1
|
Tacelli M, Gentiluomo M, Biamonte P, Castano JP, Berković MC, Cives M, Kapitanović S, Marinoni I, Marinovic S, Nikas I, Nosáková L, Pedraza-Arevalo S, Pellè E, Perren A, Strosberg J, Campa D, Capurso G. Pancreatic neuroendocrine neoplasms (pNENs): Genetic and environmental biomarkers for risk of occurrence and prognosis. Semin Cancer Biol 2025; 112:112-125. [PMID: 40158764 DOI: 10.1016/j.semcancer.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Pancreatic neuroendocrine neoplasms (pNENs) are rare and heterogeneous tumors arising from neuroendocrine cells, representing approximately 10 % of all Gastro-Entero-Pancreatic neuroendocrine neoplasms. While most pNENs are sporadic, a subset is associated with genetic syndromes such as multiple endocrine neoplasia type 1 (MEN1) or von Hippel-Lindau disease (VHL). pNENs are further classified into functioning and non-functioning tumors, with distinct clinical behaviors, prognoses, and treatment approaches. This review explores genetic and environmental biomarkers that influence the risk, prognosis, and therapeutic responses in pNENs. The epidemiology of pNENs reveals an increasing incidence, primarily due to advancements in imaging techniques. Genetic factors play a pivotal role, with germline mutations in MEN1, VHL, and other genes contributing to familial pNENs. Somatic mutations, including alterations in the mTOR pathway and DNA maintenance genes such as DAXX and ATRX, are critical in sporadic pNENs. These mutations, along with epigenetic dysregulation and transcriptomic alterations, underpin the diverse clinical and molecular phenotypes of pNENs. Emerging evidence suggests that epigenetic changes, including DNA methylation profiles, can stratify pNEN subtypes and predict disease progression. Environmental and lifestyle factors, such as diabetes, smoking, and chronic pancreatitis, have been linked to an increased risk of sporadic pNENs. While the association between these factors and tumor progression is still under investigation, their potential role in influencing therapeutic outcomes warrants further study. Advances in systemic therapies, including somatostatin analogs, mTOR inhibitors, and tyrosine kinase inhibitors, have improved disease management. Biomarkers such as Ki-67, somatostatin receptor expression, and O6-methylguanine-DNA methyltransferase (MGMT) status are being evaluated for their predictive value. Novel approaches, including the use of circulating biomarkers (NETest, circulating tumor cells, and ctDNA) and polygenic risk scores, offer promising avenues for non-invasive diagnosis and monitoring. Despite these advancements, challenges remain, including the need for large, well-annotated datasets and validated biomarkers. Future research should integrate multi-omics approaches and leverage liquid biopsy technologies to refine diagnostic, prognostic, and therapeutic strategies. Interdisciplinary collaborations and global consortia are crucial for overcoming current limitations and translating research findings into clinical practice. These insights hold promise for improving prevention, early detection, and tailored treatments, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Matteo Tacelli
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Paolo Biamonte
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Justo P Castano
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Maja Cigrovski Berković
- Department for Sport and Exercise Medicine, Faculty of Kinesiology University of Zagreb, Zagreb 10000, Croatia
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy; Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Sanja Kapitanović
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilaria Marinoni
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Sonja Marinovic
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb 10000, Croatia
| | - Ilias Nikas
- Medical School, University of Cyprus, Nicosia, Cyprus
| | - Lenka Nosáková
- Clinic of Internal Medicine - Gastroenterology, JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Bratislava, Slovakia
| | - Sergio Pedraza-Arevalo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Reina Sofia University Hospital, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Eleonora Pellè
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Aurel Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jonathan Strosberg
- Department of GI Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
2
|
Moon JM, Lee SW, Jang YS, Lee SA, Jung SH, Kim SK, Park BK, Park YS, Kim BS, Yang MS, Jung JY. Gossypin induces apoptosis and autophagy via the MAPK/JNK pathway in HT‑29 human colorectal cancer cells. Int J Mol Med 2025; 56:107. [PMID: 40376978 PMCID: PMC12101101 DOI: 10.3892/ijmm.2025.5548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/09/2025] [Indexed: 05/18/2025] Open
Abstract
Gossypin, a flavone found in Hibiscus vitifolius, exhibits antioxidant, antidiabetic, anti‑inflammatory and anticancer effects. The present study investigated the potential of gossypin to induce apoptosis and autophagy in HT‑29 human colorectal cancer (CRC) cells, and assessed its association with the MAPK/JNK pathway. Cell viability assays, DAPI staining, flow cytometry, acridine orange staining, western blotting, hematoxylin and eosin staining, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and immunohistochemistry were performed. The results revealed an increased number of apoptotic bodies, higher apoptosis rates and enhanced autophagy in gossypin‑treated HT‑29 cells. To investigate autophagy during cell death, the effects of the early autophagy inhibitor 3‑methyladenine (3‑MA) and the late autophagy inhibitor hydroxychloroquine on cell viability and the expression of apoptosis‑related proteins were assessed. Significant increases in cell viability were observed following 3‑methyladenine pretreatment, as well as a decrease in the expression levels of Bcl‑2 and an increase in Bax. The analysis of MAPK pathway proteins following treatment with gossypin revealed that the levels of phosphorylated (p‑)JNK and p‑p38 were significantly increased in a concentration‑dependent manner. The JNK inhibitor SP600125 was used to confirm the role of the JNK pathway in gossypin‑induced apoptosis and autophagy. Moreover, gossypin reduced the volume of HT‑29 tumors in mice, and western blotting indicated the induction of apoptosis and autophagy in these tumors in vivo. Finally, TUNEL and immunohistochemistry experiments confirmed the induction of apoptosis and p‑JNK upregulation in these tumors in vivo. In conclusion, the present study suggested that gossypin may induce MAPK/JNK‑mediated apoptosis and autophagy in HT‑29 CRC cells, highlighting the potential of gossypin as an anticancer agent.
Collapse
Affiliation(s)
- Jun-Mo Moon
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Sang-Woo Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Yun-Seo Jang
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Su-A Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Sang-Ki Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
- Research Institute for Natural Products, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Byung-Kwon Park
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
- Research Institute for Natural Products, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Young-Seok Park
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
- Research Institute for Natural Products, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Byeong-Soo Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
- Research Institute for Natural Products, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Myeon-Sik Yang
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
- Research Institute for Natural Products, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
- Research Center of Crop Breeding for Omics and Artificial Intelligence, Kongju National University, Yesan, Chungcheongnam 32439, Republic of Korea
| |
Collapse
|
3
|
Raeisi H, Patrizia S, Sadeghi A, Barbanti F, Tillotson G, Safarpour H, Zali M, Nazemalhosseini Mojarad E. Risk factors and outcomes of Clostridioides difficile infection in patients with colorectal cancer: critical perspective in management. Gut Pathog 2025; 17:44. [PMID: 40517235 DOI: 10.1186/s13099-025-00717-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 05/30/2025] [Indexed: 06/16/2025] Open
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer worldwide, causing a serious threat to global health and social burden. Clostridioides difficile infection (CDI) is one of the most important nosocomial infections and has a higher incidence in cancerous population compared with non-cancerous cases. Different risk factors, including gut microbiota dysbiosis, extensive surgery, chemotherapy, prolonged hospitalization, and antimicrobial therapy, compromise host defenses against CDI and contribute to cancer patients' susceptibility to this infection. The emergence of CDI in patients with CRC creates conditions for therapy escalation and prolonged hospitalization, highlighting the need for correct and effective CDI management in these patients. Here, common risk factors associated with CDI in patients with CRC are discussed. In addition, different available techniques for the prevention, detection, and treatment of CDI with the lowest impact on gut microbiota diversity are summarized. This review aims to improve the understanding of the interplay between CDI and CRC and provide new insights into restoring and maintaining gut microbiota balance during CDI management in patients with CRC.
Collapse
Affiliation(s)
- Hamideh Raeisi
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Spigaglia Patrizia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fabrizio Barbanti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy
| | | | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohamadreza Zali
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
4
|
Mansoori R, Ashrafpour M, Asghari MH, Golchoobian R, Hosseini SM, Reiter RJ, Karim B, Moghadamnia AA, Kazemi S. Protective effects of melatonin against 5-fluorouracil-induced cardiotoxicity in rats: A comprehensive evaluation of oxidative, inflammatory, and apoptotic pathways. Toxicol Appl Pharmacol 2025; 499:117343. [PMID: 40239743 DOI: 10.1016/j.taap.2025.117343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Cardiotoxicity is a serious adverse effect of 5-fluorouracil (5-FU) a common chemotherapeutic agent. This study aimed to evaluate the protective effects of melatonin (MLT) against 5-fluorouracil (5-FU)-induced cardiotoxicity in rats, focusing on oxidative stress, inflammatory pathways, gene expression, electrocardiographic and histopathological changes. MATERIALS AND METHODS Twenty-five male Wistar rats were divided into five groups. The animals received either MLT at doses of 2.5, 5, or 10 mg/kg/day, 5-FU at 50 mg/kg (i.p.), or a combination of both treatments. Cardiotoxicity was assessed through electrocardiography, cardiac enzymes, oxidative stress markers, and histopathology. RESULTS 5-FU treatment significantly increased oxidative stress markers and inflammatory mediators while causing histopathological damage in heart tissues. Co-administration of MLT with 5-FU significantly mitigated these effects by reducing oxidative damage, as evidenced by lower levels of malondialdehyde (MDA), nitric oxide (NO), and myeloperoxidase (MPO). Additionally, MLT enhanced antioxidant activity, as reflected by increased levels of superoxide dismutase (SOD) and glutathione peroxidase (GPx) in heart tissues. Gene expression analysis further confirmed that MLT treatment reduced the elevated levels of COX-2 and VEGF, which are critical players in the inflammatory process. Histopathological examination demonstrated that MLT preserved the structural integrity of myocardial tissues, reducing 5-FU-induced damage score in a dose-dependent manner. Furthermore, MLT co-administration significantly attenuated the rise in cardiac biomarkers, including LDH, AST, and CK-MB, associated with 5-FU-induced cardiotoxicity. CONCLUSION These findings highlight that MLT, through its antioxidant and anti-inflammatory properties, exerts a protective effect against 5-FU-induced toxicity, suggesting its therapeutic potential for improving cardiovascular health during chemotherapy.
Collapse
Affiliation(s)
- Razieh Mansoori
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Manoochehr Ashrafpour
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Hossien Asghari
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ravieh Golchoobian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Bardia Karim
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Pharmaceutical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
5
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
6
|
Meier J, Murimwa G, Nehrubabu M, Yopp A, DiMartino L, Singal AG, Zeh HJ, Polanco P. Defining the Role of Social Vulnerability in the Treatment and Survival of Localized Colon Cancer: A Retrospective Cohort Study. Ann Surg 2025; 281:1055-1062. [PMID: 38545790 PMCID: PMC11436472 DOI: 10.1097/sla.0000000000006282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
OBJECTIVE To determine whether variations in the Social Vulnerability Index (SVI) are associated with disparities in colon cancer surgery and mortality. BACKGROUND Colon cancer mortality is influenced by health care access, which is affected by individual and community-level factors. Prior studies have not used the SVI to compare surgical access and survival in localized colon cancer patients. Further, it is unclear whether those >65 years are more vulnerable to variations in SVI. METHODS We queried the Texas and California Cancer Registries from 2004 to 2017 to identify patients with localized colonic adenocarcinoma and categorized patients into <65 and ≥65 years. Our outcomes were survival and access to surgical intervention. The independent variable was census tract SVI, with higher scores indicating more social vulnerability. We used multivariable logistic regression and Cox proportional hazards for analysis. RESULTS We included 73,923 patients with a mean age of 68.6 years (SD: 13.0), mean SVI of 47.2 (SD: 27.6), and 51.1% males. After adjustment, increasing SVI was associated with reduced odds of undergoing surgery (odds ratio: 0.996; 95% CI: 0.995-0.997; P < 0.0001) and increased mortality (hazard ratio: 1.002; 95% CI: 1.001-1.002; P < 0.0001). Patients <65 years were more sensitive to variation in SVI. CONCLUSIONS Increased social vulnerability was associated with reduced odds of receiving surgery for early-stage colon cancer, as well as increased mortality. These findings amplify the need for policy changes at the local, state, and federal levels to address community-level vulnerability to improve access to surgical care and reduce mortality.
Collapse
Affiliation(s)
- Jennie Meier
- Department of Surgery, University of Texas Southwestern, Dallas, TX
| | - Gilbert Murimwa
- Department of Surgery, University of Texas Southwestern, Dallas, TX
| | - Mithin Nehrubabu
- Department of Mathematics, University of Texas at Dallas, Dallas, TX
| | - Adam Yopp
- Department of Surgery, University of Texas Southwestern, Dallas, TX
| | - Lisa DiMartino
- Department of Public Health, University of Texas Southwestern, Peter O'Donnell Jr. School of Public Health, Dallas, TX
| | - Amit G Singal
- Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern, Dallas, TX
| | - Herbert J Zeh
- Department of Surgery, University of Texas Southwestern, Dallas, TX
| | - Patricio Polanco
- Department of Surgery, University of Texas Southwestern, Dallas, TX
| |
Collapse
|
7
|
Kim S, Kim S, Ko C, Lee W, Kim HD. A microfluidic electrochemical immunosensor for detection of CEA and Ki67 in 3D tumor spheroids. Mater Today Bio 2025; 32:101768. [PMID: 40290895 PMCID: PMC12022681 DOI: 10.1016/j.mtbio.2025.101768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/27/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Microfluidic chip-based electrochemical sensors have been developed to detect cancer biomarkers and monitor changes in the tumor microenvironment. However, the limitation of detecting only a single biomarker restricts their utility as accurate diagnostic tools. Simultaneous detection of multiple tumor biomarkers is important for early diagnosis of cancer. In this work, we report the development of a microfluidic-based electrochemical immunosensor platform capable of simultaneously observing multiple biomarkers expressed by three dimensions (3D) cell spheroids. The sensor platform employs a nanocomposite electrode material consisting of gold nanoparticles and carbon nanotubes, which enables sensitive and selective detection. The sensor was fabricated using 3D and printed circuit boards (PCB) printing techniques, demonstrating the feasibility of cost-effective manufacturing. The developed platform was able to quantitatively detect two key cancer biomarkers, carcinoembryonic antigen (CEA) and Ki67, with limits of detection of 0.97 ng/mL for each. Furthermore, the sensor was successfully utilized to observe the knockdown of these biomarkers, showcasing its potential for both diagnostic and therapeutic monitoring applications. These results suggest that the presented electrochemical sensor platform provides a promising lab-on-a-chip technology for comprehensive 3D cell spheroid-based cancer biomarker analysis, which could have significant implications for future clinical diagnostics and personalized medicine.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Seonyeop Kim
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Chanjin Ko
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Wonseok Lee
- Department of IT-Energy Convergence (BK21 Four), Chemical Industry Institute, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
- Department of Electrical Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Hwan Drew Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| |
Collapse
|
8
|
Wang R, Ren B, Zhang X, Liu B, Zhou W. Identification of AKTIP as a biomarker for fibrolamellar carcinoma using WGCNA and machine learning. 3 Biotech 2025; 15:181. [PMID: 40417661 PMCID: PMC12095112 DOI: 10.1007/s13205-025-04323-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/11/2025] [Indexed: 05/27/2025] Open
Abstract
Fibrolamellar carcinoma (FLC) is a rare form of liver carcinoma with limited diagnostic and therapeutic options. In this study, we utilized the GSE57727 and E-MTAB-1503 datasets, downloaded from GEO and ArrayExpress, respectively, to explore hub genes for FLC diagnosis and potential therapeutic agents. Through the integration of multiple machine learning approaches and drug sensitivity databases, we identified AKTIP as a potential diagnostic biomarker for FLC. AKTIP exhibited markedly elevated expression in FLC compared to non-FLC, demonstrating superior diagnostic and prognostic performance over other FLC-specific biomarkers. Four compounds (PI-103, BVT-948, Digitoxigenin, and SB-218078) were identified as potential therapeutic agents targeting AKTIP. Molecular docking analysis revealed strong binding affinities of these compounds to AKTIP, and molecular dynamics simulations further validated the reliability and rationality of the molecular docking results. Pan-cancer analysis indicated that AKTIP expression varies across different tissues and is significantly associated with patient prognosis. qRT-PCR analysis confirmed that AKTIP mRNA levels were markedly overexpressed in normal liver epithelial cells compared to human hepatocellular carcinoma cell lines. In conclusion, AKTIP was successfully identified as a diagnostic and prognostic biomarker for FLC, and four compounds were proposed as potential therapeutic agents. This study uncovers new perspectives on diagnosing and managing of this rare type of liver carcinoma, offering promising avenues for future research and clinical applications. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04323-4.
