1
|
Lanting V, Oskam M, Wilmink H, Kamphuisen PW, van Es N. The role of germline and somatic mutations in predicting cancer-associated thrombosis: a narrative review. Curr Opin Hematol 2025; 32:138-145. [PMID: 39851266 PMCID: PMC11957438 DOI: 10.1097/moh.0000000000000861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
PURPOSE OF REVIEW Patients with cancer have an increased risk of venous thromboembolism (VTE). Guidelines suggest to use risk assessment tools to guide decisions about thromboprophylaxis, but current tools have modest discriminatory ability. Genetic information from the germline or tumor has the potential to improve VTE prediction. Here, we provide a clinical overview of the current role of genetics in cancer-associated VTE. RECENT FINDINGS Germline mutations, such as factor V Leiden and prothrombin G20210A, are associated with a 2- to 2.5-fold increased VTE risk in patients with cancer. Tumor-specific somatic mutations also contribute to VTE risk, such as ALK rearrangements increasing the risk in nonsmall cell lung cancer and IDH1 mutations decreasing the risk in gliomas. Other somatic mutations associated with VTE independent of tumor type include KRAS , STK11 , MET , KEAP1 , CTNNB1 , and CDKN2B . Incorporating data on germline or somatic mutations in risk scores improves discriminatory ability compared with the Khorana score. SUMMARY Specific germline and somatic mutations are associated with an increased VTE risk in patients with cancer and potentially improve performance of clinical risk scores. The increasing and widespread use of genetic testing in cancer care provides an opportunity for further development of prediction models incorporating genetic predictors.
Collapse
Affiliation(s)
- Vincent Lanting
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
- Tergooi MC, department of Internal Medicine, Hilversum
| | - Merel Oskam
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
| | - Hanneke Wilmink
- Amsterdam UMC, University of Amsterdam, department of Oncology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Pieter W. Kamphuisen
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
- Tergooi MC, department of Internal Medicine, Hilversum
| | - Nick van Es
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
| |
Collapse
|
2
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [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: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
3
|
Guo P, Song S, Niu Y, Kuang X, Zhou D, Zhou Z, Zhang Y, Ma X. Alternative splicing of bunched confers a dual role in hippo pathway-dependent growth and tumorigenesis. Oncogene 2025:10.1038/s41388-025-03348-6. [PMID: 40175650 DOI: 10.1038/s41388-025-03348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 04/04/2025]
Abstract
Alternative splicing is a fundamental mechanism that generates functionally distinct proteins from individual genes, contributing to gene regulation and proteomic diversity. In Drosophila, the bunched (bun) gene, a member of the TSC-22 domain gene family, undergoes alternative splicing, yielding diverse protein isoforms involved in crucial biological processes. Nevertheless, the specific roles and regulatory mechanisms of each isoform remain elusive. Here, we employed CRISPR/Cas9 technology to introduce targeted deletions within the endogenous locus of the bun gene, resulting in the removal of either long or short isoforms. We discovered that the short isoforms demonstrated a growth-suppressive role, whereas the long isoforms exhibited a growth-promoting effect. Surprisingly, the long isoforms exhibited a remarkable dual functionality, as both deletion and amplification of long isoform expression impede the excess growth induced by Hippo pathway inactivation. Mechanistically, ectopically expressed Bun long isoforms act as the transcriptional suppressor by competitively binding to targets' promoter regions in conjunction with Yorkie/Scalloped (Yki/Sd), thereby inhibiting its transcriptional outputs and ultimately leading to the growth suppression. These findings unveil the intricate interaction between distinct spliced isoforms of Bun and oncogenic outcomes, highlighting Bun long isoforms as the critical transcription suppressor regulating Hippo pathway inactivation-mediated growth and tumorigenesis in Drosophila.
Collapse
Affiliation(s)
- Pengjuan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Sha Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Yuxiao Niu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Xiaoyu Kuang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Dafa Zhou
- College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Zizhang Zhou
- College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Yanxiao Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
| |
Collapse
|
4
|
Bahari F, Ahangari Cohan R, Montazeri H. Element-specific estimation of background mutation rates in whole cancer genomes through transfer learning. NPJ Precis Oncol 2025; 9:92. [PMID: 40155429 PMCID: PMC11953285 DOI: 10.1038/s41698-025-00871-3] [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: 06/02/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
Mutational burden tests are essential for detecting signals of positive selection in cancer driver discovery by comparing observed mutation rates with background mutation rates (BMRs). However, accurate BMR estimation is challenging due to the diversity of mutational processes across genomes, complicating driver discovery efforts. Existing methods rely on various genomic regions and features for BMR estimation but lack a model that integrates both intergenic intervals and functional genomic elements on a comprehensive set of genomic features. Here, we introduce eMET (element-specific Mutation Estimator with boosted Trees), which employs 1372 (epi)genomic features from intergenic data and fine-tunes it with element-specific data through transfer learning. Applied to PCAWG somatic mutations, eMET significantly improves BMR accuracy and has potential to enhance driver discovery. Additionally, we provide an extensive analysis of BMR estimation, examining different machine learning models, genomic interval strategies, feature categories, and dimensionality reduction techniques.
Collapse
Affiliation(s)
- Farideh Bahari
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran.
| | - Hesam Montazeri
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
5
|
Jomova K, Alomar SY, Valko R, Liska J, Nepovimova E, Kuca K, Valko M. Flavonoids and their role in oxidative stress, inflammation, and human diseases. Chem Biol Interact 2025; 413:111489. [PMID: 40147618 DOI: 10.1016/j.cbi.2025.111489] [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: 06/02/2024] [Revised: 02/23/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Oxidative stress and chronic inflammation are important drivers in the pathogenesis and progression of many chronic diseases, such as cancers of the breast, kidney, lung, and others, autoimmune diseases (rheumatoid arthritis), cardiovascular diseases (hypertension, atherosclerosis, arrhythmia), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease), mental disorders (depression, schizophrenia, bipolar disorder), gastrointestinal disorders (inflammatory bowel disease, colorectal cancer), and other disorders. With the increasing demand for less toxic and more tolerable therapies, flavonoids have the potential to effectively modulate the responsiveness to conventional therapy and radiotherapy. Flavonoids are polyphenolic compounds found in fruits, vegetables, grains, and plant-derived beverages. Six of the twelve structurally different flavonoid subgroups are of dietary significance and include anthocyanidins (e.g. pelargonidin, cyanidin), flavan-3-ols (e.g. epicatechin, epigallocatechin), flavonols (e.g. quercetin, kaempferol), flavones (e.g. luteolin, baicalein), flavanones (e.g. hesperetin, naringenin), and isoflavones (daidzein, genistein). The health benefits of flavonoids are related to their structural characteristics, such as the number and position of hydroxyl groups and the presence of C2C3 double bonds, which predetermine their ability to chelate metal ions, terminate ROS (e.g. hydroxyl radicals formed by the Fenton reaction), and interact with biological targets to trigger a biological response. Based on these structural characteristics, flavonoids can exert both antioxidant or prooxidant properties, modulate the activity of ROS-scavenging enzymes and the expression and activation of proinflammatory cytokines (e.g., interleukin-1beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α)), induce apoptosis and autophagy, and target key signaling pathways, such as the nuclear factor erythroid 2-related factor 2 (Nrf2) and Bcl-2 family of proteins. This review aims to briefly discuss the mutually interconnected aspects of oxidative and inflammatory mechanisms, such as lipid peroxidation, protein oxidation, DNA damage, and the mechanism and resolution of inflammation. The major part of this article discusses the role of flavonoids in alleviating oxidative stress and inflammation, two common components of many human diseases. The results of epidemiological studies on flavonoids are also presented.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Richard Valko
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Jan Liska
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, 811 08, Bratislava, Slovakia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic; Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic
| | - Kamil Kuca
- Center of Advanced Innovation Technologies, VSB-Technical University of Ostrava, Ostrava-Poruba, 708 00, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, 5005, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia.
| |
Collapse
|
6
|
Yi S, Xie M. DriverMEDS: Cancer driver gene identification using mutual exclusivity from embeded features and driver mutation scoring. Methods 2025; 239:22-29. [PMID: 40113153 DOI: 10.1016/j.ymeth.2025.03.010] [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: 10/11/2024] [Revised: 01/24/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
Efficiently identifying cancer driver genes plays a key role in the cancer development, diagnosis and treatment. Current unsupervised driver gene identification methods typically integrate multi-omics data into gene function networks and employ network embedding algorithms to learn gene features. Additionally, they consider mutual exclusivity and mutation frequency as crucial concepts in identifying driver genes. However, existing approaches neglect the possible important implications of mutual exclusivity in the embedding space. Furthermore, they simply assume that all driver genes exhibit high mutation frequencies. Fortunately, we explored the mutual exclusivity implanted in the learned features and have verified that the Euclidean distances between learned features are strongly related to the mutual exclusivity and they can reveal more information for the mutual exclusivity. Thus, we designed an unsupervised driver gene predicting framework DriverMEDS based on the above idea and a novel driver mutation scoring strategy. First, we design a feature clustering algorithm to generate gene modules. In each module, the Euclidean distances of learned features are used to calculate a module importance score for each gene based on the related mutual exclusivity. Then, following the fact that most of driver genes have intermediate mutation frequencies, a driver mutation scoring function is designed for each gene to optimize the existing mutation frequency scoring strategy. Finally, the weighted sum of the module importance score and the driver mutation score is used to prioritize the genes. The experiment results and analysis show that DriverMEDS could detect novel cancer driver genes and relevant function modules, and outperforms other five state-of-the-art methods for cancer driver identification.
Collapse
Affiliation(s)
- Sichen Yi
- Key Laboratory of Computing and Stochastic Mathematics (Ministry of Education), School of Mathematics and Statistics, Hunan Normal University, Changsha 410081, China.
| | - Minzhu Xie
- Key Laboratory of Computing and Stochastic Mathematics (Ministry of Education), School of Mathematics and Statistics, Hunan Normal University, Changsha 410081, China; College of Information Science and Engineering, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
7
|
Bruhm DC, Vulpescu NA, Foda ZH, Phallen J, Scharpf RB, Velculescu VE. Genomic and fragmentomic landscapes of cell-free DNA for early cancer detection. Nat Rev Cancer 2025:10.1038/s41568-025-00795-x. [PMID: 40038442 DOI: 10.1038/s41568-025-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
Genomic analyses of cell-free DNA (cfDNA) in plasma are enabling noninvasive blood-based biomarker approaches to cancer detection and disease monitoring. Current approaches for identification of circulating tumour DNA typically use targeted tumour-specific mutations or methylation analyses. An emerging approach is based on the recognition of altered genome-wide cfDNA fragmentation in patients with cancer. Recent studies have revealed a multitude of characteristics that can affect the compendium of cfDNA fragments across the genome, collectively called the 'cfDNA fragmentome'. These changes result from genomic, epigenomic, transcriptomic and chromatin states of an individual and affect the size, position, coverage, mutation, structural and methylation characteristics of cfDNA. Identifying and monitoring these changes has the potential to improve early detection of cancer, especially using highly sensitive multi-feature machine learning approaches that would be amenable to broad use in populations at increased risk. This Review highlights the rapidly evolving field of genome-wide analyses of cfDNA characteristics, their comparison to existing cfDNA methods, and recent related innovations at the intersection of large-scale sequencing and artificial intelligence. As the breadth of clinical applications of cfDNA fragmentome methods have enormous public health implications for cancer screening and personalized approaches for clinical management of patients with cancer, we outline the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Yang H, Zhang L, Kang X, Si Y, Song P, Su X. Reaction Pathway Differentiation Enabled Fingerprinting Signal for Single Nucleotide Variant Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412680. [PMID: 39903775 PMCID: PMC11948007 DOI: 10.1002/advs.202412680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/17/2025] [Indexed: 02/06/2025]
Abstract
Accurate identification of single-nucleotide variants (SNVs) is paramount for disease diagnosis. Despite the facile design of DNA hybridization probes, their limited specificity poses challenges in clinical applications. Here, a differential reaction pathway probe (DRPP) based on a dynamic DNA reaction network is presented. DRPP leverages differences in reaction intermediate concentrations between SNV and WT groups, directing them into distinct reaction pathways. This generates a strong pulse-like signal for SNV and a weak unidirectional increase signal for wild-type (WT). Through the application of machine learning to fluorescence kinetic data analysis, the classification of SNV and WT signals is automated with an accuracy of 99.6%, significantly exceeding the 80.7% accuracy of conventional methods. Additionally, sensitivity for variant allele frequency (VAF) is enhanced down to 0.1%, representing a ten-fold improvement over conventional approaches. DRPP accurately identified D614G and N501Y SNVs in the S gene of SARS-CoV-2 variants in patient swab samples with accuracy over 99% (n = 82). It determined the VAF of ovarian cancer-related mutations KRAS-G12R, NRAS-G12C, and BRAF-V600E in both tissue and blood samples (n = 77), discriminating cancer patients and healthy individuals with significant difference (p < 0.001). The potential integration of DRPP into clinical diagnostics, along with rapid amplification techniques, holds promise for early disease diagnostics and personalized diagnostics.
