1
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
2
|
Huang M, Wu Y, Wei X, Cheng L, Fu L, Yan H, Wei W, Li B, Ru H, Mo X, Tang W, Su Z, Yan L. Trifluridine/tipiracil induces ferroptosis by targeting p53 via the p53-SLC7A11 axis in colorectal cancer 3D organoids. Cell Death Dis 2025; 16:255. [PMID: 40188162 PMCID: PMC11972347 DOI: 10.1038/s41419-025-07541-z] [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: 09/19/2024] [Revised: 02/15/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
Trifluridine/Tipiracil (FTD/TPI, TAS102) has been approved for the treatment of patients with colorectal cancer (CRC) for its promising anticancer activity enabled by its incorporation into double strands during DNA synthesis. However, the mechanisms underlying the anticancer targets of FTD/TPI remain not fully understood. Here we report our observation of the activation of ferroptosis in CRC by FTD/TPI. Mechanistically, FTD/TPI directly promotes the ubiquitination and degradation of MDM2, thereby stabilizing the p53. Nuclear accumulation of p53 subsequently downregulates SLC7A11 expression, leading to ferroptosis. Furthermore, we observed that FTD/TPI combined with sulfasalazine (SAS), a system Xc- inhibitor, works in a synergistic manner to induce ferroptosis and further inhibit the proliferation of CRC cells. Finally, we confirmed the synergistic effect of SAS and FTD/TPI on patient-derived organoids in vitro and patient-derived xenograft mouse models in vivo. Our findings are the first to reveal that FTD/TPI induces ferroptosis via the p53-SLC7A11 axis and that SAS enhances the sensitivity and therapeutic effect of FTD/TPI. These findings suggest that the synergistic effect of FTD/TPI and SAS may represent a new therapeutic strategy for patients with CRC.
Collapse
Affiliation(s)
- Maosen Huang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Yancen Wu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiaoxia Wei
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Linyao Cheng
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lihua Fu
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Haochao Yan
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Wene Wei
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Bo Li
- Liaoning Provincial Engineering Laboratory of Anti-tumor Immunity and Molecular Theranostics Technology, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Haiming Ru
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xianwei Mo
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zijie Su
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Linhai Yan
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
3
|
Liu H, Chen L, Chen Y, Jin Y, Chen X, Ma N, Yang F, Bi H, Wen X, Xu S, Chen J, Lin Y, Yang Y, Wu Y, Chen Y. TCP1 promotes the progression of malignant tumours by stabilizing c-Myc through the AKT/GSK-3β and ERK signalling pathways. Commun Biol 2025; 8:563. [PMID: 40185866 PMCID: PMC11971430 DOI: 10.1038/s42003-025-07867-6] [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: 10/29/2023] [Accepted: 03/03/2025] [Indexed: 04/07/2025] Open
Abstract
The chaperonin tailless complex polypeptide 1 (TCP1) is a key subunit of chaperonin containing TCP1 (CCT) that regulates the folding and stability of proteins during cancer progression. Here, the prognostic significance of TCP1 was explored mainly in patients with hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC). We showed that TCP1 expression was significantly greater in clinically malignant tumour tissues than in normal tissues and that high TCP1 expression was associated with poor prognosis. TCP1 suppression not only decreased the proliferation and invasion of cancer cells in vitro but also inhibited tumour growth and metastasis in vivo. The underlying mechanisms were determined by ubiquitination assays and Co-IP (Co-Immunoprecipitation) experiments, and it was found that TCP1 regulated the stability of c-Myc through the RAC-alpha serine/threonine-protein kinase (AKT) /Glycogen synthase kinase 3β (GSK-3β) and extracellular regulated protein kinases (ERK) signalling pathways. Moreover, TCP1 knock-in (TCP1-KI) dramatically promoted the occurrence of diethylnitrosamine (DEN) -induced primary HCC in mice. Our results highlight the critical role of TCP1 in HCC and PDAC and reveal a novel mechanism to suppress HCC and PDAC by targeting c-Myc via the TCP1-induced promotion of the AKT/GSK-3β and ERK signalling pathways. TCP1 is able to modulate the stability of target proteins by multiple pathways, thus promoting the progression of HCC and PDAC. Our study identifies TCP1 as a prognostic novel marker and therapeutic target of HCC and PDAC.
