101
|
Zeeuw M, Bereska J, Strampel M, Wagenaar L, Janssen B, Marquering H, Kemna R, van Waesberghe JH, van den Bergh J, Nota I, Moos S, Nio Y, Kop M, Kist J, Struik F, Wesdorp N, Nelissen J, Rus K, de Sitter A, Stoker J, Huiskens J, Verpalen I, Kazemier G. Evaluation of a deep-learning segmentation model for patients with colorectal cancer liver metastases (COALA) in the radiological workflow. Insights Imaging 2025; 16:110. [PMID: 40410643 PMCID: PMC12102431 DOI: 10.1186/s13244-025-01984-w] [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: 12/23/2024] [Accepted: 04/27/2025] [Indexed: 05/25/2025] Open
Abstract
OBJECTIVES For patients with colorectal liver metastases (CRLM), total tumor volume (TTV) is prognostic. A deep-learning segmentation model for CRLM to assess TTV called COlorectal cAncer Liver metastases Assessment (COALA) has been developed. This study evaluated COALA's performance and practical utility in the radiological picture archiving and communication system (PACS). A secondary aim was to provide lessons for future researchers on the implementation of artificial intelligence (AI) models. METHODS Patients discussed between January and December 2023 in a multidisciplinary meeting for CRLM were included. In those patients, CRLM was automatically segmented in portal-venous phase CT scans by COALA and integrated with PACS. Eight expert abdominal radiologists completed a questionnaire addressing segmentation accuracy and PACS integration. They were also asked to write down general remarks. RESULTS In total, 57 patients were evaluated. Of those patients, 112 contrast-enhanced portal-venous phase CT scans were analyzed. Of eight radiologists, six (75%) evaluated the model as user-friendly in their radiological workflow. Areas of improvement of the COALA model were the segmentation of small lesions, heterogeneous lesions, and lesions at the border of the liver with involvement of the diaphragm or heart. Key lessons for implementation were a multidisciplinary approach, a robust method prior to model development and organizing evaluation sessions with end-users early in the development phase. CONCLUSION This study demonstrates that the deep-learning segmentation model for patients with CRLM (COALA) is user-friendly in the radiologist's PACS. Future researchers striving for implementation should have a multidisciplinary approach, propose a robust methodology and involve end-users prior to model development. CRITICAL RELEVANCE STATEMENT Many segmentation models are being developed, but none of those models are evaluated in the (radiological) workflow or clinically implemented. Our model is implemented in the radiological work system, providing valuable lessons for researchers to achieve clinical implementation. KEY POINTS Developed segmentation models should be implemented in the radiological workflow. Our implemented segmentation model provides valuable lessons for future researchers. If implemented in clinical practice, our model could allow for objective radiological evaluation.
Collapse
Affiliation(s)
- Michiel Zeeuw
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam, The Netherlands.
| | - Jacqueline Bereska
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marius Strampel
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Luuk Wagenaar
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Boris Janssen
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Henk Marquering
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruby Kemna
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jan Hein van Waesberghe
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Janneke van den Bergh
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Irene Nota
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Shira Moos
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Yung Nio
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marnix Kop
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jakob Kist
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Femke Struik
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nina Wesdorp
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jules Nelissen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Katinka Rus
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Alexandra de Sitter
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaap Stoker
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joost Huiskens
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Inez Verpalen
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Geert Kazemier
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
102
|
Bolduan F, Müller-Bötticher N, Debnath O, Eichhorn I, Giesecke Y, Wetzel A, Sahay S, Zemojtel T, Jaeger M, Ungethuem U, Roderburg C, Kunze CA, Lehmann A, Horst D, Tacke F, Eils R, Wiedenmann B, Sigal M, Ishaque N. Small intestinal neuroendocrine tumors lack early genomic drivers, acquire DNA repair defects and harbor hallmarks of low REST expression. Sci Rep 2025; 15:17969. [PMID: 40410286 PMCID: PMC12102166 DOI: 10.1038/s41598-025-01912-4] [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: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
The tumorigenesis of small intestinal neuroendocrine tumors (siNETs) is not understood and comprehensive genomic and transcriptomic data sets are limited. Therefore, we performed whole genome and transcriptome analysis of 39 well differentiated siNET samples. Our genomic data revealed a lack of recurrent driver mutations and demonstrated that multifocal siNETs from individual patients can arise genetically independently. We detected germline mutations in Fanconi anemia DNA repair pathway (FANC) genes, involved in homologous recombination (HR) DNA repair, in 9% of patients and found mutational signatures of defective HR DNA repair in late-stage tumor evolution. Furthermore, transcriptomic analysis revealed low expression of the transcriptional repressor REST. Summarizing, we identify a novel common transcriptomic signature of siNETs and demonstrate that genomic alterations alone do not explain initial tumor formation, while impaired DNA repair likely contributes to tumor evolution and represents a potential pharmaceutical target in a subset of patients.
Collapse
Affiliation(s)
- Felix Bolduan
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353, Berlin, Germany
- BIH Charité Junior Digital Clinician Scientist Program, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Charitéplatz 1, 10117, Berlin, Germany
| | - Niklas Müller-Bötticher
- Center of Digital Health, Berlin Institute of Health at Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Olivia Debnath
- Center of Digital Health, Berlin Institute of Health at Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Ines Eichhorn
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353, Berlin, Germany
| | - Yvonne Giesecke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353, Berlin, Germany
| | - Alexandra Wetzel
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353, Berlin, Germany
| | - Shashwat Sahay
- Center of Digital Health, Berlin Institute of Health at Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Tomasz Zemojtel
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marten Jaeger
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Ute Ungethuem
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Catarina Alisa Kunze
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - Annika Lehmann
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 10117, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, CCCC (Campus Mitte), Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353, Berlin, Germany
| | - Roland Eils
- Center of Digital Health, Berlin Institute of Health at Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Bertram Wiedenmann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353, Berlin, Germany.
| | - Michael Sigal
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353, Berlin, Germany.
- Berlin Institute for Medical Systems Biology, Hannoversche Straße 28, 10115, Berlin, Germany.
| | - Naveed Ishaque
- Center of Digital Health, Berlin Institute of Health at Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
103
|
Yuan R, Song A, Zhang Y, Sha Y, Wang O, Jiang Y, Li M, Xia W, Zeng X, Xing X. High prevalence of hyperuricemia and its risk factors in a Chinese cohort of primary hyperparathyroidism patients. Endocrine 2025:10.1007/s12020-025-04276-x. [PMID: 40405058 DOI: 10.1007/s12020-025-04276-x] [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: 03/05/2025] [Accepted: 05/03/2025] [Indexed: 05/24/2025]
Abstract
PURPOSE Primary hyperparathyroidism (PHPT) is characterized by autonomous and excessive parathyroid hormone (PTH) secretion due to parathyroid gland lesions. Patients can be complicated with hyperuricemia (HUA), however, data about HUA in Chinese PHPT patients are lacking. This study aimed to explore the prevalence of HUA, factors influencing serum uric acid (SUA) level and the impacts of parathyroidectomy (PTX). METHODS 328 inpatients diagnosed with PHPT were included in total. Clinical data were collected and compared between HUA and non-HUA group. Multivariate linear regression was used to explore the factors affecting preoperative SUA and its variation after PTX. The impacts of SUA on bone mineral density (BMD) under different models were also analyzed. RESULTS The prevalence of HUA in PHPT patients was nearly 32%. Compared with non-HUA group, HUA group had significantly higher serum calcium (Ca) (2.92 [2.75, 3.26] vs. 2.78 [2.62, 3.03] mmol/L, P < 0.001), while PTH increased without significance. Gender, body mass index, serum Ca, triglycerides and renal function were independently associated with SUA. SUA decreased after PTX (340.00 [291.50, 421.00] vs. 315.00 [270.50, 375.00] μmol/L, P = 0.018), with change of SUA significantly associating with changes in serum Ca and phosphate. Presurgical SUA was positively related with femoral neck, total hip and lumbar spine BMDs after adjusting for confounders. CONCLUSION A relatively high proportion of HUA was found in our study. Besides the common risk factors, hypercalcemia may contribute to the increased SUA in PHPT patients, which could be significantly improved by curative PTX. SUA might have a protective effect on bone loss in PHPT patients.
Collapse
Affiliation(s)
- Runxue Yuan
- Department of Family Medicine & Division of General Internal Medicine, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Peking Union Medical College, Beijing, China
| | - An Song
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yun Zhang
- Department of Family Medicine & Division of General Internal Medicine, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Peking Union Medical College, Beijing, China
| | - Yue Sha
- Department of Family Medicine & Division of General Internal Medicine, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Peking Union Medical College, Beijing, China.
| | - Ou Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yan Jiang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Mei Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Weibo Xia
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xuejun Zeng
- Department of Family Medicine & Division of General Internal Medicine, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases (Peking Union Medical College Hospital), Peking Union Medical College, Beijing, China
| | - Xiaoping Xing
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
| |
Collapse
|
104
|
Gao Y, Liu D, Xiao Q, Huang S, Li L, Xie B, Zhou L, Qi Y, Liu Y. Exploration of Pathogenesis and Cutting-Edge Treatment Strategies of Sarcopenia: A Narrative Review. Clin Interv Aging 2025; 20:659-684. [PMID: 40438271 PMCID: PMC12117577 DOI: 10.2147/cia.s517833] [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: 01/21/2025] [Accepted: 05/12/2025] [Indexed: 06/01/2025] Open
Abstract
Sarcopenia a progressive and multifactorial musculoskeletal syndrome characterized by loss of muscle mass and function, poses a significant global health challenge, particularly in aging populations. Epidemiological studies reveal that sarcopenia affects approximately 5-10% of the general population, with prevalence rates escalating dramatically after age 60 to reach 10-27% in older adults. This age-associated increase contributes significantly to healthcare burdens by elevating risks of disability, frailty, and mortality. Despite its profound impact, current clinical approaches to sarcopenia remain limited. While resistance exercise and protein supplementation form the cornerstone of management, their efficacy is often constrained by poor long-term adherence and variable individual responses, highlighting the urgent need for more comprehensive and personalized treatment strategies. The pathogenesis of sarcopenia is complex and influenced by various factors, including aging, inflammation, nutritional deficits, physical inactivity, and mitochondrial dysfunction. However, the precise molecular mechanisms underlying this condition are still not fully understood. Recent research has made significant strides in elucidating the intricate mechanisms contributing to sarcopenia, revealing novel insights into its molecular and cellular underpinnings. Notably, emerging evidence points to the pivotal role of mitochondrial dysfunction, altered myokine profiles, and neuromuscular junction degeneration in sarcopenia progression. Additionally, breakthroughs in stem cell therapy, exosome-based treatments, and precision nutrition offer promising avenues for clinical intervention. This review aims to synthesize the latest advancements in sarcopenia research, focusing on the novel contributions to its pathogenesis and treatment strategies. We explore emerging trends such as the role of cellular senescence, epigenetic regulation, and targeted therapeutic interventions that could reshape future approaches to managing sarcopenia. By highlighting recent breakthroughs and cutting-edge research, we hope to advance the understanding of sarcopenia and foster the translation of these findings into effective clinical therapies.
Collapse
Affiliation(s)
- Yin Gao
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Di Liu
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Qixian Xiao
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
| | - Shan Huang
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Li Li
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Baocheng Xie
- Department of Pharmacy, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan, 52305, People’s Republic of China
| | - Limin Zhou
- Department of Pharmacy, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan, 52305, People’s Republic of China
| | - Yi Qi
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Yanzhi Liu
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
| |
Collapse
|
105
|
Peng L, Ma W, Zhang X, Zhang F, Ma F, Ai K, Ma X, Jia Y, Ou-Yang H, Pei S, Wang T, Zhu Y, Wang L. Predictive value of combined DCE-MRI perfusion parameters and clinical features nomogram for microsatellite instability in colorectal cancer. Discov Oncol 2025; 16:892. [PMID: 40410525 PMCID: PMC12102045 DOI: 10.1007/s12672-025-02705-x] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025] Open
Abstract
OBJECTIVES To develop a nomogram that combines dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) perfusion parameters, ADC values and clinical features to preoperatively identify microsatellite instability (MSI) in patients with colorectal cancer (CRC). METHODS This retrospective study included 63 CRC patients who underwent preoperative DCE-MRI and had immunohistochemistry results available. Two radiologists, in a double-blind manner, placed two circular regions of interests in the area with the highest perfusion intensity on the DCE-MRI perfusion map and the corresponding area on the ADC map. Perfusion parameters and ADC values were measured, and the average values from both radiologists were used for subsequent analysis. Univariate analysis was performed to identify independent risk factors for MSI. A nomogram was then constructed by combining the most significant clinical risk factors with DCE-MRI perfusion parameters. The model's performance was evaluated using receiver operating characteristic (ROC) curves. Calibration curves, decision curve analysis (DCA), and clinical impact curves (CIC) were used to assess the nomogram's clinical utility and net benefit. RESULTS The nomogram prediction model, which combined PLT, LNM, Ktrans, Kep, iAUC, and ADC, demonstrated good predictive performance. The combined model had an AUC of 0.951 (95% CI: 0.903-0.998), an accuracy of 0.873, a sensitivity of 1.000, and a specificity of 0.818. Both the DCA and CIC demonstrated good clinical applicability and net benefit. CONCLUSION The nomogram method demonstrated good potential in the preoperative individualized identification of MSI status in CRC patients. This tool can assist clinicians in adopting appropriate treatment strategies and optimizing personalized stratification for CRC patients.
Collapse
Affiliation(s)
- Leping Peng
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Wenting Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Xiuling Zhang
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Fan Zhang
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Fang Ma
- Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Kai Ai
- Department of Clinical and Technical Support, Philips Healthcare, Xi'an, 710065, Shanxi, China
| | - Xiaomei Ma
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Yingmei Jia
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Hong Ou-Yang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Shengting Pei
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Tao Wang
- Department of Colorectal Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
| | - Yuanhui Zhu
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.
| | - Lili Wang
- Department of Radiology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
106
|
Zaiki Y, Yap PG, Gan CY, Rani MFA, Traini D, Wong TW. "Actual" peptide properties required for nanoparticle development in precision cancer therapeutic delivery. J Control Release 2025:113866. [PMID: 40412661 DOI: 10.1016/j.jconrel.2025.113866] [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: 02/19/2025] [Revised: 04/27/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Functionalizing nanoparticles with peptides (3-30 amino acids) reduces premature clearance and increases colloidal stability and targeting capacity of cancer therapeutics. Glutamate/lysine-rich zwitterionic and hydrophilic/neutral peptides minimize reticuloendothelial digestion of nanomedicine through reducing particle hydrophobicity and depressing plasma anti-PEG immunoglobulin that disrupts the PEG-based particle stealth. Anionic peptides negate protein corona formation and subsequent particle aggregation in vivo enabling efficient nanoparticles biodistribution and drug targeting by facilitating their endothelial/extracellular matrix pore diffusion. Cationic and hydrophobic peptides display a strong affinity for anionic cancer cell membrane and mediate membrane porosification or receptor binding leading to particle uptake and endocytosis. The peptide ionic and hydrophobicity/hydrophilicity attributes collectively facilitate endosomal escape, and nuclear and mitochondria targeting of nanoparticles. Peptides are required to present with different physicochemical attributes from administration site, through blood and extracellular matrix, to cancer site of action. Charge/hydrophilicity-hydrophobicity switching and projection of receptor-specific domain of peptides are attainable through pH-pKa interplay and labile bond hydrolysis of "unwanted" domain to give rise to new functional domains in response to pH, thermal and enzymatic stimuli. Co-introducing all functional attributes on a single peptide is challenging. Use of peptide blends risks leaching during nanoparticles production. Peptides-nanoparticles conjugation risks peptide conformational alterations and loss of acidic/basic termini affecting their roles in nanoparticle stabilization, targeting, membrane permeabilization and subcellular delivery.