Collapse
Affiliation(s)
- Huixiao Yang
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
| | - Linghao Zhang
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
| | - Xinmiao Kang
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
| | - Yunpei Si
- School of Biomedical EngineeringZhangjiang Institute for Advanced Study and National Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Ping Song
- School of Biomedical EngineeringZhangjiang Institute for Advanced Study and National Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Xin Su
- State Key Laboratory of Organic‐Inorganic CompositesBeijing Key Laboratory of BioprocessBeijing Advanced Innovation Center for Soft Matter Science and EngineeringCollege of Life Science and TechnologyBeijing University of Chemical TechnologyBeijing100029China
- State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijing100191China
| |
Collapse
|
9
|
Li L, Li H, Zhang K, Zhao C, Wang F, Sun J, Wang J. The role and mechanism of hepatocyte nuclear factor 1β in the occurrence and development of different human tumors: A pan-cancer analysis. ENVIRONMENTAL TOXICOLOGY 2025; 40:471-480. [PMID: 39887605 DOI: 10.1002/tox.24254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 02/01/2025]
Abstract
Carcinomatosis is one of the leading threats to human public fitness. HNF1B is a critical transcription element in vertebrate proliferation and oncogenesis, which has been shown to play roles in reactive oxygen species (ROS) metabolism. Our previous results have identified HNF1B as a tumor suppressor that could inhibit the malignant progression of prostate cancer. Yet there is no pan-carcinomatosis analysis of HNF1B, which could help us better understand common and unique underlying mechanisms in mankind knubs to enhance novel and competent treatment. Here, in our research, we evaluated the utterance pattern and explored the function of HNF1B in 33 knub categories using the data from the Cancer Genome Atlas Program (TCGA), Gene Expression Omnibus (GEO), and CLNICAL PROTEOMICTUMOR ANALYSIS CONSORTIUM (CPTAC) dataset. We found different HNF1B roles in various cancer types. HNF1B was upregulated in CHOL, STAD, KIRP, and THCA, and was downregulated in GBM, KICH, COAD, KIRC, LUSC, SARC, PAAD, and TGCT. Prognostic analyses indicated that higher HNF1B displayed better illness outcomes in BLCA, READ, and PRAD, while poorer outcomes in LUSC and THYM. HNF1B mutation was most frequent in endometrial cancer but was not associated with disease prognosis. It was discovered that HNF1B utterance relevant to endothelial cell penetration status in BLCA, ESCA, LUAD, LUSC, and TGCT, and carcinomatosis-associated fibroblast infiltration was observed in ESCA, KIRC, LIHC, and TGCT. Moreover, functional enrichment analysis disclosed that metabolism-related functions were implicated in the function of HNF1B. Taken together, our pan- carcinomatosis analysis showed the complicated roles of HNF1B in a variety of carcinomatoses, being able to improve the extensive comprehension of HNF1B's role in tumorigenesis.
Collapse
Affiliation(s)
- Liang Li
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Haikun Li
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Ke Zhang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Chunchun Zhao
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Fei Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jian Sun
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jianqing Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
10
|
Zhou W, Chi H, Zhao X, Tao G, Gan J. A mutational signature and ARID1A mutation associated with outcome in hepatocellular carcinoma. Clin Transl Oncol 2025; 27:1166-1175. [PMID: 39179940 DOI: 10.1007/s12094-024-03669-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
OBJECTIVE The prognosis of hepatocellular carcinoma (HCC) is poor and there is no stable and reliable molecular biomarker for evaluation. This study attempted to find reliable prognostic markers from tumor mutational profiles. METHODS A total of 362 HCC samples with whole-exome sequencing were collected as discovery datasets, and 200 samples with targeted sequencing were used for validation of the relevant results. All HCC samples were obtained from previously published studies. Bayesian non-negative matrix factorization was used to extract mutational signatures, and multivariate Cox regression models were utilized to identify the prognostic role of mutational factors. Gene set enrichment analysis was employed to discover potential signaling pathways associated with specific mutational groups. RESULTS In the HCC discovery dataset, a total of four mutational signatures (i.e., signatures 4, 6, 16, and 22) were extracted, of which signature 16 characterized by T>C mutations was observed to be associated with favorable HCC prognosis, and this correlation was also found in the validation dataset. Further analysis showed that patients with ARID1A mutations exhibited inferior survival outcomes in both discovery and validation datasets. Mechanistic exploration revealed that the presence of signature 16 was associated with better immune infiltration and tumor immunogenicity, while patients with ARID1A mutations were away from these favorable immunological features. CONCLUSION By integrating somatic mutation data and clinical information of HCC, this study identified that signature 16 and ARID1A mutations were associated with better and worse outcomes respectively, providing a basis for prognosis prediction and clinical treatment strategies of HCC.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Hao Chi
- Department of Medical Laboratory, Huaian Hospital of Huaian City, Huaian, China
| | - Xiaohu Zhao
- Department of Infectious Diseases, Huaian Hospital of Huaian City, Huaian, China
| | - Guangrong Tao
- Department of Infectious Diseases, Huaian Hospital of Huaian City, Huaian, China
| | - Jianhe Gan
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
11
|
Pouyan A, Ghorbanlo M, Eslami M, Jahanshahi M, Ziaei E, Salami A, Mokhtari K, Shahpasand K, Farahani N, Meybodi TE, Entezari M, Taheriazam A, Hushmandi K, Hashemi M. Glioblastoma multiforme: insights into pathogenesis, key signaling pathways, and therapeutic strategies. Mol Cancer 2025; 24:58. [PMID: 40011944 DOI: 10.1186/s12943-025-02267-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor in adults, characterized by a poor prognosis and significant resistance to existing treatments. Despite progress in therapeutic strategies, the median overall survival remains approximately 15 months. A hallmark of GBM is its intricate molecular profile, driven by disruptions in multiple signaling pathways, including PI3K/AKT/mTOR, Wnt, NF-κB, and TGF-β, critical to tumor growth, invasion, and treatment resistance. This review examines the epidemiology, molecular mechanisms, and therapeutic prospects of targeting these pathways in GBM, highlighting recent insights into pathway interactions and discovering new therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Ashkan Pouyan
- Department of Neurosurgery, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Masoud Ghorbanlo
- Department of Anesthesiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Eslami
- Department of Neurosurgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Jahanshahi
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Ziaei
- Department of Neurosurgery, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Salami
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Koorosh Shahpasand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Tohid Emami Meybodi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Functional Neurosurgery Research Center, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Epidemiology, University of Tehran, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
12
|
Wu S, Thawani R. Tumor-Agnostic Therapies in Practice: Challenges, Innovations, and Future Perspectives. Cancers (Basel) 2025; 17:801. [PMID: 40075649 PMCID: PMC11899253 DOI: 10.3390/cancers17050801] [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: 12/31/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
This review comprehensively analyzes the current landscape of tumor-agnostic therapies in oncology. Tumor-agnostic therapies are designed to target specific molecular alterations rather than the primary site of the tumor, representing a shift in cancer treatment. We discuss recent approvals by regulatory agencies such as the FDA and EMA, highlighting therapies that have demonstrated efficacy across multiple cancer types sharing common alterations. We delve into the trial methodologies that underpin these approvals, emphasizing innovative designs such as basket trials and umbrella trials. These methodologies present unique advantages, including increased efficiency in patient recruitment and the ability to assess drug efficacy in diverse populations rapidly. However, they also entail certain challenges, including the need for robust biomarkers and the complexities of regulatory requirements. Moreover, we examine the promising prospects for developing therapies for rare cancers that exhibit common molecular targets typically associated with more prevalent malignancies. By synthesizing these insights, this review underscores the transformative potential of tumor-agnostic therapies in oncology. It offers a pathway for personalized cancer treatment that transcends conventional histology-based classification.
Collapse
Affiliation(s)
| | - Rajat Thawani
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA;
| |
Collapse
|
13
|
Wang CJ, Cui L, Li SS, Ma HH, Wang D, Lian HY, Zhao YZ, Zhang LP, Li WJ, Zhang Q, Zhao XX, Yang Y, Huang XT, Liu W, Wang YZ, Wu WS, Wang TY, Zhang R, Li ZG. Genetic Landscape and Its Prognostic Impact in Children With Langerhans Cell Histiocytosis. Arch Pathol Lab Med 2025; 149:175-190. [PMID: 38749502 DOI: 10.5858/arpa.2023-0236-oa] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 01/29/2025]
Abstract
CONTEXT.— Langerhans cell histiocytosis (LCH) is a rare myeloid neoplasm that predominantly affects young children. OBJECTIVE.— To investigate genetic alterations and their correlation with clinical characteristics and prognosis in pediatric LCH. DESIGN.— We performed targeted sequencing to detect mutations in LCH lesions from pediatric patients. RESULTS.— A total of 30 genomic alterations in 5 genes of the MAPK pathway were identified in 187 of 223 patients (83.9%). BRAF V600E (B-Raf proto-oncogene, serine/threonine kinase) was the most common mutation (51.6%), followed by MAP2K1 (mitogen-activated protein kinase kinase 1) alterations (17.0%) and other BRAF mutations (13.0%). ARAF (A-Raf proto-oncogene, serine/threonine kinase) and KRAS (KRAS proto-oncogene, GTPase) mutations were relatively rare (2.2% and 0.9%, respectively). Additionally, FNBP1 (formin-binding protein 1)::BRAF fusion and MAP3K10 (mitogen-activated protein kinase kinase 10) mutations A17T and R823C were identified in 1 case each, with possible constitutive activation of ERK1/2 phosphorylation. BRAF V600E was more frequent in patients with risk organ involvement, while MAP2K1 mutation was more prevalent in patients with single-system LCH (P = .001). BRAF V600E was associated with craniofacial bone, skin, liver, spleen, and ear involvement (all P < .05). Patients with other BRAF mutations had a higher proportion of spinal column involvement (P = .006). Univariate analysis showed a significant difference in progression-free survival among the 4 molecular subgroups for patients treated with first-line therapy (P = .02). According to multivariate analysis, risk organ involvement was the strongest independent adverse prognostic factor (hazard ratio, 8.854; P < .001); BRAF or MAP2K1 mutation was not an independent prognostic factor. CONCLUSIONS.— Most pediatric patients with LCH carry somatic mutations involving the MAPK pathway, correlating with clinical characteristics and outcomes for first-line chemotherapy.