Collapse
Affiliation(s)
- Rui Wang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030 China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730030 China
| | - Bo Ren
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030 China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730030 China
| | - Xijie Zhang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030 China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730030 China
| | - Bo Liu
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030 China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730030 China
| | - Wence Zhou
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030 China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, 730030 China
- Gansu Province Hepatobiliary Pancreatic Disease Precision Diagnosis and Treatment Engineering Research Center, Lanzhou, 730030 China
| |
Collapse
|
9
|
Abdelrahim M, Esmail A, Kim RD, Arora SP, Arshad J, Kournoutas IA, O’Donnell CD, Totev TI, Tan A, Mu F, Seshasayee SM, Hernandez S, Tran NH. Real-World Experiences Using Atezolizumab + Bevacizumab for the Treatment of Unresectable Hepatocellular Carcinoma: A Multicenter Study. Cancers (Basel) 2025; 17:1814. [PMID: 40507295 PMCID: PMC12153728 DOI: 10.3390/cancers17111814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/21/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Objective: 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. Methods: 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). Results: 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 (n = 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. Conclusions: 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.
Collapse
Affiliation(s)
- Maen Abdelrahim
- Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Abdullah Esmail
- Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Richard D. Kim
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Sukeshi Patel Arora
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| | - Junaid Arshad
- University of Arizona Cancer Center, Tucson, AZ 85719, USA;
| | | | | | - Todor I. Totev
- Analysis Group, Inc., Boston, MA 02199, USA; (T.I.T.); (F.M.); (S.M.S.)
| | - Amie Tan
- Genentech, South San Francisco, CA 94080, USA; (A.T.); (S.H.)
| | - Fan Mu
- Analysis Group, Inc., Boston, MA 02199, USA; (T.I.T.); (F.M.); (S.M.S.)
| | | | - Sairy Hernandez
- Genentech, South San Francisco, CA 94080, USA; (A.T.); (S.H.)
| | - Nguyen H. Tran
- Mayo Clinic, Rochester, MN 55905, USA; (I.A.K.); (N.H.T.)
| |
Collapse
|
10
|
Shaernejad S, Nosrat A, Baeeri M, Hashemi Goradel N, SeyedSadeghi M, Akbariani M, Arabzadeh A, Rahimifard M, Haghi-Aminjan H. Role of hesperidin/hesperetin against chemotherapy-induced cardiotoxicity: a systematic review of non-clinical studies. Cancer Cell Int 2025; 25:186. [PMID: 40405281 PMCID: PMC12100833 DOI: 10.1186/s12935-025-03828-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 05/11/2025] [Indexed: 05/24/2025] Open
Abstract
Despite the undeniable role of chemotherapeutics in cancer treatment, their administration may be associated with various side effects. Cardiac injury is among the most crucial side effects related to the induction of chemotherapeutic agents. Since the heart is a vital organ, cardiotoxicity often prevents clinicians from continuing chemotherapy. Hesperidin and hesperetin, flavonoids derived from citrus fruits, possess several pharmaceutical properties. This study firstly explores the cardioprotective effects of hesperidin and hesperetin against chemotherapy-induced cardiotoxicity mechanisms, emphasizing their potential as adjunctive therapies. Key literature gaps are identified, and further mechanistic studies will be proposed. The findings underscore the translational potential of these flavonoids, advocating for rigorous preclinical optimization and clinical trials to validate their efficacy and safety. This review lays a foundation for integrating natural compounds into cardioprotective strategies in oncology. A systematic search was conducted in databases (PubMed, Scopus, ISI) until May 2025, according to PRISMA principles. The search terms were chosen according to our research objective and queried in the title and abstract. Following the screening of 82 papers, twelve articles were selected based on our inclusion and exclusion criteria. Based on the evaluated results, chemotherapy adversely affects cardiac tissue, leading to elevated risks of morbidity and mortality. Co-administration of hesperidin and hesperetin with chemotherapy prevents heart injury and preserves cardiac function, maintaining it almost like a normal heart. The protective role of hesperidin and hesperetin is based on their ability to fight free radicals, reduce inflammation, and stop cell death. Nonclinical investigations indicate that hesperidin and hesperetin ameliorate chemotherapy-induced cardiotoxicity. Nonetheless, they may influence the efficacy of anticancer medications, which primarily function by elevating oxidants, inflammation, and apoptosis. This indicates that meticulously designed trials are necessary to evaluate the efficacy and safety of this combination along with the synergistic potential of them in preventing chemotherapy-induced cardiotoxicity while maintaining anticancer effectiveness.
Collapse
Affiliation(s)
- Sina Shaernejad
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Nosrat
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Baeeri
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mirsalim SeyedSadeghi
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mostafa Akbariani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - AmirAhmad Arabzadeh
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mahban Rahimifard
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Hamed Haghi-Aminjan
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
11
|
Chen E, Zhou W. Immunotherapy in microsatellite-stable colorectal cancer: Strategies to overcome resistance. Crit Rev Oncol Hematol 2025; 212:104775. [PMID: 40409481 DOI: 10.1016/j.critrevonc.2025.104775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025] Open
Abstract
Colorectal cancer (CRC) is among the foremost causes of cancer-related mortality worldwide; however, individuals with microsatellite-stable (MSS) disease-who constitute most CRC diagnoses-derive limited benefit from existing immunotherapeutic approaches. Here, we outline emerging methods designed to address the inherent resistance of MSS CRC to immune checkpoint inhibitors (ICIs). Recent findings emphasize how the immunosuppressive tumor microenvironment (TME) in MSS CRC, marked by diminished immunogenicity and high levels of regulatory T cells and myeloid-derived suppressor cells, restricts effective antitumor immune activity. Combination regimens that merge ICIs with chemotherapy, anti-angiogenic agents, or targeted blockade of pathways such as TGF-β and VEGF have shown encouraging early outcomes, including enhanced antigen presentation and T-cell penetration. Novel immunomodulatory platforms-such as epigenetic modifiers, oncolytic viruses, and engineered probiotic vaccines-are under assessment to further reprogram the TME and boost therapeutic efficacy. Concurrently, progress in adoptive cell therapies (for example, chimeric antigen receptor (CAR) T cells) and the development of cancer vaccines targeting tumor-associated and neoantigens promise to extend immune control over MSS CRC. In parallel, improving patient selection through predictive biomarkers-from circulating tumor DNA (ctDNA) to gene expression signatures and specific molecular subtypes-could refine individualized treatment strategies. Finally, interventions that alter the gut microbiome, including probiotics and fecal transplantation, serve as complementary tools to strengthen ICI responses. Taken together, these insights and combined treatment strategies lay the foundation for more successful immunotherapeutic interventions in MSS CRC, ultimately aiming to provide sustained clinical benefits to a broader spectrum of patients.
Collapse
Affiliation(s)
- Engeng Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou 310016, China
| | - Wei Zhou
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
12
|
Throat S, Bhattacharya S. The Role of RS Type 2 (High-Amylose Maize Starch) in the Inhibition of Colon Cancer: A Comprehensive Review of Short-Chain Fatty Acid (SCFA) Production and Anticancer Mechanisms. Mol Nutr Food Res 2025:e70107. [PMID: 40392033 DOI: 10.1002/mnfr.70107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/30/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Dietary fiber, especially resistant starch (RS) Type 2 (RS2) found in high-amylose maize starch (HAMS), is vital for gut health and helps prevent colon cancer. In contrast to most nutrients, dietary fiber is not degraded by the intestinal enzymes; it reaches the distal parts of the gut, where it is fermented by the gut microbiota into short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate. SCFAs energize colonocytes, reduce inflammation, and enhance gut immunity. HAMS is absorbed in the colon, where it ferments to create SCFAs that feed good gut flora and have antiinflammatory and antiproliferative effects. RS2 in HAMS modulates gene signaling, activates tumor-suppressor genes like tumor suppressor protein (p53), exhibits antidiabetic, cholesterol-lowering, and antiinflammatory effects. Incorporation of RS2-rich sources enhances gut barriers, decreases colorectal cancer biomarkers, and counteracts the negative impacts of low-fiber Western diets, making HAMS a promising functional food for chronic disease prevention and health promotion.
Collapse
Affiliation(s)
- Siddhi Throat
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Sankha Bhattacharya
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| |
Collapse
|
13
|
Pedersen RS, Hettich A, Thorlacius-Ussing J, Langholm LL, Crespo-Bravo M, Chen IM, Hansen CP, Johansen JS, Diab HMH, Jorgensen LN, Karsdal M, Willumsen N. Proteolytic degradation of Beta-Ig H3 (βigH3/TGFBI) can be quantified non-invasively in serum and predicts prognosis in patients with advanced pancreatic ductal adenocarcinoma. BMC Cancer 2025; 25:905. [PMID: 40394523 DOI: 10.1186/s12885-025-14283-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
The extracellular matrix (ECM) protein Beta-Ig H3 (βigH3, also known as transforming growth factor β induced protein (TGFBI)) is related to poor prognosis in patients with pancreatic ductal adenocarcinoma (PDAC). Proteolytic cleavage of βigH3 has been shown to result in release of the N-terminal fragment covering amino acid 1 to 137, but whether the degradation of βigH3 is associated to prognosis has yet to be determined. In this study we developed an ELISA targeting a collagenase generated fragment of βigH3 (cβigH3) in human serum to use the fragment as a biomarker reflecting degradation of βigH3. We demonstrated that the assay was specific to the cleaved fragment (cβigH3) and confirmed the generation of cβigH3 from degradation of fibroblast generated matrices. Moreover, higher levels of cβigH3 were released upon degradation of matrices produced by TGF-β stimulated pancreatic fibroblast compared to matrices produced by pancreatic fibroblast without TGF-β stimulation, indicating an association of the biomarker with degradation of fibrotic matrix. To evaluate the clinical relevance, we first measured cβigH3 in a cohort of 220 patients with different types of cancer with detectable levels for all 11 cancer types. We then measured the cβigH3 biomarker in pre-treatment serum from a cohort of 469 patients with locally advanced or metastatic PDAC and found that high levels of cβigH3 were associated with longer overall survival independently of age, disease stage, performance status, carbohydrate antigen 19-9 (CA19-9), and the tumor fibrosis biomarker PRO-C3 as compared to patients with high levels of cβigH3 (HR 0.78, 95% CI: 0.0.61-0.98, p = 0.04). In conclusion, cβigH3 reflects proteolytic degradation of βigH3 and shows potential as an independent prognostic biomarker for patients with advanced PDAC.
Collapse
Affiliation(s)
- Rasmus S Pedersen
- Nordic Bioscience A/S, 2730, Herlev, Denmark.
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Annika Hettich
- Nordic Bioscience A/S, 2730, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | | | | | - Marina Crespo-Bravo
- Nordic Bioscience A/S, 2730, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, 2730, Herlev, Denmark
| | - Carsten P Hansen
- Department of Surgery, Copenhagen University Hospital - Rigshospitalet, 2100, Copenhagen, Denmark
| | - Julia S Johansen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, 2730, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital - Herlev and Gentofte, 2730, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Hadi M H Diab
- Digestive Disease Center, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400, Copenhagen, Denmark
| | - Lars N Jorgensen
- Digestive Disease Center, Copenhagen University Hospital - Bispebjerg and Frederiksberg, 2400, Copenhagen, Denmark
| | | | | |
Collapse
|
14
|
Lyhne JD, Hansen VB, Vestergaard LD, Hosbond SE, Busk M, Gnanaganesh M, Maae E, Havelund BM, Hansen TF, Timm S, Jensen LH, Lyhne MD. Primary prevention of cardiotoxicity in cancer patients treated with fluoropyrimidines: a randomized controlled trial. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:48. [PMID: 40382663 DOI: 10.1186/s40959-025-00344-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Fluoropyrimidines (FP) are the third most used chemotherapeutic drugs administered in solid tumors but have cardiotoxic side effects. We aimed to determine whether pre-chemotherapeutic cardiological assessment and management of cardiovascular risk factors could prevent FP-induced cardiotoxicity and if the coronary artery calcium (CAC) score was predictive of chest pain. METHODS This was a randomized, controlled, single center trial of patients with various cancer types who were treated with FP and had no known ischemic heart disease. All patients had CAC score obtained by cardiac CT scan. Patients were randomized to pre-chemotherapeutic cardiological management or standard care. Cardiological management included risk reduction based on electro- and echocardiographic evaluation and blood samples. Primary composite endpoint included hospital admission for chest pain, acute coronary syndrome, coronary angiography intervention, or all-cause mortality. Secondary outcome was chest pain. Follow-up was 6 months. Data were analyzed using Kaplan-Meier survival function with log-rank test and ROC-analyses. RESULTS Of the 192 patients included, the primary endpoint occurred in 9/95 (9.5%) patients in the intervention group and 15/97 (15.5%) patients in the control group (log-rank p = 0.19) with an incidence rate ratio (IRR) of 0.57 (95% CI [0.22 - 1.39]). Chest pain occurred in 6/95 (6.3%) patients in the intervention group and 13/97 (13.4%) in the control group, yielding an IRR of 0.44 (95% CI [0.14 - 1.23]). CAC score did not predict chest pain occurrence. CONCLUSIONS Cardiological management of cardiovascular risk factors prior to treatment with fluoropyrimidines resulted in half as many cardiotoxic events but the study did not reach statistical significance. Further studies are needed to investigate the optimal strategies to prevent fluoropyrimidine-induced cardiotoxicity in cancer patients. TRIAL REGISTRATION ClinicalTrials.gov Identifyer NCT03486340.