Collapse
Affiliation(s)
- Yazid Zaiki
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia
| | - Pei Gee Yap
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia, University Innovation Incubator Building, SAINS@USM campus, Lebuh Bukit Jambul, Bayan Lepas, 11900, Penang, Malaysia
| | - Chee Yuen Gan
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia, University Innovation Incubator Building, SAINS@USM campus, Lebuh Bukit Jambul, Bayan Lepas, 11900, Penang, Malaysia
| | | | - Daniela Traini
- Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe, Sydney 2037, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Campus Macquarie Park, Sydney 2019, Australia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
107
|
Delgado-López PD, Cárdenas Montes M, Troya García J, Ocaña-Tienda B, Cepeda S, Martínez Martínez R, Corrales-García EM. Artificial intelligence in neuro-oncology: methodological bases, practical applications and ethical and regulatory issues. Clin Transl Oncol 2025:10.1007/s12094-025-03948-4. [PMID: 40402414 DOI: 10.1007/s12094-025-03948-4] [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: 03/12/2025] [Accepted: 04/26/2025] [Indexed: 05/23/2025]
Abstract
Artificial Intelligence (AI) is transforming neuro-oncology by enhancing diagnosis, treatment planning, and prognosis prediction. AI-driven approaches-such as CNNs and deep learning-have improved the detection and classification of brain tumors through advanced imaging techniques and genomic analysis. Explainable AI methods mitigate the "black box" problem, promoting model transparency and clinical trust. Mechanistic models complement AI by integrating biological principles, enabling precise tumor growth predictions and treatment response assessments. AI applications also include the creation of digital twins for personalized therapy optimization, virtual clinical trials, and predictive modeling for estimation of tumor resection and pattern of recurrence. However, challenges such as data bias, ethical concerns, and regulatory compliance persist. The European Artificial Intelligence Act and the Health Data Space Regulation impose strict data protection and transparency requirements. This review explores AI's methodological foundations, clinical applications, and ethical challenges in neuro-oncology, emphasizing the need for interdisciplinary collaboration and regulatory adaptation.
Collapse
Affiliation(s)
- Pedro David Delgado-López
- Servicio de Neurocirugía, Hospital Universitario de Burgos, Avda Islas Baleares 3, 09006, Burgos, Spain.
| | - Miguel Cárdenas Montes
- Departamento de Investigación Básica, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Jesús Troya García
- Servicio de Medicina Interna, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Beatriz Ocaña-Tienda
- Centro Nacional de Investigaciones Oncológicas (CNIO), Unidad de Bioinformática, Madrid, Spain
| | - Santiago Cepeda
- Servicio de Neurocirugía, Hospital Universitario Rio Hortega, Valladolid, Spain
- Grupo Especializado en Imagen Biomédica y Análisis Computacional (GEIBAC), Instituto de Investigación Biosanitaria de Valladolid (IBioVall), Valladolid, Spain
| | - Ricard Martínez Martínez
- Facultad de Derecho, Cátedra de Privacidad y Transformación Digital de la Universidad de Valencia, Valencia, Spain
| | | |
Collapse
|
108
|
Dou YN, Wang J. Advancing Oncology Drug Development in the US: The Interplay between Innovations and Regulatory Science. Ther Innov Regul Sci 2025:10.1007/s43441-025-00800-3. [PMID: 40405050 DOI: 10.1007/s43441-025-00800-3] [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: 09/11/2024] [Accepted: 05/07/2025] [Indexed: 05/24/2025]
Abstract
The landscape of drug development has evolved with the adoption of new therapeutic modalities, cutting-edge technology platforms, emerging scientific insights, and modern patient-centric clinical trial designs. In this review, we investigate the interplay between innovation and regulatory science in cancer drug development in the United States. As new innovations emerge, regulatory science adapts to integrate new discoveries and technologies, ensuring alignment with established regulations and safety standards. This fuels additional innovations through data and evidence generation, potentially expediting the development of revolutionary treatments and advancing patient access to novel, promising therapies. Early and frequent engagement with regulators is vital for drug developers aiming to successfully apply innovative approaches.
Collapse
Affiliation(s)
- Yannan Nancy Dou
- Oncology Regulatory Science, Strategy & Excellence, AstraZeneca, Gaithersburg, MD, USA.
| | - Jian Wang
- Oncology Regulatory Science, Strategy & Excellence, AstraZeneca, Gaithersburg, MD, USA.
| |
Collapse
|
109
|
Gebauer N, Wang SS. The Role of EBV in the Pathogenesis of Diffuse Large B-Cell Lymphoma. Curr Top Microbiol Immunol 2025. [PMID: 40399571 DOI: 10.1007/82_2025_296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
There are multiple established risk factors for DLBCL; these risk factors share an underlying biology, which generally cause immune dysfunction, spanning immunosuppression to chronic inflammation. EBV is an established risk factor for DLBCL and approximately 10% of DLBCLs are EBV-positive. EBV is a ubiquitous infection, and it is thus among populations that are immunocompromised, by age or medically defined, where EBV-positive DLBCLs arise. In this chapter, we review the current classification, epidemiology, clinical, pathology, and molecular characteristics of EBV-positive DLBCL, and discuss the role of EBV in lymphoma tumorigenesis. We further discuss current and novel treatments aimed at the NFκB pathway and other targets.
Collapse
Affiliation(s)
- Niklas Gebauer
- Klinik für Hämatologie und Onkologie, UKSH Campus Lübeck, Lübeck, Germany
| | - Sophia S Wang
- Division of Computational and Quantitative Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
110
|
Leberle J, Perona M, Choudhury A, Bellora N, Ibañez IL, Biolatti LV. Alternative splicing in human cells exposed to ionizing radiation: a comprehensive review of ex vivo and in vivo studies. Int J Radiat Biol 2025:1-10. [PMID: 40402085 DOI: 10.1080/09553002.2025.2505524] [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: 10/14/2024] [Revised: 03/02/2025] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE This study reviews how ionizing radiation (IR) induces alternative splicing (AS) in non-tumor and tumor cells under both ex vivo and in vivo irradiation conditions. The relevance and limitations of IR-induced AS in identifying potential biomarkers are highlighted for two main applications: biodosimetry and radiotherapy. CONCLUSIONS Radiation promotes alterations in AS, which may differentially affect the response in both tumor and non-tumor cells. This response can occur in genes that change their overall expression as well as in those that remain unaltered in response to IR. Although cis-regulators modulate AS, trans-regulators like splicing factors are more involved in the IR response. Variants of key genes involved in the DNA damage response (DDR) are regulated in non-tumor cells while they are often deregulated in tumor cells favoring radioresistance. Identifying IR-induced AS variants could enhance the sensitivity of biodosimeters for dose estimation and biomarkers for radiosensitivity, offering potential strategies to personalize radiotherapy and improve outcomes. New and advanced sequencing technologies will allow variant identification important for the field of radiobiological research.
Collapse
Affiliation(s)
- Jerónimo Leberle
- Laboratory of Radiobiology and Biodosimetry, Nuclear Medicine and Radiotherapy Center, Institute of Nuclear Technologies for Health (INTECNUS), National Atomic Energy Commission (CNEA), S.C. de Bariloche, Río Negro, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Marina Perona
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Department of Radiobiology, Nuclear Biochemistry Division, National Atomic Energy Commission (CNEA), San Martín, Buenos Aires, Argentina
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Nicolas Bellora
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Laboratory of Computational Genomics, Nuclear Medicine and Radiotherapy Center, Institute of Nuclear Technologies for Health (INTECNUS), National Atomic Energy Commission (CNEA), S.C. de Bariloche, Río Negro, Argentina
| | - Irene L Ibañez
- Technology and Applications of Accelerators Assistant Management, Research and Applications Management, National Atomic Energy Commission (CNEA), Constituyentes Atomic Center, San Martín, Buenos Aires, Argentina
- Institute of Nanosciences and Nanotechnology (INN), National Atomic Energy Commission (CNEA) - National Scientific and Technical Research Council (CONICET), San Martín, Buenos Aires, Argentina
| | - Luisa V Biolatti
- Division of Cancer Sciences, The University of Manchester, The Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
111
|
Chen F, Zhang Y, Li W, Sedlazeck FJ, Shen L, Creighton CJ. Global DNA methylation differences involving germline structural variation impact gene expression in pediatric brain tumors. Nat Commun 2025; 16:4713. [PMID: 40399292 PMCID: PMC12095544 DOI: 10.1038/s41467-025-60110-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: 11/18/2024] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
The extent of genetic variation and its influence on gene expression across multiple tissue and cellular contexts is still being characterized, with germline Structural Variants (SVs) being historically understudied. DNA methylation also represents a component of normal germline variation across individuals. Here, we combine germline SVs (by short-read sequencing) with tumor DNA methylation across 1292 pediatric brain tumor patients. For thousands of methylation probes for CpG Islands (CGIs) or enhancers, rare and common SV breakpoints upstream or downstream associate with differential methylation in tumors spanning various histologic types, a significant subset involving genes with SV-associated differential expression. Cancer predisposition genes involving SV-associated differential methylation and expression include MSH2, RSPA, and PALB2. SV breakpoints falling within CGIs or histone marks H3K36me3 or H3K9me3 associate with differential CGI methylation. Genes with SVs and CGI methylation associated with patient survival include POLD4. Our results capture a class of normal phenotypic variation having disease implications.
Collapse
Affiliation(s)
- Fengju Chen
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Computer Science, Rice University, Houston, TX, 77005, USA
| | - Lanlan Shen
- USDA Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
112
|
Gatenby RA, Teer JK, Tsai KY, Brown JS. Parallel and convergent dynamics in the evolution of primary breast and lung adenocarcinomas. Commun Biol 2025; 8:775. [PMID: 40399443 PMCID: PMC12095661 DOI: 10.1038/s42003-025-08123-7] [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: 10/31/2024] [Accepted: 04/23/2025] [Indexed: 05/23/2025] Open
Abstract
Cancer development requires an evolutionary transformation from mammalian cells fully regulated by and integrated into multicellular tissue to cancer cells that, as single cell protists, are individually subject to Darwinian selection. Through genetic and epigenetic mechanisms of inheritance, the evolving cancer phenotype must acquire independence from host controls, downregulate differentiated functions that benefit the host but not individual cells, and generate phenotypic traits that increase fitness in the context of the selection forces within the local microenvironment. Here, we investigate this evolutionary transition in breast (BRCA) and lung (LUAD, without EGFR, KRAS or BRAF driver mutations) adenocarcinomas using bulk mutation and expression data from the TCGA database. We define evolution selection for genes and molecular pathways based on 1) changes in gene expression compared to normal tissue, and 2) significantly larger or smaller observed mutation rates compared to those expected based on the gene size. We find BRCA and LUAD disable different genes and gene pathways associated with tissue-specific signaling and differentiated functions but promote common molecular pathways associated with cell cycle, cell-cell interactions, cytoskeleton, voltage gated ion channels, and microenvironmental niche construction. Thus, tissue-specific parallel evolution in early cancer development is followed by convergence to a common cancer phenotype.
Collapse
Affiliation(s)
- Robert A Gatenby
- Cancer Biology and Evolution Program, Tampa, FL, USA.
- Integrated Mathematical Oncology Department, Tampa, FL, USA.
| | - Jamie K Teer
- Biostatistics and Bioinformatics Department, Tampa, FL, USA
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, Tampa, FL, USA
- Pathology Department Moffitt Cancer Center, Tampa, FL, USA
| | - Joel S Brown
- Cancer Biology and Evolution Program, Tampa, FL, USA
- Integrated Mathematical Oncology Department, Tampa, FL, USA
| |
Collapse
|
113
|
Carr A, Jackson JB, Coldren C, Chandra P, Koohestani F, Shiller M, Auber R. Tumor diagnosis recharacterization enabled by comprehensive genomic profiling to guide precision medicine strategy. NPJ Precis Oncol 2025; 9:149. [PMID: 40399445 PMCID: PMC12095656 DOI: 10.1038/s41698-025-00942-5] [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: 10/02/2024] [Accepted: 05/11/2025] [Indexed: 05/23/2025] Open
Abstract
Comprehensive genomic profiling (CGP) via next-generation sequencing is standard clinical practice for advanced and metastatic cancers in the U.S. and can help identify clinically actionable alterations in patients who may benefit from targeted therapies. CGP can also complement clinicopathological findings and in certain cases, may lead to diagnostic recharacterization resulting in more precise therapeutic strategies. Here, we highlight examples where molecular findings resulted in tumor re-evaluation and subsequent recharacterization. Twenty-eight cases where CGP results were inconsistent with initial pathological diagnosis and clinical presentation were selected for secondary clinicopathological review to explore alternative diagnostic explanations more consistent with the genomic results. Genomic profiling identified clinically actionable and prognostic variants leading to more accurate therapeutic recommendations based on the updated diagnoses highlighting the value of CGP beyond biomarker detection for therapy selection and supporting its complementary use in diagnostic confirmation to unveil opportunities for precision medicine strategies.