Collapse
Affiliation(s)
- Chan-Juan Wang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Lei Cui
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | | | - Hong-Hao Ma
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Dong Wang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Hong-Yun Lian
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Yun-Ze Zhao
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Li-Ping Zhang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Wei-Jing Li
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Qing Zhang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Xiao-Xi Zhao
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Ying Yang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Xiao-Tong Huang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Wei Liu
- the Department of Hematology Oncology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China (Liu)
| | - Yi-Zhuo Wang
- the Department of Pediatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China (Y-Z Wang)
| | - Wan-Shui Wu
- and the Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China (Wu)
| | - Tian-You Wang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Rui Zhang
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| | - Zhi-Gang Li
- From the Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute (C-J Wang, Cui, W-J Li, Q Zhang, X-X Zhao, Huang, Z-G Li) and Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Center (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li), Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- National Key Discipline of Pediatrics, Capital Medical University, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China (C-J Wang, Cui, Ma, D Wang, Lian, Y-Z Zhao, L-P Zhang, W-J Li, Q Zhang, X-X Zhao, Yang, Huang, T-Y Wang, R Zhang, Z-G Li)
| |
Collapse
|
14
|
Janiszewska J, Kostrzewska-Poczekaj M, Wierzbicka M, Brenner JC, Giefing M. HPV-driven oncogenesis-much more than the E6 and E7 oncoproteins. J Appl Genet 2025; 66:63-71. [PMID: 38907809 PMCID: PMC11761861 DOI: 10.1007/s13353-024-00883-y] [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: 11/28/2023] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
High-risk human papillomaviruses are well-established drivers of several cancer types including cervical, head and neck, penile as well as anal cancers. While the E6 and E7 viral oncoproteins have proven to be critical for malignant transformation, evidence is also beginning to emerge suggesting that both host pathways and additional viral genes may also be pivotal for malignant transformation. Here, we focus on the role of host APOBEC genes, which have an important role in molecular editing including in the response to the viral DNA and their role in HPV-driven carcinogenesis. Further, we also discuss data developed suggesting the existence of HPV-derived miRNAs in HPV + tumors and their potential role in regulating the host transcriptome. Collectively, while recent advances in these two areas have added complexity to the working model of papillomavirus-induced oncogenesis, these discoveries have also shed a light onto new areas of research that will be required to fully understand the process.
Collapse
Affiliation(s)
- J Janiszewska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - M Kostrzewska-Poczekaj
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - M Wierzbicka
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
- Research & Development Centre, Regional Specialist Hospital Wroclaw, Wroclaw, Poland
- Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - J C Brenner
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - M Giefing
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland.
| |
Collapse
|
15
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
16
|
Fan JJ, Erickson AW, Carrillo-Garcia J, Wang X, Skowron P, Wang X, Chen X, Shan G, Dou W, Bahrampour S, Xiong Y, Dong W, Abeysundara N, Francisco MA, Pusong RJ, Wang W, Li M, Ying E, Suárez RA, Farooq H, Holgado BL, Wu X, Daniels C, Dupuy AJ, Cadiñanos J, Bradley A, Bagchi A, Moriarity BS, Largaespada DA, Morrissy AS, Ramaswamy V, Mack SC, Garzia L, Dirks PB, Li X, Wanggou S, Egan S, Sun Y, Taylor MD, Huang X. A forward genetic screen identifies potassium channel essentiality in SHH medulloblastoma maintenance. Dev Cell 2025:S1534-5807(25)00001-2. [PMID: 39862856 DOI: 10.1016/j.devcel.2025.01.001] [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: 09/02/2024] [Revised: 10/28/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025]
Abstract
Distinguishing tumor maintenance genes from initiation, progression, and passenger genes is critical for developing effective therapies. We employed a functional genomic approach using the Lazy Piggy transposon to identify tumor maintenance genes in vivo and applied this to sonic hedgehog (SHH) medulloblastoma (MB). Combining Lazy Piggy screening in mice and transcriptomic profiling of human MB, we identified the voltage-gated potassium channel KCNB2 as a candidate maintenance driver. KCNB2 governs cell volume of MB-propagating cells (MPCs), with KCNB2 depletion causing osmotic swelling, decreased plasma membrane tension, and elevated endocytic internalization of epidermal growth factor receptor (EGFR), thereby mitigating proliferation of MPCs to ultimately impair MB growth. KCNB2 is largely dispensable for mouse development and KCNB2 knockout synergizes with anti-SHH therapy in treating MB. These results demonstrate the utility of the Lazy Piggy functional genomic approach in identifying cancer maintenance drivers and elucidate a mechanism by which potassium homeostasis integrates biomechanical and biochemical signaling to promote MB aggression.
Collapse
Affiliation(s)
- Jerry J Fan
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anders W Erickson
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Julia Carrillo-Garcia
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xin Wang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Patryk Skowron
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xian Wang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Xin Chen
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China
| | - Guanqiao Shan
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Shahrzad Bahrampour
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Yi Xiong
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Weifan Dong
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Namal Abeysundara
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Michelle A Francisco
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ronwell J Pusong
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Wei Wang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Miranda Li
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Elliot Ying
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Raúl A Suárez
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hamza Farooq
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Borja L Holgado
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiaochong Wu
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Texas Children's Cancer and Hematology Center, Houston, TX 77030, USA; Department of Pediatrics, Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Craig Daniels
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Texas Children's Cancer and Hematology Center, Houston, TX 77030, USA; Department of Pediatrics, Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Adam J Dupuy
- Department of Anatomy & Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Juan Cadiñanos
- Instituto de Medicina Oncológica y Molecular de Asturias (IMOMA), Oviedo 33193, Spain
| | - Allan Bradley
- T-Therapeutics Ltd. One Riverside, Granta Park, Cambridge CB21 6AD, UK
| | - Anindya Bagchi
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Branden S Moriarity
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - David A Largaespada
- Masonic Cancer Center, Department of Pediatrics, and Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - A Sorana Morrissy
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4Z6, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T8, Canada
| | - Vijay Ramaswamy
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Paediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Stephen C Mack
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Developmental Neurobiology, Neurobiology and Brain Tumor Program, Center of Excellence in Neuro-Oncology Sciences, St Jude Children's Hospital, Memphis, TN 38105, USA
| | - Livia Garzia
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal, QC H4A 3J1, Canada; Cancer Research Program, RI-MUHC, Montreal, QC H4A 3J1, Canada
| | - Peter B Dirks
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Siyi Wanggou
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Sean Egan
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada; Texas Children's Cancer and Hematology Center, Houston, TX 77030, USA; Department of Pediatrics, Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Texas Children's Hospital, Houston, TX 77030, USA; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Xi Huang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
17
|
Zeng G, Zhao C, Li G, Huang Z, Zhuang J, Liang X, Yu X, Fang S. Identifying somatic driver mutations in cancer with a language model of the human genome. Comput Struct Biotechnol J 2025; 27:531-540. [PMID: 39968174 PMCID: PMC11833646 DOI: 10.1016/j.csbj.2025.01.011] [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: 11/21/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
Somatic driver mutations play important roles in cancer and must be precisely identified to advance our understanding of tumorigenesis and its promotion and progression. However, identifying somatic driver mutations remains challenging in Homo sapiens genomics due to the random nature of mutations and the high cost of qualitative experiments. Building on the powerful sequence interpretation capabilities of language models, we propose a self-attention-based contextualized pretrained language model for somatic driver mutation identification. We pretrained the model with the Homo sapiens reference genome to equip it with the ability to understand genome sequences and then fine-tuned it for oncogene and tumor suppressor gene prediction tasks, enabling it to extract features related to driver genes from the original genome sequence. The fine-tuned model was used to obtain the mutations' carcinogenic effect characteristics to further identify whether the mutation is a driver or a passenger. Compared with other computational algorithms, our method achieved excellent somatic driver mutation identification performance on the test set, with an absolute improvement of 4.31% in AUROC over the best comparison method. The strong performance of our method indicates that it can provide new insights into the discovery of cancer drivers.
Collapse
Affiliation(s)
- Guangjian Zeng
- School of Biomedical Engineering, Shenzhen University, Shenzhen, China
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Chengzhi Zhao
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Guanpeng Li
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Zhengyang Huang
- School of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Jinhu Zhuang
- Shenzhen Health Development Research and Data Management Center, Guangdong, China
| | - Xiaohua Liang
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaxia Yu
- School of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Shenying Fang
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
18
|
Dong JH, Shen XY, Chen YN, Liu Y, Xue CY, Zhang RH, Liu YH, Zhou YL, Zhang XX. Glycosylase Pretreatment with Chemical Labeling-Assisted HPLC-MS/MS: An Ultrasensitive and Reliable Strategy for Quantification of 8-Oxo-7,8-dihydro-2'-deoxyguanosine in Genomic DNA. Anal Chem 2025; 97:365-372. [PMID: 39707940 DOI: 10.1021/acs.analchem.4c04339] [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: 12/23/2024]
Abstract
8-Oxo-7,8-dihydro-2'-deoxyguanosine (dOG), the dominant oxidative product of 2'-deoxyguanosine (dG) under high levels of reactive oxygen species, usually serves as a biomarker for oxidative stress and a risk assessment factor for various diseases. Due to the extremely low abundance of dOG and the susceptibility of dOG detection to the interference of spurious oxidation, research on related biological processes is limited by insufficient sensitivity and specificity. In this work, an ultrasensitive and reliable approach for genome-wide dOG quantification was developed through chemical labeling-assisted high-performance liquid chromatography-tandem mass spectrometry with the introduction of glycosylase pretreatment. Upon derivatization by a novel labeling reagent rhodamine B ethylenediamine, the detection sensitivity of dOG was enhanced by 100-fold, and the detection limit was as low as 25 amol, which was superior to those of reported mass spectrometry-based methods. Potassium ferricyanide, as a single-electron oxidant, was shown to possess strong selectivity for dOG versus dG, improving the labeling specificity and reducing the interference from dG. The spurious oxidation during sample pretreatment was systematically explored and minimized, and a control assay of glycosylase pretreatment was proposed to further improve the quantitative accuracy of dOG. Precise quantification of endogenous dOG in different cells was achieved with less than 500 ng of genomic DNA. This method was successfully applied to the assessment of the overall level of oxidative damage under the treatment of glycosylase inhibitors, potentially contributing to the exploration of the complex role of dOG in physiological status and disease phenotype.
Collapse
Affiliation(s)
- Jia-Hui Dong
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Xu-Yang Shen
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yu-Nan Chen
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Global Manufacturing Business Unit, WuXi Biologics, Wuxi 214091, China
| | - Ying Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chen-Yu Xue
- Key Laboratory of Forensic Toxicology, Ministry of Public Security, Beijing 100191, China
| | - Run-Hong Zhang
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Ya-Hong Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Ying-Lin Zhou
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xin-Xiang Zhang
- Beijing National Laboratory for Molecular Sciences (BNLMS), MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
19
|
Mazzocca A, Ferraro G, Misciagna G. The systemic evolutionary theory of the origin of cancer (SETOC): an update. Mol Med 2025; 31:12. [PMID: 39806272 PMCID: PMC11730465 DOI: 10.1186/s10020-025-01069-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025] Open
Abstract
The Systemic Evolutionary Theory of the Origin of Cancer (SETOC) is a recently proposed theory founded on two primary principles: the cooperative and endosymbiotic process of cell evolution as described by Lynn Margulis, and the integration of complex systems operating in eukaryotic cells, which is a core concept in systems biology. The SETOC proposes that malignant transformation occurs when cells undergo a continuous adaptation process in response to long-term injuries, leading to tissue remodeling, chronic inflammation, fibrosis, and ultimately cancer. This process involves a maladaptive response, wherein the 'endosymbiotic contract' between the nuclear-cytoplasmic system (derived from the primordial archaeal cell) and the mitochondrial system (derived from the primordial α-proteobacterium) gradually breaks down. This ultimately leads to uncoordinated behaviors and functions in transformed cells. The decoupling of the two cellular subsystems causes transformed cells to acquire phenotypic characteristics analogous to those of unicellular organisms, as well as certain biological features of embryonic development that are normally suppressed. These adaptive changes enable cancer cells to survive in the harsh tumor microenvironment characterized by low oxygen concentrations, inadequate nutrients, increased catabolic waste, and increased acidity. De-endosymbiosis reprograms the sequential metabolic functions of glycolysis, the TCA cycle, and oxidative phosphorylation (OxPhos). This leads to increased lactate fermentation (Warburg effect), respiratory chain dysfunction, and TCA cycle reversal. Here, we present an updated version of the SETOC that incorporates the fundamental principles outlined by this theory and integrates the epistemological approach used to develop it.
Collapse
Affiliation(s)
- Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124, Bari, Italy.
