 2-3, median DFS: 29.8 vs. 14.2 months, <i>p</i> = 0.0081) and receipt of ACT (Yes vs. No, median DFS: 22.2 vs. 12.4 months, <i>p</i> < 0.0001) demonstrated significant associations with superior DFS. Multivariable analysis identified receipt of ACT as an independent predictor of superior DFS (HR 0.55, 95% CI: 0.39-0.78, <i>p</i> = 0.0007) and OS (HR 0.49, 95% CI: 0.33-0.71, <i>p</i> = 0.0002). However, the NACT regimen (mFOLFIRINOX vs. GEM-NAB) and the transition between neoadjuvant and adjuvant therapy (de-escalation vs. continuation vs. change) did not correlate with DFS or OS. The duration of perioperative chemotherapy showed a trend toward improved survival outcomes, though not statistically significant (6 months vs. <6 months: DFS, 19.4 vs. 16.2 months, <i>p</i> = 0.1448; OS, 49.6 vs. 30.4 months, <i>p</i> = 0.0623). In the following subgroup analyses, receipt of ACT provided DFS/OS benefits in patients who did not achieve a major pathologic response, pN0, or R0 resection (DFS: <i>p</i> = 0.0003; OS: <i>p</i> < 0.0001). However, it did not provide DFS/OS benefits in those who achieved a major pathologic response with pN0/R0 to NACT (DFS: <i>p</i> = 0.8036; OS: <i>p</i> = 0.1877). <b>Conclusions</b>: In resected pancreatic adenocarcinoma following NACT, receiving ACT was associated with favorable survival outcomes. Additional ACT appears to benefit patients who did not achieve a major pathologic response (pN0 or R0) to neoadjuvant therapy, with limited benefit for those who achieved a major response with pN0/R0. The specific NACT regimen (mFOLFIRINOX vs. GEM-NAB) and changes in ACT from NACT did not significantly influence survival outcomes in our cohort.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153592/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282475","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-28DOI: 10.3390/cancers17111796
Ramesh Paudyal, Alfonso Lema-Dopico, Akash Deelip Shah, Vaios Hatzoglou, Muhammad Awais, Eric Aliotta, Victoria Yu, Thomas L Chenevert, Dariya I Malyarenko, Lawrence H Schwartz, Nancy Lee, Amita Shukla-Dave
{"title":"Correction of Gradient Nonlinearity Bias in Apparent Diffusion Coefficient Measurement for Head and Neck Cancers Using Single- and Multi-Shot Echo Planar Diffusion Imaging.","authors":"Ramesh Paudyal, Alfonso Lema-Dopico, Akash Deelip Shah, Vaios Hatzoglou, Muhammad Awais, Eric Aliotta, Victoria Yu, Thomas L Chenevert, Dariya I Malyarenko, Lawrence H Schwartz, Nancy Lee, Amita Shukla-Dave","doi":"10.3390/cancers17111796","DOIUrl":"10.3390/cancers17111796","url":null,"abstract":"<p><p><b>Background/Objectives:</b> This work prospectively evaluates the vendor-provided Low Variance (LOVA) apparent diffusion coefficient (ADC) gradient nonlinearity correction (GNC) technique for primary tumors, neck nodal metastases, and normal masseter muscles in patients with head and neck cancers (HNCs). <b>Methods</b>: Multiple b-value diffusion-weighted (DW)-MR images were acquired on a 3.0 T scanner using a single-shot echo planar imaging (SS-EPI) and multi-shot (MS)-EPI for diffusion phantom materials (20% and 40% polyvinylpyrrolidone (PVP) in water). Pretreatment DW-MRI acquisitions were performed for sixty HNC patients (n = 60) who underwent chemoradiation therapy. ADC values with and without GNC were calculated offline using a monoexponential diffusion model over all b-values, relative percentage (r%) changes (Δ) in ADC values with and without GNC were calculated, and the ADC histograms were analyzed. <b>Results</b>: Mean ADC values calculated using SS-EPI DW data with and without GNC differed by ≤1% for both PVP20% and PVP40% at the isocenter, whereas off-center differences were ≤19.6% for both concentrations. A similar trend was observed for these materials with MS-EPI. In patients, the mean rΔADC (%) values measured with SS-EPI differed by 4.77%, 3.98%, and 5.68% for primary tumors, metastatic nodes, and masseter muscle. MS-EPI exhibited a similar result with 5.56%, 3.95%, and 4.85%, respectively. <b>Conclusions</b>: This study showed that the GNC method improves the robustness of the ADC measurement, enhancing its value as a quantitative imaging biomarker used in HNC clinical trials.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153906/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282482","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CancersPub Date : 2025-05-28DOI: 10.3390/cancers17111810
Nabiha Midhat Ansari, Usman Khalid, Daniel Markov, Kristian Bechev, Vladimir Aleksiev, Galabin Markov, Elena Poryazova
{"title":"AI-Augmented Advances in the Diagnostic Approaches to Endometrial Cancer.","authors":"Nabiha Midhat Ansari, Usman Khalid, Daniel Markov, Kristian Bechev, Vladimir Aleksiev, Galabin Markov, Elena Poryazova","doi":"10.3390/cancers17111810","DOIUrl":"10.3390/cancers17111810","url":null,"abstract":"<p><strong>Background: </strong>Endometrial cancer (EC) is the most common gynecological malignancy in developed countries, with diagnostic accuracy and early detection being critical to patient outcomes. Recent advances in artificial intelligence (AI) offer new opportunities to enhance diagnostic precision and clinical decision-making.</p><p><strong>Objectives: </strong>This literature review aims to explore recent developments in AI-augmented diagnostic approaches for EC, with a focus on applications in histopathology, imaging, and multi-omics, and to assess their clinical impact and future potential.</p><p><strong>Methods: </strong>A non-systematic literature review was conducted to examine recent advances in artificial intelligence applications for the diagnosis of EC. Relevant studies were identified through searches on PubMed and Google Scholar, focusing on the integration of AI techniques in histopathology, imaging, and multi-omics data.</p><p><strong>Conclusions: </strong>AI-driven diagnostic tools have shown high performance in detecting and characterizing EC across multiple modalities, often matching or exceeding expert-level accuracy. These technologies hold promise for earlier detection, better risk assessment, and more personalized treatment planning. However, further research and validation are needed to address current limitations and support their broader integration into clinical workflows.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"17 11","pages":""},"PeriodicalIF":4.5,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12153548/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282471","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}