Collapse
Affiliation(s)
- Johanne D Lyhne
- Department of Oncology, Lillebaelt Hospital -Vejle, University Hospital of Southern Denmark, Beriderbakken 4, Vejle, 7100, Denmark.
| | - Vibeke B Hansen
- Department of Cardiology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Lone D Vestergaard
- Department of Cardiology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Susanne E Hosbond
- Department of Cardiology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Martin Busk
- Department of Cardiology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Mayooran Gnanaganesh
- Department of Cardiology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Else Maae
- Department of Oncology, Lillebaelt Hospital -Vejle, University Hospital of Southern Denmark, Beriderbakken 4, Vejle, 7100, Denmark
| | - Birgitte M Havelund
- Department of Oncology, Lillebaelt Hospital -Vejle, University Hospital of Southern Denmark, Beriderbakken 4, Vejle, 7100, Denmark
| | - Torben F Hansen
- Department of Oncology, Lillebaelt Hospital -Vejle, University Hospital of Southern Denmark, Beriderbakken 4, Vejle, 7100, Denmark
| | - Signe Timm
- Department of Oncology, Lillebaelt Hospital -Vejle, University Hospital of Southern Denmark, Beriderbakken 4, Vejle, 7100, Denmark
| | - Lars H Jensen
- Department of Oncology, Lillebaelt Hospital -Vejle, University Hospital of Southern Denmark, Beriderbakken 4, Vejle, 7100, Denmark
| | - Mads D Lyhne
- Department of Cardiology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Anello MP, Ward DD, Garrison OM, Gode A, Ioachimescu OC, Friedland DR, Shaker R. Public health emergency accelerated research response-the Clinical and Translational Science Institute of Southeast Wisconsin COVID-19 research initiative. Front Public Health 2025; 13:1529121. [PMID: 40416685 PMCID: PMC12098277 DOI: 10.3389/fpubh.2025.1529121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/04/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction In March 2020, the National Center for Advancing Translational Sciences-Clinical and Translational Science Awards (CTSA) Program issued an urgent "Call to Action," requesting CTSA hubs to accelerate clinical and translational research (C&TR) in response to the COVID-19 public health emergency. The Clinical and Translational Science Institute of Southeast Wisconsin (CTSI) quickly responded by launching a regional research initiative among its eight academic and healthcare partner institutions to nucleate teams around COVID-19 C&TR. Methods A comprehensive search of COVID-19 funding opportunities, combined with suggestions from CTSI leadership and C&TR investigators, produced a list of 31 distinct C&TR questions that were used to nucleate investigators into teams. A survey was shared with the faculty of all eight partner institutions to solicit interest in joining the teams. Multidisciplinary team formation was based on a novel CTSI model, called the "Team Science-Guided Integrated Clinical and Research Ensemble (Ensemble)." In this model, teams are formed around an unmet patient medical need, based on the intentional recruitment of members from three domains: (1) the clinical and translational research enterprise, (2) the health care systems, and (3) the community of stakeholders. The teams were provided no funding, but received substantial CTSI research and administrative support. Results Forty-one teams were formed, and 243 investigators participated during the first year of the initiative. Team efforts resulted in the submission of 21 grant proposals, totaling $32,528,297. Three grant proposals were funded, totaling $609,888. The research initiative generated eight publications and had a significant impact on patient health, involving a combined total of 456 research participants. The initiative led to several systemic improvements, by (1) exposing investigators to team science-guided C&TR (Ensembles), (2) increasing inter-institutional and inter-departmental collaborations, (3) creating new partnerships with community organizations, and (4) providing qualitative data on lessons learned. Conclusion The COVID-19 regional research initiative provided a compelling model of how basic science, clinical/translational, and community researchers can be mobilized for accelerated C&TR to address a public health threat. The initiative demonstrated that the fundamentals of the novel CTSI Ensemble team concept can be leveraged to expedite the formation of highly efficient teams.
Collapse
Affiliation(s)
- Michael P. Anello
- Clinical and Translational Science Institute of Southeast Wisconsin, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Doriel D. Ward
- Clinical and Translational Science Institute of Southeast Wisconsin, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Orsolya M. Garrison
- Clinical and Translational Science Institute of Southeast Wisconsin, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Amit Gode
- Clinical and Translational Science Institute of Southeast Wisconsin, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Octavian C. Ioachimescu
- Clinical and Translational Science Institute of Southeast Wisconsin, The Medical College of Wisconsin, Milwaukee, WI, United States
- Clement J. Zablocki Veteran Affairs Medical Center, Milwaukee, WI, United States
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - David R. Friedland
- Clinical and Translational Science Institute of Southeast Wisconsin, The Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Otolaryngology and Communication Sciences, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - Reza Shaker
- Clinical and Translational Science Institute of Southeast Wisconsin, The Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Division of Gastroenterology and Hepatology, The Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
16
|
Zhao K, Muralidharan V, Brown S, Upton A, Alshimirti M, Cooray PD. Neoadjuvant Pembrolizumab Enables Successful Downstaging and Resection of Borderline Resectable MSI-H/dMMR Pancreatic Ductal Adenocarcinoma: A Case Report and Literature Review. J Gastrointest Cancer 2025; 56:112. [PMID: 40341577 PMCID: PMC12062155 DOI: 10.1007/s12029-025-01237-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a poor prognosis. While immunotherapy has shown limited efficacy in most PDAC cases due to an immunosuppressive tumour microenvironment, tumours with microsatellite instability-high (MSI-H) or deficient mismatch repair (dMMR) status exhibit increased responsiveness to immune checkpoint inhibitors. CASE PRESENTATION We report the case of a 45-year-old woman with Lynch syndrome who was diagnosed with MSI-H/dMMR PDAC during routine surveillance. Given the borderline resectable nature of her tumour and previous chemotherapy-related neurotoxicity, she was treated with neoadjuvant pembrolizumab instead of conventional chemotherapy. Following four cycles of pembrolizumab, imaging revealed a marked metabolic response, allowing for successful R0 pancreatoduodenectomy. Postoperative histology confirmed a significant reduction in tumour size, and immunohistochemical analysis demonstrated increased CD8 + T cell infiltration, supporting an enhanced anti-tumour immune response. The patient continues adjuvant pembrolizumab therapy without complications. CONCLUSION This case highlights the potential role of neoadjuvant pembrolizumab in MSI-H/dMMR PDAC, demonstrating successful tumour downstaging and facilitating surgical resection. Our findings support further investigation into the integration of immunotherapy as a neoadjuvant strategy for select PDAC patients.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Female
- Middle Aged
- Neoadjuvant Therapy/methods
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/surgery
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/surgery
- Pancreatic Neoplasms/genetics
- Microsatellite Instability
- DNA Mismatch Repair
- Antineoplastic Agents, Immunological/therapeutic use
- Pancreaticoduodenectomy
- Colorectal Neoplasms, Hereditary Nonpolyposis
Collapse
Affiliation(s)
- Kevin Zhao
- University of Melbourne School of Medicine, Melbourne, Australia
| | - Vijayaragavan Muralidharan
- Prometheus Research Collaborative, Department of Surgery, Austin Precinct, The University of Melbourne, Austin Health, Melbourne, Australia
| | | | | | | | - Prasad D Cooray
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, Australia.
| |
Collapse
|
17
|
Cann C, LaPelusa M, Cimino S, Cancelliere V, Dow-Hillgartner E, Zhao Z, Deming D, Eng C. Biweekly dosing of trifluridine-tipiracil reduces rates of myelosuppression while maintaining efficacy in patients with metastatic colorectal cancer. Oncologist 2025; 30:oyaf099. [PMID: 40421955 PMCID: PMC12107540 DOI: 10.1093/oncolo/oyaf099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 04/09/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Trifluridine-tipiracil (TAS-102) is approved as monotherapy and in combination with bevacizumab for refractory unresectable metastatic colorectal cancer (mCRC). The recommended dose is 35 mg/m2 twice daily on days 1-5 and days 8-12 of 28-day cycles commonly resulting in grade 3-4 neutropenia, dose delays/reductions, and requiring GCSF support. To maintain efficacy and reduce toxicity, we analyzed a biweekly dosing schedule (days 1-5 and days 15-19 q28 days). PATIENTS AND METHODS A retrospective analysis was performed in patients with mCRC and appendiceal cancer who completed >12 days of TAS-102 therapy and underwent surveillance imaging every 8-12 weeks. ECOG performance status (PS), prior lines of therapy, use of bevacizumab, CTCAE grade of treatment-related myelotoxicity, dose reductions/delays, and use of GCSF were assessed. Among patients with mCRC, survival analyses were performed. RESULTS 61 patients met inclusion criteria, with mCRC:appendiceal CA ratio of 56:5. Median ECOG PS = 1; median number of prior therapies = 3; Reduction in grade ≥3 neutropenia (16.3%, 1 patient with grade 4) and grade ≥3 anemia (8.2%) relative to the historic controls of the RECOURSE and SUNLIGHT trials. No neutropenic fever was noted; GCSF was not required. Eight patients required a dose delay. In the mCRC patients, the median progression-free survival (PFS) was 4.2 months, with a median overall survival (OS) of 9.2 months. CONCLUSIONS Biweekly dosing of TAS-102 demonstrated a reduction in myelosuppression, with similar PFS and OS. With an improved toxicity profile, this alternative dosing schedule may potentially broaden the utilization of TAS-102 in patients with borderline PS and provide a favorable option for future combination studies.
Collapse
Affiliation(s)
- Christopher Cann
- Fox Chase Cancer Center, Department of Hematology and Oncology, Philadelphia, PA, USA
| | - Michael LaPelusa
- Vanderbilt University Medical Center, Division of Hematology and Oncology, Nashville, TN, USA
| | - Sarah Cimino
- Vanderbilt University Medical Center, Division of Hematology and Oncology, Nashville, TN, USA
| | - Victoria Cancelliere
- University of Wisconsin Carbone Cancer Center, Division of Hematology, Medical Oncology and Palliative Care, Madison, WI, USA
| | - Elizabeth Dow-Hillgartner
- University of Wisconsin Carbone Cancer Center, Division of Hematology, Medical Oncology and Palliative Care, Madison, WI, USA
| | - Zhiguo Zhao
- Vanderbilt University Medical Center, Division of Hematology and Oncology, Nashville, TN, USA
| | - Dustin Deming
- University of Wisconsin Carbone Cancer Center, Division of Hematology, Medical Oncology and Palliative Care, Madison, WI, USA
| | - Cathy Eng
- Vanderbilt University Medical Center, Division of Hematology and Oncology, Nashville, TN, USA
| |
Collapse
|
18
|
Verdaguer J, Chouchana L, Robert M, Bergeron S, Montastruc F, Barus R. Ischemic cardiopathy induced by capecitabine in gastric cancer: The role of dihydropyrimidine dehydrogenase metabolites. Therapie 2025; 80:305-309. [PMID: 39438243 DOI: 10.1016/j.therap.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/24/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Fluoropyrimidine-based therapies, 5-fluorouracil (5-FU) and its oral prodrugs, capecitabine and tegafur/oteracil/gimeracil (S-1), are pivotal drugs to treat gastric cancer. Fluoropyrimidines are associated with cardiotoxicity including ischemic cardiopathy. The mechanisms of ischemic cardiopathy are considered to be multifactorial, potentially involving metabolites of 5-FU generated by the dihydropyrimidine dehydrogenase (DPD). By using Vigibase®, the World Health Organization pharmacovigilance database, we aimed to investigate the implication of the 5-FU metabolites induced by DPD in the occurrence of ischemic cardiopathy in patients with gastric cancer using capecitabine. METHODS In Vigibase®, we included serious reports of ischemic cardiopathy with capecitabine and S-1 from January 1st, 2013, to September 16th, 2023. Among patients with gastric cancer, we calculated the reporting odds ratio (ROR) of ischemic cardiopathy to compare capecitabine (a prodrug without DPD antagonist) with S-1 (a prodrug associated with a DPD antagonist). The ROR was also calculated regardless of the drug indication. An ancillary analysis based on the French pharmacovigilance database was also performed. We evaluated the ROR of serious cardiac disorders induced by 5-FU intravenous infusion according to the DPD status (no deficiency versus complete or partial deficiency). RESULTS In gastric cancer, 1843 reports (including 23 ischemic cardiopathy) for capecitabine and 2225 reports (including 17 ischemic cardiopathy) for S-1 were included. Median time-to-onset was 7 (3-26) days for capecitabine and 22 (13.25-30) days for S-1. Capecitabine was associated with an increased ROR of ischemic cardiopathy compared with S-1 in gastric cancer (ROR=1.6; [95% CI=1.5-1.8]) and regardless of the indication (7.3; [95% CI=6.6-8.0]). In the ancillary analysis, among 5-FU users, the lack of DPD deficiency increased the ROR for cardiac disorders (2.1; [95% CI=1.9-2.3]) compared to the DPD deficiency. CONCLUSION This work supports the role of toxic 5-FU metabolites generated by dihydropyrimidine dehydrogenase in the occurrence of ischemic cardiopathy among patients with gastric cancer using capecitabine.