Collapse
Affiliation(s)
- Ann Carr
- PathGroup, Nashville, TN, 37217, USA.
| | | | | | | | | | | | | |
Collapse
|
114
|
Hu D, Xu B, Huang G, Hu X, Li J, Chen Z, Liu W, Wen Z. CALB2 facilitates macrophage M2 polarization to promote the growth and metastasis of pancreatic adenocarcinoma. Cell Signal 2025; 134:111887. [PMID: 40409389 DOI: 10.1016/j.cellsig.2025.111887] [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: 02/14/2025] [Revised: 05/02/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Tumor-associated macrophages mainly differentiate into M2 phenotypes, which secrete cytokines that reshape the tumor microenvironment and promote tumor progression. This study was to explore the mechanism of CALB2 in M2 polarization and pancreatic adenocarcinoma (PAAD). Clinical tissue samples of PAAD were collected, followed by detection of WTAP, FOSL1, and CALB2 expression. The correlation between WTAP and FOSL1 or between FOSL1 and CALB2 was analyzed. THP1 cells were induced into M0 macrophages, followed by plasmid transfection and induction of M2-type macrophages. After macrophages were co-cultured with PAAD cells, functional experiments were designed to evaluate PAAD cell malignant behaviors. A transplantation tumor model and a liver metastasis model were established to assess tumor growth and metastasis. High expression of WTAP, FOSL1, and CALB2 was found in PAAD tissues and M2-type macrophages. WTAP positively linked with FOSL1, so as FOSL1 and CALB2. Mechanistically, WTAP enhanced m6A modification of FOSL1 to promote its expression, and FOSL1 promoted CALB2 transcription. Knockdown of WTAP, FOSL1, or CALB2 in macrophages inhibited PAAD cell malignant behaviors, which could be reversed by CALB2 upregulation. WTAP knockdown restrained the growth and metastasis of PAAD in nude mice via the FOSL1/CALB2 axis. In conclusion, WTAP increased the m6A level of FOSL1, activated CALB2 transcription, and promoted M2 polarization of macrophages, thereby promoting the growth and metastasis of PAAD.
Collapse
Affiliation(s)
- Dongwei Hu
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang 325000, PR China
| | - Bo Xu
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Guoyu Huang
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ 85259, USA
| | - Xiaowei Hu
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang 325000, PR China
| | - Jinjie Li
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Zongjing Chen
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Wei Liu
- Institute of Digestive Disease, China Three Gorges University, Yichang, PR China.
| | - Zhengde Wen
- Department of Hepato-pancreato-biliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China.
| |
Collapse
|
115
|
Kang YJ, Pan L, Liu Y, Rong Z, Liu J, Liu F. GEPIA3: Enhanced drug sensitivity and interaction network analysis for cancer research. Nucleic Acids Res 2025:gkaf423. [PMID: 40396370 DOI: 10.1093/nar/gkaf423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/18/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
The GEPIA series has provided robust and widely used tools for pan-cancer analysis of gene expression data. In the post-genomic era, a major challenge lies in deconvoluting complex regulatory relationship influenced by multiple factors and discovering gene-based precision therapeutics. Here we present GEPIA3, an advanced version of GEPIA that provides a comprehensive analysis of gene/protein interactions across various cancer types. This version facilitates the investigation of treatment sensitivity utilizing both real-world patient data and cell line screens for over 1000 therapeutic agents, as well as the integration of RNA alterations derived from the pan-cancer analysis of whole genomes project. GEPIA3 represents a significant enhancement of the original platform, enabling in-depth exploration of gene regulation and cancer phenotypes, thereby supporting the identification of novel biomarkers and therapeutic targets. GEPIA3 is publicly accessible at https://gepia3.bioinfoliu.com.
Collapse
Affiliation(s)
- Yu-Jian Kang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Lingjie Pan
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Yiyu Liu
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhengqin Rong
- Wuxi Yiou Biotechnology Co., Ltd., Wuxi 214000, China
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P. R. China
| | - Jiaxi Liu
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Fenglin Liu
- School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
116
|
Huang WJ, Xie HB, Liu PP, Liu L, Liu ZY, Wang QJ, Li YZ, Meng QW, Wang RT. Pericardial Fat and Primary Tumor Radiomics for Predicting Occult N2 Disease and Survival in Clinical Stage I Non-Small Cell Lung Cancer: Multicenter Study With Biologic Correlation. AJR Am J Roentgenol 2025. [PMID: 40397555 DOI: 10.2214/ajr.25.32861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Background: Occult N2 disease significantly affects clinical stage I non-small cell lung cancer (NSCLC) prognosis. Pericardial fat characteristics also have prognostic associations. Objective: To develop and test a model incorporating pericardial fat and tumor radiomic features on CT for detecting occult N2 disease in clinical stage I NSCLC, explore the model's prognostic role, and investigate its biologic basis through radiogenomics analyses. Methods: This retrospective study included patients who underwent clinical stage I NSCLC resection at three hospitals [center 1 (January 2016 to December 2022), stratified randomly by 6:2:2 ratio into training, tuning, and internal test sets; centers 2 and 3 (January 2019 to December 2023), serving as external test sets]. Pericardial fat and primary tumors were segmented on preoperative CT to extract radiomic features and generate tumor and fat rad-scores, respectively. Multivariable analysis was performed to create a hybrid model for predicting occult N2 disease at surgery. Performance was evaluated in external test sets. Associations with recurrence-free survival (RFS) and overall survival (OS) were evaluated using log-rank tests in the internal test set; follow-up data were unavailable in external test sets. Biologic mechanisms were explored through RNA and gene expression analysis in a separate set of patients with NSCLC obtained from a public radiogenomics database. Results: From the three centers, 1662 patients (mean age, 58.6 years; 663 men, 999 women) were included. Following multivariable analysis, the hybrid model included nodule density, fat rad-score, and tumor rad-score. The model had AUC, accuracy, sensitivity, and specificity for occult N2 disease of 0.921, 89.7%, 59.3%, and 93.1%, and 0.913, 91.8%, 56.2%, and 95.5% in external test sets 1 and 2, respectively. High-risk compared with low-risk patients, applying the model in the internal test set, showed worse RFS (p<.001) and OS (p<.001). In 122 patients in radiogenomics analysis, high-risk status was associated with activation of molecular pathways and increased activated dendritic cell and mast cell infiltration. Conclusion: A model incorporating tumor and pericardial fat radiomics showed good performance in predicting occult N2 disease as well as associations with survival and with RNA and gene expression. Clinical Impact: The model could help guide NSCLC management.
Collapse
Affiliation(s)
- Wen-Juan Huang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Han-Bing Xie
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Ping-Ping Liu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Le Liu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Zeng-Yao Liu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Department of Interventional Medicine, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Qiu-Jun Wang
- Department of General Practice, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, 150086, China
| | - Yuan-Zhou Li
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Qing-Wei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| |
Collapse
|
117
|
Filis P, Papagiannopoulos CK, Markozannes G, Chalitsios CV, Zerdes I, Valachis A, Papandreou C, Christakoudi S, Tsilidis KK. Associations of sarcopenia, sarcopenia components and sarcopenic obesity with cancer incidence: A prospective cohort study of 414,094 participants in UK Biobank. Int J Cancer 2025. [PMID: 40396701 DOI: 10.1002/ijc.35480] [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: 08/14/2024] [Revised: 04/17/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025]
Abstract
Sarcopenia is characterised by low grip strength, muscle quantity or quality, and physical performance. This study investigated the associations of sarcopenia and its components with cancer incidence. A prospective cohort study was conducted utilising data from the UK Biobank. Sarcopenia and its components were defined according to the European Working Group on Sarcopenia in Older People criteria (EWGSOP2 2019). Cox proportional hazard models adjusted for sociodemographic, lifestyle, and health-related factors were performed. Overall, 63,379 out of 414,094 study participants had an incident diagnosis of cancer during a median follow-up of 11.7 years. In total, 32,286 participants had probable sarcopenia and 934 confirmed/severe sarcopenia at recruitment. Combined probable, confirmed, and severe sarcopenia was associated with a higher risk of liver (hazard ratio [HR] = 1.65, 95% confidence interval [CI]: 1.17-2.33), haematological (HR = 1.22, 95% CI: 1.01-1.46), and colorectal cancer (HR = 1.21, 95% CI: 1.04-1.41) in males, but not in females. The components of sarcopenia were associated with a higher risk of several cancers, including low grip strength (with liver, haematological and colorectal cancer in males), low muscle mass index (oesophageal in females and oral cancer in males), and slow walking pace (liver and lung in males, lung and overall cancer in females). Compared to participants with non-sarcopenic obesity, those with sarcopenic obesity had a higher risk of colorectal cancer in males (HR = 1.31, 95% CI: 1.03-1.68). Our study suggests that sarcopenia, sarcopenia components, and sarcopenic obesity can be associated with risk for several cancers, mainly of the gastrointestinal tract and in males. Thus, early identification of sarcopenia components may benefit cancer prevention.
Collapse
Affiliation(s)
- Panagiotis Filis
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
- Department of Medical Oncology, University of Ioannina School of Medicine, Ioannina, Greece
| | | | - Georgios Markozannes
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Christos V Chalitsios
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Ioannis Zerdes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Christopher Papandreou
- Institut d'Investigació Sanitària Pere Virgili (IISPV), NeuroÈpia Group, Hospital Universitari Sant Joan de Reus, Reus (Tarragona), Spain
- Department of Nutrition and Dietetics Sciences, School of Health Sciences, Hellenic Mediterranean University (HMU), Siteia, Greece
| | - Sofia Christakoudi
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Konstantinos K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
118
|
Alberge JB, Dutta AK, Poletti A, Coorens THH, Lightbody ED, Toenges R, Loinaz X, Wallin S, Dunford A, Priebe O, Dagan J, Boehner CJ, Horowitz E, Su NK, Barr H, Hevenor L, Towle K, Beesam R, Beckwith JB, Perry J, Cordas Dos Santos DM, Bertamini L, Greipp PT, Kübler K, Arndt PF, Terragna C, Zamagni E, Boyle EM, Yong K, Morgan G, Walker BA, Dimopoulos MA, Kastritis E, Hess J, Sklavenitis-Pistofidis R, Stewart C, Getz G, Ghobrial IM. Genomic landscape of multiple myeloma and its precursor conditions. Nat Genet 2025:10.1038/s41588-025-02196-0. [PMID: 40399554 DOI: 10.1038/s41588-025-02196-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/15/2025] [Indexed: 05/23/2025]
Abstract
Reliable strategies to capture patients at risk of progression from precursor stages of multiple myeloma (MM) to overt disease are still missing. We assembled a comprehensive collection of MM genomic data comprising 1,030 patients (218 with precursor conditions) that we used to identify recurrent coding and non-coding candidate drivers as well as significant hotspots of structural variation. We used those drivers to define and validate a simple 'MM-like' score, which we could use to place patients' tumors on a gradual axis of progression toward active disease. Our MM precursor genomic map provides insights into the time of initiation and cell-of-origin of the disease, order of acquisition of genomic alterations and mutational processes found across the stages of transformation. Taken together, we highlight here the potential of genome sequencing to better inform risk assessment and monitoring of MM precursor conditions.
Collapse
Affiliation(s)
- Jean-Baptiste Alberge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ankit K Dutta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrea Poletti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Tim H H Coorens
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elizabeth D Lightbody
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rosa Toenges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xavi Loinaz
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sofia Wallin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew Dunford
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Oliver Priebe
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Johnathan Dagan
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cody J Boehner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Erica Horowitz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nang K Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hadley Barr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laura Hevenor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine Towle
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rashmika Beesam
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jenna B Beckwith
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacqueline Perry
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David M Cordas Dos Santos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Luca Bertamini
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Patricia T Greipp
- Department of Laboratory Medicine and Pathology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, USA
| | - Kirsten Kübler
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center of Functional Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Krantz Family Center for Cancer Research and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Peter F Arndt
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Carolina Terragna
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Elena Zamagni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Eileen M Boyle
- Department of Haematology, Cancer Institute, University College London Cancer Institute, London, UK
| | - Kwee Yong
- Department of Haematology, Cancer Institute, University College London Cancer Institute, London, UK
| | - Gareth Morgan
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Brian A Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology and Oncology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | | | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Julian Hess
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Romanos Sklavenitis-Pistofidis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Chip Stewart
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gad Getz
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Krantz Family Center for Cancer Research and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Pathology, Harvard Medical School, Boston, MA, USA.
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
119
|
Gustafsson OJR, Wilkinson SR, Bacall F, Soiland-Reyes S, Leo S, Pireddu L, Owen S, Juty N, Fernández JM, Brown T, Ménager H, Grüning B, Capella-Gutierrez S, Coppens F, Goble C. WorkflowHub: a registry for computational workflows. Sci Data 2025; 12:837. [PMID: 40399296 PMCID: PMC12095652 DOI: 10.1038/s41597-025-04786-3] [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: 11/05/2024] [Accepted: 03/07/2025] [Indexed: 05/23/2025] Open
Abstract
The rising popularity of computational workflows is driven by the need for repetitive and scalable data processing, sharing of processing know-how, and transparent methods. As both combined records of analysis and descriptions of processing steps, workflows should be reproducible, reusable, adaptable, and available. Workflow sharing presents opportunities to reduce unnecessary reinvention, promote reuse, increase access to best practice analyses for non-experts, and increase productivity. In reality, workflows are scattered and difficult to find, in part due to the diversity of available workflow engines and ecosystems, and because workflow sharing is not yet part of research practice. WorkflowHub provides a unified registry for all computational workflows that links to community repositories, and supports both the workflow lifecycle and making workflows findable, accessible, interoperable, and reusable (FAIR). By interoperating with diverse platforms, services, and external registries, WorkflowHub adds value by supporting workflow sharing, explicitly assigning credit, enhancing FAIRness, and promoting workflows as scholarly artefacts. The registry has a global reach, with hundreds of research organisations involved, and more than 800 workflows registered.
Collapse
Grants
- European Commission Horizon 2020 programme: H2020-INFRAEOSC-2018-2 824087, H2020-INFRADEV-2019-2 871118 European Commission Horizon Europe programme: HORIZONINFRA-2021-EOSC-01-04 101057388, HORIZONINFRA-2021-EMERGENCY-01 101046203, HORIZON-INFRA-2021-EOSC-01-05 101057344, HORIZON-INFRA-2021-TECH-01-01 101057437, HORIZON-INFRA-2021-SERV-01-02 101058020, HORIZON-INFRA-2022-TECH-01 101094287 UKRI Innovate UK under the UK Government’s Horizon Europe funding guarantee: 10038963, 10038992, 10038930, 10038927, 10048146
- NCRIS via Bioplatforms Australia
- Office of Science of the U.S. Department of Energy DE-AC05-00OR22725.
- Sardinian Regional Government XData Project Italian Ministry of Health, POS T3 LIFEMAP project
- European Commission Horizon 2020 programme: H2020-INFRAEOSC-2018-2 824087 European Commission Horizon Europe programme: HORIZONINFRA-2021-EMERGENCY-01 101046203
- HORIZON-INFRA-2021-TECH-01-01 101057437
- German Federal Ministry of Education and Research BMBF grant 031 A538A de.NBI-RBC The Ministry of Science, Research and the Arts Baden-W¨urttemberg (MWK) LIBIS/de.NBI Freiburg.