- Association for Systems Science, Via S. Stefano, 42, I-75100, Matera, Italy.
| | - Giovanni Ferraro
- Association for Systems Science, Via S. Stefano, 42, I-75100, Matera, Italy
| | - Giovanni Misciagna
- Association for Systems Science, Via S. Stefano, 42, I-75100, Matera, Italy
| |
Collapse
|
20
|
Funk JS, Klimovich M, Drangenstein D, Pielhoop O, Hunold P, Borowek A, Noeparast M, Pavlakis E, Neumann M, Balourdas DI, Kochhan K, Merle N, Bullwinkel I, Wanzel M, Elmshäuser S, Teply-Szymanski J, Nist A, Procida T, Bartkuhn M, Humpert K, Mernberger M, Savai R, Soussi T, Joerger AC, Stiewe T. Deep CRISPR mutagenesis characterizes the functional diversity of TP53 mutations. Nat Genet 2025; 57:140-153. [PMID: 39774325 PMCID: PMC11735402 DOI: 10.1038/s41588-024-02039-4] [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: 03/17/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
The mutational landscape of TP53, a tumor suppressor mutated in about half of all cancers, includes over 2,000 known missense mutations. To fully leverage TP53 mutation status for personalized medicine, a thorough understanding of the functional diversity of these mutations is essential. We conducted a deep mutational scan using saturation genome editing with CRISPR-mediated homology-directed repair to engineer 9,225 TP53 variants in cancer cells. This high-resolution approach, covering 94.5% of all cancer-associated TP53 missense mutations, precisely mapped the impact of individual mutations on tumor cell fitness, surpassing previous deep mutational scan studies in distinguishing benign from pathogenic variants. Our results revealed even subtle loss-of-function phenotypes and identified promising mutants for pharmacological reactivation. Moreover, we uncovered the roles of splicing alterations and nonsense-mediated messenger RNA decay in mutation-driven TP53 dysfunction. These findings underscore the power of saturation genome editing in advancing clinical TP53 variant interpretation for genetic counseling and personalized cancer therapy.
Collapse
Affiliation(s)
- Julianne S Funk
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Maria Klimovich
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Ole Pielhoop
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Pascal Hunold
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Anna Borowek
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Maxim Noeparast
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Michelle Neumann
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Dimitrios-Ilias Balourdas
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Katharina Kochhan
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Nastasja Merle
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Imke Bullwinkel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Michael Wanzel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Julia Teply-Szymanski
- Institute of Pathology, Philipps-University, Marburg University Hospital, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps-University, Marburg, Germany
| | - Tara Procida
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Marek Bartkuhn
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Justus-Liebig-University, Giessen, Germany
| | - Katharina Humpert
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
- Bioinformatics Core Facility, Philipps-University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Lung Microenvironmental Niche in Cancerogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thierry Soussi
- Centre de Recherche Saint-Antoine UMRS_938, INSERM, Sorbonne Université, Paris, France
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Clinical Genetics, Uppsala University Hospital, Uppsala, Sweden
| | - Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.
- Genomics Core Facility, Philipps-University, Marburg, Germany.
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.
- Bioinformatics Core Facility, Philipps-University, Marburg, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
21
|
Ngoi NYL, Gallo D, Torrado C, Nardo M, Durocher D, Yap TA. Synthetic lethal strategies for the development of cancer therapeutics. Nat Rev Clin Oncol 2025; 22:46-64. [PMID: 39627502 DOI: 10.1038/s41571-024-00966-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Synthetic lethality is a genetic phenomenon whereby the simultaneous presence of two different genetic alterations impairs cellular viability. Importantly, targeting synthetic lethal interactions offers potential therapeutic strategies for cancers with alterations in pathways that might otherwise be considered undruggable. High-throughput screening methods based on modern CRISPR-Cas9 technologies have emerged and become crucial for identifying novel synthetic lethal interactions with the potential for translation into biologically rational cancer therapeutic strategies as well as associated predictive biomarkers of response capable of guiding patient selection. Spurred by the clinical success of PARP inhibitors in patients with BRCA-mutant cancers, novel agents targeting multiple synthetic lethal interactions within DNA damage response pathways are in clinical development, and rational strategies targeting synthetic lethal interactions spanning alterations in epigenetic, metabolic and proliferative pathways have also emerged and are in late preclinical and/or early clinical testing. In this Review, we provide a comprehensive overview of established and emerging technologies for synthetic lethal drug discovery and development and discuss promising therapeutic strategies targeting such interactions.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Gallo
- Repare Therapeutics, Inc., Montreal, Quebec, Canada
| | - Carlos Torrado
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirella Nardo
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-024-2770-x. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
23
|
Sears TJ, Pagadala MS, Castro A, Lee KH, Kong J, Tanaka K, Lippman SM, Zanetti M, Carter H. Integrated Germline and Somatic Features Reveal Divergent Immune Pathways Driving Response to Immune Checkpoint Blockade. Cancer Immunol Res 2024; 12:1780-1795. [PMID: 39255339 PMCID: PMC11612627 DOI: 10.1158/2326-6066.cir-24-0164] [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: 02/16/2024] [Revised: 06/13/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
Immune checkpoint blockade (ICB) has revolutionized cancer treatment; however, the mechanisms determining patient response remain poorly understood. Here, we used machine learning to predict ICB response from germline and somatic biomarkers and interpreted the learned model to uncover putative mechanisms driving superior outcomes. Patients with higher infiltration of T-follicular helper cells had responses even in the presence of defects in the MHC class-I (MHC-I). Further investigation uncovered different ICB responses in tumors when responses were reliant on MHC-I versus MHC-II neoantigens. Despite similar response rates, MHC II-reliant responses were associated with significantly longer durable clinical benefits (discovery: median overall survival of 63.6 vs. 34.5 months; P = 0.0074; validation: median overall survival of 37.5 vs. 33.1 months; P = 0.040). Characteristics of the tumor immune microenvironment reflected MHC neoantigen reliance, and analysis of immune checkpoints revealed LAG3 as a potential target in MHC II-reliant but not MHC I-reliant responses. This study highlights the value of interpretable machine learning models in elucidating the biological basis of therapy responses.
Collapse
Affiliation(s)
- Timothy J. Sears
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California
| | - Meghana S. Pagadala
- Biomedical Sciences Program, University of California San Diego, La Jolla, California
| | - Andrea Castro
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, United Kingdom
| | - Ko-han Lee
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California
| | - JungHo Kong
- Division of Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, California
| | - Kairi Tanaka
- School of Biological Sciences, University of California San Diego, La Jolla, California
| | - Scott M. Lippman
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Maurizio Zanetti
- Moores Cancer Center, University of California San Diego, La Jolla, California
- The Laboratory of Immunology, Moores Cancer Center and Department of Medicine, University of California San Diego, La Jolla, California
| | - Hannah Carter
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California
- Moores Cancer Center, University of California San Diego, La Jolla, California
- The Laboratory of Immunology, Moores Cancer Center and Department of Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
24
|
Bukkuri A, Adler FR. Of criminals and cancer: The importance of social bonds and innate morality on cellular societies. Cells Dev 2024; 180:203964. [PMID: 39151750 DOI: 10.1016/j.cdev.2024.203964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
The current dogma in cancer biology contends that cancer is an identity problem: mutations in a cell's DNA cause it to "go rogue" and proliferate out of control. However, this largely ignores the role of cell-cell interaction and fails to explain phenomena such as cancer reversion, the existence of cancers without mutations, and foreign-body carcinogenesis. In this proof-of-concept paper, we draw on criminology to propose that cancer may alternatively be conceptualized as a relational problem: Although a cell's genetics is essential, the influence of its interaction with other cells is equally important in determining its phenotype. We create a simple agent-based network model of interactions among normal and cancer cells to demonstrate this idea. We find that both high mutation rates and low levels of connectivity among cells can promote oncogenesis. Viewing cancer as a breakdown in communication networks among cells in a tissue complements the gene-centric paradigm nicely and provides a novel perspective for understanding and treating cancer.
Collapse
Affiliation(s)
- Anuraag Bukkuri
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| | - Frederick R Adler
- School of Biological Sciences, University of Utah, Salt Lake City, UT, United States; Department of Mathematics, University of Utah, Salt Lake City, UT, United States; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
25
|
Alkafaas SS, Khedr SA, ElKafas SS, Hafez W, Loutfy SA, Sakran M, Janković N. Targeting JNK kinase inhibitors via molecular docking: A promising strategy to address tumorigenesis and drug resistance. Bioorg Chem 2024; 153:107776. [PMID: 39276490 DOI: 10.1016/j.bioorg.2024.107776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/13/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024]
Abstract
Among members of the mitogen-activated protein kinase (MAPK) family, c-Jun N-terminal kinases (JNKs) are vital for cellular responses to stress, inflammation, and apoptosis. Recent advances have highlighted their important implications in cancer biology, where dysregulated JNK signalling plays a role in the growth, progression, and metastasis of tumors. The present understanding of JNK kinase and its function in the etiology of cancer is summarized in this review. By modifying a number of downstream targets, such as transcription factors, apoptotic regulators, and cell cycle proteins, JNKs exert diverse effects on cancer cells. Apoptosis avoidance, cell survival, and proliferation are all promoted by abnormal JNK activation in many types of cancer, which leads to tumor growth and resistance to treatment. JNKs also affect the tumour microenvironment by controlling the generation of inflammatory cytokines, angiogenesis, and immune cell activity. However, challenges remain in deciphering the context-specific roles of JNK isoforms and their intricate crosstalk with other signalling pathways within the complex tumor environment. Further research is warranted to delineate the precise mechanisms underlying JNK-mediated tumorigenesis and to develop tailored therapeutic strategies targeting JNK signalling to improve cancer management. The review emphasizes the role of JNK kinases in cancer biology, as well as their potential as pharmaceutical targets for precision oncology therapy and cancer resistance. Also, this review summarizes all the available promising JNK inhibitors that are suggested to promote the responsiveness of cancer cells to cancer treatment.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, 31527, Egypt.
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Sara Samy ElKafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt; Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, Russia
| | - Wael Hafez
- NMC Royal Hospital, 16th St - Khalifa City - SE-4 - Abu Dhabi, United Arab Emirates; Department of Internal Medicine, Medical Research and Clinical Studies Institute, The National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate 12622, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Sakran
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Nenad Janković
- Institute for Information Technologies Kragujevac, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia.