Collapse
Affiliation(s)
- Joaquim Verdaguer
- Department of Medical and Clinical Pharmacology, Faculty of Medicine, Centre of PharmacoVigilance and Pharmacoepidemiology, Toulouse University Hospital, 31000 Toulouse, France
| | - Laurent Chouchana
- Department of Perinatal, Pediatric and Adult Pharmacology, Centre of Pharmacovigilance, Cochin Hospital, AP-HP.centre-université Paris Cité, 75000 Paris, France
| | - Marion Robert
- Department of Medical and Clinical Pharmacology, CHU de Grenoble, 38000 Grenoble, France
| | - Sandrine Bergeron
- Department of Medical and Clinical Pharmacology, CHU de Lille, 59000 Lille, France
| | - François Montastruc
- Department of Medical and Clinical Pharmacology, Faculty of Medicine, Centre of PharmacoVigilance and Pharmacoepidemiology, Toulouse University Hospital, 31000 Toulouse, France; Inserm, UMR 1027 Pharmacoepidemiology, Assessment of Drug Utilization and Drug Safety, CIC 1426 - University Paul Sabatier Toulouse, 31000 Toulouse, France
| | - Romain Barus
- Department of Medical and Clinical Pharmacology, Faculty of Medicine, Centre of PharmacoVigilance and Pharmacoepidemiology, Toulouse University Hospital, 31000 Toulouse, France.
| |
Collapse
|
19
|
Hao X, Zhang K, Hou Z, Guo J, Yang L, Sun S. Advances in natural polysaccharide/protein-based bioadhesive formulations for the potential application in esophagus: A review. Int J Biol Macromol 2025; 308:142513. [PMID: 40147657 DOI: 10.1016/j.ijbiomac.2025.142513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
The esophagus is susceptible to various injuries or disorders, which can significantly impact quality of life and pose potentially life-threatening risks. The unique anatomical and physiological characteristics of the esophagus present challenges in achieving optimal bioavailability and efficacy during diagnosis and treatment. To address these challenges, polysaccharide- and protein-based bioadhesive formulations have been developed to adhere to esophageal tissue, thereby prolonging residence time and enhancing diagnostic accuracy and therapeutic outcomes. Natural polysaccharides and proteins have garnered attention in the medical field owing to their exceptional properties, including biocompatibility, bioavailability, biodegradability, and low toxicity. A substantial body of research has demonstrated the significant potential of polysaccharides and proteins in clinical applications for the esophagus. The objective of this review is to discuss the structural characteristics and biological activities of various polysaccharides, including chitosan, hyaluronic acid, alginate, cellulose, guar gum, gellan gum, and xanthan gum, as well as proteins such as gelatin and fibrin, and their utilization in esophageal bioadhesive formulations. The practical challenges and prospects associated with implementing polysaccharide and protein-based bioadhesives on the esophagus are also discussed.
Collapse
Affiliation(s)
- Xuanyu Hao
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zhipeng Hou
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Jintao Guo
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Liqun Yang
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| | - Siyu Sun
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
20
|
Kaplan Z, Prezioso E, Jain A, Lavu H, Yeo CJ, Bowne WB, Nevler A. Clinical Implications of Mismatch Repair Deficiency in Pancreatic Ductal Adenocarcinoma. Cancer Med 2025; 14:e70960. [PMID: 40366030 PMCID: PMC12076359 DOI: 10.1002/cam4.70960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive and lethal disease, characterized by a limited response to chemotherapy and overall poor prognosis. Pancreatic cancers with a distinct mismatch repair deficiency, although relatively rare, have been shown to be associated with markedly better outcomes in comparison. Furthermore, whereas pancreatic cancers are generally unresponsive to current immunotherapy, this specific group of tumors has been shown to have a notable susceptibility to immune checkpoint inhibitors. AIMS In this review, we aim to summarize the relevant literature regarding mismatch-repair associated pancreatic cancers, the impacted biological mechanisms, and the resulting vulnerabilities for potential opportunistic immunotherapeutic treatment approaches. We will also review the current clinical studies assessing survival outcomes of mismatch repair deficient pancreatic cancers and ongoing clinical trials in this emerging field. RESULTS AND CONCLUSIONS Patients with dMMR/MSI-H pancreatic cancers harbor a distinct phenotype that has increased immune activation, greater responsiveness to immune checkpoint inhibitor therapy and better overall survival when compared to other pancreatic cancers. Although this molecular subtype makes up a small minority of cases, emerging data suggest immunotherapy may offer benefit to these patients.
Collapse
Affiliation(s)
- Zachary Kaplan
- Sidney Kimmel Medical CollegePhiladelphiaPennsylvaniaUSA
| | | | - Aditi Jain
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Harish Lavu
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Charles J. Yeo
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Wilbur B. Bowne
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Avinoam Nevler
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
21
|
Eikenboom EL, Nasar N, Seier K, Gönen M, Spaander MCW, O'Reilly EM, Jarnagin WR, Drebin J, D'Angelica MI, Kingham TP, Balachandran VP, Soares KC, Wagner A, Wei AC. Survival of Patients with Resected Microsatellite Instability-High, Mismatch Repair Deficient, and Lynch Syndrome-Associated Pancreatic Ductal Adenocarcinomas. Ann Surg Oncol 2025; 32:3568-3577. [PMID: 39656390 DOI: 10.1245/s10434-024-16621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/19/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a challenging disease due to its aggressiveness, late-stage diagnosis, and limited treatment options. Microsatellite instability-high (MSI-H) cancers are susceptible to immune checkpoint inhibitors. Survival outcomes for patients with MSI-H PDAC are unknown as the disease is rare. METHODS This study included patients with PDACs surgically resected from 1990 to 2023, and those with germline or sporadic pathogenic variants in DNA mismatch repair genes were identified. The study matched MSI-H, mismatch repair-deficient (MMRd), and Lynch syndrome (LS)-associated PDAC cases (on age, gender, and year of surgery) with microsatellite-stable (MSS), mismatch repair-proficient, or non-LS-associated PDAC cases in a 1:2 ratio. A generalized estimating equation Cox model with a robust sandwich estimator was used to compare overall survival (OS) in the matched cohorts. RESULTS Of 936 cases, 18 were included. Eight cases were MSI-H/MMRd, two were MSI/IHC-indeterminate, seven were MSS, and one was not tested for MSI. Nine patients had LS (MLH1 [n = 1], MSH2 [n = 4], MSH6 [n = 1], PMS2 [n = 3]), and nine patients had sporadic pathogenic variants in DNA MMR genes (MLH1 [n = 4], MSH6 [n = 5]). After matching to 36 control patients, the MSI-H/MMRd/LS PDACs had a significantly better OS (hazard ratio [HR], 0.36 [95% confidence interval [CI], 0.18-0.73; p = 0.005]; 5-year OS: MSI-H 77% [95% CI 58-100%] vs. MSS 27% [95% CI 15-51%]). CONCLUSION Before routine use of immune checkpoint inhibitors, the patients with MSI-H, MMRd, and LS-associated PDACs displayed significantly better survival than the patients with MSS, MMR-proficient, non-LS-associated PDACs. It is expected that survival for this cohort will further improve with increased availability of immunotherapy.
Collapse
Affiliation(s)
- Ellis L Eikenboom
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Clinical Genetics, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Naaz Nasar
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Seier
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William R Jarnagin
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Drebin
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael I D'Angelica
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Peter Kingham
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin C Soares
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alice C Wei
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
22
|
Akkus E, Yasar HA, Rimassa L, Lamarca A. Efficacy and Toxicity of Pemigatinib in Advanced Cholangiocarcinoma Harboring FGFR Fusions or Rearrangements: A Systematic Review and Meta-analysis. Target Oncol 2025; 20:389-403. [PMID: 40223038 DOI: 10.1007/s11523-025-01142-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND The efficacy and safety of pemigatinib in advanced cholangiocarcinoma (aCCA) were presented in phase I-II trials and retrospective reports, with small sample sizes and variable results. METHODS A systematic literature search included studies investigating the efficacy/safety of pemigatinib in aCCA harboring FGFR fusions/rearrangements. Primary outcomes were objective response rate (ORR) and treatment-related adverse events (AEs). A pooled proportion meta-analysis was performed. RESULTS Three hundred and twenty-seven patients in eight studies were included (three phase-II, one phase-I/II, two phase-I, and two retrospective). In the pooled analyses, the median age was 58.9 years (95% confidence interval (CI): 51.9-65.8); 33.4% (95% CI: 28.1-39.0) were male. Pemigatinib was the second-line treatment in 58.5% (95% CI: 52.7-64.1) and was beyond second-line in the remaining. ORR was 42.2% (95% CI: 35.9-48.7) (I2:48.4%) and disease control rate (DCR) was 86.5% (95% CI: 81.6-90.5) (I2: 58.8%). Median progression-free survival (PFS) was 7.8 months (95% CI: 6.2-9.4) (I2: 11.6%). Two studies reported overall survival (OS) (median 17.5 and 17.1 months). The most common AEs (any grade) were hyperphosphatemia (46%), dysgeusia (33.2%), alopecia (31.4%), fatigue (30.9%), stomatitis (28.5%), and diarrhea (27.5%). Cumulative eye and nail toxicities were observed in 32.5% and 40.9%, and retinal detachment in 5.5%. CONCLUSION This analysis emphasizes the FGFR alteration testing and pemigatinib use in the second-line and beyond treatment of aCCA. REGISTRATION ID (PROSPERO) CRD42024627459.
Collapse
Affiliation(s)
- Erman Akkus
- Department of Medical Oncology, Faculty of Medicine, Ankara University, Ankara, Turkey
- Ankara University Cancer Research Institute, Ankara, Turkey
| | - Hatime Arzu Yasar
- Department of Medical Oncology, Faculty of Medicine, Ankara University, Ankara, Turkey
- Ankara University Cancer Research Institute, Ankara, Turkey
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Angela Lamarca
- Department of Oncology, OncoHealth Institute, Instituto de Investigaciones Sanitarias FJD, Fundación Jiménez Díaz University Hospital, Avda Reyes Catolicos 2, 28040, Madrid, Spain.
| |
Collapse
|
23
|
Melehy A, Agopian VG. Role of Liver Transplant in Primary and Secondary Liver Malignancies. Clin Liver Dis 2025; 29:217-234. [PMID: 40287268 DOI: 10.1016/j.cld.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Hepatocellular carcinoma (HCC) and cholangiocarcinoma are the primary hepatic malignancies with established pathways to transplantation and model for end-stage liver disease (MELD) exception points. Other tumors managed with liver transplantation (LT) include hepatic epithelioid hemangioendothelioma and fibrolamellar HCC. LT for metastatic neuroendocrine tumor has been established with patient selection criteria and a path to MELD exception points. Additionally, recent data on LT for patients with unresectable hepatic colorectal metastases demonstrate increasingly encouraging initial results.
Collapse
Affiliation(s)
- Andrew Melehy
- Department of Surgery, Dumont-UCLA Transplant and Liver Cancer Centers, David Geffen School of Medicine at University of California, Los Angeles, CA, USA
| | - Vatche G Agopian
- Division of Liver and Pancreas Transplantation, Department of Surgery, Dumont-UCLA Transplant and Liver Cancer Centers, David Geffen School of Medicine at University of California, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Liu X, Liang C, Ding L, Zhang Q, Liu Y, Wang W. Analysis of the clinical application value of cfDNA methylation and fragmentation in early diagnosis of esophageal cancer. Genomics 2025; 117:111034. [PMID: 40188889 DOI: 10.1016/j.ygeno.2025.111034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND This study explores the clinical value of cfDNA methylation and fragmentation for the early diagnosis of esophageal cancer using liquid biopsy. METHODS Whole genome bisulfite sequencing and low-pass whole genome sequencing were utilized to detect cfDNA biomarkers, comparing 30 esophageal cancer patients with 10 healthy controls. RESULTS Significant differences in cfDNA methylation and fragmentation were observed between cancerous and non-cancerous samples (p < 0.05). A volcano plot identified 822 differentially methylated markers (817 upregulated, 5 downregulated), with SOX17, SOX1, ZNF382, ZNF667-AS1, and TFPI2 highly associated with esophageal cancer. Fragmentation markers (EDM, FSD, FSR, TFBS, CNV) showed 95 % specificity and sensitivity, with EDM demonstrating the best performance. CONCLUSION Our study highlights the clinical potential of cfDNA methylation and fragmentation biomarkers for the early diagnosis of esophageal cancer.
Collapse
Affiliation(s)
- Xin Liu
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China; Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, China
| | - Chen Liang
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, China
| | - Lingwen Ding
- Department of Vaccination clinic, Xiangyang Center for Disease Control and Prevention, Xiangyang, Hubei, China
| | - Qian Zhang
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yi Liu
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Wei Wang
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China.
| |
Collapse
|
25
|
Sanz-Garcia E, Peinado P, Peláez A, Lopez-Aranda EG, Álvarez-Gallego R, Muñoz C, Toledano C, Mihic L, Ortega J, Lazaro A, Montesino BM, Bressel C, Gonzalo A, Hernando O, Lopez M, Rubio C, Fabra I, Quijano Y, Vicente E, Cubillo A. Circulating tumor DNA using a plasma-only assay predicts survival in patients with oligometastatic colorectal cancer after definitive therapy. J Gastrointest Oncol 2025; 16:580-590. [PMID: 40386606 PMCID: PMC12078829 DOI: 10.21037/jgo-24-819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/23/2025] [Indexed: 05/20/2025] Open
Abstract
Background Patients with colorectal cancer (CRC) and oligometastases are usually treated with surgery and radiation. The addition of adjuvant chemotherapy is controversial. The detection of circulating tumor DNA (ctDNA) may provide further insight in prognosis as well as help in decision of adjuvant therapies. We aim to show that detection of ctDNA after definitive therapy in oligometastatic CRC is associated with worse outcomes. Methods A single centre prospective study included patients with oligometastatic CRC treated with surgery or radiation with definitive intent. Plasma samples were collected before procedure and 4 weeks after, prior to adjuvant chemotherapy. Plasma samples were analyzed using a tumor-naive assay focusing on genomic and methylation alterations (Guardant Reveal). Disease-free survival (DFS) and overall survival (OS) were estimated using Kaplan Meier method. Results A total of 25 patients were included: 19 were evaluated at baseline and post-treatment. ctDNA detection at baseline was not associated with any clinicopathological characteristics, neither OS nor DFS. In contrast, patients with ctDNA detection post-treatment had worse OS [hazard ratio (HR): 11.28; 95% confidence interval (CI): 1.31-97.05] and a trend to shorter DFS (HR: 2.97; 95% CI: 0.97-9.06). Patients who were persistently negative or cleared ctDNA had similar outcomes. Conclusions ctDNA detection after surgery/radiation in oligometastatic CRC predicts worse OS and DFS. ctDNA could help to guide the decision regarding need of adjuvant chemotherapy in this population.
