{"title":"Clonal evolution and therapy resistance in the era of precision cancer medicine: evolutionary trajectories in pediatric cancer","authors":"Jenny Karlsson , Mayen Briggs , Aditi Vedi , David Gisselsson","doi":"10.1016/j.semcancer.2025.10.001","DOIUrl":"10.1016/j.semcancer.2025.10.001","url":null,"abstract":"<div><div>Pediatric cancers exhibit complex evolutionary dynamics driven by genetic instability, clonal selection, epigenetic reprogramming, and microenvironmental interactions. Tumor evolution is typically characterized by branching phylogenies, where subclones with unique genetic profiles emerge at different locations and time points. Partly as a result of this diversity, advanced cancers often develop resistance to multiple therapies, complicating treatment strategies. This review explores recurrent themes in clonal evolution across childhood cancers as well as evolution-inspired strategies to counter treatment resistance. A model of dynamic decision making is suggested to accommodate emerging methods for monitoring shifts in a cancer’s clonal landscape and the widening repertoire of precision drugs. A larger drug toolbox will allow for more sophisticated evolutionary stratagems to cure or stabilize cancer, using methods such as adaptive therapy, extinction therapy, and reflexive control therapies. However, there are also inherent limits in predicting and controlling cancer evolution- emphasizing the need for early detection, particularly in the setting of predicting relapse, which disproportionately contributes to cancer related mortality in childhood. Understanding the evolutionary trajectories of pediatric cancers can inform more effective, personalized treatment protocols, ultimately enhancing survival rates and quality of life for young patients.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 121-134"},"PeriodicalIF":15.7,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145259036","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The complex interplay of lipoprotein(a) and cancer in the era of emerging therapeutics","authors":"Vanessa Bianconi , Massimo R. Mannarino , Filippo Figorilli , Matteo Pirro , Amirhossein Sahebkar","doi":"10.1016/j.semcancer.2025.09.008","DOIUrl":"10.1016/j.semcancer.2025.09.008","url":null,"abstract":"<div><div>In the last decades, the biological properties of Lp(a) have attracted increasing attention for their possible involvement in a wide range of clinical conditions other than atherosclerotic cardiovascular disease. To date, whether a pathogenic interplay may exist between Lp(a) and cancer remains unclear. Indeed, experimental studies mainly show a protective effect of Lp(a) toward cancer, while results of clinical studies are highly contradictory. Nonetheless, the confirmation of any link between Lp(a) metabolism and cancer may be highly impactful for its translational implications in the current era of a renewed scientific interest in this lipoprotein. Indeed, the increasing availability of laboratory assays for the routine assessment of plasma Lp(a) levels could be proposed as an additional tool for cancer diagnosis and prognostic stratification. In addition, the tumultuous development of anti-Lp(a) therapeutics, if a pro-cancerogenic Lp(a) activity will be confirmed, could have an impact on the natural history of cancer and on its pharmacological management. This review resumes current knowledge on the relationship between Lp(a) and cancer as well as on its possible impact on the oncological field.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 96-107"},"PeriodicalIF":15.7,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145186713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
M C Martinez-Campanario, Marlies Cortés, Lu Han, Agnese Brischetto, Paloma Rivero-Soriano, Joan Font-Diaz, Antonio Postigo
{"title":"Lipid-laden macrophages in atherosclerosis and cancer.","authors":"M C Martinez-Campanario, Marlies Cortés, Lu Han, Agnese Brischetto, Paloma Rivero-Soriano, Joan Font-Diaz, Antonio Postigo","doi":"10.1016/j.semcancer.2025.09.007","DOIUrl":"https://doi.org/10.1016/j.semcancer.2025.09.007","url":null,"abstract":"<p><p>This review explores the dual roles of lipid-laden macrophages (LL-Macs) in inflammation, atherosclerosis and cancer, emphasizing both their shared and divergent functions across physiological and pathological conditions. Lipid metabolism regulates the polarization of macrophages in homeostasis and inflammation. In atherosclerosis, LL-Macs contribute to plaque formation and inflammation, while in cancer, LL-Macs play crucial roles in immune suppression and tumor progression. The article outlines the molecular mechanisms driving macrophage lipid accumulation in each of these scenarios and how the process is influenced by the distinct local microenvironments in inflammation, atherosclerosis and cancer. Single-cell RNA sequencing (scRNA-seq) studies have identified both common and unique gene signatures between LL-Macs in atherosclerosis and cancer, reflecting the varying microenvironmental cues that shape macrophage function and disease outcomes. Finally, we examine lipid-modulating strategies in atherosclerosis, assess their potential in cancer treatment, and highlight research gaps for developing new LL-TAM-targeted therapies.</p>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":" ","pages":""},"PeriodicalIF":15.7,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145177796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiaqi Li, Lin Chen, Xiaolin Liu, Mei Zhang, Xiaoling Liu