Collapse
Affiliation(s)
| | - Sean R Wilkinson
- Oak Ridge Leadership Computing Facility, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Finn Bacall
- Department of Computer Science, The University of Manchester, Manchester, UK
| | - Stian Soiland-Reyes
- Department of Computer Science, The University of Manchester, Manchester, UK
- Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Simone Leo
- Center for Advanced Studies, Research, and Development in Sardinia (CRS4), Pula, Cagliari, Italy
| | - Luca Pireddu
- Center for Advanced Studies, Research, and Development in Sardinia (CRS4), Pula, Cagliari, Italy
| | - Stuart Owen
- Department of Computer Science, The University of Manchester, Manchester, UK
| | - Nick Juty
- Department of Computer Science, The University of Manchester, Manchester, UK
| | - José M Fernández
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Spanish National Bioinformatics Institute (INB), Barcelona, Spain
| | - Tom Brown
- Leibniz Institute for Zoo- and Wildlife Research, Berlin, Germany
| | - Hervé Ménager
- Institut Pasteur, Université Paris Cité, Bioinformatics of Biostatistics Hub, Paris, 75015, France
- CNRS, UMS 3601, Institut Français de Bioinformatique, Evry, France
| | - Björn Grüning
- Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Salvador Capella-Gutierrez
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
- Spanish National Bioinformatics Institute (INB), Barcelona, Spain
| | | | - Carole Goble
- Department of Computer Science, The University of Manchester, Manchester, UK.
| |
Collapse
|
120
|
Ruan Z, Tian H, Li R, Zhang J, Cao S, Yang Z, Chen X, Li D, Miao Q. Joint association of frailty index and biological age with chronic obstructive pulmonary disease: a cohort study from CHARLS. Sci Rep 2025; 15:17616. [PMID: 40399410 PMCID: PMC12095576 DOI: 10.1038/s41598-025-99682-6] [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: 02/04/2025] [Accepted: 04/22/2025] [Indexed: 05/23/2025] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is associated with frailty and ageing, but there is insufficient evidence from existing longitudinal studies. This research explored the longitudinal association between frailty, ageing and COPD. We used the China Health and Retirement Longitudinal Study (CHARLS) data to perform a cohort study. The study population was non-COPD patients in wave 1 (2011), and the outcome was the occurrence of COPD at the end of follow-up (wave 4-wave 5). Frailty was assessed using the CHARLS modified frailty index (CMFI), and ageing was evaluated using the biological age (BA). We used multivariate logistic regression to examine the longitudinal associations between CMFI and BA with COPD. Fitted curves were used to analyze the dose-response relationship of CMFI and BA with COPD. A 3D surface diagram was used to analyze the association between BA and CMFI with COPD. In addition, subgroup and sensitivity analyses were performed. 6452 non-COPD patients were enrolled in the study, and after follow-up, 616 participants were diagnosed with COPD. Logistic regression and fitted curves showed a positive correlation between CMFI and BA and the development of COPD. The risk of COPD increased by 19% for every one standard deviation (SD) increase in BA and 32% for every one SD increase in CMFI. A 3D surface diagram shows a joint association between CMFI and BA with the COPD. Subgroup and sensitivity analysis results are stable. This study found a joint association between CMFI and BA with COPD, suggesting that CMFI and BA are risk factors for the development of COPD.
Collapse
Affiliation(s)
- Zhishen Ruan
- Xiyuan Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyan Tian
- Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, Shandong, China
| | - Rui Li
- Xiyuan Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Jinzhi Zhang
- Xiyuan Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Sheng Cao
- Xiyuan Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Zi Yang
- Xiyuan Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyan Chen
- Xiyuan Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Li
- Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Miao
- Xiyuan Hospital of Chinese Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
121
|
Consul S, Robertson J, Vikalo H. XVir: A Transformer-Based Architecture for Identifying Viral Reads from Cancer Samples. J Comput Biol 2025. [PMID: 40392695 DOI: 10.1089/cmb.2025.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
It is estimated that approximately 15% of cancers worldwide can be linked to viral infections. The viruses that can cause or increase the risk of cancer include human papillomavirus, hepatitis B and C viruses, Epstein-Barr virus, and human immunodeficiency virus, to name a few. The computational analysis of the massive amounts of tumor DNA data, whose collection is enabled by the advancements in sequencing technologies, has allowed studies of the potential association between cancers and viral pathogens. However, the high diversity of oncoviral families makes reliable detection of viral DNA difficult, and the training of machine learning models that enable such analysis computationally challenging. We introduce XVir, a data pipeline that deploys a transformer-based deep learning architecture to reliably identify viral DNA present in human tumors. XVir is trained on a mix of sequencing reads coming from viral and human genomes, resulting in a model capable of robust detection of potentially mutated viral DNA across a range of experimental settings. Results on semi-experimental data demonstrate that XVir is able to achieve high classification accuracy, generally outperforming state-of-the-art competing methods. In particular, it retains high accuracy even when faced with diverse viral populations while being significantly faster to train than other large deep learning-based classifiers.
Collapse
Affiliation(s)
- Shorya Consul
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - John Robertson
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Haris Vikalo
- Chandra Family Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
122
|
Rebello RJ, Posner A, Dong R, Prall OWJ, Sivakumaran T, Mitchell CB, Flynn A, Caneborg A, Mitchell C, Kanwal S, Fedele C, Webb S, Fisher K, Wong HL, Balachander S, Zhu W, Nicolson S, Dimitriadis V, Wilcken N, DeFazio A, Gao B, Singh M, Collins IM, Steer C, Warren M, Karanth N, Xu H, Fellowes A, Hicks RJ, Stewart KP, Shale C, Priestley P, Dawson SJ, Vissers JHA, Fox SB, Schofield P, Bowtell D, Hofmann O, Grimmond SM, Mileshkin L, Tothill RW. Whole genome sequencing improves tissue-of-origin diagnosis and treatment options for cancer of unknown primary. Nat Commun 2025; 16:4422. [PMID: 40393956 PMCID: PMC12092688 DOI: 10.1038/s41467-025-59661-x] [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: 08/08/2024] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
Genomics can inform both tissue-of-origin (TOO) and precision treatments for patients with cancer of unknown primary (CUP). Here, we use whole genome and transcriptome sequencing (WGTS) for 72 patients and show diagnostic superiority of WGTS over panel testing (386-523 genes) in 71 paired cases. WGTS detects all reportable DNA features found by panel as well as additional mutations of diagnostic or therapeutic relevance in 76% of cases. Curated WGTS features and a CUP prediction algorithm (CUPPA) trained on WGTS data of known cancer types informs TOO in 71% of cases otherwise undiagnosed by clinicopathology review. WGTS informs treatments for 79% of patients, compared to 59% by panel testing. Finally, WGS of cell-free DNA (cfDNA) from patients with a high cfDNA tumour fraction (>7%), enables high-likelihood CUPPA predictions in 41% of cases. WGTS is therefore superior to panel testing, broadens treatment options, and is feasible using routine pathology samples and cfDNA.
Collapse
Affiliation(s)
- Richard J Rebello
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Atara Posner
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Ruining Dong
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Owen W J Prall
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Tharani Sivakumaran
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Camilla B Mitchell
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Aidan Flynn
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Alex Caneborg
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sehrish Kanwal
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Clare Fedele
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Samantha Webb
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Krista Fisher
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Hui-Li Wong
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Shiva Balachander
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Wenying Zhu
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Shannon Nicolson
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Voula Dimitriadis
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Nicholas Wilcken
- The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Anna DeFazio
- The Westmead Institute for Medical Research, Sydney, NSW, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Bo Gao
- Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
| | - Madhu Singh
- Department of Medical Oncology, Barwon Health Cancer Services, Geelong, VIC, Australia
| | - Ian M Collins
- Department of Medical Oncology, Southwest HealthCare, Warrnambool and Deakin University, Geelong, VIC, Australia
| | - Christopher Steer
- Border Medical Oncology, Albury Wodonga Regional Cancer Centre, Albury NSW, Australia and UNSW School of Clinical Medicine, Rural Clinical Campus, Albury, NSW, Australia
| | - Mark Warren
- Department of Medical Oncology, Bendigo Health, Bendigo, VIC, Australia
| | - Narayan Karanth
- Division of Medicine, Alan Walker Cancer Centre, Darwin, NT, Australia
| | - Huiling Xu
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Andrew Fellowes
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rodney J Hicks
- The St Vincent's Hospital Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Kym Pham Stewart
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | | | | | - Sarah-Jane Dawson
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Joseph H A Vissers
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Penelope Schofield
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Psychology, and Iverson Health Innovation Research Institute, Swinburne University, Melbourne, VIC, Australia
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, VIC, Australia
| | - David Bowtell
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Oliver Hofmann
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Sean M Grimmond
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Linda Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Richard W Tothill
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia.
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
123
|
Vescovo M, Raspollini MR, Nibid L, Castiglione F, Nardi E, de Biase D, Massari F, Giunchi F, Pepe F, Troncone G, Malapelle U, Carosi M, Casini B, Melucci E, Fassan M, Toffolatti L, Guerini-Rocco E, Conversano F, Rappa A, Tommasi S, Coppola CA, Zeppa P, Caputo A, Gaeta S, Pagni F, Seminati D, Vecchione A, Scarpino S, Righi D, Taffon C, Prata F, Perrone G. Storage Time and DNA Quality Determine BRCA1/2 Sequencing Success in Prostate Cancer: A Multicentre Analysis with Therapeutic Implications. Cancers (Basel) 2025; 17:1705. [PMID: 40427202 PMCID: PMC12110138 DOI: 10.3390/cancers17101705] [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: 04/08/2025] [Revised: 05/04/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Approximately 25.0% of metastatic prostate cancer patients harbour DNA damage repair mutations, including BRCA1 and BRCA2, which are actionable targets for poly(ADP-ribose) polymerase (PARP) inhibitors. Accurate detection of BRCA1/2 mutations is critical for guiding targeted therapies, but crucial pre-analytical factors, such as tissue storage duration and DNA fragmentation, drastically affect the reliability of next-generation sequencing (NGS) using real-world diagnostic specimens. METHODS This multicentre study analysed 954 formalin-fixed paraffin-embedded tissue samples from 11 centres, including 559 biopsies and 395 surgical specimens. This study examined the impact of storage duration (<1 year, 1-2 years, and >2 years) and DNA parameters (concentration and fragmentation index) on NGS success rates. Logistic regression and Cox regression analyses were used to assess correlations between these factors and sequencing outcomes. RESULTS NGS success rates decreased significantly with longer storage, from 87.8% (<1 year) to 69.1% (>2 years). Samples with higher DNA concentrations and fragmentation indexes had higher success rates (p < 0.001). Surgical specimens had superior success rates (83.3%) compared with biopsies (72.8%) due to better DNA quality. The DNA degradation rate was more pronounced in older samples, underscoring the negative impact of extended storage. CONCLUSIONS Timely testing of BRCA1/2 mutations is critical for optimizing the identification of prostate cancer patients eligible for PARP inhibitors. Surgical specimens provide more reliable results than biopsies and minimizing the storage duration significantly enhances testing outcomes. Standardizing pre-analytical and laboratory procedures across centres is essential to ensure personalized treatments and improve patient outcomes.
Collapse
Affiliation(s)
- Mariavittoria Vescovo
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
| | - Maria Rosaria Raspollini
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Via Pieraccini, 6, 50129 Florence, Italy; (M.R.R.); (F.C.); (E.N.)
| | - Lorenzo Nibid
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Francesca Castiglione
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Via Pieraccini, 6, 50129 Florence, Italy; (M.R.R.); (F.C.); (E.N.)
| | - Eleonora Nardi
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Via Pieraccini, 6, 50129 Florence, Italy; (M.R.R.); (F.C.); (E.N.)
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (F.P.); (G.T.); (U.M.)
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (F.P.); (G.T.); (U.M.)
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, 80131 Naples, Italy; (F.P.); (G.T.); (U.M.)
| | - Mariantonia Carosi
- Molecular Diagnostic Laboratory, Pathology Department, Advanced Diagnostics Research and Technological Innovation Department, Regina, Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (M.C.); (B.C.); (E.M.)
| | - Beatrice Casini
- Molecular Diagnostic Laboratory, Pathology Department, Advanced Diagnostics Research and Technological Innovation Department, Regina, Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (M.C.); (B.C.); (E.M.)
| | - Elisa Melucci
- Molecular Diagnostic Laboratory, Pathology Department, Advanced Diagnostics Research and Technological Innovation Department, Regina, Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy; (M.C.); (B.C.); (E.M.)
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy;
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy
| | - Luisa Toffolatti
- Surgical Pathology Unit, Ca’ Foncello General Hospital, ULSS2 Marca Trevigiana, 31100 Treviso, Italy;
| | - Elena Guerini-Rocco
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy;
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (F.C.); (A.R.)
| | - Federica Conversano
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (F.C.); (A.R.)
| | - Alessandra Rappa
- Division of Pathology, European Institute of Oncology, IRCCS, 20141 Milan, Italy; (F.C.); (A.R.)
| | - Stefania Tommasi
- Pharmacogenetics and Molecular Diagnostics Unit, IRCCS Istituto Tumori Giovanni Paolo II Bari, 70124 Bari, Italy; (S.T.); (C.A.C.)
| | - Claudio Antonio Coppola
- Pharmacogenetics and Molecular Diagnostics Unit, IRCCS Istituto Tumori Giovanni Paolo II Bari, 70124 Bari, Italy; (S.T.); (C.A.C.)
| | - Pio Zeppa
- Department of Medicine, Surgery, and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84084 Baronissi, Italy; (P.Z.); (A.C.)
| | - Alessandro Caputo
- Department of Medicine, Surgery, and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84084 Baronissi, Italy; (P.Z.); (A.C.)
| | - Sara Gaeta
- Department of Pathology, University Hospital of Salerno, 84081 Salerno, Italy;
| | - Fabio Pagni
- Department of Medicine and Surgery, University Milan Bicocca and Fondazione IRCCS San Gerardo dei Tintori Monza, 20126 Milan, Italy; (F.P.)
| | - Davide Seminati
- Department of Medicine and Surgery, University Milan Bicocca and Fondazione IRCCS San Gerardo dei Tintori Monza, 20126 Milan, Italy; (F.P.)
| | - Andrea Vecchione
- Unit of Pathology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (A.V.); (S.S.)
| | - Stefania Scarpino
- Unit of Pathology, Department of Clinical and Molecular Medicine, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (A.V.); (S.S.)
| | - Daniela Righi
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
| | - Chiara Taffon
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Francesco Prata
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
| | - Giuseppe Perrone
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (L.N.); (D.R.); (C.T.); (G.P.)