Collapse
Affiliation(s)
- Enrique Sanz-Garcia
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Paloma Peinado
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- HM Hospitales Health Research Institute, Madrid, Spain
- Department of Medicine, HM Health Faculty, Camilo José Cela University, Boadilla del Monte, Spain
| | - Adrián Peláez
- Respiratory Diseases Networking Biomedical REsearch Centre (CIBERES), Carlos III Health Institute (ISCIII), Madrid, Spain
- Data Analysis Department, HM Hospitales Research Foundation, HM Hospitales, Madrid, Spain
| | | | - Rafael Álvarez-Gallego
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- HM Hospitales Health Research Institute, Madrid, Spain
- Department of Medicine, HM Health Faculty, Camilo José Cela University, Boadilla del Monte, Spain
| | - César Muñoz
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- HM Hospitales Health Research Institute, Madrid, Spain
- Department of Medicine, HM Health Faculty, Camilo José Cela University, Boadilla del Monte, Spain
| | - Carmen Toledano
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- HM Hospitales Health Research Institute, Madrid, Spain
- Department of Medicine, HM Health Faculty, Camilo José Cela University, Boadilla del Monte, Spain
| | - Luka Mihic
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- HM Hospitales Health Research Institute, Madrid, Spain
- Department of Medicine, HM Health Faculty, Camilo José Cela University, Boadilla del Monte, Spain
| | - Justo Ortega
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- HM Hospitales Health Research Institute, Madrid, Spain
- Department of Medicine, HM Health Faculty, Camilo José Cela University, Boadilla del Monte, Spain
| | | | | | | | - Alicia Gonzalo
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Ovidio Hernando
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Mercedes Lopez
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Carmen Rubio
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Isabel Fabra
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Yolanda Quijano
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Emilio Vicente
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Antonio Cubillo
- HM CIOCC MADRID (Clara Campal Comprehensive Cancer Centre), Oncology Department, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
- HM Hospitales Health Research Institute, Madrid, Spain
- Department of Medicine, HM Health Faculty, Camilo José Cela University, Boadilla del Monte, Spain
| |
Collapse
|
26
|
Wang Y, Pan S, Tian J, Wang J, Yu Y, Wang S, Li F, Yang L, Liu X, Shen Y, Qiu Q, Luan J, Jia M, Xiong C, Duan X, Wang FS, Meng F. Cadonilimab, a PD-1/CTLA-4 bispecific antibody in unresectable hepatocellular carcinoma: a real-world study. Cancer Immunol Immunother 2025; 74:186. [PMID: 40293533 PMCID: PMC12037968 DOI: 10.1007/s00262-025-04038-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/26/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE This study retrospectively evaluated the safety and efficacy of cadonilimab combined with tyrosine kinase inhibitors (TKI) for the treatment of unresectable hepatocellular carcinoma (uHCC). PATIENTS AND METHODS Seventy-eight patients who received cadonilimab + TKI were included; 42 and 36 received it as first-line (1 L) and second-line and above (≥ 2 L) systemic treatment, respectively. Besides, ninety-five patients who received PD-1 inhibitor + TKI as first-line treatments were included. Safety was the primary endpoint; secondary endpoints were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). RESULTS Treatment-related adverse events (TRAEs) of any grade occurred in 84.6% of the patients, with grade ≥ 3 in 20.5%. In patients with a Child-Pugh score of ≥ 8 (CP ≥ 8), any grade TRAEs occurred in 88.2%, and grade ≥ 3 in 20.6%. The overall cohort's median progression-free survival (mPFS) was 3.6 months, whereas the median overall survival (mOS) was 8.8 months. In the 1 L group, mPFS was 6.7 months versus 2.3 months in ≥ 2 L. In the 1 L group, mOS was 13.7 months versus 3.2 months in ≥ 2 L. For CP < 8, 1 L mPFS was 7.6 months, mOS not reached; CP ≥ 8 had mPFS of 5.2 months, mOS of 5.6 months. For CP < 8 in ≥ 2 L, mPFS was 3.1 months, mOS 8.8 months; CP ≥ 8 had mPFS of 1.4 months, mOS of 2.2 months. After propensity score matching (PSM), the incidence of TRAEs of any grade was 77.1%, with grade ≥ 3 accounting for 17.1% in the PD-1 group. In the PD-1/CTLA-4 group, the incidence of TRAEs of any grade was 80.0%, and that of grade ≥ 3 TRAEs was 17.1%. The mPFS was 6.7 months in the PD-1/CTLA-4 group versus 3.3 months in the PD-1 group. The mOS was 13.7 months in the PD-1/CTLA-4 group versus 6.7 months in the PD-1 group. CONCLUSION Cadonilimab + TKI showed a favorable trend in safety and efficacy, especially when applied as first-line systemic therapy for uHCC. This study offers a clinical reference for its use in systemic uHCC therapy, particularly in patients with advanced liver dysfunction.
Collapse
Affiliation(s)
- Yilin Wang
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Shida Pan
- Capital Medical University, Beijing, 100069, China
| | - Jiahe Tian
- Peking University 302 Clinical Medical School, Beijing, 100191, China
| | - Jianing Wang
- Peking University 302 Clinical Medical School, Beijing, 100191, China
| | - Yingying Yu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Siyu Wang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Fengyi Li
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Luo Yang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Xiaomeng Liu
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Yingjuan Shen
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Qin Qiu
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Junqing Luan
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Mengdie Jia
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China
| | - Chuyue Xiong
- Peking University 302 Clinical Medical School, Beijing, 100191, China
| | - Xuanxuan Duan
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China
| | - Fu-Sheng Wang
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China.
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China.
| | - Fanping Meng
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China.
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China.
| |
Collapse
|
27
|
Hoang T, Choi MK, Oh JH, Kim J. Utility of circulating tumor DNA to detect minimal residual disease in colorectal cancer: A systematic review and network meta-analysis. Int J Cancer 2025. [PMID: 40293388 DOI: 10.1002/ijc.35442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025]
Abstract
Circulating tumor DNA (ctDNA) is a promising biomarker for predicting minimal residual disease (MRD) and guiding treatment decisions in patients with colorectal cancer (CRC). This study aimed to examine the study designs and settings of ongoing clinical trials that use ctDNA to guide treatment decisions and to determine the best timing for detecting MRD in non-metastatic CRC. We searched PubMed, Embase, Web of Science, Cochrane Library, and clinicaltrials.gov for English language records. The ctDNA settings from the clinical trials were categorized by randomization to ctDNA testing, treatment options based on ctDNA results, and the timing of ctDNA testing relative to adjuvant therapy. For prospective studies, a network meta-analysis using a frequentist approach was conducted to examine the pairwise associations between different ctDNA timing strategies and MRD, defined as recurrence, relapse, and progression. The main approaches in ctDNA-based interventional trial designs were categorized as ctDNA-guided treatment, ctDNA-by-treatment, ctDNA-guided surveillance, and ctDNA-enriched adjuvant therapy for guiding treatment decisions, including both escalation and de-escalation strategies, and surveillance. Overall, both preoperative and postoperative ctDNA detection were linked to higher risks of progression, with pooled hazard ratios (95% confidence intervals) of 5.23 (2.10-13.00) and 7.95 (5.30-11.91), respectively. Among the timing strategies, ctDNA testing after adjuvant therapy was the most effective for identifying high-risk patients, strongly suggesting the presence of residual disease. This study comprehensively reviewed the clinical settings of ctDNA testing in ongoing trials and provided evidence supporting the selection of post-adjuvant therapy as the optimal timing for ctDNA testing.
Collapse
Affiliation(s)
- Tung Hoang
- Department of Cancer AI & Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Republic of Korea
- Faculty of Pharmacy, University of Health Sciences, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Moon Ki Choi
- Center for Colorectal Cancer, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jae Hwan Oh
- Center for Colorectal Cancer, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jeongseon Kim
- Department of Cancer AI & Digital Health, National Cancer Center Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
28
|
Doi T, Ishikawa T, Moriguchi M, Itoh Y. Current status of cancer genome medicine for pancreatic ductal adenocarcinoma. Jpn J Clin Oncol 2025; 55:443-452. [PMID: 39893577 DOI: 10.1093/jjco/hyaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis; however, advancements in cancer genome profiling using next-generation sequencing have provided new perspectives. KRAS mutations are the most frequently observed genomic alterations in patients with PDAC. However, until recently, it was not considered a viable therapeutic target. Although KRAS G12C mutations for which targeted therapies are already available are infrequent in PDAC, treatments targeting KRAS G12D and pan-KRAS are still under development. Similarly, new treatment methods for KRAS, such as chimeric antigen receptor T-cell therapy, have been developed. Several other potential therapeutic targets have been identified for KRAS wild-type PDAC. For instance, immune checkpoint inhibitors have demonstrated efficacy in PDAC treatment with microsatellite instability-high/deficient mismatch repair and tumor mutation burden-high profiles. However, for other PDAC cases with low immunogenicity, combination therapies that enhance the effectiveness of immune checkpoint inhibitors are being considered. Additionally, homologous recombination repair deficiencies, including BRCA1/2 mutations, are prevalent in PDAC and serve as important biomarkers for therapies involving poly (adenosine diphosphate-ribose) polymerase inhibitors and platinum-based therapies. Currently, olaparib is available for maintenance therapy of BRCA1/2 mutation-positive PDAC. Further therapeutic developments are ongoing for genetic abnormalities involving BRAF V600E and the fusion genes RET, NTRK, NRG, ALK, FGFR2, and ROS1. Overcoming advanced PDAC remains a formidable challenge; however, this review outlines the latest therapeutic strategies that are expected to lead to significant advancements.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Cancer Genome Medical Center, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Medical Oncology Unit, University Hospital, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
29
|
Ciner A, Hosein PJ, Jiang Y, Rassool F. The Interplay Between DNA Repair and the Immune Microenvironment in Pancreatic Cancer. Biomedicines 2025; 13:1031. [PMID: 40426860 PMCID: PMC12108561 DOI: 10.3390/biomedicines13051031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 05/29/2025] Open
Abstract
This narrative review describes the relationship between DNA repair and the immune microenvironment in pancreatic cancer and its potential clinical relevance. Pancreatic cancer is a devastating disease, often diagnosed at an advanced and incurable stage. BRCA or PALB2 mutations occur in a small subset, disabling accurate DNA double-strand break repair and sensitizing tumors to platinum-based chemotherapy and poly-ADP ribose polymerase inhibitors. While immune checkpoint blockade targeting PD1 and CTLA4 is ineffective for most patients, accumulating translational work indicates that those with BRCA or PALB2 mutations harbor a distinct and more permissive immune microenvironment. The phase 2 TAPUR study and retrospective series demonstrate that combined PD1 and CTLA4 inhibition can be effective for this subgroup of patients. In this manuscript, we review the current treatment landscape, the underlying mechanisms for immune resistance, and the interplay between defective DNA repair and the immune microenvironment in pancreatic cancer.
Collapse
Affiliation(s)
- Aaron Ciner
- Greenebaum Comprehensive Cancer Center, School of Medicine Baltimore, University of Maryland, Baltimore, MD 21201, USA
| | - Peter J. Hosein
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA;
| | - Yixing Jiang
- Greenebaum Comprehensive Cancer Center, School of Medicine Baltimore, University of Maryland, Baltimore, MD 21201, USA
| | - Feyruz Rassool
- Greenebaum Comprehensive Cancer Center, School of Medicine Baltimore, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
30
|
Gutiu AG, Zhao L, Marrah AJ, Maher AJ, Voss BB, Mayberry TG, Cowan BC, Wakefield MR, Fang Y. Promising immunotherapeutic treatments for colon cancer. Med Oncol 2025; 42:175. [PMID: 40266497 DOI: 10.1007/s12032-025-02724-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Colon cancer continues to predominate as one of the most common causes of morbidity and mortality worldwide due to its subtle symptomology and multifactorial etiology. Currently, the standard treatment approach involves surgical resection for localized tumors and adjuvant chemotherapy (AC) for cancers that have spread or invaded locally. Unfortunately, this treatment strategy may not be effective for advanced, metastatic stages of the disease or in instances of recurrence. However, advancements in immunotherapy have demonstrated promising results in the field of medical oncology, which could redefine the way patients and clinicians view the disease. These treatments have shown benefit and have the potential to be superior to the current interventions available for afflicted patients. The application of immunotherapy in conjunction with traditional treatments may improve the outcomes of patients suffering from colon cancer and decrease the burden this disease has on patients. Furthermore, more effective treatments in the form of conjunction immunological, surgical, and chemotherapeutic techniques may shorten treatment courses and lead to better long-term effects. This review paper investigates immunotherapeutic interventions for colon cancer, which include immune checkpoint inhibitors, oncolytic virotherapy, cancer vaccines, cytokine therapy, and CAR-T. Such a study might provide more information for clinicians to treat patients with colon cancer in the advanced stages or in recurrence.
Collapse
Affiliation(s)
- Arturo G Gutiu
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA, 50266, USA
| | - Lei Zhao
- Department of Respiratory Medicine, The 2nd People's Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Austin J Marrah
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Austin J Maher
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Brady B Voss
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Trenton G Mayberry
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Braydon C Cowan
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA, 50266, USA.
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.
- Department of Microbiology, Immunology & Pathology, Des Moines University College of Osteopathic Medicine, West Des Moines, IA, 50266, USA.
| |
Collapse
|
31
|
Zhang L, Gatlin V, Gupta S, Salinas ML, Romero S, Cai JJ, Chapkin RS, Safe S. Expression of Prooncogenic Nuclear Receptor 4A (NR4A)-Regulated Genes β1-Integrin and G9a Inhibited by Dual NR4A1/2 Ligands. Int J Mol Sci 2025; 26:3909. [PMID: 40332813 PMCID: PMC12028307 DOI: 10.3390/ijms26083909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Bis-indole-derived compounds including 1,1-bis(3'-indolyl)-1-(3,5-disubstitutedphenyl)methane (DIM-3,5) analogs bind both orphan nuclear receptors 4A1 (NR4A1) and NR4A2, and DIM-3,5 compounds act as dual receptor inverse agonists and inhibit both NR4A1- and NR4A2-regulated responses. Chromatin immunoprecipitation assays show that β1-integrin and the methyltransferase gene G9a are regulated by both NR4A1 and NR4A2 acting as cofactors for Sp1- and Sp4-dependent gene expression. DIM-3,5 treatment results in the loss of one or more of these nuclear factors from the β1-integrin and G9a promoters. Single-cell and RNAseq analyses show that both receptors regulate common (<10%) and unique genes in SW480 colon cancer cells; however, functional enrichment analysis of the differentially expressed genes converges to several common pathways and gene ontology terms.
Collapse
MESH Headings
- Humans
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Integrin beta1/genetics
- Integrin beta1/metabolism
- Cell Line, Tumor
- Ligands
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/agonists
- Histocompatibility Antigens/genetics
- Histocompatibility Antigens/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Promoter Regions, Genetic
- Indoles/pharmacology
Collapse
Affiliation(s)
- Lei Zhang
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA;
| | - Victoria Gatlin
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (V.G.); (S.G.); (S.R.); (J.J.C.)
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX 77843, USA; (M.L.S.); (R.S.C.)
| | - Shreyan Gupta
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (V.G.); (S.G.); (S.R.); (J.J.C.)
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX 77843, USA; (M.L.S.); (R.S.C.)
| | - Michael L. Salinas
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX 77843, USA; (M.L.S.); (R.S.C.)
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Selim Romero
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (V.G.); (S.G.); (S.R.); (J.J.C.)
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX 77843, USA; (M.L.S.); (R.S.C.)
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (V.G.); (S.G.); (S.R.); (J.J.C.)
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX 77843, USA; (M.L.S.); (R.S.C.)