Collapse
Affiliation(s)
- Hekun Liu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Linying Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, No. 20, Chazhong Road, 350005, Fuzhou, Fujian, China
| | - Yuwen Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yiyi Jin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Xiance Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Nengjun Ma
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Fan Yang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Huixia Bi
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Xinxin Wen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Shenmin Xu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Juan Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yanping Lin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yinghong Yang
- Department of Pathology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Yong Wu
- Fujian Institute of Haematology, Fujian Key Laboratory on Haematology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, 350001, Fuzhou, Fujian, China.
| | - Yuanzhong Chen
- Fujian Institute of Haematology, Fujian Key Laboratory on Haematology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, 350001, Fuzhou, Fujian, China.
| |
Collapse
|
4
|
Han S, Li Y, Chen D, Si Z, Xu T, Du Y, Xing N. Comprehensive Genetic Profile of Chinese Muscle-Invasive Bladder Cancer Cohort. Clin Genitourin Cancer 2025; 23:102280. [PMID: 39817975 DOI: 10.1016/j.clgc.2024.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/23/2024] [Indexed: 01/18/2025]
Abstract
OBJECTIVE The aim of our study was to characterize the spectrum of mutations in muscle-invasive bladder cancer (MIBC) in the Chinese population, identifying mutational features and exploring potential therapeutic targets. METHODS We collected samples from 62 Chinese patients with MIBC. For each patient, tumor tissues or blood samples were collected and sequenced by whole exome sequencing. RESULTS Our findings revealed the most frequently mutated genes included TP53 (41%), TTN (41%), HYDIN (34%), FRG1 (33%), ZNF717 (23%), AHNAK2 (21%), MUC4 (21%), KMT2D (20%), CDC27 (18%) and IGSF3 (18%). The most frequently mutated DNA damage repair (DDR) genes were TP53 (49%), SMARCA4 (10%), ERCC2 (8%), BRAC2 (6%), HERC2 (6%), HLTF (6%), PALB2 (6%) and POLG (6%). Additionally, our analysis confirmed an association between DDR mutations and high TMB (P = .022). Significant differences in MSI were observed between smokers and nonsmokers (P = .022), drinkers and nondrinkers (P = .018). By analyzing the data of 323 white MIBC samples from TCGA database, we identified frequently mutated driver genes in both our cohort and TCGA white cohort, including TP53, KMT2D, KMT2C, and FGFR3. Our study also revealed genes with distinct mutation frequencies compared to the TCGA white cohort, including FRG1, CDC27, IGSF3, MUC16, and ARID1A. CONCLUSIONS Our study provided comprehensive insights into genomic alterations in a cohort of Chinese MIBC, which could provide potential clues for clinical applications.
Collapse
Affiliation(s)
- Sujun Han
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yining Li
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Dong Chen
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhannan Si
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Yiqing Du
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Frediani E, Anceschi C, Ruzzolini J, Ristori S, Nerini A, Laurenzana A, Chillà A, Germiniani CEZ, Fibbi G, Del Rosso M, Mocali A, Venturin M, Battaglia C, Giovannelli L, Margheri F. Divergent regulation of long non-coding RNAs H19 and PURPL affects cell senescence in human dermal fibroblasts. GeroScience 2025; 47:2079-2097. [PMID: 39438391 DOI: 10.1007/s11357-024-01399-3] [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: 03/07/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Cellular senescence is a permanent cell growth arrest that occurs in response to various intrinsic and extrinsic stimuli and is associated with cellular and molecular changes. Long non-coding RNAs (lncRNAs) are key regulators of cellular senescence by affecting the expression of many important genes involved in senescence-associated pathways and processes. Here, we evaluated a panel of lncRNAs associated with senescence for their differential expression between young and senescent human dermal fibroblasts (NHDFs) and studied the effect of a known senomorphic compound, resveratrol, on the expression of lncRNAs in senescent NHDFs. As markers of senescence, we evaluated cell growth, senescence-associated (SA)-β-Gal staining, and the expression of p21, Lamin B1 and γH2AX. We found that H19 and PURPL were the most altered lncRNAs in replicative, in doxorubicin (DOXO) and ionising radiation (IR)-induced senescence models. We then investigated the function of H19 and PURPL in cell senescence by siRNA-mediated silencing in young and senescent fibroblasts, respectively. Our results showed that H19 knockdown reduced cell viability and induced cell senescence and autophagy of NHDFs through the regulation of the PI3K/AKT/mTOR pathway; conversely, PURPL silencing reversed senescence by reducing (SA)-β-Gal staining, recovering cell proliferation with an increase of S-phase cells, and reducing the p53-dependent DNA damage response. Overall, our data highlighted the role of H19 and PURPL in the senescent phenotype and suggested that these lncRNAs may have important implications in senescence-related diseases.