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| |
Collapse
|
124
|
Otlu B, Alexandrov LB. Evaluating topography of mutational signatures with SigProfilerTopography. Genome Biol 2025; 26:134. [PMID: 40394581 PMCID: PMC12093824 DOI: 10.1186/s13059-025-03612-8] [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: 01/08/2024] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
The mutations found in a cancer genome are shaped by diverse processes, each displaying a characteristic mutational signature that may be influenced by the genome's architecture. While prior analyses have evaluated the effect of topographical genomic features on mutational signatures, there has been no computational tool that can comprehensively examine this interplay. Here, we present SigProfilerTopography, a Python package that allows evaluating the effect of chromatin organization, histone modifications, transcription factor binding, DNA replication, and DNA transcription on the activities of different mutational processes. SigProfilerTopography elucidates the unique topographical characteristics of mutational signatures, unveiling their underlying biological and molecular mechanisms.
Collapse
Affiliation(s)
- Burçak Otlu
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, 06800, Turkey
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA.
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA.
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA.
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
125
|
Loeb S, Vadaparampil ST, Giri VN. Germline testing for prostate cancer: current state and opportunities for enhanced access. EBioMedicine 2025; 116:105705. [PMID: 40398351 DOI: 10.1016/j.ebiom.2025.105705] [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: 11/10/2024] [Revised: 02/16/2025] [Accepted: 04/02/2025] [Indexed: 05/23/2025] Open
Abstract
Germline Testing (GT) for prostate cancer (PCA) is now central to PCA care and hereditary cancer assessment, with a rising role in PCA screening approaches. Guidelines have significantly expanded to include testing patients with metastatic PCA, advanced PCA or with high-risk features, and for males with or without PCA with a strong family cancer history to identify hereditary cancer syndromes for patients and their families. However, the expansion of GT has overwhelmed genetic counselling programs, necessitating the development and evaluation of alternate genetic delivery models. Furthermore, disparities in engagement in PCA GT are of major concern for impacting PCA-related and overall cancer-related outcomes for patients and their families. This review focuses on integrating PCA GT guidelines with implementation strategies and addressing PCA GT disparities to help inform current and future strategies to enhance the benefits of GT across populations.
Collapse
Affiliation(s)
- Stacy Loeb
- Department of Urology and Population Health, New York University Langone Health, New York, NY, USA
| | - Susan T Vadaparampil
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | - Veda N Giri
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
126
|
Zhang Y, Dali R, Blanchette M. RobusTAD: reference panel based annotation of nested topologically associating domains. Genome Biol 2025; 26:129. [PMID: 40390127 PMCID: PMC12087246 DOI: 10.1186/s13059-025-03568-9] [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: 09/29/2023] [Accepted: 04/04/2025] [Indexed: 05/21/2025] Open
Abstract
Topologically associating domains (TADs) are fundamental units of 3D genomes and play essential roles in gene regulation. Hi-C data suggests a hierarchical organization of TADs. Accurately annotating nested TADs from Hi-C data remains challenging, both in terms of the precise identification of boundaries and the correct inference of hierarchies. While domain boundary is relatively well conserved across cells, few approaches have taken advantage of this fact. Here, we present RobusTAD to annotate TAD hierarchies. It incorporates additional Hi-C data to refine boundaries annotated from the study sample. RobusTAD outperforms existing tools at boundary and domain annotation across several benchmarking tasks.
Collapse
Affiliation(s)
- Yanlin Zhang
- School of Computer Science, Mcgill University, Montréal, Canada
| | - Rola Dali
- School of Computer Science, Mcgill University, Montréal, Canada
| | | |
Collapse
|
127
|
Han J, Meng X, Kong H, Li X, Chen P, Zhang XA. Links between short-chain fatty acids and osteoarthritis from pathology to clinic via gut-joint axis. Stem Cell Res Ther 2025; 16:251. [PMID: 40390010 PMCID: PMC12090658 DOI: 10.1186/s13287-025-04386-3] [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: 02/17/2025] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
Short-chain fatty acids (SCFAs), the primary metabolites produced by the microbial fermentation of dietary fibers in the gut, have a key role in protecting gut health. Increasing evidence indicates SCFAs can exert effects on distant tissues and organs beyond the gut via blood circulation. Osteoarthritis (OA) is a chronic inflammatory joint disease that severely diminishes the physical function and quality of life. However, effective clinical treatments for OA remain elusive. Recent studies have shown that SCFAs can exert beneficial effects on damaged joints in OA. SCFAs can mitigate OA progression by preserving intestinal barrier function and maintaining the integrity of cartilage and subchondral bone, suggesting that they have substantial potential to be the adjunctive treatment strategy for OA. This review described the SCFAs in the human body and their cellular signaling mechanism, and summarized the multiple effects of SCFAs (especially butyrate, propionate, and acetate) on the prevention and treatment of OA by regulating the gut-joint axis, providing novel insights into their promising clinical applications.
Collapse
Affiliation(s)
- Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Xin Meng
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Hui Kong
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Xinran Li
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China
| | - Peijie Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, 110100, China.
| |
Collapse
|
128
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [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: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
129
|
Zhang S, Jiang Y, Wang H, Liu Q, He X, Pan M, Chen Y, Xiong J, Chen Z, Liu P, Huang H, Wang X, Wang L, Tan J, Fang C, Fu Q, Li X, Zhang Q, Lu Y, Li X. Associations between plant-based diets and cardiovascular disease, frailty, and cognitive dysfunction in middle and old age: a systematic review and meta-analysis of cohort studies. Food Funct 2025; 16:4061-4084. [PMID: 40298944 DOI: 10.1039/d4fo03384a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background: Evidence suggests that plant-based diets are associated with multiple health outcomes. However, in previous pooled studies, the lack of a standardized definition of plant-based diet has led to conflicting evidence regarding its relationship with health outcomes in middle-aged and elderly people. Methods and findings: Searches were conducted on PubMed, Embase, Cochrane Library and Web of Science databases from inception until July 10, 2024. We included studies that examined the association between (1) the standardized healthy plant-based diet index (hPDI) and unhealthy plant-based diet index (uPDI) and (2) cardiovascular disease (CVD), cardiovascular mortality (CVD mortality), frailty, and cognitive dysfunction. Dose-response analysis was performed to assess the relationships between (1) hPDI and uPDI and (2) CVD and frailty. Statistical analyses were performed using an inverse variance random-effects model, and results are reported as risk ratio (RR) with 95% confidence interval (CI) by combining the most adjusted RR, odds ratio (OR) and hazard ratio (HR) under the rare outcome assumption. A total of 25 cohort studies were included. Higher hPDI scores were associated with lower risks of CVD (RR = 0.81, 95% CI 0.71-0.93), CVD mortality (RR = 0.83, 95% CI 0.75-0.90), coronary heart disease (CHD) (RR = 0.79, 95% CI 0.70-0.88), stroke (RR = 0.91, 95% CI 0.86-0.96), ischemic stroke (RR = 0.86, 95% CI 0.79-0.94), cognitive dysfunction (RR = 0.75, 95% CI 0.66-0.84) and frailty (RR = 0.72, 95% CI 0.60-0.86). By contrast, higher uPDI scores were associated with increased risks of CVD (RR = 1.16, 95% CI 1.09-1.24), CVD mortality (RR = 1.14, 95% CI 1.05-1.23), frailty (RR = 1.52, 95% CI 1.10-2.08) and cognitive dysfunction (RR = 1.24, 95% CI 1.11-1.38). Dose-response analyses showed that increasing adherence to hPDI was associated with reduced risks of CVD and frailty, whereas increasing intake of hPDI was linked to increased risk of CVD and frailty. Conclusions: Our study suggests that adherence to hPDI is associated with favourable health outcomes in middle-aged and elderly people, except for hemorrhagic stroke. These findings highlight the potential benefits of hPDI for promoting healthy ageing.
Collapse
Affiliation(s)
- Shipeng Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Yanjie Jiang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 157 Daming Road, Nanjing 210022, China.
| | - Hanyu Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Qiqi Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Xingyi He
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Moshen Pan
- Shanghai University of Finance and Economics, Shanghai, China
| | - Yuecan Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiahui Xiong
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Ze Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Pin Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Honglu Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Xiaocui Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Lu Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Junwen Tan
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Caishan Fang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Qinwei Fu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Xinrong Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| | - Qinxiu Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- World Health Organization Collaborating Centre (WHOCC), CHN-56, Chengdu, 610041, China
| | - Yan Lu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 157 Daming Road, Nanjing 210022, China.
| | - Xueying Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, P.R. China.
| |
Collapse
|
130
|
Lee CW, Myung SK. Consumption of fruit juice and risk of type 2 diabetes mellitus: A systematic review and meta-analysis of prospective cohort studies: Fruit Juice and Risk of Type 2 Diabetes. Am J Med 2025:S0002-9343(25)00303-1. [PMID: 40393612 DOI: 10.1016/j.amjmed.2025.05.021] [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: 03/11/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Previous observational studies on the association between the consumption of fruit juice and the risk of type 2 diabetes mellitus have reported inconsistent findings. We investigated the association using a meta-analysis of prospective cohort studies. METHODS Studies were identified through PubMed and EMBASE searches from inception to August 3, 2024. We calculated pooled relative risks (RRs) and 95% confidence intervals (CIs). The consumption of fruit juice was categorized into 100% fruit juice and non-100% fruit juice. The primary outcome was the incidence of type 2 diabetes mellitus. RESULTS Out of 1591 articles, 14 prospective cohort studies were included in the final analysis. In the meta-analysis of all studies, there was no significant association between the consumption of overall fruit juice and the risk of type 2 diabetes mellitus (RR, 1.06 [95% CI, 0.98-1.15], P = 0.170). In the subgroup meta-analysis by juice type, non-100% fruit juice was statistically significantly associated with an increased risk of type 2 diabetes mellitus (RR, 1.15 [95% CI, 1.03-1.28], P = 0.012), while there was no significant association between the consumption of 100% fruit juice and the risk of type 2 diabetes mellitus. An increased risk of type 2 diabetes mellitus by fruit juice was observed only in Asian populations (RR, 1.17 [95% CI 1.02-1.34], P = 0.023). CONCLUSION The consumption of non-100% fruit juice increased the risk of type 2 diabetes mellitus. Unlike whole fruit consumption, 100% fruit juice had no beneficial effect on the risk of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Chung-Woo Lee
- Department of Family Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Seung-Kwon Myung
- Department of Public Health & AI, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Korea; Department of Family Medicine, National Cancer Center, Goyang, Korea.
| |
Collapse
|
131
|
Zeng X, Peng F, Wang Z, Teng Q, Sha Y, Leung RKK, Christopher LAIKC, Li G, Huang X, Lin S. New insights into tumor microenvironment and HPV integrations in cervical cancer pathogenesis revealed by single-cell transcriptome data. Hum Mol Genet 2025; 34:920-933. [PMID: 40151001 DOI: 10.1093/hmg/ddaf027] [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: 05/28/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 03/29/2025] Open
Abstract
HPV infection is common among women and can result in serious illnesses. This research utilizes single-cell RNA-sequencing (scRNA-seq) to study the connection between cellular heterogeneity and HPV integrations in cervical histopathology. scRNA-seq was used to examine heterogeneity among normal patients and those in three disease stages: high-grade squamous intraepithelial lesions (HSIL), microinvasive carcinoma (MIC), and cervical squamous epithelium carcinoma cancer (CSCC) tissues. A method was developed to identify HPV integration events from scRNA-seq data. Our results indicated an increase in squamous epithelial cells and a decrease in columnar epithelial cells as the disease progressed from normal to CSCC. We discovered HPV genes that were differentially expressed across normal patients and those in the three disease stages. Notably, HPV integration events were more common in squamous epithelial cells at the single-cell level. The ratio of HPV-integrated cells increased as the disease progressed from normal tissue to CSCC, eventually stabilizing. Several genes, such as EGR1, S100A11, S100A8, KRT5, RPL34, ATP1B1, RPS4X and EEF2, were frequently integrated by HPV across patients. In contrast, genes like PAN3, BABAM2, SPEN, TCIM-SIRLNT, TEX41-PABPC1P2 and KCNV1-LINC01608 showed frequent integration events across cells. KRT5, ATP1B1, RPS4X, PAN3 and SPEN were novel recurrent HPV-integrated genes we observed at the patient or cell level in this study. Additionally, we found that HPV genes from various HPV types exhibited integration preferences in various samples and disease stages. This provides a valuable insight into the mechanism of HPV-induced cervical cancer from a single-cell standpoint, highlighting its clinical relevance.
Collapse
Affiliation(s)
- Xi Zeng
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, China
| | - Fang Peng
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, China
| | - Ziying Wang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, China
| | - Qiuli Teng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xilu, Jinan, Shandong 250012, PR China
| | - Ying Sha
- Engineering Research Center of Intelligent Technology for Agriculture, Ministry of Education, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, China
| | - Ross Ka-Kit Leung
- S.H. Ho Research Centre for Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
- Hebei Maternity Hospital, #27 Shifeng Road, Qiaoxi Strict, Shijiazhuang, Hebei 050000, China
- Sansure Biotech Inc., No. 680 Lusong Road, Changsha, Hunan 410205, China
| | - L A I Koon Chi Christopher
- S.H. Ho Research Centre for Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
- Department of microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
| | - Guoliang Li
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan 430070, China
| | - Xiaoyuan Huang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 JieFang Avenue, Wuhan 430000, China
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 JieFang Avenue, Wuhan 430000, Hubei, China
| | - Shitong Lin
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 JieFang Avenue, Wuhan 430000, China
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, 430022, China
| |
Collapse
|
132
|
Li X, Liu N. Advances in understanding LINE-1 regulation and function in the human genome. Trends Genet 2025:S0168-9525(25)00103-9. [PMID: 40382218 DOI: 10.1016/j.tig.2025.04.011] [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: 02/21/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
LINE-1 (long interspersed nuclear element 1, L1) retrotransposons constitute ~17% of human DNA (~0.5 million genomic L1 copies) and exhibit context-dependent expression in different cell lines. Recent studies reveal that L1 is under multilayered control by diverse factors that either collaborate or compete with each other to ensure precise L1 activity. Remarkably, L1s have been co-opted as various transcription-dependent regulatory elements, such as promoters, enhancers, and topologically associating domain (TAD) boundaries, that regulate gene expression in zygotic genome activation, aging, cancer, and other disorders. This review highlights the regulation of L1 and its regulatory functions that influence disease and development.