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Robert S. Chapkin
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX 77843, USA; (M.L.S.); (R.S.C.)
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
32
|
Ghoreyshi N, Heidari R, Farhadi A, Chamanara M, Farahani N, Vahidi M, Behroozi J. Next-generation sequencing in cancer diagnosis and treatment: clinical applications and future directions. Discov Oncol 2025; 16:578. [PMID: 40253661 PMCID: PMC12009796 DOI: 10.1007/s12672-025-01816-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/15/2025] [Indexed: 04/22/2025] Open
Abstract
Next-generation sequencing (NGS) has emerged as a pivotal technology in the field of oncology, transforming the approach to cancer diagnosis and treatment. This paper provides a comprehensive overview of the integration of NGS into clinical settings, emphasizing its significant contributions to precision medicine. NGS enables detailed genomic profiling of tumors, identifying genetic alterations that drive cancer progression and facilitating personalized treatment plans targeting specific mutations, thereby improving patient outcomes. This capability facilitates the development of personalized treatment plans targeting specific mutations, leading to improved patient outcomes and the potential for better prognosis. The application of NGS extends beyond identifying actionable mutations; it is instrumental in detecting hereditary cancer syndromes, thus aiding in early diagnosis and preventive strategies. Furthermore, NGS plays a crucial role in monitoring minimal residual disease, offering a sensitive method to detect cancer recurrence at an early stage. Its use in guiding immunotherapy by identifying biomarkers that predict response to treatment is also highlighted. Ethical issues related to genetic testing, such as concerns around patient consent and data privacy, are also important considerations that need to be addressed for the broader implementation of NGS. These include the complexities of data interpretation, the need for robust bioinformatics support, cost considerations, and ethical issues related to genetic testing. Addressing these challenges is essential for the widespread adoption of NGS. Looking forward, advancements such as single-cell sequencing and liquid biopsies promise to further enhance the precision of cancer diagnostics and treatment. This review emphasizes the transformative impact of NGS in oncology and advocates for its incorporation into routine clinical practice to promote molecularly driven cancer care.
Collapse
Affiliation(s)
- Nima Ghoreyshi
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Heidari
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Arezoo Farhadi
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohsen Chamanara
- Department of Clinical Pharmacy, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Nastaran Farahani
- Department of Genetics and Biotechnology, Faculty of Life Science, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Mahmood Vahidi
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran.
- Department of Medical Laboratory Sciences, School of Allied Health Medicine, AJA University of Medical Sciences, Tehran, Iran.
| | - Javad Behroozi
- Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran.
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
33
|
Cosgrove D, Tan R, Osterland AJ, Hernandez S, Ogale S, Mahrus S, Murphy J, Wilson T, Patton G, Loaiza-Bonilla A, Singal AG. Atezolizumab Plus Bevacizumab in Patients with Unresectable Hepatocellular Carcinoma: Real-World Experience From a US Community Oncology Network. J Hepatocell Carcinoma 2025; 12:791-804. [PMID: 40271535 PMCID: PMC12015733 DOI: 10.2147/jhc.s492881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 04/25/2025] Open
Abstract
Purpose Atezolizumab plus bevacizumab (atezo-bev) is a preferred first-line (1L) systemic therapy option for unresectable hepatocellular carcinoma (uHCC). However, evidence of its effectiveness in real-world clinical practice, including in patients with impaired liver function, remains limited. Patients and Methods This retrospective observational study included adult patients who initiated 1L atezo-bev for uHCC within The US Oncology Network between 1/1/2019 and 8/31/2022 using structured and unstructured electronic health records data. Overall survival (OS) and real-world progression-free survival (rwPFS) were assessed using Kaplan-Meier methods for the overall cohort and in a subgroup of "trial-like" patients with characteristics that were consistent with those of the IMbrave150 Trial (ECOG performance status 0-1, Child-Pugh class A, albumin-bilirubin grade 1-2). Results Overall, 374 patients met eligibility criteria (mean age 68.8 years, 78.9% male, 31% Child-Pugh class B-C among reported, 18% ECOG performance status ≥2 among reported), of whom 132 patients comprised the trial-like subgroup. At a median follow-up of 5.6 months, median (95% CI) OS was 13.2 (9.5, 15.9) months and rwPFS was 6.4 (5.1, 7.7) months. In the trial-like subgroup, median (95% CI) OS was 16.5 (13.2, NR) months and rwPFS was 9.4 (5.7, 12.5) months. Conclusion Atezo-bev was used as 1L systemic therapy for HCC in a diverse patient population across US community oncology settings. Real-world effectiveness of atezo-bev among trial-like patients is comparable to that reported in the Phase 3 study. These data can help guide selection of appropriate treatment candidates and maximize the benefits of atezo-bev in routine clinical practice.
Collapse
Affiliation(s)
- David Cosgrove
- Medical Oncology, Compass Oncology/The US Oncology Network, Vancouver, WA, USA
| | | | | | | | | | | | - John Murphy
- Real-World Research, Ontada, Boston, MA, USA
| | | | | | | | - Amit G Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
34
|
Gadour E, Miutescu B, Hassan Z, Aljahdli ES, Raees K. Advancements in the diagnosis of biliopancreatic diseases: A comparative review and study on future insights. World J Gastrointest Endosc 2025; 17:103391. [PMID: 40291132 PMCID: PMC12019128 DOI: 10.4253/wjge.v17.i4.103391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/19/2025] [Accepted: 03/08/2025] [Indexed: 04/14/2025] Open
Abstract
Owing to the complex and often asymptomatic presentations, the diagnosis of biliopancreatic diseases, including pancreatic and biliary malignancies, remains challenging. Recent technological advancements have remarkably improved the diagnostic accuracy and patient outcomes in these diseases. This review explores key advancements in diagnostic modalities, including biomarkers, imaging techniques, and artificial intelligence (AI)-based technologies. Biomarkers, such as cancer antigen 19-9, KRAS mutations, and inflammatory markers, provide crucial insights into disease progression and treatment responses. Advanced imaging modalities include enhanced computed tomography (CT), positron emission tomography-CT, magnetic resonance cholangiopancreatography, and endoscopic ultrasound. AI integration in imaging and pathology has enhanced diagnostic precision through deep learning algorithms that analyze medical images, automate routine diagnostic tasks, and provide predictive analytics for personalized treatment strategies. The applications of these technologies are diverse, ranging from early cancer detection to therapeutic guidance and real-time imaging. Biomarker-based liquid biopsies and AI-assisted imaging tools are essential for non-invasive diagnostics and individualized patient management. Furthermore, AI-driven models are transforming disease stratification, thus enhancing risk assessment and decision-making. Future studies should explore standardizing biomarker validation, improving AI-driven diagnostics, and expanding the accessibility of advanced imaging technologies in resource-limited settings. The continued development of non-invasive diagnostic techniques and precision medicine approaches is crucial for optimizing the detection and management of biliopancreatic diseases. Collaborative efforts between clinicians, researchers, and industry stakeholders will be pivotal in applying these advancements in clinical practice.
Collapse
Affiliation(s)
- Eyad Gadour
- Multiorgan Transplant Centre of Excellence, Liver Transplantation Unit, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
- Internal Medicine, Zamzam University College, School of Medicine, Khartoum 11113, Sudan
| | - Bogdan Miutescu
- Department of Gastroenterology and Hepatology, Victor Babes University of Medicine and Pharmacy, Timisoara 300041, Romania
- Advanced Regional Research Center in Gastroenterology and Hepatology, Victor Babes University of Medicine and Pharmacy, Timisoara 30041, Romania
| | - Zeinab Hassan
- Department of Internal Medicine, Stockport Hospitals NHS Foundation Trust, Manchester SK2 7JE, United Kingdom
| | - Emad S Aljahdli
- Gastroenterology Division, King Abdulaziz University, Faculty of Medicine, Jeddah 21589, Saudi Arabia
- Gastrointestinal Oncology Unit, King Abdulaziz University Hospital, Jeddah 22252, Saudi Arabia
| | - Khurram Raees
- Department of Gastroenterology and Hepatology, Royal Blackburn Hospital, Blackburn BB2 3HH, United Kingdom
| |
Collapse
|
35
|
Lindskrog SV, Strandgaard T, Nordentoft I, Galsky MD, Powles T, Agerbæk M, Jensen JB, Alix-Panabières C, Dyrskjøt L. Circulating tumour DNA and circulating tumour cells in bladder cancer - from discovery to clinical implementation. Nat Rev Urol 2025:10.1038/s41585-025-01023-9. [PMID: 40234713 DOI: 10.1038/s41585-025-01023-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2025] [Indexed: 04/17/2025]
Abstract
Liquid biopsies, indicating the sampling of body fluids rather than solid-tissue biopsies, have the potential to revolutionize cancer care through personalized, noninvasive disease detection and monitoring. Circulating tumour DNA (ctDNA) and circulating tumour cells (CTCs) are promising blood-based biomarkers in bladder cancer. Results from several studies have shown the clinical potential of ctDNA and CTCs in bladder cancer for prognostication, treatment-response monitoring, and early detection of minimal residual disease and disease recurrence. Following successful clinical trial evaluation, assessment of ctDNA and CTCs holds the potential to transform the therapeutic pathway for patients with bladder cancer - potentially in combination with the analysis of urinary tumour DNA - through tailored treatment guidance and optimized disease surveillance.
Collapse
Affiliation(s)
- Sia V Lindskrog
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Iver Nordentoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Matthew D Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Mads Agerbæk
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Catherine Alix-Panabières
- Laboratory of Rare Circulating Human Cells - Liquid Biopsy Laboratory, Site Unique de Biology, University Medical Center of Montpellier, Montpellier, France
- CREEC/CANECEV MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
36
|
Tillman L, Margalef Rieres J, Ahjem E, Bishop-Guest F, McGrath M, Hatrick H, Pranjol MZI. Thinking Outside the Therapeutic Box: The Potential of Polyphenols in Preventing Chemotherapy-Induced Endothelial Dysfunction. Cells 2025; 14:566. [PMID: 40277892 PMCID: PMC12026109 DOI: 10.3390/cells14080566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
The numerous side effects and adverse health implications associated with chemotherapies have long plagued the field of cancer care. Whilst in some cases a curative measure, this highly toxic intervention consistently scores poorly on quantitative measures of tolerability and safety. Of these side effects, cardiac and microvascular defects pose the greatest health risk and are the leading cause of death amongst cancer survivors who do not succumb to relapse. In fact, in many low-grade cancers, the risk of recurrence is far outweighed by the cardiovascular risk of morbidity. As such, there is a pressing need to improve outcomes within these populations. Polyphenols are a group of naturally occurring metabolites that have shown potential vasoprotective effects. Studies suggest they possess antioxidant and anti-inflammatory activities, in addition to directly modulating vascular signalling pathways and gene expression. Leveraging these properties may help counteract the vascular toxicity induced by chemotherapy. In this review, we outline the main mechanisms by which the endothelium is damaged by chemotherapeutic agents and discuss the ability of polyphenols to counteract such side effects. We suggest future considerations that may help overcome some of the published limitations of these compounds that have stalled their clinical success. Finally, we briefly explore their pharmacological properties and how novel approaches could enhance their efficacy while minimising treatment-related side effects.
Collapse
Affiliation(s)
- Luke Tillman
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Jaume Margalef Rieres
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Elena Ahjem
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Fynn Bishop-Guest
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Meghan McGrath
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | - Helena Hatrick
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (L.T.); (J.M.R.); (M.M.); (H.H.)
| | | |
Collapse
|
37
|
Jamal A, Aldreiwish AD, Banawas SS, Alqurashi YE, Kamal MA, Ahmad F. The paths toward immunotherapy of esophageal cancer: An overview of clinical trials. Int Immunopharmacol 2025; 151:114261. [PMID: 40015204 DOI: 10.1016/j.intimp.2025.114261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
As the seventh-leading contributor to global cancer-related deaths, esophageal cancer (EC) is one of the most challenging types of cancer. Despite advancements in conventional therapies, including surgery, chemotherapy, and radiotherapy, the five-year survival rate remains low, underscoring the need for the development of more efficacious treatment approaches. Immunotherapy has emerged as a promising treatment approach, offering new hope for EC patients. This review provides an in-depth examination of the latest immunotherapeutic strategies for EC, focusing on immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and oncolytic virotherapy. We critically analyze the current clinical data to highlight the progress and pitfalls of each immunotherapeutic approach for EC. Additionally, we explore the potential for combination therapies, which could overcome the resistance often seen with monotherapies. Finally, we discuss the limitations of current treatments and outline key areas for future research to improve patient outcomes and survival.
Collapse
Affiliation(s)
- Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Allolo D Aldreiwish
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Saeed S Banawas
- Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Yaser E Alqurashi
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh 13713, Saudi Arabia
| |
Collapse
|
38
|
Okano N, Pirozzi A, Abidoye O, Hoyek C, Eslinger C, Zheng-Lin B, Jamal F, Sahwan O, Sonbol MB, Uson Junior PLS, Hayashi M, Sato T, Nishioka M, Nagashima F, Bekaii-Saab T, Borad MJ, Hironaka S. Systemic therapy for pretreated advanced biliary tract cancer: past developments and recent advances. Jpn J Clin Oncol 2025:hyaf052. [PMID: 40173029 DOI: 10.1093/jjco/hyaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025] Open
Abstract
Biliary tract cancer (BTC) remains among the most challenging malignancies with a poor prognosis and limited treatment options, particularly in pretreated patients. As most patients experience disease progression after first-line treatment, effective second-line and subsequent treatments are required. Although the addition of modified FOLFOX (fluorouracil, leucovorin, and oxaliplatin) to active symptom control improved the overall survival of patients with progressing advanced BTC despite gemcitabine plus cisplatin treatment, its efficacy was modest. Moreover, most clinical trials demonstrated modest efficacy of molecular-targeted agents for molecularly unselected pretreated advanced BTC. Patients with advanced BTC carry a relatively high druggable genetic alteration rate and have shown promising responses to molecular-matched therapies targeting gene alterations such as FGFR2 fusions/rearrangements, IDH1 mutation, and HER2 overexpression/amplification. Additionally, tumor-agnostic approaches, including BRAF V600E, NTRK fusion, and RET fusion, have expanded the treatment options for some patients. Immune checkpoint inhibitors have shown limited efficacy as mono- or combination therapy in patients with pretreated advanced BTC. Therefore, developmental efforts have shifted to immune checkpoint inhibitor and other combinations such as vascular endothelial growth factor receptor inhibitors or radiation. In addition to refining combination strategies to enhance the therapeutic potential of immune checkpoint inhibitor, future research should focus on elucidating the tumor microenvironment. This review delineates the evolution of systemic therapies in patients with pretreated advanced BTC. By examining past developments and recent advances through prospective trials, it highlights novel approaches that may improve outcomes in this challenging disease.