| |
Collapse
|
26
|
Farahani N, Alimohammadi M, Raei M, Nabavi N, Aref AR, Hushmandi K, Daneshi S, Razzaghi A, Taheriazam A, Hashemi M. Exploring the dual role of endoplasmic reticulum stress in urological cancers: Implications for tumor progression and cell death interactions. J Cell Commun Signal 2024; 18:e12054. [PMID: 39691874 PMCID: PMC11647052 DOI: 10.1002/ccs3.12054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 12/19/2024] Open
Abstract
The endoplasmic reticulum (ER) is crucial for maintaining calcium balance, lipid biosynthesis, and protein folding. Disruptions in ER homeostasis, often due to the accumulation of misfolded or unfolded proteins, lead to ER stress, which plays a significant role in various diseases, especially cancer. Urological cancers, which account for high male mortality worldwide, pose a persistent challenge due to their incurability and tendency to develop drug resistance. Among the numerous dysregulated biological mechanisms, ER stress is a key factor in the progression and treatment response of these cancers. This review highlights the dual role of aberrant ER stress activation in urologic cancers, affecting both tumor growth and therapeutic outcomes. While ER stress can support tumor growth through pro-survival autophagy, it primarily inhibits cancer progression via apoptosis and pro-death autophagy. Interestingly, ER stress can paradoxically aid cancer progression through mechanisms such as exosome-mediated immune evasion. Additionally, the review examines how pharmacological interventions, particularly with phytochemicals, can stimulate ER stress-mediated tumor suppression. Key regulators, including PERK, IRE1α, and ATF6, are discussed for their roles in upregulating CHOP levels and triggering apoptosis. In conclusion, a deeper understanding of ER stress in urological cancers not only clarifies the complex interactions between cellular stress and cancer progression but also provides new opportunities for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mina Alimohammadi
- Department of ImmunologySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Mehdi Raei
- Health Research CenterLife Style InstituteBaqiyatallah University of Medical SciencesTehranIran
| | | | - Amir Reza Aref
- Department of SurgeryMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kiavash Hushmandi
- Nephrology and Urology Research CenterClinical Sciences InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - Salman Daneshi
- Department of Public HealthSchool of HealthJiroft University of Medical SciencesJiroftIran
| | - Alireza Razzaghi
- Social Determinants of Health Research CenterResearch Institute for Prevention of Non‐Communicable DiseasesQazvin University of Medical SciencesQazvinIran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of OrthopedicsFaculty of MedicineTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| |
Collapse
|
27
|
Wang X, Guo Y, Lin P, Yu M, Song S, Xu W, Kong D, Wang Y, Zhang Y, Lu F, Xie Q, Ma X. Nuclear receptor E75/NR1D2 promotes tumor malignant transformation by integrating Hippo and Notch pathways. EMBO J 2024; 43:6336-6363. [PMID: 39516282 PMCID: PMC11649922 DOI: 10.1038/s44318-024-00290-3] [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: 02/21/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Hormone therapy resistance and the ensuing aggressive tumor progression present a significant clinical challenge. However, the mechanisms underlying the induction of tumor malignancy upon inhibition of steroid hormone signaling remain poorly understood. Here, we demonstrate that Drosophila malignant epithelial tumors show a similar reduction in ecdysone signaling, the main steroid hormone pathway. Our analysis of ecdysone-induced downstream targets reveals that overexpression of the nuclear receptor E75, particularly facilitates the malignant transformation of benign tumors. Genome-wide DNA binding profiles and biochemistry data reveal that E75 not only binds to the transcription factors of both Hippo and Notch pathways, but also exhibits widespread co-binding to their target genes, thus contributing to tumor malignancy. We further validated these findings by demonstrating that depletion of NR1D2, the mammalian homolog of E75, inhibits the activation of Hippo and Notch target genes, impeding glioblastoma progression. Together, our study unveils a novel mechanism by which hormone inhibition promotes tumor malignancy, and describes an evolutionarily conserved role of the oncogene E75/NR1D2 in integration of Hippo and Notch pathway activity during tumor progression.
Collapse
Affiliation(s)
- Xianping Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Yifan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Peng Lin
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Min Yu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sha Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wenyan Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Du Kong
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Yin Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute of City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Yanxiao Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Fei Lu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Qi Xie
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
28
|
Graham S, Dmitrieva M, Vendramini-Costa DB, Francescone R, Trujillo MA, Cukierman E, Wood LD. From precursor to cancer: decoding the intrinsic and extrinsic pathways of pancreatic intraepithelial neoplasia progression. Carcinogenesis 2024; 45:801-816. [PMID: 39514554 DOI: 10.1093/carcin/bgae064] [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: 08/09/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
This review explores the progression of pancreatic intraepithelial neoplasia (PanIN) to pancreatic ductal adenocarcinoma through a dual lens of intrinsic molecular alterations and extrinsic microenvironmental influences. PanIN development begins with Kirsten rat sarcoma viral oncogene (KRAS) mutations driving PanIN initiation. Key additional mutations in cyclin-dependent kinase inhibitor 2A (CDKN2A), tumor protein p53 (TP53), and mothers against decapentaplegic homolog 4 (SMAD4) disrupt cell cycle control and genomic stability, crucial for PanIN progression from low-grade to high-grade dysplasia. Additional molecular alterations in neoplastic cells, including epigenetic modifications and chromosomal alterations, can further contribute to neoplastic progression. In parallel with these alterations in neoplastic cells, the microenvironment, including fibroblast activation, extracellular matrix remodeling, and immune modulation, plays a pivotal role in PanIN initiation and progression. Crosstalk between neoplastic and stromal cells influences nutrient support and immune evasion, contributing to tumor development, growth, and survival. This review underscores the intricate interplay between cell-intrinsic molecular drivers and cell-extrinsic microenvironmental factors, shaping PanIN predisposition, initiation, and progression. Future research aims to unravel these interactions to develop targeted therapeutic strategies and early detection techniques, aiming to alleviate the severe impact of pancreatic cancer by addressing both genetic predispositions and environmental influences.
Collapse
Affiliation(s)
- Sarah Graham
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Mariia Dmitrieva
- Cancer Signaling & Microenvironment Program, M&C Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA 19111, United States
| | - Debora Barbosa Vendramini-Costa
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, United States
| | - Ralph Francescone
- Henry Ford Pancreatic Cancer Center, Henry Ford Health, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, United States
| | - Maria A Trujillo
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, M&C Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Lewis Katz School of Medicine, Temple Health, Philadelphia, PA 19111, United States
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD 21231, United States
| |
Collapse
|
29
|
Ding G, Ma T, Zhang K, Chen G, Shen J, Zhang S, Li K, Zhao C, Wang F, Sun J, Wang J. A pan-cancer analysis of the role of WDFY2 in human tumors. Biotechnol Genet Eng Rev 2024; 40:1456-1471. [PMID: 36971139 DOI: 10.1080/02648725.2023.2194077] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
WDFY2 is a protein that may provide valuable insights into the mechanisms underlying human tumors and aid in the development of novel therapies. Despite its potential importance, the role of WDFY2 in pan-cancer has not been systematically investigated. In this study, we comprehensively explored the expression pattern and function of WDFY2 across 33 cancers using various databases, including TCGA, CPTAC and GEO datasets. Our results indicate that WDFY2 is downregulated in most cancer types, including BRCA, KIRP, KICH, LUAD, KIRC, PCPG, PRAD, THCA, ACC, OV, TGCT and UCS, while it is upregulated in CESC, CHOL, COAD, HNSC, LUSC, READ, STAD and UCEC. Prognostic analyses showed that higher levels of WDFY2 were associated with worse disease outcomes in ACC, BLCA, COAD, READ, SARC, MESO and OV. WDFY2 mutations were most frequent in colorectal cancer but were not associated with disease prognosis. We also found that WDFY2 expression correlated with monocyte infiltration status in SKCM, endothelial cell infiltration in COAD, KIRC, MESO, OV and THCA, and cancer-associated fibroblast infiltration in COAD, LUAD and OV. Additionally, functional enrichment analysis revealed that WDFY2 is involved in metabolism. Overall, our comprehensive analysis sheds light on the role of WDFY2 in various cancers, providing a better understanding of its role in tumorigenesis.
Collapse
Affiliation(s)
- Guanxiong Ding
- Department of Urology, Huashan Hospital, Fudan University
| | - Tianyan Ma
- Department of Nursing, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke Zhang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Gang Chen
- Department of Vascular Interventional, Binzhou Medical University Hospital, Binzhou, China
| | - Jingjing Shen
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Sijie Zhang
- Department of Integrated Sciences, University of British Columbia, Vancouver, Canada
| | - Kai Li
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Chunchun Zhao
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Fei Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jian Sun
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jianqing Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| |
Collapse
|
30
|
Aqil A, Li Y, Wang Z, Islam S, Russell M, Kallak TK, Saitou M, Gokcumen O, Masuda N. Switch-like Gene Expression Modulates Disease Susceptibility. RESEARCH SQUARE 2024:rs.3.rs-4974188. [PMID: 39315271 PMCID: PMC11419265 DOI: 10.21203/rs.3.rs-4974188/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
A fundamental challenge in biomedicine is understanding the mechanisms predisposing individuals to disease. While previous research has suggested that switch-like gene expression is crucial in driving biological variation and disease susceptibility, a systematic analysis across multiple tissues is still lacking. By analyzing transcriptomes from 943 individuals across 27 tissues, we identified 1,013 switch-like genes. We found that only 31 (3.1%) of these genes exhibit switch-like behavior across all tissues. These universally switch-like genes appear to be genetically driven, with large exonic genomic structural variants explaining five (~18%) of them. The remaining switch-like genes exhibit tissue-specific expression patterns. Notably, tissue-specific switch-like genes tend to be switched on or off in unison within individuals, likely under the influence of tissue-specific master regulators, including hormonal signals. Among our most significant findings, we identified hundreds of concordantly switched-off genes in the stomach and vagina that are linked to gastric cancer (41-fold, p<10-4) and vaginal atrophy (44-fold, p<10-4), respectively. Experimental analysis of vaginal tissues revealed that low systemic levels of estrogen lead to a significant reduction in both the epithelial thickness and the expression of the switch-like gene ALOX12. We propose a model wherein the switching off of driver genes in basal and parabasal epithelium suppresses cell proliferation therein, leading to epithelial thinning and, therefore, vaginal atrophy. Our findings underscore the significant biomedical implications of switch-like gene expression and lay the groundwork for potential diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Alber Aqil
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Yanyan Li
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhiliang Wang
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Saiful Islam
- Institute for Artificial Intelligence and Data Science, State University of New York at Buffalo, Buffalo, NY, USA
| | - Madison Russell
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
| | | | - Marie Saitou
- Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
| | - Omer Gokcumen
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Naoki Masuda
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
- Institute for Artificial Intelligence and Data Science, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
31
|
Stanescu LS, Ghemigian A, Ciobica ML, Nistor C, Ciuche A, Radu AM, Sandru F, Carsote M. Thyroid Malignancy and Cutaneous Lichen Amyloidosis: Key Points Amid RET Pathogenic Variants in Medullary Thyroid Cancer/Multiple Endocrine Neoplasia Type 2 (MEN2). Int J Mol Sci 2024; 25:9765. [PMID: 39337252 PMCID: PMC11431960 DOI: 10.3390/ijms25189765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
We aimed to provide an updated narrative review with respect to the RET pathogenic variants and their implications at the clinical and molecular level in the diagnosis of medullary thyroid cancer (MTC)/multiple endocrine neoplasia (MEN) type 2, particularly with respect to the presence of cutaneous lichen amyloidosis (CLA). We searched English-language, in extenso original articles with no timeline nor study design restriction that were published on PubMed. A traditional interplay stands for CLA and MTC in MEN2 (not MEN3) confirmation. While the connection has been reported for more than three decades, there is still a large gap in understanding and addressing it. The majority of patients with MEN2A-CLA have RET pathogenic variants at codon 634; hence, it suggests an involvement of this specific cysteine residue in both disorders (most data agree that one-third of C634-positive subjects have CLA, but the ranges are between 9% and 50%). Females seem more prone to MEN2-CLA than males. Non-C634 germline RET pathogenic variants included (at a low level of statistical evidence) the following: RET V804M mutation in exon 14 for MTC-CLA (CLA at upper back); RET S891A mutation in exon 15 binding OSMR variant G513D (familial MTC and CLA comprising the lower legs to thighs, upper back, shoulders, arms, and forearms); and C611Y (CLA at interscapular region), respectively. Typically, CLA is detected at an early age (from childhood until young adulthood) before the actual MTC identification unless RET screening protocols are already applied. The time frame between CLA diagnosis and the identification of RET pathogenic variants was between 5 and 60 years according to one study. The same RET mutation in one family is not necessarily associated with the same CLA presentation. In MTC/MEN2 subjects, the most affected CLA area was the scapular region of the upper back. Alternatively, another hypothesis highlighted the fact that CLA is secondary to long-term prurit/notalgia paresthetica (NP) in MTC/MEN2. OSMR p. G513D may play a role in modifying the evolutionary processes of CLA in subjects co-harboring RET mutations (further studies are necessary to sustain this aspect). Awareness in CLA-positive patients is essential, including the decision of RET testing in selected cases.