Collapse
Affiliation(s)
- Xiufeng Li
- State Key Laboratory of Green Biomanufacturing, Tsinghua University-Peking University Joint Center for Life Sciences, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nian Liu
- State Key Laboratory of Green Biomanufacturing, Tsinghua University-Peking University Joint Center for Life Sciences, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
133
|
Gill GS, Kharb S, Goyal G, Das P, Kurdia KC, Dhar R, Karmakar S. Immune Checkpoint Inhibitors and Immunosuppressive Tumor Microenvironment: Current Challenges and Strategies to Overcome Resistance. Immunopharmacol Immunotoxicol 2025:1-45. [PMID: 40376861 DOI: 10.1080/08923973.2025.2504906] [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: 02/22/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to improve cancer treatment effectiveness by boosting the immune system of the patient. Nevertheless, the unique and highly suppressive TME poses a significant challenge, causing heterogeneity of response or resistance in a considerable number of patients. This review focuses on the evasive attributes of the TME. Immune evasion mechanism in TME include immunosuppressive cells, cytokine and chemokine signaling, metabolic alterations and overexpression of immune checkpoint molecules such as PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, BTLA and their interactions within the TME. In addition, this review focuses on the overcoming resistance by targeting immunosuppressive cells, normalizing tumor blood vessels, blocking two or three checkpoints simultaneously, combining vaccines, oncolytic viruses and metabolic inhibitors with ICIs or other therapies. This review also focuses on the necessity of finding predictive markers for the stratification of patients and to check response of ICIs treatment. It remains to be made certain by new research and intelligent innovations how these discoveries of the TME and its interplay facilitate ICI treatment and change the face of cancer treatment.
Collapse
Affiliation(s)
- Gurpreet Singh Gill
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Simmi Kharb
- Department of Biochemistry, Pt. B.D. Sharma Postgraduate Institute of Medical Sciences, Rohtak, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kailash Chand Kurdia
- Department of GI Surgery & Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
134
|
Viot J, Loyon R, Adib N, Laurent-Puig P, de Reyniès A, André F, Monnien F, André T, Svrcek M, Turpin A, Selmani Z, Arnould L, Guyard L, Gilbert N, Boureux A, Adotevi O, Vienot A, Abdeljaoued S, Vernerey D, Borg C, Gautheret D. Deciphering human endogenous retrovirus expression in colorectal cancers: exploratory analysis regarding prognostic value in liver metastases. EBioMedicine 2025; 116:105727. [PMID: 40381378 PMCID: PMC12145686 DOI: 10.1016/j.ebiom.2025.105727] [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: 08/09/2024] [Revised: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Human Endogenous RetroVirus (HERV) expression in tumours reflects epigenetic dysregulation of cancer and an oncogenic factor through promoter/enhancer action on genes. While more than 50% of colorectal cancers develop liver metastases, HERV has not been studied in this context. METHODS We collected 400 RNA-seq samples from over 200 patients with primary and liver metastases, including public data and a novel set of 200 samples. FINDINGS We observed global stability of HERV expression between liver metastases and primary colorectal cancers, suggesting an early oncogenic footprint. We identified a list of 17 HERV loci for liver metastatic colorectal cancer (lmCRC) characterization; with tumour-specificity validated in single-cell metastatic colorectal cancer data and normal tissue bulk RNA-seq. Eleven loci produced HERV-derived peptides as per tandem mass spectrometry from primary colorectal cancer. Six loci were associated with the risk of relapse after lmCRC surgery. Four, HERVH_Xp22.32a, HERVH_20p11.23b, HERVH_13q33.3, HERVH_13q31.3, had adverse prognostic value (log-rank p-value 0.028, 0.0083, 9e-4, 0.05, respectively) while two, HERVH_Xp22.2c (log-rank p-value 0.032) and HERVH_8q21.3b (in multivariable models) were associated with better prognosis. Moreover, the markers showed a cumulative effect on survival when expressed. Some were associated with decreased cytotoxic immune cells and most of them correlated with cell cycle pathways. INTERPRETATION These findings provide insights into the lmCRC transcriptome landscape by suggesting prognostic markers and potential therapeutic targets. FUNDING This work was supported by funding from institutional grants from Inserm, EFS, University of Bourgogne Franche-Comté, national found "Agence Nationale de la Recherche - ANR-JCJC: Projet HERIC and ANR-22-CE45-0007", and "La ligue contre le cancer".
Collapse
Affiliation(s)
- Julien Viot
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.
| | - Romain Loyon
- Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Nawfel Adib
- Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Pierre Laurent-Puig
- Department of Biology, Institut du Cancer Paris CARPEM, APHP, APHP.Centre-Université Paris Cité, Hôpital Européen G. Pompidou, Paris, France; Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, EPIGENETEC, Paris 75006, France
| | - Aurélien de Reyniès
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, EPIGENETEC, Paris 75006, France
| | - Fabrice André
- Paris-Saclay University, Gustave Roussy, Villejuif, France; Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Franck Monnien
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Thierry André
- Department of Medical Oncology, Sorbonne University, Saint-Antoine Hospital, AP-HP, Paris, France
| | - Magali Svrcek
- Department of Pathology, Saint-Antoine Hospital, AP-HP, Sorbonne Université, Paris, France
| | - Anthony Turpin
- Department of Oncology, Lille University Hospital, France; CNRS UMR9020, INSERM UMR1277, University of Lille, Institut Pasteur, Lille, France
| | - Zohair Selmani
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Laurent Arnould
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, Dijon, France; CCRB Ferdinand Cabanne de Dijon, France
| | - Laura Guyard
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, Dijon, France; CCRB Ferdinand Cabanne de Dijon, France
| | - Nicolas Gilbert
- IRMB, INSERM U1183, Hopital Saint-Eloi, Universite de Montpellier, Montpellier, France
| | - Anthony Boureux
- IRMB, INSERM U1183, Hopital Saint-Eloi, Universite de Montpellier, Montpellier, France
| | - Olivier Adotevi
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Angélique Vienot
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Syrine Abdeljaoued
- Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Dewi Vernerey
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France
| | - Christophe Borg
- Département d'Oncologie Médicale, CHU Besançon, Besançon 25000, France; Université Marie et Louis Pasteur, INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Daniel Gautheret
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CNRS, CEA, Gif-sur-Yvette 91190, France
| |
Collapse
|
135
|
Tan HL, Zhao Y, Chua DW, Goh BKP, Koh YX. SEER-based evaluation of lymph node yield as a prognostic indicator of cancer-specific survival in nonmetastatic pancreatic ductal adenocarcinoma. Pancreatology 2025:S1424-3903(25)00093-6. [PMID: 40410047 DOI: 10.1016/j.pan.2025.05.008] [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: 03/07/2025] [Revised: 05/10/2025] [Accepted: 05/15/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND/OBJECTIVES Although the American Joint Committee on Cancer (AJCC) 8th edition recommends harvesting at least 12 lymph nodes for optimal staging in pancreatic ductal adenocarcinoma (PDAC), the precise lymph node yield (LNY) needed for accurate prognostication in different treatment settings remains unclear. This study aimed to identify subgroup-specific LNY cutoffs and evaluate their prognostic significance in nonmetastatic PDAC. METHODS We analyzed 5609 patients with nonmetastatic PDAC from the Surveillance, Epidemiology, and End Results (SEER) database undergoing pancreatectomy. Patients were categorized by nodal status (N0 vs. N+) and receipt of neoadjuvant therapy (NAT) or upfront surgery (UPS). We used maximum selected rank statistics and a conditional inference tree approach to determine optimal LNY cutoffs for each subgroup. Kaplan-Meier curves and Cox proportional hazards models were employed to assess cancer-specific survival (CSS) and identify independent prognostic factors. RESULTS Distinct LNY thresholds were identified for N0 (>13) and N+ (>10) cohorts, with the highest cutoffs in N0-NAT subgroups (>27). Across all analyses, patients exceeding these LNY cutoffs demonstrated significantly prolonged CSS. The N0-NAT group with LNY >27 achieved the longest median survival (60 months), whereas N+ patients undergoing UPS with LNY ≤10 had the poorest outcomes (16 months). Multivariate Cox regressions consistently showed that higher LNY was an independent predictor of improved survival. CONCLUSIONS Higher LNY thresholds than the current AJCC standard of 12 appear beneficial for more accurate staging and improved survival in resected PDAC. Tailoring LNY goals based on nodal status and treatment modality may further refine prognostic stratification and guide more effective therapeutic strategies.
Collapse
Affiliation(s)
- Hwee Leong Tan
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Academia, 20 College Road, 169856, Singapore; Duke-National University of Singapore Medical School, Singapore
| | - Yun Zhao
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Academia, 20 College Road, 169856, Singapore
| | - Darren Weiquan Chua
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Academia, 20 College Road, 169856, Singapore; Duke-National University of Singapore Medical School, Singapore; Liver Transplant Service, SingHealth Duke-National University of Singapore Transplant Centre, Singapore
| | - Brian Kim Poh Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Academia, 20 College Road, 169856, Singapore; Duke-National University of Singapore Medical School, Singapore; Liver Transplant Service, SingHealth Duke-National University of Singapore Transplant Centre, Singapore
| | - Ye Xin Koh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Academia, 20 College Road, 169856, Singapore; Duke-National University of Singapore Medical School, Singapore; Liver Transplant Service, SingHealth Duke-National University of Singapore Transplant Centre, Singapore.
| |
Collapse
|
136
|
Pierpoint M, Floyd W, Wisdom AJ, Luo L, Ma Y, Dickson BC, Waitkus MS, Kirsch DG. Loss of function of Atrx recapitulates phenotypes of alternative lengthening of telomeres in a primary mouse model of sarcoma. iScience 2025; 28:112357. [PMID: 40292316 PMCID: PMC12033954 DOI: 10.1016/j.isci.2025.112357] [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: 12/05/2023] [Revised: 07/02/2024] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
The development of a telomere maintenance mechanism is essential for immortalization in human cancer. While most cancers elongate their telomeres by expression of telomerase, 10-15% of human cancers utilize a pathway known as alternative lengthening of telomeres (ALT). ALT is commonly associated with loss-of-function mutations in ATRX. Here, we developed a genetically engineered primary mouse model of sarcoma in CAST/EiJ mice to investigate the extent to which telomerase deficiency and Atrx-inactivation lead to ALT induction. We observed increases in multiple ALT-associated phenotypic indicators in tumors with loss of function mutations of Atrx. Furthermore, we found that loss of Atrx leads to an increase in telomeric instability and telomere sister chromatid exchange. However, Atrx-deficient tumors did not show productive telomere length maintenance in the absence of telomerase. This primary mouse model of sarcoma could facilitate future investigations into the molecular features of ALT in vivo.
Collapse
Affiliation(s)
- Matthew Pierpoint
- Duke Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Warren Floyd
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy J. Wisdom
- Harvard Radiation Oncology Program, Boston, MA 02115, USA
| | - Lixia Luo
- Duke Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yan Ma
- Duke Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Brendan C. Dickson
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Matthew S. Waitkus
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
- The Preston Robert Tisch Brain Tumor Center at Duke, Durham, NC 27710, USA
| | - David G. Kirsch
- Duke Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
- Duke Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5G 2M9, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
137
|
Ardila CM, González-Arroyave D, Ramírez-Arbeláez J. Artificial intelligence as a predictive tool for gastric cancer: Bridging innovation, clinical translation, and ethical considerations. World J Gastrointest Oncol 2025; 17:103275. [DOI: 10.4251/wjgo.v17.i5.103275] [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: 11/13/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 05/15/2025] Open
Abstract
With gastric cancer ranking among the most prevalent and deadly malignancies worldwide, early detection and individualized prognosis remain essential for improving patient outcomes. This letter discusses recent advancements in artificial intelligence (AI)-driven predictive tools for gastric cancer, emphasizing a computed tomography-based radiomic model that achieved a predictive accuracy of area under the curve of 0.893 for treatment response in advanced cases undergoing neoadjuvant immunochemotherapy. AI offers promising avenues for predictive accuracy and personalized treatment planning in gastric oncology. Additionally, this letter highlights the comparison of these AI tools with traditional methodologies, demonstrating their potential to streamline clinical workflows and address existing gaps in risk stratification and early detection. Furthermore, this letter addresses the ethical considerations and the need for robust clinical-AI collaboration to achieve reliable, transparent, and unbiased outcomes. Strengthening cross-disciplinary efforts will be vital for the responsible and effective deployment of AI in this critical area of oncology.
Collapse
Affiliation(s)
- Carlos M Ardila
- Department of Basic Sciences, Biomedical Stomatology Research Group, Faculty of Dentistry, Universidad de Antioquia U de A, Medellín 050010, Antioquia, Colombia
- Department of Periodontics, Saveetha Dental College, and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Saveetha, Saveetha 600077, India
| | | | - Jaime Ramírez-Arbeláez
- Department of Transplantation, Hospital San Vicente Fundación, Rionegro 054047, Antioquia, Colombia
| |
Collapse
|
138
|
Much C, Rajkumar SM, Chen L, Cohen JM, Gade AR, Pitt GS, Long Y. Macromolecular interactions dictate Polycomb-mediated epigenetic repression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.15.654236. [PMID: 40463101 PMCID: PMC12132310 DOI: 10.1101/2025.05.15.654236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The dynamic regulation of epigenetic states relies on complex macromolecular interactions. PRC2, the methyltransferase complex responsible for depositing H3K27me3, interacts with distinct accessory proteins to form the mutually exclusive subcomplexes PHF1-PRC2.1, MTF2-PRC2.1, PHF19-PRC2.1, and PRC2.2. The functions of these subcomplexes are unclear and thought to be highly redundant. Here we show that PRC2 subcomplexes have distinct roles in epigenetic repression of lineage-specific genes and stem cell differentiation. Using a human pluripotent stem cell model, we engineered a comprehensive set of separation-of-function mutants to dissect the roles of individual protein-protein and DNA-protein interactions. Our results show that PRC2.1 and PRC2.2 deposit H3K27me3 locus-specifically, resulting in opposing outcomes in cardiomyocyte differentiation. We find that MTF2 stimulates PRC2.1-mediated repression in stem cells and cardiac differentiation through its interaction with DNA and H3K36me3, while PHF19 antagonizes it. Furthermore, MTF2-PRC2.1 maintains normal cardiomyocyte function. Together, these results reveal the importance and specificity of individual macromolecular interactions in Polycomb-mediated epigenetic repression in human stem cells and differentiation.
Collapse
Affiliation(s)
- Christian Much
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sandy M. Rajkumar
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
- These authors contributed equally
| | - Liming Chen
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
- These authors contributed equally
| | - John M. Cohen
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| | - Aravind R. Gade
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Geoffrey S. Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Yicheng Long
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
139
|
Lin KH, Kao TH, Wang LC, Kuo CT, Chen PCH, Chu YC, Yeh YC. Benchmarking large language models GPT-4o, llama 3.1, and qwen 2.5 for cancer genetic variant classification. NPJ Precis Oncol 2025; 9:141. [PMID: 40369023 PMCID: PMC12078457 DOI: 10.1038/s41698-025-00935-4] [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: 01/18/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
Classifying cancer genetic variants based on clinical actionability is crucial yet challenging in precision oncology. Large language models (LLMs) offer potential solutions, but their performance remains underexplored. This study evaluates GPT-4o, Llama 3.1, and Qwen 2.5 in classifying genetic variants from the OncoKB and CIViC databases, as well as a real-world dataset derived from FoundationOne CDx reports. GPT-4o achieved the highest accuracy (0.7318) in distinguishing clinically relevant variants from variants of unknown clinical significance (VUS), outperforming Qwen 2.5 (0.5731) and Llama 3.1 (0.4976). LLMs demonstrated better concordance with expert annotations for variants with strong clinical evidence but exhibited greater inconsistencies for those with weaker evidence. All three models showed a tendency to assign variants to higher evidence levels, suggesting a propensity for overclassification. Prompt engineering significantly improved accuracy, while retrieval-augmented generation (RAG) further enhanced performance. Stability analysis across 100 iterations revealed greater consistency with the CIViC system than with OncoKB. These findings highlight the promise of LLMs in cancer genetic variant classification while underscoring the need for further optimization to improve accuracy, consistency, and clinical applicability.