Collapse
Affiliation(s)
- Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Angelo Pirozzi
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan 20072, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan 20089, Italy
| | - Oluseyi Abidoye
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Celine Hoyek
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Cody Eslinger
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Binbin Zheng-Lin
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Fares Jamal
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Oudai Sahwan
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Mohamad Bassam Sonbol
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Pedro Luiz Serrano Uson Junior
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Avenida Albert Einstein 627, São Paulo 05652900, Brazil
| | - Masato Hayashi
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Taro Sato
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
- Department of Gastroenterology and Hepatology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Mariko Nishioka
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Fumio Nagashima
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Tanios Bekaii-Saab
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Mitesh J Borad
- Division of Hematology and Oncology, Mayo Clinic, 5881 E Mayo Blvd, Phoenix, AZ 85054, United States
| | - Shuichi Hironaka
- Department of Medical Oncology, Kyorin University Faculty of Medicine, 6-20-2, Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| |
Collapse
|
39
|
Zhang M, Chen X, Zhou Q, Guo N, Cao B, Zeng H, Chen W, Sun F. The global progress and quality assessment of research on the association between circulating tumor DNA and clinical prognosis: a systematic review. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:156-166. [PMID: 40265099 PMCID: PMC12010383 DOI: 10.1016/j.jncc.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/24/2024] [Accepted: 10/30/2024] [Indexed: 04/24/2025] Open
Abstract
Objective Circulating tumor DNA (ctDNA) has shown potential as a prognostic biomarker in patients with solid tumors. This study aimed to systematically summarize the global application of ctDNA in the prognostic management of solid tumor patients and to evaluate the quality of the current studies. Methods PubMed, Web of Science, Embase, Cochrane Library, Scopus, and clinical trials.gov databases were searched to collect cohort studies on ctDNA in the prognosis of solid tumor patients from January 2016 to May 2022. The language was limited to English. Information including general information, participants and cancer characteristics, ctDNA and outcome information were extracted. The quality of the studies was assessed using the Newcastle-Ottawa Scale checklist. Results A total of 214 studies were included in the final analysis, encompassing 21,076 patients. The number of studies has increased annually from 2016 to 2022. The most common types of solid tumors studied were colorectal cancer (27.10 %), lung cancer (20.09 %), pancreatic cancer (16.82 %), and breast cancer (14.02 %). The top three journals by number of publications had an impact factor in 2023 greater than 10. Of the studies, the median sample size was 69 (interquartile range: 41-111), 69.81 % had a sample size <100, 68.92 % had a median/mean age ≥60 years, and 74.05 % were from developed countries. Multi-center studies accounted for 40.36 %. Additionally, 29.82 % of the studies had a bias risk score ≤6. Only 16.67 % of studies on liver cancer had a bias risk score >6. The primary criteria not met by the studies included "Adequacy of follow-up of cohorts" (33.33 %), "Assessment of outcome" (32.16 %) and "Representativeness of the exposed cohort" (27.49 %). Conclusions The prognostic value of ctDNA in patients with solid tumors is gaining increasing attention, leading to a steady rise in the number of studies. However, many studies still suffer from small sample sizes and a lack of representativeness. Furthermore, details regarding ctDNA detection methods and results reporting are often insufficiently described. There is an urgent need to improve the quality of such research.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xiaowei Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Qingxin Zhou
- Tianjin Centers for Disease Control and Prevention, Tianjin, China
| | - Nana Guo
- Hebei Centers for Disease Control and Prevention, Shijiazhuang, China
| | - Baoshan Cao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Hongmei Zeng
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
- Xinjiang Medical University, Urumqi, China
| |
Collapse
|
40
|
Kobayashi S, Nakamura Y, Hashimoto T, Bando H, Oki E, Karasaki T, Horinouchi H, Ozaki Y, Iwata H, Kato T, Miyake H, Ohba A, Ikeda M, Chiyoda T, Hasegawa K, Fujisawa T, Matsuura K, Namikawa K, Yajima S, Yoshino T, Hasegawa K. Japan society of clinical oncology position paper on appropriate clinical use of molecular residual disease (MRD) testing. Int J Clin Oncol 2025; 30:605-654. [PMID: 39920551 PMCID: PMC11946966 DOI: 10.1007/s10147-024-02683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 12/09/2024] [Indexed: 02/09/2025]
Abstract
Although the 5-year relative survival rates for resectable solid tumors have improved over the past few years, the risk of postoperative recurrence necessitates effective monitoring strategies. Recent advancements in molecular residual disease (MRD) testing based on circulating tumor DNA (ctDNA) analysis have shown considerable promise in the context of predicting recurrence; however, significant barriers to widespread clinical implementation remain-mainly, low awareness among healthcare professionals, high costs, and lack of standardized assays and comprehensive evidence. This position paper, led by the Japan Society of Clinical Oncology, aims to establish a common framework for the appropriate clinical use of MRD testing in a tumor type-agnostic manner. It synthesizes currently available evidence, reviews region-specific clinical trends, addresses critical clinical questions related to MRD testing, and offers recommendations to guide healthcare professionals, biotechnology and pharmaceutical companies, and regulatory authorities. These recommendations were developed based on a voting process involving 15 expert members, ensuring a consensus-driven approach. These findings underscore the importance of collaborative efforts among various stakeholders in enhancing the clinical utility of MRD testing. This project aimed to foster consensus and provide clear guidelines to support the advancement of precision medicine in oncology and improve patient outcomes in the context of perioperative care.
Collapse
Affiliation(s)
- Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
- Perioperative Treatment Development Promotion Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
| | - Yoshiaki Nakamura
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
- International Research Promotion Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan.
| | - Tadayoshi Hashimoto
- Perioperative Treatment Development Promotion Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Hideaki Bando
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Karasaki
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yukinori Ozaki
- Department of Breast Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroji Iwata
- Department of Advanced Clinical Research and Development, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hideaki Miyake
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akihiro Ohba
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Takao Fujisawa
- Translational Research Support Office, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kazuto Matsuura
- Department of Head and Neck Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shugo Yajima
- Department of Urology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
- Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kiyoshi Hasegawa
- Department of Surgery, Graduate School of Medicine, Hepato-Biliary-Pancreatic Surgery Division, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Ebner R, Sheikh GT, Brendel M, Ricke J, Cyran CC. ESR Essentials: staging and restaging with FDG-PET/CT in oncology-practice recommendations by the European Society for Hybrid, Molecular and Translational Imaging. Eur Radiol 2025; 35:1894-1902. [PMID: 39384589 PMCID: PMC11914360 DOI: 10.1007/s00330-024-11094-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 10/11/2024]
Abstract
Positron emission tomography (PET) stands as the paramount clinical molecular imaging modality, especially in oncology. Unlike conventional anatomical-morphological imaging methods such as computed tomography (CT) and magnetic resonance imaging (MRI), PET provides detailed visualizations of internal activity at the molecular and cellular levels. 18-fluorine-fluorodeoxyglucose ([18F]FDG)-PET combined with contrast-enhanced CT (ceCT) significantly improves the detection of various cancers. Appropriate patient selection is crucial, and physicians should carefully assess the appropriateness of [18F]FDG-PET/CT based on specific clinical criteria and evidence. Due to its high diagnostic accuracy, [18F]FDG-PET/CT is indispensable for evaluating the extent of disease, staging, and restaging known malignancies, and assessing the response to therapy. PET/CT imaging offers significant advantages in patient management, particularly by identifying occult metastases that might otherwise go undetected. This can help prevent unnecessary surgeries, allowing many patients to be redirected to systemic chemotherapy instead. However, it is important to note that the gold standard for surgical planning remains CT and/or MRI, depending on the body region. These imaging modalities, with or without associated angiography, provide superior contrast and spatial resolution, essential for detailed surgical preparation and planning. [18F]FDG-PET/CT has a central role in the precise and early diagnosis of cancer, contributing significantly to personalized treatment plans. However, it has limitations, including non-tumor-specific uptake and the potential to inaccurately capture the metabolic activity of certain tumor types due to low uptake in some well-differentiated tumor cell lines. Therefore, it should be utilized in clinical scenarios where it offers crucial diagnostic insights not readily available with other imaging modalities. KEY POINTS: Use [18F]FDG-PET/CT selectively based on clinical appropriateness criteria and existing evidence to optimize resource utilization and minimize patient exposure. Employ [18F]FDG-PET/CT in treatment planning and monitoring, particularly for assessing chemotherapy or radiotherapy response in FDG-avid lymphoma and solid tumors. When available, [18F]FDG-PET/CT can be integrated with other diagnostic tools, such as MRI, to enhance overall diagnostic accuracy.
Collapse
Affiliation(s)
- Ricarda Ebner
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany.
| | - Gabriel T Sheikh
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
42
|
Dalbeni A, Cattazzo F, Natola LA, Zoncapè M, Faccincani D, Stefanini B, Ravaioli F, Villani R, Auriemma A, Sacerdoti D. What can real-world data teach us about treating patients with unresectable hepatocellular carcinoma? Expert Rev Gastroenterol Hepatol 2025; 19:389-398. [PMID: 40042586 DOI: 10.1080/17474124.2025.2476541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) remains a major global health concern, as it is the most common primary liver cancer and the fourth leading cause of cancer-related mortality. AREAS COVERED Immune checkpoint inhibitors (ICIs) have significantly shifted the treatment paradigm, offering promising survival outcomes. However, the controlled conditions of randomized clinical trials (RCTs) often fail to reflect real-world complexities, emphasizing the necessity for strong real-world evidence (RWE). RWE, in most cases derived from observational studies, provides critical insights into the effectiveness, safety, and tolerability of systemic therapies across diverse populations and settings. The authors searched MEDLINE, Ovid Embase, and Scopus for full-text published articles in any language from the inception to 30 June 2024.This review evaluates RWE on systemic therapies for advanced HCC, including tyrosine kinase inhibitors (TKIs) like sorafenib and lenvatinib, ICIs such as nivolumab and pembrolizumab, and combination therapies like atezolizumab/bevacizumab and durvalumab/tremelimumab. EXPERT OPINION Studies reveal discrepancies in treatment efficacy and adverse event profiles between RCTs and routine clinical practice, underscoring the need for individualized treatment strategies. RWE highlights the influence of liver disease etiology, liver function, and tumor burden on treatment outcomes, guiding therapy selection.
Collapse
Affiliation(s)
- A Dalbeni
- Unit of General Medicine C, Medicine Department, University of Verona and Hospital Trust (AOUI) of Verona, Verona, Italy
- Liver Unit, Medicine Department, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - F Cattazzo
- Liver Unit, Medicine Department, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - L A Natola
- Unit of General Medicine C, Medicine Department, University of Verona and Hospital Trust (AOUI) of Verona, Verona, Italy
- Liver Unit, Medicine Department, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - M Zoncapè
- Liver Unit, Medicine Department, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - D Faccincani
- Unit of General Medicine C, Medicine Department, University of Verona and Hospital Trust (AOUI) of Verona, Verona, Italy
- Liver Unit, Medicine Department, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - B Stefanini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - F Ravaioli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - R Villani
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - A Auriemma
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - D Sacerdoti
- Liver Unit, Medicine Department, University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| |
Collapse
|
43
|
Xu X, Ao W, Wang J. Artificial intelligence based on imaging data to predict rectal cancer recurrence: A meta-analysis. Cancer Radiother 2025; 29:104617. [PMID: 40250036 DOI: 10.1016/j.canrad.2025.104617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 04/20/2025]
Abstract
PURPOSE The purpose of this study was to evaluate the diagnostic performance of artificial intelligence based on imaging data to predict rectal cancer recurrence using a meta-analysis system. MATERIALS AND METHODS Medline, Embase, Cochrane Library, Web of Science, and other databases were searched for all articles on artificial intelligence prediction of rectal cancer recurrence based on imaging data published publicly from the establishment of the library to December 31, 2023. The quality of the articles was assessed using Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2). Meta-analysis was performed by the software Revman 5.4 and Statistics data (Stata), and sensitivity analysis was used to detect potential sources of heterogeneity and test to assess the presence of publication bias. We evaluated how well imaging-based data can predict recurrence in patients with rectal cancer by analysing the pooled sensitivity, specificity, and area under the curve. RESULTS Ten studies were included. The pooled sensitivity, specificity, and area under the curve of imaging-based data for recurrence in patients with rectal cancer were respectively 0.84 (95 % confidence interval [CI]: 0.74-0.91), 0.87 (95 % CI: 0.82-0.91) and 0.92 (95 % CI: 0.89-0.94). Based on QUADAS-2, the quality of the article is acceptable. We found the causes of heterogeneity through meta-regression: recurrence time predesign Lasso. Based on Deeks' funnel plot, no publication bias was detected. CONCLUSION Artificial intelligence based on imaging data has a high predictive ability for rectal cancer recurrence.
Collapse
Affiliation(s)
- Xiaoling Xu
- Graduate School, Zhejiang Chinese Medical University, Hangzhou Zhejiang, China; Department of Radiology, The Affiliated Hospital of Shao Xing University (Shao Xing Municipal Hospital), Shaoxing Zhejiang, China
| | - Weiqun Ao
- Department of Radiology, Tongde Hospital of Zhejiang Province Afflicted to Zhejiang Chinese Medical University (Tongde hospital of Zhejiang Province), Hangzhou Zhejiang, China
| | - Jian Wang
- Department of Radiology, Tongde Hospital of Zhejiang Province Afflicted to Zhejiang Chinese Medical University (Tongde hospital of Zhejiang Province), Hangzhou Zhejiang, China.
| |
Collapse
|
44
|
Mozooni Z, Amiri KK, Golestani N, Shahmohammadi A, Minaeian S, Bahadorizadeh L. Relationships between MMP-2, MMP-9, and ADAMDEC1 serum and tissue levels in patients with colorectal cancer. Ann Coloproctol 2025; 41:136-144. [PMID: 40313127 PMCID: PMC12046409 DOI: 10.3393/ac.2024.00227.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 11/15/2024] [Accepted: 02/10/2025] [Indexed: 05/03/2025] Open
Abstract
PURPOSE Colorectal cancer (CRC) is the most common malignancy of the gastrointestinal system globally. Identifying specific gene expression patterns indicative of early-stage CRC could enable early diagnosis and rapid treatment initiation. Matrix metalloproteinases (MMPs) play crucial roles in extracellular matrix degradation and tissue remodeling. Among them, MMP-2 and MMP-9 have been found to be upregulated in various cancers, including CRC, and are associated with tumor invasion, metastasis, and angiogenesis. In contrast, a disintegrin and metalloproteinase like decysin 1 (ADAMDEC1) is a relatively newly discovered gene with demonstrated involvement in immune response and inflammation. This study investigated serum levels of MMP-2 and MMP-9, along with tissue expression of MMP-2, MMP-9, and ADAMDEC1, and explored potential associations with pathological and clinical factors in patients with CRC. METHODS This study included 100 patients with CRC and 100 control participants. Tissue and blood samples were collected. Serum MMP-2 and MMP-9 levels were analyzed using the enzyme-linked immunosorbent assay. Quantitative real-time polymerase chain reaction was employed to assess the expression levels of MMP-2, MMP-9, and ADAMDEC1 in CRC tissue samples compared to adjacent control tissue. RESULTS The expression levels of MMP-2, MMP-9, and ADAMDEC1 were significantly upregulated in CRC relative to adjacent control tissues. Analysis of clinicopathological features revealed statistically significant differences in the expression levels of MMP-2, MMP-9, and ADAMDEC1 between patients with CRC with and without lymphovascular invasion (P<0.001). Based on receiver operating characteristic curve analysis, these genes represent promising candidate diagnostic biomarkers for CRC. CONCLUSION MMP-2, MMP-9, and ADAMDEC1 levels may serve as potential diagnostic biomarkers for CRC.