Collapse
Affiliation(s)
- Elena Frediani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Cecilia Anceschi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Sara Ristori
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Alice Nerini
- Department of Neurofarba (Department of Neurosciences, Drug Research and Child Health), University of Florence, Viale Pieraccini, 6 - 50139, Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Claudia Elena Zoe Germiniani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32 - 20133, Milan, Italy
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Alessandra Mocali
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy
| | - Marco Venturin
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32 - 20133, Milan, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli, 32 - 20133, Milan, Italy
| | - Lisa Giovannelli
- Department of Neurofarba (Department of Neurosciences, Drug Research and Child Health), University of Florence, Viale Pieraccini, 6 - 50139, Florence, Italy.
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni, 50 - 50134, Florence, Italy.
| |
Collapse
|
6
|
Zhang S, Hong HI, Mak VCY, Zhou Y, Lu Y, Zhuang G, Cheung LWT. Vertical inhibition of p110α/AKT and N-cadherin enhances treatment efficacy in PIK3CA-aberrated ovarian cancer cells. Mol Oncol 2025; 19:1132-1154. [PMID: 39543937 DOI: 10.1002/1878-0261.13761] [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: 03/22/2024] [Revised: 08/14/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha [PIK3CA, encoding PI3Kalpha (also known as p110α)] is one of the most commonly aberrated genes in human cancers. In serous ovarian cancer, PIK3CA amplification is highly frequent but PIK3CA point mutation is rare. However, whether PIK3CA amplification and PIK3CA driver mutations have the same functional impact in the disease is unclear. Here, we report that both PIK3CA amplification and E545K mutation are tumorigenic. While the protein kinase B (AKT) signaling axis was activated in both E545K knock-in cells and PIK3CA-overexpressing cells, the mitogen-activated protein kinase 3/1 (ERK1/2) pathway was induced selectively by E545K mutation but not PIK3CA amplification. Intriguingly, AKT signaling in these PIK3CA-aberrated cells increased transcriptional coactivator YAP1 (YAP) Ser127 phosphorylation and thereby cytoplasmic YAP levels, which in turn increased cell migration through Ras-related C3 botulinum toxin substrate 1 (RAC1) activation. In addition to the altered YAP signaling, AKT upregulated N-cadherin expression, which also contributed to cell migration. Pharmacological inhibition of N-cadherin reduced cell migratory potential. Importantly, co-targeting N-cadherin and p110α/AKT caused additive reduction in cell migration in vitro and metastases formation in vivo. Together, this study reveals the molecular pathways driven by the PIK3CA aberrations and the exploitable vulnerabilities in PIK3CA-aberrated serous ovarian cancer cells.