Collapse
Affiliation(s)
- Laura-Semonia Stanescu
- PhD Doctoral School, "Carol Davila" University of Medicine and Pharmacy, 0505474 Bucharest, Romania
- Department of Clinical Endocrinology V, C.I. Parhon National Institute of Endocrinology, 011863 Bucharest, Romania
| | - Adina Ghemigian
- Department of Clinical Endocrinology V, C.I. Parhon National Institute of Endocrinology, 011863 Bucharest, Romania
- Department of Endocrinology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihai-Lucian Ciobica
- Department of Internal Medicine and Gastroenterology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Internal Medicine I and Rheumatology, "Dr. Carol Davila" Central Military University Emergency Hospital, 010825 Bucharest, Romania
| | - Claudiu Nistor
- Department 4-Cardio-Thoracic Pathology, Thoracic Surgery II Discipline, "Carol Davila" University of Medicine and Pharmacy, 0505474 Bucharest, Romania
- Thoracic Surgery Department, "Dr. Carol Davila" Central Emergency University Military Hospital, 010825 Bucharest, Romania
| | - Adrian Ciuche
- Department 4-Cardio-Thoracic Pathology, Thoracic Surgery II Discipline, "Carol Davila" University of Medicine and Pharmacy, 0505474 Bucharest, Romania
- Thoracic Surgery Department, "Dr. Carol Davila" Central Emergency University Military Hospital, 010825 Bucharest, Romania
| | - Andreea-Maria Radu
- Department of Dermatovenerology, Elias University Emergency Hospital, 011461 Bucharest, Romania
- Department of Dermatovenerology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Florica Sandru
- Department of Dermatovenerology, Elias University Emergency Hospital, 011461 Bucharest, Romania
- Department of Dermatovenerology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mara Carsote
- Department of Clinical Endocrinology V, C.I. Parhon National Institute of Endocrinology, 011863 Bucharest, Romania
- Department of Endocrinology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
32
|
Ke R, Kumar S, Singh SK, Rana A, Rana B. Molecular insights into the role of mixed lineage kinase 3 in cancer hallmarks. Biochim Biophys Acta Rev Cancer 2024; 1879:189157. [PMID: 39032538 DOI: 10.1016/j.bbcan.2024.189157] [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: 12/22/2023] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Mixed-lineage kinase 3 (MLK3) is a serine/threonine kinase of the MAPK Kinase kinase (MAP3K) family that plays critical roles in various biological processes, including cancer. Upon activation, MLK3 differentially activates downstream MAPKs, such as JNK, p38, and ERK. In addition, it regulates various non-canonical signaling pathways, such as β-catenin, AMPK, Pin1, and PAK1, to regulate cell proliferation, apoptosis, invasion, and metastasis. Recent studies have also uncovered other potentially diverse roles of MLK3 in malignancy, which include metabolic reprogramming, cancer-associated inflammation, and evasion of cancer-related immune surveillance. The role of MLK3 in cancer is complex and cancer-specific, and an understanding of its function at the molecular level aligned specifically with the cancer hallmarks will have profound therapeutic implications for diagnosing and treating MLK3-dependent cancers. This review summarizes the current knowledge about the effect of MLK3 on the hallmarks of cancer, providing insights into its potential as a promising anticancer drug target.
Collapse
Affiliation(s)
- Rong Ke
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Hospital and Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
33
|
Aqil A, Li Y, Wang Z, Islam S, Russell M, Kallak TK, Saitou M, Gokcumen O, Masuda N. Switch-like Gene Expression Modulates Disease Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.24.609537. [PMID: 39229158 PMCID: PMC11370615 DOI: 10.1101/2024.08.24.609537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
A fundamental challenge in biomedicine is understanding the mechanisms predisposing individuals to disease. While previous research has suggested that switch-like gene expression is crucial in driving biological variation and disease susceptibility, a systematic analysis across multiple tissues is still lacking. By analyzing transcriptomes from 943 individuals across 27 tissues, we identified 1,013 switch-like genes. We found that only 31 (3.1%) of these genes exhibit switch-like behavior across all tissues. These universally switch-like genes appear to be genetically driven, with large exonic genomic structural variants explaining five (~18%) of them. The remaining switch-like genes exhibit tissue-specific expression patterns. Notably, tissue-specific switch-like genes tend to be switched on or off in unison within individuals, likely under the influence of tissue-specific master regulators, including hormonal signals. Among our most significant findings, we identified hundreds of concordantly switched-off genes in the stomach and vagina that are linked to gastric cancer (41-fold, p<10-4) and vaginal atrophy (44-fold, p<10-4), respectively. Experimental analysis of vaginal tissues revealed that low systemic levels of estrogen lead to a significant reduction in both the epithelial thickness and the expression of the switch-like gene ALOX12. We propose a model wherein the switching off of driver genes in basal and parabasal epithelium suppresses cell proliferation therein, leading to epithelial thinning and, therefore, vaginal atrophy. Our findings underscore the significant biomedical implications of switch-like gene expression and lay the groundwork for potential diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Alber Aqil
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Yanyan Li
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhiliang Wang
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Saiful Islam
- Institute for Artificial Intelligence and Data Science, State University of New York at Buffalo, Buffalo, NY, USA
| | - Madison Russell
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
| | | | - Marie Saitou
- Faculty of Biosciences, Norwegian University of Life Sciences, Aas, Norway
| | - Omer Gokcumen
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Naoki Masuda
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY, USA
- Institute for Artificial Intelligence and Data Science, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
34
|
Neagu AI, Poalelungi DG, Fulga A, Neagu M, Fulga I, Nechita A. Enhanced Immunohistochemistry Interpretation with a Machine Learning-Based Expert System. Diagnostics (Basel) 2024; 14:1853. [PMID: 39272638 PMCID: PMC11394116 DOI: 10.3390/diagnostics14171853] [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: 06/13/2024] [Revised: 07/26/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND In recent decades, machine-learning (ML) technologies have advanced the management of high-dimensional and complex cancer data by developing reliable and user-friendly automated diagnostic tools for clinical applications. Immunohistochemistry (IHC) is an essential staining method that enables the identification of cellular origins by analyzing the expression of specific antigens within tissue samples. The aim of this study was to identify a model that could predict histopathological diagnoses based on specific immunohistochemical markers. METHODS The XGBoost learning model was applied, where the input variable (target variable) was the histopathological diagnosis and the predictors (independent variables influencing the target variable) were the immunohistochemical markers. RESULTS Our study demonstrated a precision rate of 85.97% within the dataset, indicating a high level of performance and suggesting that the model is generally reliable in producing accurate predictions. CONCLUSIONS This study demonstrated the feasibility and clinical efficacy of utilizing the probabilistic decision tree algorithm to differentiate tumor diagnoses according to immunohistochemistry profiles.
Collapse
Affiliation(s)
- Anca Iulia Neagu
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania
- Saint John Clinical Emergency Hospital for Children, 800487 Galati, Romania
| | - Diana Gina Poalelungi
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Ana Fulga
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Marius Neagu
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Iuliu Fulga
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania
- Saint Apostle Andrew Emergency County Clinical Hospital, 177 Brailei St., 800578 Galati, Romania
| | - Aurel Nechita
- Faculty of Medicine and Pharmacy, Dunarea de Jos University of Galati, 35 AI Cuza St., 800010 Galati, Romania
- Saint John Clinical Emergency Hospital for Children, 800487 Galati, Romania
| |
Collapse
|
35
|
Tomuleasa C, Tigu AB, Munteanu R, Moldovan CS, Kegyes D, Onaciu A, Gulei D, Ghiaur G, Einsele H, Croce CM. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct Target Ther 2024; 9:201. [PMID: 39138146 PMCID: PMC11323831 DOI: 10.1038/s41392-024-01899-w] [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: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Receptor tyrosine kinases (RTKs), a category of transmembrane receptors, have gained significant clinical attention in oncology due to their central role in cancer pathogenesis. Genetic alterations, including mutations, amplifications, and overexpression of certain RTKs, are critical in creating environments conducive to tumor development. Following their discovery, extensive research has revealed how RTK dysregulation contributes to oncogenesis, with many cancer subtypes showing dependency on aberrant RTK signaling for their proliferation, survival and progression. These findings paved the way for targeted therapies that aim to inhibit crucial biological pathways in cancer. As a result, RTKs have emerged as primary targets in anticancer therapeutic development. Over the past two decades, this has led to the synthesis and clinical validation of numerous small molecule tyrosine kinase inhibitors (TKIs), now effectively utilized in treating various cancer types. In this manuscript we aim to provide a comprehensive understanding of the RTKs in the context of cancer. We explored the various alterations and overexpression of specific receptors across different malignancies, with special attention dedicated to the examination of current RTK inhibitors, highlighting their role as potential targeted therapies. By integrating the latest research findings and clinical evidence, we seek to elucidate the pivotal role of RTKs in cancer biology and the therapeutic efficacy of RTK inhibition with promising treatment outcomes.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania.
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Cristian-Silviu Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
36
|
Baráti L, Maász A, Mikó A, Bércesi É, Kalbani SA, Bene J, Kovács S, Mangel L, Hadzsiev K. Molecular genetic investigation of hereditary breast and ovarian cancer patients in the Southern Transdanubian region: widening the mutation spectrum and searching for new pathogenic variants using next-generation methods. Pathol Oncol Res 2024; 30:1611813. [PMID: 39148954 PMCID: PMC11324426 DOI: 10.3389/pore.2024.1611813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Hereditary breast and ovarian cancer is a well-known genetic condition, inherited mainly in an autosomal dominant way, which elevates the risk of developing malignancies at a young age in heterozygous carriers. Advances in new generation sequencing have enabled medical professionals to determine whether a patient is harbouring mutations in moderate- or high penetrance susceptibility genes. We conducted a retrospective analysis among 275 patients who underwent genetic counselling and multigene panel testing for hereditary breast and ovarian cancer syndrome in our department. From these patients 74.5% (205/275) were affected by some type of malignancy, while the remaining 25.5% (70/275) had a positive family history of different cancers, suggesting a genetic predisposition. These tests confirmed a genetic variant in 29.8% and 28.6% of these patient groups respectively. The results also mirrored our general knowledge concerning the genetic background of hereditary breast and ovarian cancer, as variants in either one of the BRCA1 and BRCA2 genes proved to be the most common cause among our patients with 41.5%. Our test also detected a novel mutation in the CDH1 gene and three patients with double heterozygosity in two different susceptibility genes. This study demonstrates the relevance of genetic counselling and non-BRCA gene sequencing among cancer patients and patients who fulfil the criteria for genetic testing, while also providing important details about the genetic profile of Hungarian patients.
Collapse
Affiliation(s)
- László Baráti
- Department of Medical Genetics, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
| | - Anita Maász
- Department of Medical Genetics, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
| | - Alexandra Mikó
- Department of Medical Genetics, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Éva Bércesi
- Department of Oncotherapy, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
| | - Sultan Al Kalbani
- Department of Medical Genetics, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
| | - Judit Bene
- Department of Medical Genetics, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
| | - Sebestyén Kovács
- Urology Clinic, Clinical Centre, University of Pécs, Pécs, Hungary
| | - László Mangel
- Department of Oncotherapy, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
| | - Kinga Hadzsiev
- Department of Medical Genetics, Clinical Centre, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
37
|
Herbert A. Osteogenesis imperfecta type 10 and the cellular scaffolds underlying common immunological diseases. Genes Immun 2024; 25:265-276. [PMID: 38811682 DOI: 10.1038/s41435-024-00277-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
Osteogenesis imperfecta type 10 (OI10) is caused by loss of function codon variants in the gene SERPINH1 that encodes heat shock protein 47 (HSP47), rather than in a gene specifying bone formation. The HSP47 variants disrupt the folding of both collagen and the endonuclease IRE1α (inositol-requiring enzyme 1α) that splices X-Box Binding Protein 1 (XBP1) mRNA. Besides impairing bone development, variants likely affect osteoclast differentiation. Three distinct biochemical scaffold play key roles in the differentiation and regulated cell death of osteoclasts. These scaffolds consist of non-templated protein modifications, ordered lipid arrays, and protein filaments. The scaffold components are specified genetically, but assemble in response to extracellular perturbagens, pathogens, and left-handed Z-RNA helices encoded genomically by flipons. The outcomes depend on interactions between RIPK1, RIPK3, TRIF, and ZBP1 through short interaction motifs called RHIMs. The causal HSP47 nonsynonymous substitutions occur in a novel variant leucine repeat region (vLRR) that are distantly related to RHIMs. Other vLRR protein variants are causal for a variety of different mendelian diseases. The same scaffolds that drive mendelian pathology are associated with many other complex disease outcomes. Their assembly is triggered dynamically by flipons and other context-specific switches rather than by causal, mendelian, codon variants.