Collapse
Affiliation(s)
- Zahra Mozooni
- Antimicrobial Resistance Research Center, Institute of Immunology, and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Kiana Khajeh Amiri
- Department of Internal Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Golestani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Sara Minaeian
- Antimicrobial Resistance Research Center, Institute of Immunology, and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Leyla Bahadorizadeh
- Antimicrobial Resistance Research Center, Institute of Immunology, and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
- Department of Internal Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Xu L, Qiu X, He H, Liu L, He Q, Sun J. TAS-102 in combination with bevacizumab for second-line treatment of metastatic colorectal cancer with a hypertensive elderly patient: a case report. Front Oncol 2025; 15:1558470. [PMID: 40224173 PMCID: PMC11985432 DOI: 10.3389/fonc.2025.1558470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Background Colorectal cancer (CRC) is a common malignant tumor worldwide. Approximately 20%-25% of patients have metastases at the time of initial diagnosis, and nearly half eventually develop metastatic cancer. The standard first- and second-line treatments for unresectable metastatic CRC are full-dose two-/three-agent chemotherapy with or without a combination of molecularly targeted agents. However, many patients are ineligible for intensive therapy due to poor performance status or advanced age. TAS-102 (trifluridine/tipiracil) in combination with bevacizumab may provide a new treatment strategy for patients with advanced CRC who are ineligible for intensive therapy. Case report description We report a case of a 91-year-old woman diagnosed with stage IV adenocarcinoma of the rectosigmoid junction in the presence of multiple metastases. The patient had a history of hypertension, had suffered from deep vein thrombosis of the left lower extremity, and was allergic to several drugs. Genetic testing showed multiple mutations in Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS), Phosphatidylinositol-4,5-bisphosphate 3-Kinase Catalytic Subunit Alpha (PIK3CA), and Tumor Protein p53 (TP53); microsatellite stability; and a tumor mutational burden of 4.5 Mut/Mb. The patient was diagnosed with stage IV adenocarcinoma of the rectosigmoid junction in May 2020, with a clinical stage of cTxNxM1. Without surgery, the patient received first-line treatment with capecitabine in combination with bevacizumab, which was changed to second-line treatment with TAS-102 in combination with bevacizumab after disease progression, with a progression-free survival of 10 months, achieving a significant survival benefit. Later, due to the patient's poor physical condition, no further medication was administered, and the patient died on 1 September 2022. Conclusion TAS-102 in combination with bevacizumab for the treatment of elderly patients with metastatic CRC who are ineligible for intensive therapy is a promising treatment option.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinghua Sun
- Department of Medical Oncology, Second Affiliated Hospital of Dalian Medical
University, Dalian, China
| |
Collapse
|
46
|
Abiodun AT, Ju C, Welch CA, Lai J, Tyrer F, Chambers P, Paley L, Vernon S, Deanfield J, de Belder M, Rutherford MJ, Lambert PC, Slater S, Shiu KK, Wei L, Peake MD, Adlam D, Manisty C. Fluoropyrimidine Chemotherapy and the Risk of Death and Cardiovascular Events in Patients With Gastrointestinal Cancer. JACC CardioOncol 2025:S2666-0873(25)00089-4. [PMID: 40202479 DOI: 10.1016/j.jaccao.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Fluoropyrimidine chemotherapy is administered first-line for many gastrointestinal cancers. However, patients with cardiovascular disease commonly receive alternative treatment due to cardiotoxicity concerns. OBJECTIVES The study sought to assess the risks of all-cause mortality and acute cardiovascular events with fluoropyrimidine treatment. METHODS We conducted an observational cohort study applying a target trial emulation framework to linked national cancer, cardiac, and hospitalization registry data from the Virtual Cardio-Oncology Research Initiative. Adults diagnosed with tumors eligible for fluoropyrimidine-based chemotherapy as first-line therapy were included. All-cause mortality and a composite of hospitalization for acute cardiovascular events (acute coronary syndrome, heart failure, cardiac arrhythmia, cardiac intervention, cardiac arrest, and cardiac death) were compared in patients treated with fluoropyrimidine-based chemotherapy vs alternative management. Adjusted, weighted pooled logistic regression models were used to estimate the 1-year risk difference (RD). RESULTS Among 103,110 patients (mean age 69.7 years, 59% male), the absolute risk of death at 1 year was significantly lower in fluoropyrimidine-treated patients (RD: -7.7%; 95% CI: -8.7% to -6.7%) with a small increased risk of acute cardiovascular events (RD: 0.9%; 95% CI: 0.0% to 1.9%). This was primarily due to arrhythmias (RD: 0.8%; 95% CI: 0.1% to 1.6%) and cardiac arrest (RD: 0.3%; 95% CI: 0.1% to 0.5%), with no increased risk of acute coronary syndromes including in the subgroup of patients with pre-existing coronary artery disease. CONCLUSIONS The markedly improved overall survival with fluoropyrimidines in patients with gastrointestinal cancer significantly outweighs the small risk of cardiac arrhythmia and arrest. Oncologists should take this into consideration for decision making to avoid undue clinical conservatism, particularly in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Aderonke T Abiodun
- Institute of Cardiovascular Science, University College London, United Kingdom; Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom; National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom
| | - Chengsheng Ju
- Institute of Cardiovascular Science, University College London, United Kingdom; National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom; Research Department of Practice and Policy, School of Pharmacy, University College London, United Kingdom
| | - Catherine A Welch
- National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom; Department of Population Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Jennifer Lai
- National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom
| | - Freya Tyrer
- National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom; Department of Population Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Pinkie Chambers
- Research Department of Practice and Policy, School of Pharmacy, University College London, United Kingdom; Cancer Division, University College London Hospitals NHS Foundation Trust, London, United Kingdom; Centre for Medicines Optimization Research and Education, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Lizz Paley
- National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom
| | - Sally Vernon
- National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom
| | - John Deanfield
- Institute of Cardiovascular Science, University College London, United Kingdom; National Institute of Cardiovascular Outcomes Research, NHS Arden and Greater East Midlands Commissioning Support Unit, Leicester, United Kingdom
| | - Mark de Belder
- National Institute of Cardiovascular Outcomes Research, NHS Arden and Greater East Midlands Commissioning Support Unit, Leicester, United Kingdom
| | - Mark J Rutherford
- Department of Population Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Paul C Lambert
- Department of Population Health Sciences, University of Leicester, Leicester, United Kingdom; Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Sarah Slater
- Barts Cancer Centre, Barts Health NHS Trust, London, United Kingdom
| | - Kai-Keen Shiu
- Cancer Division, University College London Hospitals NHS Foundation Trust, London, United Kingdom; Cancer Institute, University College London, London, United Kingdom
| | - Li Wei
- Research Department of Practice and Policy, School of Pharmacy, University College London, United Kingdom; Centre for Medicines Optimization Research and Education, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Michael D Peake
- Department of Respiratory Medicine, University of Leicester, Glenfield Hospital, Leicester, United Kingdom; Cancer Research UK, London, England
| | - David Adlam
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
| | - Charlotte Manisty
- Institute of Cardiovascular Science, University College London, United Kingdom; Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom; National Disease Registration Service, NHS England, Wellington Place, Leeds, United Kingdom.
| |
Collapse
|
47
|
Laowichuwakonnukul K, Soontornworajit B, Arunpanichlert J, Rotkrua P. Simultaneous targeted delivery of doxorubicin and KRAS suppression by a hybrid molecule containing miR-143 and AS1411 aptamer. Sci Rep 2025; 15:10590. [PMID: 40148451 PMCID: PMC11950302 DOI: 10.1038/s41598-025-94159-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Hybrid molecules can be engineered to target tumors by merging drugs with the same or distinct mechanisms of action. The coexistence of multiple pharmacologically active entities within the cancer cell enhances the therapeutic efficacy of the hybrid molecule compared to single-target inhibitors. KRAS is considered the most common oncogenic gene in human cancers and is targeted by tumor suppressor miR-143. Therefore, an increase in miR-143 expression is a promising way to inhibit CRC cell growth. This research aims to develop a hybrid anticancer drug carrier by combining miR-143 and AS1411 aptamers through a hybridization strand (MAH) and loading doxorubicin (Dox), a chemotherapy drug. The uptake capability of MAH into the SW480 CRC cells was confirmed by detecting fluorescence intensity with a fluorescence microscope. After treatment of MAH in SW480 cells, the level of miR-143 was increased, but KRAS expression was decreased for both mRNA and protein. KRAS downstream target proteins, ERK and AKT, were downregulated as well. Furthermore, it was confirmed that DOX could be gradually released from MAH, with approximately 95% released over 72 h. Treating cells with Dox-MAH resulted in the inhibition of cell proliferation and induction of apoptosis. The protein expression of procaspase-3 and Bcl-2 was decreased, while Bax was increased, confirming that Dox-MAH triggered the cell apoptosis. The success of this research proposed a new strategy for a drug delivery system, which has multiple functions simultaneously; CRC cell-specificity, Dox carrier, and miR-143 delivery.
Collapse
Affiliation(s)
- Khanittha Laowichuwakonnukul
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Boonchoy Soontornworajit
- Department of Chemistry, Faculty of Science and Technology, Thammasat University, Pathumthani, 12120, Thailand
- Thammasat University Research Unit in Innovation of Molecular Hybrid for Biomedical Application, Pathumthani, Thailand
| | - Jiraporn Arunpanichlert
- Department of Chemistry, Faculty of Science and Technology, Thammasat University, Pathumthani, 12120, Thailand
- Thammasat University Research Unit in Innovation of Molecular Hybrid for Biomedical Application, Pathumthani, Thailand
| | - Pichayanoot Rotkrua
- Division of Biochemistry, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Thammasat University Research Unit in Innovation of Molecular Hybrid for Biomedical Application, Pathumthani, Thailand.
| |
Collapse
|
48
|
Sonbol MB, Bekaii-Saab T, Larsen CM. Fluoropyrimidine Therapy in Gastrointestinal Cancer: Balancing Survival Benefits and Cardiotoxicity Risks. JACC CardioOncol 2025:S2666-0873(25)00088-2. [PMID: 40202480 DOI: 10.1016/j.jaccao.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 04/10/2025] Open
Affiliation(s)
- Mohamad Bassam Sonbol
- Hematology and Medical Oncology, Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.
| | - Tanios Bekaii-Saab
- Hematology and Medical Oncology, Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | | |
Collapse
|
49
|
Bovinder Ylitalo E, Vidman L, Harlid S, Van Guelpen B. mRNA extracted from frozen buffy coat samples stored long term in tubes with no RNA preservative shows promise for downstream sequencing analyses. PLoS One 2025; 20:e0318834. [PMID: 40106499 PMCID: PMC11922291 DOI: 10.1371/journal.pone.0318834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/23/2025] [Indexed: 03/22/2025] Open
Abstract
Transcriptomics is an important OMICs method that is often unavailable in biobank research. Frozen blood samples are routinely collected and stored in medical biobanks, but transcriptional studies have been limited due to technical difficulties of extracting high-quality RNA from blood frozen in standard tubes (without RNA preservatives). We aimed to determine whether biobanked buffy coat samples stored at -80°C for up to 23 years could be successfully used for mRNA sequencing. We used a CryoXtract CXT 350 to remove frozen sample cores, which were immersed in RNA preservative during thawing prior to RNA extraction. RNA sequencing was then performed on extractions from pooled samples as well as from 23 buffy coat samples from prospective colorectal cancer cases and 23 matched controls included in the population-based, prospective Northern Sweden Health and Disease Study (NSHDS). For all samples, two library preparation methods were used (Illumina TruSeq Stranded mRNA poly-A selection and Illumina Stranded Total RNA with Ribo-Zero Globin). RNA yields of over 1 µg were obtained from the majority of NSHDS samples (mean = 2.57 µg), and over 92% of samples had RIN values of ≥ 6, indicating suitability for downstream analyses. In conclusion, we developed a method for successfully extracting and sequencing high-quality mRNA from frozen buffy coat samples stored long term in tubes with no RNA preservative.
Collapse
Affiliation(s)
| | - Linda Vidman
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
| | - Sophia Harlid
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
| | - Bethany Van Guelpen
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
50
|
Zhao Y, Zhang Z, Qiu JH, Li RY, Sun ZG. Catching cancer signals in the blood: Innovative pathways for early esophageal cancer diagnosis. World J Gastroenterol 2025; 31:101838. [PMID: 40093671 PMCID: PMC11886526 DOI: 10.3748/wjg.v31.i10.101838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
In recent years, significant progress has been made in the application of DNA methylation for the early detection of esophageal cancer (EC). As an epigenetic modification, DNA methylation allows for noninvasive screening by detecting the methylation status of circulating tumor DNA. Studies have shown that the methylation of genes such as SHOX2, SEPTIN9, EPO, and RNF180 significantly improves diagnostic sensitivity and specificity. Currently, SEPTIN9 has been approved by the Food and Drug Administration for colorectal cancer screening, while SHOX2 and EPO show promising results in EC, and RNF180 has potential in gastrointestinal tumors. This editorial reviews the study by Liu et al, which demonstrated the potential of combining the methylation of these four genes for early EC screening. In addition to their roles in early diagnosis, DNA methylation markers are gaining attention because of their roles in predicting recurrence and in postoperative follow-up. By monitoring changes in methylation levels, these markers can provide valuable insights into treatment efficacy and long-term management. As research progresses, liquid biopsy technology is expected to become an essential tool in the precision diagnosis and treatment of EC, benefiting patients significantly.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan 250063, Shandong Province, China
| | - Zhan Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan 250063, Shandong Province, China
| | - Jian-Hao Qiu
- Department of Thoracic Surgery, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan 250063, Shandong Province, China
| | - Rong-Yang Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan 250063, Shandong Province, China
| | - Zhen-Guo Sun
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan 250063, Shandong Province, China
| |
Collapse
|