Collapse
Affiliation(s)
- Shibo Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Hei Ip Hong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - Victor C Y Mak
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - Yuan Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - Yiling Lu
- Division of Cancer Medicine, Department of Genomic Medicine, UT MD Anderson Cancer Centre, Houston, TX, USA
| | - Guanglei Zhuang
- State Key Laboratory of Systems Medicine for Cancer, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Lydia W T Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| |
Collapse
|
7
|
Chen Y, Fan Z, Luo Z, Kang X, Wan R, Li F, Lin W, Han Z, Qi B, Lin J, Sun Y, Huang J, Xu Y, Chen S. Impacts of Nutlin-3a and exercise on murine double minute 2-enriched glioma treatment. Neural Regen Res 2025; 20:1135-1152. [PMID: 38989952 PMCID: PMC11438351 DOI: 10.4103/nrr.nrr-d-23-00875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 12/21/2023] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00029/figure1/v/2024-07-06T104127Z/r/image-tiff Recent research has demonstrated the impact of physical activity on the prognosis of glioma patients, with evidence suggesting exercise may reduce mortality risks and aid neural regeneration. The role of the small ubiquitin-like modifier (SUMO) protein, especially post-exercise, in cancer progression, is gaining attention, as are the potential anti-cancer effects of SUMOylation. We used machine learning to create the exercise and SUMO-related gene signature (ESLRS). This signature shows how physical activity might help improve the outlook for low-grade glioma and other cancers. We demonstrated the prognostic and immunotherapeutic significance of ESLRS markers, specifically highlighting how murine double minute 2 (MDM2), a component of the ESLRS, can be targeted by nutlin-3. This underscores the intricate relationship between natural compounds such as nutlin-3 and immune regulation. Using comprehensive CRISPR screening, we validated the effects of specific ESLRS genes on low-grade glioma progression. We also revealed insights into the effectiveness of Nutlin-3a as a potent MDM2 inhibitor through molecular docking and dynamic simulation. Nutlin-3a inhibited glioma cell proliferation and activated the p53 pathway. Its efficacy decreased with MDM2 overexpression, and this was reversed by Nutlin-3a or exercise. Experiments using a low-grade glioma mouse model highlighted the effect of physical activity on oxidative stress and molecular pathway regulation. Notably, both physical exercise and Nutlin-3a administration improved physical function in mice bearing tumors derived from MDM2-overexpressing cells. These results suggest the potential for Nutlin-3a, an MDM2 inhibitor, with physical exercise as a therapeutic approach for glioma management. Our research also supports the use of natural products for therapy and sheds light on the interaction of exercise, natural products, and immune regulation in cancer treatment.
Collapse
Affiliation(s)
- Yisheng Chen
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongcheng Fan
- Department of Orthopedic Surgery, Hainan Province Clinical Medical Center, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, Hainan Province, China
| | - Zhiwen Luo
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xueran Kang
- Department of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Renwen Wan
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangqi Li
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiwei Lin
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Zhihua Han
- Department of Orthopedics, Shanghai General Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Beijie Qi
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiebin Huang
- Department of Infectious Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Shiyi Chen
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Zheng Z, Liu H, Xu Q, Cui W, Liu K. Comprehensive identification of a migrasomes-associated long non-coding RNA signature to predict the prognosis and treatment options in colon adenocarcinoma. Discov Oncol 2025; 16:409. [PMID: 40146487 PMCID: PMC11950624 DOI: 10.1007/s12672-025-02197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Migrasomes, recently discovered cellular substructures, may play a crucial role in cancer progression, treatment response, and prognosis. However, the prognostic value of migrasome-associated long non-coding RNAs (lncRNAs) in colon adenocarcinoma (COAD) remains unexplored. METHODS RNA-seq data from 459 COAD patients, including clinical characteristics and outcome information, were obtained from The Cancer Genome Atlas. A risk model was constructed through co-expression analysis of migrasome genes and lncRNAs, followed by Cox regression and least absolute shrinkage and selection operator analysis to identify prognostic lncRNAs. Functional enrichment analyses were performed to elucidate underlying biological mechanisms. Immune landscape characterization utilized ESTIMATE, CIBERSORT, Tumor Immune Estimation Resource (TIME), and single-sample Gene Set Enrichment Analysis (ssGSEA). Drug sensitivity analysis was conducted for select therapeutic agents. RESULTS Nine prognostic lncRNAs (AC010463.3, AL590483.4, AP005264.1, ZEB1-AS1, AC104088.1, PRKAR1B-AS2, AC009315.1, SUCLG2-AS1, and AC006111.2) were identified and incorporated into a risk model. Low-risk patients demonstrated significantly improved survival outcomes. The model exhibited independent prognostic capability, with AUCs of 0.783, 0.749, and 0.713 for one-, three-, and five-year survival, respectively, in the training cohort. High-risk patients displayed reduced overall survival and elevated tumor mutation burden. Additionally, these patients showed decreased sensitivity to therapeutic agents, including Oxaliplatin, Irinotecan, and 5-Fluorouracil. CONCLUSION Our novel migrasome-associated lncRNA signature demonstrates robust predictive capacity for both prognosis and chemotherapeutic sensitivity in COAD, potentially facilitating personalized treatment strategies and improved patient management.