Collapse
Affiliation(s)
- Alan Herbert
- InsideOutBio, 42 8th Street, Charlestown, MA, USA.
| |
Collapse
|
38
|
Milan A, Mioc M, Mioc A, Gogulescu A, Mardale G, Avram Ș, Maksimović T, Mara B, Șoica C. Cytotoxic Potential of Betulinic Acid Fatty Esters and Their Liposomal Formulations: Targeting Breast, Colon, and Lung Cancer Cell Lines. Molecules 2024; 29:3399. [PMID: 39064977 PMCID: PMC11279467 DOI: 10.3390/molecules29143399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Betulinic acid is a lupane-type pentacyclic triterpene mostly found in birch bark and thoroughly explored for its wide range of pharmacological activities. Despite its impressive biological potential, its low bioavailability has challenged many researchers to develop different formulations for achieving better in vitro and in vivo effects. We previously reported the synthesis of fatty acid esters of betulinic acid using butyric, stearic, and palmitic acids (But-BA, St-BA, and Pal-BA) and included them in surfaced-modified liposomes (But-BA-Lip, St-BA-Lip, Pal-BA-Lip). In the current study, we evaluated the cytotoxic effects of both fatty acid esters and their respective liposomal formulations against MCF-7, HT-29, and NCI-H460 cell line. The cytotoxic assessment of BA derivatives revealed that both the fatty esters and their liposomal formulations acted as cytotoxic agents in a dose- and time-dependent manner. But-BA-Lip exerted stronger cytotoxic effects than the parent compound, BA and its liposomal formulation, and even stronger effects than 5-FU against HT-29 cells (IC50 of 30.57 μM) and NCI-H460 cells (IC50 of 30.74 μM). BA's fatty esters and their respective liposomal formulations facilitated apoptosis in cancer cells by inducing nuclear morphological changes and increasing caspase-3/-7 activity. The HET-CAM assay proved that none of the tested compounds induced any irritative effect, suggesting that they can be used safely for local applications.
Collapse
Affiliation(s)
- Andreea Milan
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Marius Mioc
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Alexandra Mioc
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Armand Gogulescu
- Faculty of Medicine, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Gabriel Mardale
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Ștefana Avram
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| | - Tamara Maksimović
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
| | - Bogdan Mara
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Institute of Chemistry Coriolan Drăgulescu, 24 Mihai Viteazu Ave, 300223 Timișoara, Romania
| | - Codruța Șoica
- Faculty of Pharmacy, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania; (A.M.); (M.M.); (A.M.); (G.M.); (Ș.A.); (T.M.); (B.M.); (C.Ș.)
- Research Centre for Pharmaco-Toxicological Evaluation, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timișoara, Romania
| |
Collapse
|
39
|
Colussi DM, Stathopulos PB. The mitochondrial calcium uniporter: Balancing tumourigenic and anti-tumourigenic responses. J Physiol 2024; 602:3315-3339. [PMID: 38857425 DOI: 10.1113/jp285515] [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: 12/26/2023] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
Increased malignancy and poor treatability associated with solid tumour cancers have commonly been attributed to mitochondrial calcium (Ca2+) dysregulation. The mitochondrial Ca2+ uniporter complex (mtCU) is the predominant mode of Ca2+ uptake into the mitochondrial matrix. The main components of mtCU are the pore-forming mitochondrial Ca2+ uniporter (MCU) subunit, MCU dominant-negative beta (MCUb) subunit, essential MCU regulator (EMRE) and the gatekeeping mitochondrial Ca2+ uptake 1 and 2 (MICU1 and MICU2) proteins. In this review, we describe mtCU-mediated mitochondrial Ca2+ dysregulation in solid tumour cancer types, finding enhanced mtCU activity observed in colorectal cancer, breast cancer, oral squamous cell carcinoma, pancreatic cancer, hepatocellular carcinoma and embryonal rhabdomyosarcoma. By contrast, decreased mtCU activity is associated with melanoma, whereas the nature of mtCU dysregulation remains unclear in glioblastoma. Furthermore, we show that numerous polymorphisms associated with cancer may alter phosphorylation sites on the pore forming MCU and MCUb subunits, which cluster at interfaces with EMRE. We highlight downstream/upstream biomolecular modulators of MCU and MCUb that alter mtCU-mediated mitochondrial Ca2+ uptake and may be used as biomarkers or to aid in the development of novel cancer therapeutics. Additionally, we provide an overview of the current small molecule inhibitors of mtCU that interact with the Asp residue of the critical Asp-Ile-Met-Glu motif or through other allosteric regulatory mechanisms to block Ca2+ permeation. Finally, we describe the relationship between MCU- and MCUb-mediating microRNAs and mitochondrial Ca2+ uptake that should be considered in the discovery of new treatment approaches for cancer.
Collapse
Affiliation(s)
- Danielle M Colussi
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
40
|
Zhou Z, Wang J, Wang J, Yang S, Wang R, Zhang G, Li Z, Shi R, Wang Z, Lu Q. Deciphering the tumor immune microenvironment from a multidimensional omics perspective: insight into next-generation CAR-T cell immunotherapy and beyond. Mol Cancer 2024; 23:131. [PMID: 38918817 PMCID: PMC11201788 DOI: 10.1186/s12943-024-02047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Tumor immune microenvironment (TIME) consists of intra-tumor immunological components and plays a significant role in tumor initiation, progression, metastasis, and response to therapy. Chimeric antigen receptor (CAR)-T cell immunotherapy has revolutionized the cancer treatment paradigm. Although CAR-T cell immunotherapy has emerged as a successful treatment for hematologic malignancies, it remains a conundrum for solid tumors. The heterogeneity of TIME is responsible for poor outcomes in CAR-T cell immunotherapy against solid tumors. The advancement of highly sophisticated technology enhances our exploration in TIME from a multi-omics perspective. In the era of machine learning, multi-omics studies could reveal the characteristics of TIME and its immune resistance mechanism. Therefore, the clinical efficacy of CAR-T cell immunotherapy in solid tumors could be further improved with strategies that target unfavorable conditions in TIME. Herein, this review seeks to investigate the factors influencing TIME formation and propose strategies for improving the effectiveness of CAR-T cell immunotherapy through a multi-omics perspective, with the ultimate goal of developing personalized therapeutic approaches.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiahui Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Nephrology, Union Medical College Hospital, Chinese Academy of Medical Sciences, PekingBeijing, 100730, China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shuai Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruizhi Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
41
|
Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, Lin D, Wu C. Tumor initiation and early tumorigenesis: molecular mechanisms and interventional targets. Signal Transduct Target Ther 2024; 9:149. [PMID: 38890350 PMCID: PMC11189549 DOI: 10.1038/s41392-024-01848-7] [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: 01/01/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 06/20/2024] Open
Abstract
Tumorigenesis is a multistep process, with oncogenic mutations in a normal cell conferring clonal advantage as the initial event. However, despite pervasive somatic mutations and clonal expansion in normal tissues, their transformation into cancer remains a rare event, indicating the presence of additional driver events for progression to an irreversible, highly heterogeneous, and invasive lesion. Recently, researchers are emphasizing the mechanisms of environmental tumor risk factors and epigenetic alterations that are profoundly influencing early clonal expansion and malignant evolution, independently of inducing mutations. Additionally, clonal evolution in tumorigenesis reflects a multifaceted interplay between cell-intrinsic identities and various cell-extrinsic factors that exert selective pressures to either restrain uncontrolled proliferation or allow specific clones to progress into tumors. However, the mechanisms by which driver events induce both intrinsic cellular competency and remodel environmental stress to facilitate malignant transformation are not fully understood. In this review, we summarize the genetic, epigenetic, and external driver events, and their effects on the co-evolution of the transformed cells and their ecosystem during tumor initiation and early malignant evolution. A deeper understanding of the earliest molecular events holds promise for translational applications, predicting individuals at high-risk of tumor and developing strategies to intercept malignant transformation.
Collapse
Affiliation(s)
- Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyi Xiao
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yonglin Yi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyu Wang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Lingxuan Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Changping Laboratory, 100021, Beijing, China
| | - Yanrong Shen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- CAMS Oxford Institute, Chinese Academy of Medical Sciences, 100006, Beijing, China.
| |
Collapse
|
42
|
Guo D, Wang Y, Chen J, Liu X. Integration of multi-omics data for survival prediction of lung adenocarcinoma. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 250:108192. [PMID: 38701699 DOI: 10.1016/j.cmpb.2024.108192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/08/2024] [Accepted: 04/20/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND AND OBJECTIVE The morbidity of lung adenocarcinoma (LUAD) has been increasing year by year and the prognosis is poor. This has prompted researchers to study the survival of LUAD patients to ensure that patients can be cured in time or survive after appropriate treatment. There is still no fully valid model that can be applied to clinical practice. METHODS We introduced struc2vec-based multi-omics data integration (SBMOI), which could integrate gene expression, somatic mutations and clinical data to construct mutation gene vectors representing LUAD patient features. Based on the patient features, the random survival forest (RSF) model was used to predict the long- and short-term survival of LUAD patients. To further demonstrate the superiority of SBMOI, we simultaneously replaced scale-free gene co-expression network (FCN) with a protein-protein interaction (PPI) network and a significant co-expression network (SCN) to compare accuracy in predicting LUAD patient survival under the same conditions. RESULTS Our results suggested that compared with SCN and PPI network, the FCN based SBMOI combined with RSF model had better performance in long- and short-term survival prediction tasks for LUAD patients. The AUC of 1-year, 5-year, and 10-year survival in the validation dataset were 0.791, 0.825, and 0.917, respectively. CONCLUSIONS This study provided a powerful network-based method to multi-omics data integration. SBMOI combined with RSF successfully predicted long- and short-term survival of LUAD patients, especially with high accuracy on long-term survival. Besides, SBMOI algorithm has the potential to combine with other machine learning models to complete clustering or stratificational tasks, and being applied to other diseases.
Collapse
Affiliation(s)
- Dingjie Guo
- Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yixian Wang
- Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Jing Chen
- Academy for Advanced Interdisciplinary Studies, Northeast Normal University, Changchun, 130024, China
| | - Xin Liu
- Epidemiology and Statistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
43
|
Doré S, Ali M, Sorin M, McDowell SAC, Desharnais L, Breton V, Yu MW, Arabzadeh A, Ryan MI, Milette S, Quail DF, Walsh LA. Exploring the prognostic significance of arm-level copy number alterations in triple-negative breast cancer. Oncogene 2024; 43:2015-2024. [PMID: 38744952 PMCID: PMC11196216 DOI: 10.1038/s41388-024-03051-y] [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: 07/31/2023] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024]
Abstract
Somatic copy number alterations (SCNAs) are prevalent in cancer and play a significant role in both tumorigenesis and therapeutic resistance. While focal SCNAs have been extensively studied, the impact of larger arm-level SCNAs remains poorly understood. Here, we investigated the association between arm-level SCNAs and overall survival in triple-negative breast cancer (TNBC), an aggressive subtype of breast cancer lacking targeted therapies. We identified frequent arm-level SCNAs, including 21q gain and 7p gain, which correlated with poor overall survival in TNBC patients. Further, we identified the expression of specific genes within these SCNAs associated with survival. Notably, we found that the expression of RIPK4, a gene located on 21q, exhibited a strong correlation with poor overall survival. In functional assays, we demonstrated that targeting Ripk4 in a murine lung metastatic TNBC model significantly reduced tumor burden, improved survival, and increased CD4+ and CD8+ T cell infiltration. RIPK4 enhanced the survival of triple-negative breast cancer cells at secondary sites, thereby facilitating the formation of metastatic lesions. Our findings highlight the significance of arm-level SCNAs in breast cancer progression and identify RIPK4 as a putative driver of TNBC metastasis and immunosuppression.