{"title":"Molecular mechanisms and therapies of immune checkpoint inhibitor associated myocarditis: Update on recent developments","authors":"Jiaqi Li, Lin Chen, Xiaolin Liu, Mei Zhang, Xiaoling Liu","doi":"10.1016/j.semcancer.2025.09.006","DOIUrl":"10.1016/j.semcancer.2025.09.006","url":null,"abstract":"<div><div>Immune checkpoint inhibitors (ICIs) enhance systemic antitumor immune responses by improving the ability of immune cells to recognize and kill tumor cells specifically. Activated T lymphocytes may target tissues and organs outside the tumor, leading to different degrees of immune-related adverse events (irAEs). Immune checkpoint inhibitor-associated myocarditis (ICIAM) is a rare irAE, with an incidence of approximately 0.04–1.7 %. However, it is the most fatal irAE and often becomes a critical cause of short-term mortality in patients. Currently, the molecular mechanisms of ICIAM involve immune damage mediated by T-cell and macrophage infiltration, the common antigen theory, and pro-inflammatory responses driven by inflammatory factors and signaling pathways. However, the specific mechanisms remain unclear. A comprehensive understanding of the pathophysiological mechanisms of ICIAM can aid in identifying biomarkers and treatment targets for irAEs, ultimately improving the survival status of patients with cancer.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 108-120"},"PeriodicalIF":15.7,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145150621","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Àngels Solanelles Curco , Eduardo Garcia , Anna Polishchuk , Maria Teresa La chica-Lhoëst , Vicenta Llorente-Cortes
{"title":"Impaired cholesterol and LDL uptake pathways in the development of oncological and cardiovascular diseases","authors":"Àngels Solanelles Curco , Eduardo Garcia , Anna Polishchuk , Maria Teresa La chica-Lhoëst , Vicenta Llorente-Cortes","doi":"10.1016/j.semcancer.2025.09.004","DOIUrl":"10.1016/j.semcancer.2025.09.004","url":null,"abstract":"<div><div>Dietary lipids play a critical role in the development of cardiovascular disease and cancer by influencing key cellular processes. Lipoprotein and fatty acid receptors activate intracellular signaling pathways that promote tumor growth and vascular remodeling. A key event in both cancer and vascular diseases is the retention of low-density lipoproteins (LDL) and other lipoprotein particles by proteoglycans in the extracellular matrix (ECM) of atherosclerotic plaques and the tumor stroma. This retention facilitates lipoprotein modification processes. Dysregulated uptake of modified lipoproteins—particularly through receptors that are not downregulated by intracellular cholesterol levels—leads to excessive lipid accumulation within lipid droplets (LDs) and other intracellular organelles. The lipid and protein content of LDs and mitochondria determine the biophysical and functional characteristics of the crucial interactions between these suborgalles. In particular, lipid-derived mediators such as prostaglandins, leukotrienes, ceramides, oxidized fatty acids, and cholesterol can disrupt LD biogenesis and dynamics, impair mitochondrial function, and alter the expression, activity, and subcellular localization of proteins that regulate LD-mitochondria interactions. Dysfunctional communication between LDs and mitochondria contributes to the onset and progression of cancer and cardiovascular disease by disturbing cellular metabolism and energy homeostasis. Common LDL-related mechanisms in atherosclerosis and cancer have been summarized in Figure 1.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 84-95"},"PeriodicalIF":15.7,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145150579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hao Li , Fei-Yang Chen , Qing Wang , Tian-Fu Wu , Zhi-Jun Sun