Collapse
Affiliation(s)
- Kuan-Hsun Lin
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, ROC
| | - Tzu-Hang Kao
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Lei-Chi Wang
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chen-Tsung Kuo
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, ROC
| | - Paul Chih-Hsueh Chen
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yuan-Chia Chu
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
- Department of Information Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, ROC.
- Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC.
| |
Collapse
|
140
|
Yin H, Zhao Q, Yang L, Yi G, Yao W, Fang L, Bai L. A multi-tissue and -breed catalogue of chromatin conformations and their implications in gene regulation in pigs. BMC Genomics 2025; 26:484. [PMID: 40375066 PMCID: PMC12079826 DOI: 10.1186/s12864-025-11490-4] [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: 04/09/2024] [Accepted: 03/14/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Topologically associating domains (TADs) are functional units that organize chromosomes into 3D structures of interacting chromatin, and play a crucial role in regulating gene expression by constraining enhancer-promoter contacts. Evidence suggests that deletion of TAD boundaries can lead to aberrant expression of neighboring genes. In our study, we analyzed high-throughput chromatin conformation capture (Hi-C) datasets from publicly available sources, integrating 71 datasets across five tissues in six pig breeds. RESULTS Our comprehensive analysis revealed 65,843 TADs in pigs, and we found that TAD boundaries are enriched for expression Quantitative Trait Loci (eQTL), splicing Quantitative Trait Loci (sQTL), Loss-of-Function variants (LoFs), and other regulatory variants. Genes within conserved TADs are associated with fundamental biological functions, while those in dynamic TADs may have tissue-specific roles. Specifically, we observed differential expression of the NCOA2 gene within dynamic TADs. This gene is highly expressed in adipose tissue, where it plays a crucial role in regulating lipid metabolism and maintaining energy homeostasis. Additionally, differential expression of the BMPER gene within dynamic TADs is associated with its role in modulating the activities of bone morphogenetic proteins (BMPs)-critical growth factors involved in bone and cartilage development. CONCLUSION Our investigations have shed light on the pivotal roles of TADs in governing gene expression and even influencing traits. Our study has unveiled a holistic interplay between chromatin interactions and gene regulation across various tissues and pig breeds. Furthermore, we anticipate that incorporating markers, such as structural variants (SVs), and phenotypes will enhance our understanding of their intricate interactions.
Collapse
Affiliation(s)
- Hongwei Yin
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Qianyi Zhao
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Liu Yang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Guoqiang Yi
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Wenye Yao
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, 6708 PB, The Netherlands
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics (QGG), Aarhus University, Aarhus, Denmark.
| | - Lijing Bai
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China.
| |
Collapse
|
141
|
Pea A, Bevere M, Gkountakos A, Pasini D, Fiorini D, Mafficini A, Golovco S, Simbolo M, Pedron S, Sciammarella C, Mattiolo P, Mombello A, Villanova M, Franzina C, Masetto F, Ciulla C, Sperandio N, Fujikura K, Ahadi MS, Samra JS, Johns AL, Verheij J, Stommel MWJ, van Santvoort H, Schubert Santana L, Malleo G, Milella M, Brosens LAA, Wood LD, Chang DK, De Robertis R, D'Onofrio M, Gill AJ, Salvia R, Corbo V, Lawlor RT, Scarpa A, Luchini C. Mucinous cystic neoplasms and simple mucinous cysts are two distinct precursors of pancreatic cancer: clinicopathological, genomic, and transcriptomic characterization. J Pathol 2025. [PMID: 40371932 DOI: 10.1002/path.6437] [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: 01/09/2025] [Revised: 03/15/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Mucinous cystic neoplasms (MCNs) of the pancreas are macroscopic precursors of pancreatic cancer. A similar cystic lesion but lacking the ovarian-type subepithelial stroma has been recently defined as a simple mucinous cyst (SMC); however, its nature remains unclear. This study aims to define the clinicopathological and molecular profiles of a cohort of MCNs and SMCs of the pancreas and their associated invasive carcinoma. Overall, 23 cases were identified, comprising 19 MCNs and 4 SMCs with co-occurring invasive carcinoma. A multiregional (two samples from each cystic lesion and one from the adenocarcinoma) DNA and RNA sequencing approach was used. The key findings can be summarized as follows: (1) Molecular association: In 22/23 cases (95.7%), the concomitant mucinous cyst and invasive carcinoma shared specific genomic alterations, establishing for the first time that SMC is a true precursor of pancreatic cancer. (2) Clinical behavior: carcinomas arising from SMC appeared to be more aggressive than those arising from MCN. (3) Mutational profile: both cyst types showed significant similarities to conventional pancreatic ductal adenocarcinoma (PDAC), with KRAS and TP53 the most commonly altered genes. (4) Intracystic heterogeneity: while most molecular alterations were present in both analyzed cystic areas, RNF43 showed the highest heterogeneity. (5) CDKN2A: its alterations were predominantly restricted to the invasive component, suggesting a role in driving the invasion in a subset of cases. CNKN2A may also serve as a potential biomarker for identifying high-risk cysts. (6) RNAseq: most cases showed a switch from the classical to the basal transcriptome subtype during the progression from cystic neoplasms to invasive cancers. These findings establish SMCs as new precursors of pancreatic cancer and provide critical insights into the tumorigenesis of MCNs, with potential immediate implications for tumor taxonomy and clinical management. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Antonio Pea
- Department of General and Pancreatic Surgery-The Pancreas Institute, Verona University Hospital Trust, Verona, Italy
| | - Michele Bevere
- ARC-Net Research Center, University of Verona, Verona, Italy
| | | | - Davide Pasini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Denise Fiorini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Andrea Mafficini
- ARC-Net Research Center, University of Verona, Verona, Italy
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Stela Golovco
- ARC-Net Research Center, University of Verona, Verona, Italy
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Serena Pedron
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Concetta Sciammarella
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Aldo Mombello
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Manuela Villanova
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Carlotta Franzina
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | | | - Calogero Ciulla
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | | | - Kohei Fujikura
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Masha S Ahadi
- Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, and Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Jaswinder S Samra
- Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Amber L Johns
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hjalmar van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht and St Antonius Hospital, Utrecht, Netherlands
| | - Leonor Schubert Santana
- Wolfson Wohl Cancer Research Centre, Research Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Giuseppe Malleo
- Department of General and Pancreatic Surgery-The Pancreas Institute, Verona University Hospital Trust, Verona, Italy
| | - Michele Milella
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Lodewijk A A Brosens
- Department of Pathology, UMC Utrecht, Utrecht University, Utrecht, and Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Research Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Riccardo De Robertis
- Department of Diagnostics and Public Health, Section of Radiology, University of Verona, Verona, Italy
| | - Mirko D'Onofrio
- Department of Diagnostics and Public Health, Section of Radiology, University of Verona, Verona, Italy
| | - Anthony J Gill
- Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, and Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Roberto Salvia
- Department of General and Pancreatic Surgery-The Pancreas Institute, Verona University Hospital Trust, Verona, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Rita T Lawlor
- ARC-Net Research Center, University of Verona, Verona, Italy
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-Net Research Center, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Claudio Luchini
- ARC-Net Research Center, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| |
Collapse
|
142
|
Selvi S, Real CM, Gentiluomo M, Balounova K, Vokacova K, Cumova A, Mohlenikova-Duchonova B, Rizzato C, Halasova E, Vodickova L, Smolkova B, Hemminki K, Campa D, Vodicka P. Genomic instability, DNA damage response and telomere homeostasis in pancreatic cancer. Semin Cancer Biol 2025; 113:59-73. [PMID: 40378535 DOI: 10.1016/j.semcancer.2025.05.005] [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: 11/05/2024] [Revised: 04/16/2025] [Accepted: 05/04/2025] [Indexed: 05/19/2025]
Abstract
Pancreatic cancer (PC) is becoming one of the most serious health problems at present, but its causes and risk factors are still unclear. One of the drivers in pancreatic carcinogenesis is altered genomic (DNA) integrity with subsequent genomic instability in cancer cells. The latter comprises a) DNA damage response and DNA repair mechanisms, b) DNA replication and mitosis, c) epigenetic regulation, and d) telomere maintenance. In our review we addressed the above aspects in relation to the most abundant and severe form of PC, pancreatic ductal adenocarcinoma (PDAC). In summary, the interactions between the DNA damage response, telomere homeostasis and mitotic regulation are not comprehensively understood at present, including the epigenetic factors entering the trait of genomic stability maintenance. In addition, the complexity of telomere homeostasis in relation to PDAC risk, prognosis and prediction also warrants further investigations.
Collapse
Affiliation(s)
- Saba Selvi
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic; Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague 12800, Czech Republic
| | - Carmen Macías Real
- Cancer Predisposition and Biomarkers Group, Instituto de Investigacion Sanitaria de Santiago, Santiago de Compostela, Spain
| | | | - Katerina Balounova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic; Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Klara Vokacova
- Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Andrea Cumova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 84505, Slovakia
| | | | - Cosmeri Rizzato
- Department of Biology, University of Pisa, Pisa 56123, Italy
| | - Erika Halasova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia
| | - Ludmila Vodickova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, Prague 12800, Czech Republic
| | - Bozena Smolkova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava 84505, Slovakia
| | - Kari Hemminki
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, FRG 69120, Germany
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa 56123, Italy
| | - Pavel Vodicka
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4, Martin 03601, Slovakia; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, Pilsen 32300, Czech Republic; Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, Prague 12800, Czech Republic.
| |
Collapse
|
143
|
Zheng Y, Chen Y, Meng X, Zhang L, Ma Y, Zhou R, Fu S, Chen H, Xuanyuan X, Jiang R, Hou P, Song X, Wang Y, Sun J, Zhang W, Li J, Liu Z, Zhang Z, Zeng H, He Y. FADD Functions as an Oncogene in Chr11q13.3-Amplified Head and Neck Squamous Cell Carcinoma. Cancer Res 2025; 85:1909-1927. [PMID: 40370064 DOI: 10.1158/0008-5472.can-24-2562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/02/2024] [Accepted: 02/21/2025] [Indexed: 05/16/2025]
Abstract
Chromosomal 11q13.3 amplification is the most common gene copy-number variation event in head and neck squamous cell carcinoma (HNSCC) that corresponds with poor prognosis. Although cyclin D1, a G1/S phase cell-cycle regulatory protein at this locus, is considered as a key driver of malignant progression, further exploration is needed to develop more effective targets for cases with this amplification. Using CRISPR-based gene knockout screening of genes located in chr11q13.3, we found that loss of the gene encoding the Fas-associated death domain (FADD) protein, a well-recognized adapter to caspase-8 that induces cell apoptosis, significantly reduced cancer cell proliferation. FADD expression was elevated in chr11q13.3-amplified tumors and correlated with poor prognosis. RNA sequencing, mass spectrometry, and proteomics analyses revealed a direct relationship between FADD and the DNA helicase MCM5 in the S phase. FADD and cyclin D1 acted at different stages of the cell cycle to synergistically induce proliferation, and caspase-8 deficiency was required for the oncogenic activity of FADD. In a patient-derived xenograft model with chr11q13.3 amplification, combined administration of the DNA helicase complex inhibitor and CDK4/6 inhibitor effectively curtailed tumor growth. Overall, this study identified a nonclassic oncogenic role for FADD in mediating tumor progression in HNSCC and provided a feasible treatment option for patients with chr11q13.3 amplification. Significance: FADD promotes progression of tumors with chr11q13.3 amplification by binding to the DNA helicase complex, which can be targeted in combination with cyclin D1 as a viable therapeutic strategy for HNSCC patients.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yinan Chen
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Meng
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Li Zhang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanni Ma
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Zhou
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shuiting Fu
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Heng Chen
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xinyang Xuanyuan
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Jiang
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengcong Hou
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomeng Song
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yanqiu Wang
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, Nanjing, China
| | - Jingjing Sun
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wuchang Zhang
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiang Li
- Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhonglong Liu
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Hanlin Zeng
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue He
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
144
|
Pham TH, Do TTH, Nguyen TN, Luu LC, Tran HT, Nguyen TX, Nguyen TTH, Vu HTT. Characteristics of sarcopenia in patients with rheumatoid arthritis at a central university hospital in Hanoi, Vietnam: a cross-sectional study. Clin Rheumatol 2025:10.1007/s10067-025-07469-x. [PMID: 40369253 DOI: 10.1007/s10067-025-07469-x] [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: 01/08/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
AIM To determine the prevalence and factors associated with sarcopenia in rheumatoid arthritis (RA) patients at a central university hospital in Hanoi, Vietnam. METHODS Patients with RA aged ≥ 18 were enrolled in this cross-sectional study. Dual-energy X-ray absorptiometry was performed to measure appendicular skeletal muscle (ASM). Assessment of muscle function included hand grip strength (HGS) and gait speed (GS). Sarcopenia was defined according to Asian Working Group for Sarcopenia (AWGS) criteria 2019. RA disease activity was evaluated by disease activity score 28 CRP (DAS28-CRP). Medical history and previous medications including steroids, methotrexate, bDMARDs, clinical characteristics, and comorbidities were also documented. Multivariable adjusted regression was used to examine potential factors associated with sarcopenia in patients with RA. RESULTS A total of 156 patients with RA were recruited, out of which the prevalence of sarcopenia was 62.82%. Among the participants, 95.16% had a low appendicular lean mass index (ALMI), 91.67% had low hand grip strength, and 36.54% had slow gait speed. Active RA disease was significantly associated with a higher odd ratio of having sarcopenia, low muscle mass, and low hand grip strength. After adjusting for potential factors, male (aOR 6.66), RA disease activity level (aOR 3.07), and hypertension (aOR 4.06) were statistically independent factors associated with sarcopenia. CONCLUSION Effective RA management to achieve clinical remission is essential to mitigate sarcopenia risk. Further studies are essential to better understand sarcopenia and improve management in patients with RA. Key Points • Prevalence: Sarcopenia was present in 62.82% of patients with RA, especially in those with higher disease activity. • Risk Factors: Male gender, active disease, and hypertension were associated with sarcopenia. • Inflammation and disease activity: Active RA and elevated CRP levels were linked to reduced muscle mass and strength.