Collapse
Affiliation(s)
- Zhen Zheng
- Department of Chemoradiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, 57 Xingning Road, Ningbo, 315000, Zhejiang, China
| | - Hui Liu
- Department of Chemoradiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, 57 Xingning Road, Ningbo, 315000, Zhejiang, China
| | - Quan Xu
- Department of Chemoradiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, 57 Xingning Road, Ningbo, 315000, Zhejiang, China
| | - Wei Cui
- Department of Colorectal Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Kaitai Liu
- Department of Chemoradiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, 57 Xingning Road, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
9
|
Jayagopal A, Walsh RJ, Hariprasannan KK, Mariappan R, Mahapatra D, Jaynes PW, Lim D, Peng Tan DS, Tan TZ, Pitt JJ, Jeyasekharan AD, Rajan V. A multi-task domain-adapted model to predict chemotherapy response from mutations in recurrently altered cancer genes. iScience 2025; 28:111992. [PMID: 40160429 PMCID: PMC11952854 DOI: 10.1016/j.isci.2025.111992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 08/23/2024] [Accepted: 02/06/2025] [Indexed: 04/02/2025] Open
Abstract
Next-generation sequencing (NGS) is increasingly utilized in oncological practice; however, only a minority of patients benefit from targeted therapy. Developing drug response prediction (DRP) models is important for the "untargetable" majority. Prior DRP models typically use whole-transcriptome and whole-exome sequencing data, which are clinically unavailable. We aim to develop a DRP model toward the repurposing of chemotherapy, requiring only information from clinical-grade NGS (cNGS) panels of restricted gene sets. Data sparsity and limited patient drug response information make this challenging. We firstly show that existing DRPs perform equally with whole-exome versus cNGS (∼300 genes) data. Drug IDentifier (DruID) is then described, a DRP model for restricted gene sets using transfer learning, variant annotations, domain-invariant representation learning, and multi-task learning. DruID outperformed state-of-the-art DRP methods on pan-cancer data and showed robust response classification on two real-world clinical datasets, representing a step toward a clinically applicable DRP tool.
Collapse
Affiliation(s)
- Aishwarya Jayagopal
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Robert J. Walsh
- Department of Haematology-Oncology, National University Cancer Institute, NUHS Tower Block, Level 7, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Krishna Kumar Hariprasannan
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Ragunathan Mariappan
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Debabrata Mahapatra
- Department of Computer Science, School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Patrick William Jaynes
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599, Singapore
| | - Diana Lim
- Department of Pathology, National University Health System, 1E Kent Ridge Road Singapore 119228, Singapore
| | - David Shao Peng Tan
- Department of Haematology-Oncology, National University Cancer Institute, NUHS Tower Block, Level 7, 1E Kent Ridge Road, Singapore 119228, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore. 1E Kent Ridge Road, NUHS Tower Block, Level 10, Singapore 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599, Singapore
| | - Jason J. Pitt
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599, Singapore
| | - Anand D. Jeyasekharan
- Department of Haematology-Oncology, National University Cancer Institute, NUHS Tower Block, Level 7, 1E Kent Ridge Road, Singapore 119228, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Center for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599, Singapore
| | - Vaibhav Rajan
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| |
Collapse
|
10
|
Liu Y, Zhong Y, Sang Y, Zhu S, Xu K, Zhu X, Cui X, Liu X, Wang X, Chen H, Jing C, Chong W, Li L. Molecular characteristics and cancer immunity of LRP1B and its relationship with the Hedgehog signaling pathway in colorectal cancer. Front Immunol 2025; 16:1567102. [PMID: 40170839 PMCID: PMC11959038 DOI: 10.3389/fimmu.2025.1567102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 02/21/2025] [Indexed: 04/03/2025] Open
Abstract
Background Colorectal cancer (CRC) is a malignant tumor of the digestive tract that significantly impacts human health. LDL receptor-related protein 1B (LRP1B) may play a crucial role in tumorigenesis and disease progression. Methods We performed a comparative analysis of differential gene expression, mutation patterns, drug sensitivity, and cellular phenotypes across different subgroups with varying LRP1B expression levels. Cellular and molecular experiments were conducted to validate our findings. Results Our analysis implicated LRP1B as a tumor suppressor gene. Experimental results confirmed that LRP1B expression was reduced in CRC and its knockdown was associated with poor prognosis. Molecular mechanism studies revealed that LRP1B negatively regulated the Hedgehog (Hh) signaling pathway, influencing cell cycle and apoptosis processes. Single-cell analysis showed significant differences in the infiltration of T cells, B cells, epithelial cells, and myeloid cells between high and low LRP1B expression groups. Immune cell infiltration and drug sensitivity analyses demonstrated that LRP1B plays a crucial role in immunotherapy and targeted therapy, suggesting that restoring LRP1B function could be a promising treatment strategy for CRC. Conclusion Our results indicate that LRP1B may function as a tumor suppressor factor in CRC, playing a significant role in mutation, therapy, and immune infiltration. Knockdown of LRP1B activates the Hh pathway in tumor cells, leading to the inhibition of several malignant biological behaviors.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yang Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Clinical Research Center of Shandong University, Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yaodong Sang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Siqiang Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kang Xu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xingyu Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoling Cui