{"title":"Microorganisms in cancer tertiary lymphoid structure formation","authors":"Hao Li , Fei-Yang Chen , Qing Wang , Tian-Fu Wu , Zhi-Jun Sun","doi":"10.1016/j.semcancer.2025.09.005","DOIUrl":"10.1016/j.semcancer.2025.09.005","url":null,"abstract":"<div><div>Tertiary lymphoid structures (TLSs) are essential in cancer immunotherapy, markedly improving the efficacy of immune checkpoint blockade (ICB) therapy. The presence of TLSs within the tumor microenvironment (TME) is associated with improved responses to ICB therapy, making them valuable biomarkers for predicting the efficacy of ICB therapy. Recent studies have demonstrated that diverse microorganisms—including bacteria, viruses, and eukaryotes such as fungi—impact the effectiveness of ICB treatments. Notably, studies also emphasize that specific microorganisms contribute to the development of TLSs. However, the interactions between microorganisms and TLSs are complex, and the specific mechanisms by which microorganisms impact TLS formation remain largely unknown. This review summarizes emerging advances on the formation processes of TLSs, focusing on the involved signaling pathways, and discusses how microorganisms facilitate TLS formation by affecting these pathways, chemokines, antigen presentation, and immune cells. Understanding these relationships could help in identifying microorganisms, chemokines, and signaling pathways as diagnostic or prognostic biomarkers in cancer, as well as new therapeutic approaches and potential targets for cancer treatment.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 69-83"},"PeriodicalIF":15.7,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145114058","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Belen Picatoste , Heidy M. Guaman , Marina Herrero-Matesanz , Laura V. Piñeres , Alberto Benito-Martin
{"title":"Extracellular vesicles and the lipid messengers: The adipose tissue connection to cancer and metabolic disease.","authors":"Belen Picatoste , Heidy M. Guaman , Marina Herrero-Matesanz , Laura V. Piñeres , Alberto Benito-Martin","doi":"10.1016/j.semcancer.2025.09.003","DOIUrl":"10.1016/j.semcancer.2025.09.003","url":null,"abstract":"<div><div>Adipose tissue is essential for maintaining metabolic balance by serving as a major lipid depot for energy storage and release, while also secreting adipokines and extracellular vesicles that regulate inflammation and insulin sensitivity. The lipidic content of adipose tissue not only supports energy homeostasis but also contributes to its inflammatory profile, with altered lipid composition being a key factor in the pathogenesis of metabolic diseases such as obesity, type 2 diabetes, and cardiovascular disorders. Moreover, adipose tissue-derived extracellular vesicles, enriched in lipids, have emerged as significant mediators of intercellular communication, influencing local and systemic processes, including tumor progression. Adipose tissue-derived extracellular vesicles (EVs) have emerged as pivotal mediators of intercellular communication, with increasing evidences describing their role in microenvironment communication. In this review, we explore the diverse lipid species identified within EVs and examine lipidomics as a powerful and emerging technique to decode their functional roles. By analyzing the lipid cargo of these vesicles, we highlight their potential influence on cancer progression and cardiovascular disease. Furthermore, we discuss the broader implications of EV-derived lipids in metabolic regulation and disease pathophysiology. Understanding the complex interplay between EV lipid composition and pathological processes could open new avenues for biomarker discovery and therapeutic interventions in oncology and cardiovascular medicine.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 59-68"},"PeriodicalIF":15.7,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145102977","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Maria J. Monte , Laura Fàbrega , Marta R. Romero , Alvaro G. Temprano , Neil Kaplowitz , Carmen Garcia-Ruiz , Jose J.G. Marin , Jose C. Fernandez-Checa
{"title":"Bile acids in liver and gastrointestinal cancer","authors":"Maria J. Monte , Laura Fàbrega , Marta R. Romero , Alvaro G. Temprano , Neil Kaplowitz , Carmen Garcia-Ruiz , Jose J.G. Marin , Jose C. Fernandez-Checa","doi":"10.1016/j.semcancer.2025.09.002","DOIUrl":"10.1016/j.semcancer.2025.09.002","url":null,"abstract":"<div><div>Bile acids (BAs) are cholesterol-derived metabolites synthesized by hepatocytes and secreted in bile to follow an inter-organ transit to the intestine, where they facilitate fat digestion and the absorption of lipids and liposoluble vitamins. Traditionally considered mere end products of cholesterol catabolism, they are now acknowledged to play intricate roles in regulating intermediary metabolism by controlling the expression of crucial genes. Additionally, they exert a significant impact on inflammation, cytotoxicity, and carcinogenesis. Moreover, BAs have a critical impact on the crosstalk between gut microbiota and host physiology, which affects the progression of liver and gastrointestinal cancers. Clinical data and results from studies of animal models support the involvement of BAs in the development of hepatocellular carcinoma, cholangiocarcinoma, colorectal adenocarcinoma, and pancreatic cancer. On the other hand, BAs and their derivatives have been proposed as pharmacological tools in strategies to restore abnormal hepatobiliary function and target cytostatic agents to cancers of the enterohepatic circuit. In the present review, we summarize basic concepts of BA physiology and regulation, as well as new advances in this expanding field of renewed interest for cancer biology, lending further support for the key role of BAs in liver and gastrointestinal cancer.