Collapse
Affiliation(s)
- Thu Hoai Pham
- Hanoi Medical University, Hanoi, 100000, Vietnam.
- Hanoi Medical University Hospital, Hanoi Medical University, 1 Ton That Tung, Dong Da, Hanoi, 100000, Vietnam.
| | | | - Tam Ngoc Nguyen
- Hanoi Medical University, Hanoi, 100000, Vietnam
- National Geriatric Hospital, Hanoi, 100000, Vietnam
| | - Linh Canh Luu
- Hanoi Medical University Hospital, Hanoi Medical University, 1 Ton That Tung, Dong Da, Hanoi, 100000, Vietnam
| | | | - Thanh Xuan Nguyen
- Hanoi Medical University, Hanoi, 100000, Vietnam
- National Geriatric Hospital, Hanoi, 100000, Vietnam
| | - Thu Thi Hoai Nguyen
- Hanoi Medical University, Hanoi, 100000, Vietnam
- National Geriatric Hospital, Hanoi, 100000, Vietnam
| | - Huyen Thi Thanh Vu
- Hanoi Medical University, Hanoi, 100000, Vietnam
- Hanoi Medical University Hospital, Hanoi Medical University, 1 Ton That Tung, Dong Da, Hanoi, 100000, Vietnam
- National Geriatric Hospital, Hanoi, 100000, Vietnam
| |
Collapse
|
145
|
Mohsen H, Blenman K, Emani PS, Morris Q, Carrot-Zhang J, Pusztai L. Dynamic clustering of genomics cohorts beyond race, ethnicity-and ancestry. BMC Med Genomics 2025; 18:87. [PMID: 40375077 PMCID: PMC12082885 DOI: 10.1186/s12920-025-02154-z] [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: 01/10/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Recent decades have witnessed a steady decrease in the use of race categories in genomic studies. While studies that still include race categories vary in goal and type, these categories already build on a history during which racial color lines have been enforced and adjusted in the service of social and political systems of power and disenfranchisement. For early modern classification systems, data collection was also considerably arbitrary and limited. Fixed, discrete classifications have limited the study of human genomic variation and disrupted widely spread genetic and phenotypic continuums across geographic scales. Relatedly, the use of broad and predefined classification schemes-e.g. continent-based-across traits can risk missing important trait-specific genomic signals. METHODS To address these issues, we introduce a dynamic approach to clustering human genomics cohorts based on genomic variation in trait-specific loci and without using a set of predefined categories. We tested the approach on whole-exome sequencing datasets in ten cancer types and partitioned them based on germline variants in cancer-relevant genes that could confer cancer type-specific disease predisposition. RESULTS Results demonstrate clustering patterns that transcend discrete continent-based categories across cancer types. Functional analysis based on cancer type-specific clusterings also captures the fundamental biological processes underlying cancer, differentiates between dynamic clusters on a functional level, and identifies novel potential drivers overlooked by a predefined continent-based clustering. CONCLUSIONS Through a trait-based lens, the dynamic clustering approach reveals genomic patterns that transcend predefined classification categories. We propose that coupled with diverse data collection, new clustering approaches have the potential to draw a more complete portrait of genomic variation and to address, in parallel, technical and social aspects of its study.
Collapse
Affiliation(s)
- Hussein Mohsen
- Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada.
| | - Kim Blenman
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, 06511, USA
- Computer Science, Yale University, New Haven, CT, 06511, USA
| | - Prashant S Emani
- Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
| | - Quaid Morris
- Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Computational Biology and Medicine, Weill-Cornell Medical College, New York, NY, 10065, USA
| | - Jian Carrot-Zhang
- Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lajos Pusztai
- Breast Medical Oncology, School of Medicine, Yale University, New Haven, CT, 06511, USA
| |
Collapse
|
146
|
Sun Z, Li Z, Wei Y, Xu L, Hang X, Kang Y. SMARCA4 Inhibits Breast Cancer Progression and Metastasis through RHOA Suppression. Cancer Res 2025; 85:1803-1818. [PMID: 39992701 PMCID: PMC12081196 DOI: 10.1158/0008-5472.can-24-2801] [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: 08/05/2024] [Revised: 01/07/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most challenging subtype of the disease due to its aggressive nature and lack of targeted therapy options. To identify regulators of TNBC, we conducted a genome-wide CRISPR knockout screen in both three-dimensional (3D) tumor spheroid and two-dimensional cell culture models. The 3D spheroid model displayed unique potential in identifying putative tumor suppressors because of its closer mimicry of in vivo tumor growth conditions. Notably, the chromatin remodeling SWI/SNF complex emerged as a potent suppressor of tumor spheroid growth. Specifically, loss of the SWI/SNF ATPase subunit SMARCA4 promoted tumor spheroid growth with reduced compactness and enhanced primary tumor growth and metastasis across multiple TNBC models. SMARCA4 was required for the transcription of the Rho GTPase-activating factor ARHGAP29 by enhancing DNA accessibility through direct binding to its promoter. SMARCA4 loss resulted in reduced ARHGAP29 levels and hyperactive RHOA signaling, subsequently disrupting cell adhesion, facilitating the formation of a loose spheroid structure in vitro, and enhancing breast cancer growth and metastasis in vivo. These results establish SMARCA4 and SWI/SNF as tumor suppressors of TNBC through suppression of RHOA activity. Significance: CRISPR-knockout screen in 3D tumor spheroid revealed that SMARCA4, a SWI/SNF ATPase subunit, suppresses triple-negative breast cancer growth and metastasis by increasing ARHGAP29 transcription and inhibiting the RHOA signaling pathway.
Collapse
Affiliation(s)
- Zheng Sun
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Zhuo Li
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
- Present address: BeiGene Global Research, Shanghai, P.R. China
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ 08544
| | - Lillian Xu
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
- Present address: Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Xiang Hang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ 08544
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ 08544
| |
Collapse
|
147
|
Niu W, Zhang Y, Wu J. Letter to the editor on "Prognostic relevance of lymph node metastasis in pancreaticoduodenectomy for distal cholangiocarcinoma: Rational extent and number-based nodal classification for regional lymphadenectomy". Surgery 2025:109341. [PMID: 40374404 DOI: 10.1016/j.surg.2025.109341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 05/17/2025]
Affiliation(s)
- Weiqiao Niu
- Department of Hepatobiliary Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Suzhou, Jiangsu, China
| | - Yan Zhang
- Department of Hepatobiliary Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Suzhou, Jiangsu, China
| | - Jianwu Wu
- Department of Hepatobiliary Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Suzhou, Jiangsu, China.
| |
Collapse
|
148
|
Bart NK, Bianchi G, Cuddy SAM, Goyal P, Griffin JM, Hummel SL, Macdonald P, Maurer M, Montgomery E, Nanne MG, Orkaby AR, Sanchorawala V, Damluji AA, ACC Geriatric Cardiology Leadership Council. Cardiac Amyloidosis in Older Adults With a Focus on Frailty: JACC: Advances Expert Consensus. JACC. ADVANCES 2025; 4:101784. [PMID: 40373524 PMCID: PMC12144467 DOI: 10.1016/j.jacadv.2025.101784] [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: 08/15/2024] [Revised: 01/02/2025] [Accepted: 01/30/2025] [Indexed: 05/17/2025]
Abstract
Amyloidosis, which is caused by misfolded proteins that form amyloid fibrils, is predominantly diagnosed in older adults. Although previously considered a rare disease, increased awareness and noninvasive diagnostic methods have resulted in a rise in diagnoses. As a multisystem disease that affects multiple organ systems (cardiac, gastrointestinal, renal, and neurological), there is significant overlap with both geriatric conditions and common conditions in heart failure. Frailty is recognized as a distinct biological syndrome of declines across multiple physiological systems, which prevents maintenance of homeostasis and limits the ability to respond to stressors. Frailty was initially characterized as physical frailty alone; however, it is increasingly recognized that it is multidimensional with components including nutrition, cognitive, psychological, and social. Frailty in cardiovascular disease has become an important risk factor, indicator for disease severity, and can help guide decisions around intervention. In certain patients, frailty may be reversible. Given the lack of consensus definitions, tools, and implementation of frailty in both clinical and research settings in the field of amyloidosis, we convened a group of experts from cardiology, geriatric cardiology, geriatrics, hematology, and allied health to form this state-of-the-art review. There are many points of intersectionality between amyloidosis, aging, and frailty which herald a need for multidisciplinary care. This review document aims to provide guidance in how to understand and address frailty in older patients with a specific focus on cardiac amyloidosis.
Collapse
Affiliation(s)
- Nicole K Bart
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Cardiac Amyloidosis Program, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiology, St Vincent's Hospital, Sydney, NSW, Australia; The Victor Chang Cardiac Research Institute, Sydney, NSW, Australia; University of New South Wales Sydney, Sydney, NSW, Australia
| | - Giada Bianchi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Cardiac Amyloidosis Program, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, Massachusetts, USA; Division of Hematology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sarah A M Cuddy
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Cardiac Amyloidosis Program, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Parag Goyal
- Program for the Care and Study of the Aging Heart, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Jan M Griffin
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Scott L Hummel
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA; Division of Cardiovascular Medicine, Department of Internal Medicine, VA Ann Arbor Health System, Ann Arbor, Michigan, USA
| | - Peter Macdonald
- Department of Cardiology, St Vincent's Hospital, Sydney, NSW, Australia; The Victor Chang Cardiac Research Institute, Sydney, NSW, Australia; University of New South Wales Sydney, Sydney, NSW, Australia
| | - Mathew Maurer
- Cardiac Amyloidosis Program, Department of Cardiology, Columbia University Irving Medical, New York City, New York, USA
| | - Elyn Montgomery
- Department of Cardiology, St Vincent's Hospital, Sydney, NSW, Australia; The Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Michael G Nanne
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ariela R Orkaby
- New England Geriatric Research, Education, and Clinical Center (GRECC), VA Boston Healthcare System, Boston, Massachusetts, USA; Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, Massachusetts, USA; Division of Aging, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vaishali Sanchorawala
- Amyloidosis Center, Boston University Chobanian and Avedisian School of Medicine, Boston Medical Center, Boston, Massachusetts, USA
| | - Abdulla A Damluji
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Cardiovascular Center on Aging, Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA.
| | | |
Collapse
|
149
|
Wang J, Wang X, Chu Y, Li S, Hao J. Predictive factors for efficacy of oxaliplatin-based chemotherapy in advanced well-differentiated neuroendocrine tumors: an observational cohort study and meta-analysis. Front Endocrinol (Lausanne) 2025; 16:1595151. [PMID: 40438391 PMCID: PMC12116336 DOI: 10.3389/fendo.2025.1595151] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/21/2025] [Indexed: 06/01/2025] Open
Abstract
Background Oxaliplatin-based chemotherapy (OX-CT) has shown promising antitumor activity in advanced well-differentiated neuroendocrine tumors (WD-NETs). However, no meta-analysis has been conducted to explore the factors associated with ORR and PFS of OX-CT, and data are still limited in Chinese cohort. Methods We performed a retrospective cohort study with advanced WD-NETs who received OX-CT. We also conducted a systematic review and performed a meta-analysis to explore factors associated with ORR and PFS. Results A total of 27 patients were included, with 21 receiving OX-CT as first line. Furthermore, 18 were of pancreas origin, and the median Ki-67 was 30%. The ORR and DCR were 29.6% and 81.5%, respectively. The median PFS was 9.3 months (95%CI: 4.6-14.0), and OS was not reached. A Ki-67 value >10% predicted higher ORR (36.4% vs. 0.0%, p = 0.28) and better PFS (10.0 vs. 2.1 months, p = 0.06). Patients with hepatic tumor burden ≤25% had a similar ORR (33.3% vs. 22.2%, p = 0.68), but with a trend of longer PFS (10.2 vs. 4.7 months, p = 0.21) than those >25%. Both ORR and PFS were independent of MGMT status. A total of 962 patients were included in the systemic review. The pooled ORR (28.2%, p = 0.84) and DCR (82.9%, p = 0.85) were comparable with this cohort. No difference was observed between GEMOX and FOLFOX/CAPOX in both ORR (23.9% vs. 29.6%, p = 0.19) and PFS (10.5 vs. 11.8 months, p = 0.69). Enhanced ORR was seen in pNETs than epNETs (36.8% vs. 16.7%, p < 0.001) and also in G3 NETs than G1-2 NETs (45.5% vs. 24.7%, p < 0.001). The pooled median PFS and OS were 10.8 months (95%CI: 8.8-12.8) and 30.4 months (95%CI: 24.8-35.9). Conclusions Oxaliplatin-based chemotherapy could be a good option for advanced WD-NETs with high Ki-67 index and pancreatic origin.
Collapse
Affiliation(s)
| | | | | | | | - Jing Hao
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
150
|
Lakbir S, de Wit R, de Bruijn I, Kundra R, Madupuri R, Gao J, Schultz N, Meijer GA, Heringa J, Fijneman RJA, Abeln S. Tumor break load quantitates structural variant-associated genomic instability with biological and clinical relevance across cancers. NPJ Precis Oncol 2025; 9:140. [PMID: 40369102 PMCID: PMC12078582 DOI: 10.1038/s41698-025-00922-9] [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: 12/04/2024] [Accepted: 04/24/2025] [Indexed: 05/16/2025] Open
Abstract
While structural variants (SVs) are a clear sign of genomic instability, they have not been systematically quantified per patient since declining costs have only recently enabled large-scale profiling. Therefore, the biological and clinical impact of high numbers of SVs in patients is unknown. We introduce tumor break load (TBL), defined as the sum of unbalanced SVs, as a measure for SV-associated genomic instability. Using pan-cancer data from TCGA, PCAWG, and CCLE, we show that a high TBL is associated with significant changes in gene expression in 26/31 cancer types that consistently involve upregulation of DNA damage repair and downregulation of immune response pathways. Patients with a high TBL show a higher risk of recurrence and shorter median survival times for 5/15 cancer types. Our data demonstrate that TBL is a biologically and clinically relevant feature of genomic instability that may aid patient prognostication and treatment stratification. For the datasets analyzed in this study, TBL has been made available in cBioPortal.
Collapse
Affiliation(s)
- Soufyan Lakbir
- Bioinformatics Section, Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Translational Gastrointestinal Oncology Group, Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- AI Technology for Life Group, Department of Information and Computing Science; Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Renske de Wit
- Translational Gastrointestinal Oncology Group, Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- AI Technology for Life Group, Department of Information and Computing Science; Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Ino de Bruijn
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ritika Kundra
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Jianjiong Gao
- Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | | | - Gerrit A Meijer
- Translational Gastrointestinal Oncology Group, Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jaap Heringa
- Bioinformatics Section, Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Remond J A Fijneman
- Translational Gastrointestinal Oncology Group, Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Sanne Abeln
- Bioinformatics Section, Department of Computer Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- AI Technology for Life Group, Department of Information and Computing Science; Department of Biology, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|