Collapse
Affiliation(s)
- Samuel Doré
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mariam Ali
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mark Sorin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Sheri A C McDowell
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Lysanne Desharnais
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Valérie Breton
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Miranda W Yu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Azadeh Arabzadeh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Malcolm I Ryan
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Surgery, McGill University Health Center, Montreal, QC, Canada
| | - Simon Milette
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
44
|
Xue Y, Ruan Y, Wang Y, Xiao P, Xu J. Signaling pathways in liver cancer: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:20. [PMID: 38816668 PMCID: PMC11139849 DOI: 10.1186/s43556-024-00184-0] [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: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Liver cancer remains one of the most prevalent malignancies worldwide with high incidence and mortality rates. Due to its subtle onset, liver cancer is commonly diagnosed at a late stage when surgical interventions are no longer feasible. This situation highlights the critical role of systemic treatments, including targeted therapies, in bettering patient outcomes. Despite numerous studies on the mechanisms underlying liver cancer, tyrosine kinase inhibitors (TKIs) are the only widely used clinical inhibitors, represented by sorafenib, whose clinical application is greatly limited by the phenomenon of drug resistance. Here we show an in-depth discussion of the signaling pathways frequently implicated in liver cancer pathogenesis and the inhibitors targeting these pathways under investigation or already in use in the management of advanced liver cancer. We elucidate the oncogenic roles of these pathways in liver cancer especially hepatocellular carcinoma (HCC), as well as the current state of research on inhibitors respectively. Given that TKIs represent the sole class of targeted therapeutics for liver cancer employed in clinical practice, we have particularly focused on TKIs and the mechanisms of the commonly encountered phenomena of its resistance during HCC treatment. This necessitates the imperative development of innovative targeted strategies and the urgency of overcoming the existing limitations. This review endeavors to shed light on the utilization of targeted therapy in advanced liver cancer, with a vision to improve the unsatisfactory prognostic outlook for those patients.
Collapse
Affiliation(s)
- Yangtao Xue
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yeling Ruan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yali Wang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Peng Xiao
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China.
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China.
- Zhejiang University Cancer Center, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
45
|
Zhang X, Celic I, Mitchell H, Stuckert S, Vedula L, Han J. Comprehensive profiling of L1 retrotransposons in mouse. Nucleic Acids Res 2024; 52:5166-5178. [PMID: 38647072 PMCID: PMC11109951 DOI: 10.1093/nar/gkae273] [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: 11/13/2023] [Revised: 03/25/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024] Open
Abstract
L1 elements are retrotransposons currently active in mammals. Although L1s are typically silenced in most normal tissues, elevated L1 expression is associated with a variety of conditions, including cancer, aging, infertility and neurological disease. These associations have raised interest in the mapping of human endogenous de novo L1 insertions, and a variety of methods have been developed for this purpose. Adapting these methods to mouse genomes would allow us to monitor endogenous in vivo L1 activity in controlled, experimental conditions using mouse disease models. Here, we use a modified version of transposon insertion profiling, called nanoTIPseq, to selectively enrich young mouse L1s. By linking this amplification step with nanopore sequencing, we identified >95% annotated L1s from C57BL/6 genomic DNA using only 200 000 sequencing reads. In the process, we discovered 82 unannotated L1 insertions from a single C57BL/6 genome. Most of these unannotated L1s were near repetitive sequence and were not found with short-read TIPseq. We used nanoTIPseq on individual mouse breast cancer cells and were able to identify the annotated and unannotated L1s, as well as new insertions specific to individual cells, providing proof of principle for using nanoTIPseq to interrogate retrotransposition activity at the single-cell level in vivo.
Collapse
Affiliation(s)
- Xuanming Zhang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ivana Celic
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hannah Mitchell
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sam Stuckert
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Lalitha Vedula
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jeffrey S Han
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
46
|
Bugallo A, Sánchez M, Fernández-García M, Segurado M. S-phase checkpoint prevents leading strand degradation from strand-associated nicks at stalled replication forks. Nucleic Acids Res 2024; 52:5121-5137. [PMID: 38520409 PMCID: PMC11109941 DOI: 10.1093/nar/gkae192] [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: 11/06/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/25/2024] Open
Abstract
The S-phase checkpoint is involved in coupling DNA unwinding with nascent strand synthesis and is critical to maintain replication fork stability in conditions of replicative stress. However, its role in the specific regulation of leading and lagging strands at stalled forks is unclear. By conditionally depleting RNaseH2 and analyzing polymerase usage genome-wide, we examine the enzymology of DNA replication during a single S-phase in the presence of replicative stress and show that there is a differential regulation of lagging and leading strands. In checkpoint proficient cells, lagging strand replication is down-regulated through an Elg1-dependent mechanism. Nevertheless, when checkpoint function is impaired we observe a defect specifically at the leading strand, which was partially dependent on Exo1 activity. Further, our genome-wide mapping of DNA single-strand breaks reveals that strand discontinuities highly accumulate at the leading strand in HU-treated cells, whose dynamics are affected by checkpoint function and Exo1 activity. Our data reveal an unexpected role of Exo1 at the leading strand and support a model of fork stabilization through prevention of unrestrained Exo1-dependent resection of leading strand-associated nicks after fork stalling.
Collapse
Affiliation(s)
- Alberto Bugallo
- Instituto de Biología Funcional y Genómica (CSIC/USAL), Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Mar Sánchez
- Instituto de Biología Funcional y Genómica (CSIC/USAL), Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - María Fernández-García
- Instituto de Biología Funcional y Genómica (CSIC/USAL), Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Mónica Segurado
- Instituto de Biología Funcional y Genómica (CSIC/USAL), Campus Miguel de Unamuno, Salamanca 37007, Spain
- Departamento de Microbiología y Genética (USAL), Campus Miguel de Unamuno, Salamanca 37007, Spain
| |
Collapse
|
47
|
Umapathy VR, Natarajan PM, Swamikannu B. Molecular and Therapeutic Roles of Non-Coding RNAs in Oral Cancer-A Review. Molecules 2024; 29:2402. [PMID: 38792263 PMCID: PMC11123887 DOI: 10.3390/molecules29102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Oral cancer (OC) is among the most common malignancies in the world. Despite advances in therapy, the worst-case scenario for OC remains metastasis, with a 50% survival rate. Therefore, it is critical to comprehend the pathophysiology of the condition and to create diagnostic and treatment plans for OC. The development of high-throughput genome sequencing has revealed that over 90% of the human genome encodes non-coding transcripts, or transcripts that do not code for any proteins. This paper describes the function of these different kinds of non-coding RNAs (ncRNAs) in OC as well as their intriguing therapeutic potential. The onset and development of OC, as well as treatment resistance, are linked to dysregulated ncRNA expression. These ncRNAs' potentially significant roles in diagnosis and prognosis have been suggested by their differing expression in blood or saliva. We have outlined every promising feature of ncRNAs in the treatment of OC in this study.
Collapse
Affiliation(s)
- Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Dr. M.G.R. Educational and Research Institute, Thai Moogambigai Dental College and Hospital, Chennai 600107, Tamil Nadu, India
| | - Prabhu Manickam Natarajan
- Department of Clinical Sciences, Centre of Medical and Bio-Allied Health Sciences and Research Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Bhuminathan Swamikannu
- Department of Prosthodontics, Sree Balaji Dental College and Hospital, Pallikaranai, BIHER, Chennai 600100, Tamil Nadu, India;
| |
Collapse
|
48
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
49
|
Gao S, Huang Q, Wei S, Lv Y, Xie Y, Hao Y. Prognostic nomogram based on pre-treatment HALP score for patients with advanced non-small cell lung cancer. Clinics (Sao Paulo) 2024; 79:100371. [PMID: 38735175 PMCID: PMC11101934 DOI: 10.1016/j.clinsp.2024.100371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 03/27/2024] [Accepted: 04/17/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND To explore the correlation of pre-treatment Hemoglobin-Albumin-Lymphocyte-Platelet (HALP) score with the prognosis of patients with advanced Non-Small Cell Lung Cancer (NSCLC) undergoing first-line conventional platinum-based chemotherapy. METHODS In this retrospective cohort study, 203 patients with advanced NSCLC were recruited from January 2017 to December 2021. The cut-off value for the HALP score was determined by Receiver Operating Characteristic (ROC) curve analysis. The baseline characteristics and blood parameters were recorded, and the Log-rank test and Kaplan-Meier curves were applied for the survival analysis. In the univariate and multivariate analyses, the Cox regression analysis was carried out. The predictive accuracy and discriminative ability of the nomogram were determined by the Concordance index (C-index) and calibration curve and compared with a single HALP score by ROC curve analysis. RESULTS The optimal cut-off value for the HALP score was 28.02. The lower HALP score was closely associated with poorer Progression-Free Survival (PFS) and Overall Survival (OS). The male gender and other pathological types were associated with shorter OS. Disease progression and low HALP were correlated with shorter OS and PFS. In addition, nomograms were established based on HALP scores, gender, pathology type and efficacy rating, and used to predict OS. The C-index for OS prediction was 0.7036 (95% CI 0.643 to 0.7643), which was significantly higher than the C-index of HALP at 6-, 12-, and 24-months. CONCLUSION The HALP score is associated with the prognosis of advanced NSCLC patients receiving conventional platinum-based chemotherapy, and the nomogram established based on the HALP score has a better predictive capability for OS.
Collapse
Affiliation(s)
- Shan Gao
- Medical Oncology Division 1, Clinical Oncology Center, People' s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China; The First Clinical Medical College, Jinan University, Guangzhou, Guangdong, China
| | - Qin Huang
- Department of Oncology and Chemotherapy, Yulin Red Cross Hospital, Yulin, Guangxi, China
| | - Suosu Wei
- Department of Scientific Cooperation of Guangxi Academy of Medical Sciences, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yanru Lv
- Department of Scientific Cooperation of Guangxi Academy of Medical Sciences, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yanyan Xie
- Medical Oncology Division 1, Clinical Oncology Center, People' s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yanrong Hao
- Medical Oncology Division 1, Clinical Oncology Center, People' s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| |
Collapse
|
50
|
Yu J, Shen Q, Li J. Toxicology study profile of Nicotinamide mononucleotide after acute and 90-day sub chronic dosing in Wistar rats and mutagenicity tests. Curr Res Toxicol 2024; 6:100171. [PMID: 38765763 PMCID: PMC11101926 DOI: 10.1016/j.crtox.2024.100171] [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: 10/01/2023] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024] Open
Abstract
Nicotinamide mononucleotide (NMN) is an intermediate in biosynthesis pathway of Nicotinamide adenine dinucleotide (NAD+), an essential cofactor in all living cells involved in fundamental biological processes. Evidence stemming from recent studies have unveiled numerous roles of NAD+ metabolism on aging, longevity, delaying the progression of age-related diseases. A three-study genetic toxicity (genetox) battery (bacterial mutagenesis, in vitro cytogenetics, and in vivo mammalian test) is usually required to confirm safety of a new dietary ingredient and this study showed the data from in vivo mutagenicity test for the first time. The acute oral LD50 of NMN was greater than 2000 mg/kg body weight with 5000 mg/kg body weight as LD50 cut-off value and was classified under "Category 5 or Unclassified" as per Globally Harmonized System of Classification and Labelling of Chemicals (GHS). Based on 90 days repeated dose toxicity test the NOAEL was considered to be NLT 800 mg NMN/kg body weight in Wistar rats. The bacterial reverse mutation test, the in vitro and in vivo chromosomal aberration test, found NMN to be non-mutagenic. In the mammalian bone marrow chromosomal aberration test, it was concluded that NMN is non clastogenic at and up to 2,000 mg/kg body weight in all the animals tested to confirm safety of a new dietary ingredient and this study showed the data from in vivo mutagenicity test for the first time.
Collapse
Affiliation(s)
- Jianjun Yu
- Effepharm (Shanghai) Co. Ltd, Shanghai, China
| | - Qiang Shen
- Effepharm (Shanghai) Co. Ltd, Shanghai, China
| | - Jiayan Li
- Effepharm (Shanghai) Co. Ltd, Shanghai, China
| |
Collapse
|