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 45-58"},"PeriodicalIF":15.7,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145041200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ales Langer , Pavel Soucek , Veronika Vymetalkova , Cosmeri Rizzato , Stefania Bunduc , Ilias P. Nikas , Viktor Hlavac , Michal Kroupa , Riccardo Farinella , Daniele Campa , Beatrice Mohelnikova-Duchonova
{"title":"Therapeutic pancreatic cancer biomarkers and pharmacogenetics","authors":"Ales Langer , Pavel Soucek , Veronika Vymetalkova , Cosmeri Rizzato , Stefania Bunduc , Ilias P. Nikas , Viktor Hlavac , Michal Kroupa , Riccardo Farinella , Daniele Campa , Beatrice Mohelnikova-Duchonova","doi":"10.1016/j.semcancer.2025.08.002","DOIUrl":"10.1016/j.semcancer.2025.08.002","url":null,"abstract":"<div><div>FOLFIRINOX and gemcitabine plus nab-paclitaxel represent the most effective chemotherapy regimens for metastatic pancreatic cancer patients nowadays, but the median overall survival remains less than one year. Pharmacogenomics and the individualization of therapy represent a promising strategy, including identifying patients at increased risk of toxicity. This review summarizes contemporary knowledge about genetic variability and putative biomarkers with published associations to therapy responses of pancreatic cancer not only for gold standard treatment regimens (FOLFIRINOX, gemcitabine/nab-paclitaxel and nal-IRI/5-fluorouracil) but also for other therapeutic options regarding targeted therapy and immunotherapy. From the published data reviewed, it is evident that the problem is highly complex, and the ultimate profile of the drug-sensitive or resistant patient, followed by individualized therapy, is the most probable way to improve the poor prognosis of pancreatic cancer patients. Additionally, we will give a brief recap of what has been learned from genome-wide association studies, from gene candidate studies carried out in the context of large consortia such as the Pancreatic Disease Research (PANDoRA) consortium, and from studies focused on specific mutations in DNA repair genes.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 31-44"},"PeriodicalIF":15.7,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145024155","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Giuseppe Annunziata , Massimiliano Caprio , Evelyn Frias-Toral , Annamaria Colao , Luigi Barrea
{"title":"Ketogenic diets: Dietary therapeutic potential on breast cancer","authors":"Giuseppe Annunziata , Massimiliano Caprio , Evelyn Frias-Toral , Annamaria Colao , Luigi Barrea","doi":"10.1016/j.semcancer.2025.09.001","DOIUrl":"10.1016/j.semcancer.2025.09.001","url":null,"abstract":"<div><div>Among the various types of tumors, breast cancer (BC) has a high distribution in the world population and is responsible for a high mortality rate. Like other forms of cancer, BC is characterised by distinctive features such as high-energy metabolism in tumor cells, genetic mutations, and mitochondrial dysfunction that sometimes make conventional therapies less effective. However, there is a growing awareness of the vital role played by diet therapy in the overall management of the patient with BC, both by supporting standard therapy and by directly targeting aberrant biological processes involved in carcinogenesis. Among the various nutritional strategies, the ketogenic diet (KD) has recently emerged as a promising therapeutic option for BC. Emerging evidence shows that KD, whether used alone or in combination with standard care, can directly influence tumor cell metabolism and reduces the risk of metastasis, thereby enhancing the efficacy of conventional drug therapies. This may allow for drug dose optimization, leading to fewer side effects and improved patient's quality of life. Nonetheless, the lack of clear evidence on the nutritional protocol to be adopted in BC patients, underlines the need for further research. This review aims to provide an overview of the effects of KD in the management of BC, both as a nutritional treatment and as an adjuvant therapy, providing insights into the implementation of precision nutrition protocols for this and other types of cancer.</div></div>","PeriodicalId":21594,"journal":{"name":"Seminars in cancer biology","volume":"116 ","pages":"Pages 22-30"},"PeriodicalIF":15.7,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145016142","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}