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xinyu Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaohan Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Chen
- Clinical Research Center of Shandong University, Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
11
|
Spring J, Gurbuxani S, Golovkina T. Microbiota does not influence tumor development in two models of heritable cancer. mBio 2025; 16:e0386624. [PMID: 39969175 PMCID: PMC11898629 DOI: 10.1128/mbio.03866-24] [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: 12/15/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
Microbial impact on tumorigenesis of heritable cancers proximal to the gut is well-documented. Whether the microbiota influences cancers arising from inborn mutations at sites distal to the gut is undetermined. Using two models of heritable cancer, Trp53-deficient mice and Wnt1-transgenic mice, and a gnotobiotic approach, we found the microbiota to be inconsequential for tumor development. This work furthers our understanding of the degree of the microbial impact on tumor development. IMPORTANCE The influence of the microbiome on the development of cancer is well-documented with many if not all published studies reporting either a positive or a negative impact. None of the published studies, however, presented data on the influence of the microbiome on the development of heritable cancer. We find that the microbiota has no influence on cancer development in two models of spontaneous cancers driven by germline Trp53 deficiency and constitutive Wnt1 signaling.
Collapse
Affiliation(s)
- Jessica Spring
- Committee on Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Sandeep Gurbuxani
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Tatyana Golovkina
- Committee on Microbiology, University of Chicago, Chicago, Illinois, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
- Committee on Genetics, Genomics and System Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
12
|
Fan Y, Ji X, Yuan K, Wu Q, Lou M. HDAC1 Mediates Immunosuppression of the Tumor Microenvironment in Non-Small Cell Lung Cancer. J Inflamm Res 2025; 18:3333-3347. [PMID: 40078575 PMCID: PMC11900795 DOI: 10.2147/jir.s509316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Background Studies have demonstrated that histone deacetylase 1 (HDAC1) enables cancer cells to evade killing mediated by cytotoxic T lymphocytes. However, there are no studies on the immunological aspects of HDAC1 in non-small cell lung cancer (NSCLC). Methods In this research, we used the Cancer Genome Atlas (TCGA) public database combined with tissue microarray (TMA) to investigate HDAC1 expression and prognosis in NSCLC. According to the median value of HDAC1 expression in the TCGA dataset, samples of patients with NSCLC were classified into high- and low-expression cohorts. Subsequently, the biological characteristics of HDAC1 in high- and low-expression cohorts in terms of signaling pathways, immune cell infiltration, immune cell function, and genomic stability were investigated to better understand the effect of HDAC1 in the tumor microenvironment (TME) of NSCLC. Additionally, we selected tissue samples with HDAC1 overexpression in TMA2 and performed immunohistochemical staining of CD8+ T cells to observe the distribution of CD8+ T cells in the tumor. Results The findings revealed that overexpression of HDAC1 in NSCLC was associated with poor prognosis. Analysis of signaling pathway enrichment indicated that HDAC1 downregulated immune-related signaling pathways in NSCLC. Immune cell infiltration, immune cell function, and genomic stability analyses suggested that the TME alteration mediated by HDAC1 in the high-expression cohort was consistent with the "immune desert" phenotype. Furthermore, CD8+ T immunohistochemical staining experiments of tissue samples with HDAC1 overexpression in NSCLC revealed few CD8+ T cells distributed in the tumor parenchyma and interstitium. Conclusion Conclusively, our findings from several biological analyses revealed that HDAC1 is overexpressed in NSCLC and induces TME immunosuppression.
Collapse
Affiliation(s)
- Yongfei Fan
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Xiang Ji
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, People’s Republic of China
| | - Kai Yuan
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Qiyong Wu
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| | - Ming Lou
- Department of Thoracic Surgery, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
- Heart and Lung Disease Laboratory, The second People’s Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, People’s Republic of China
| |
Collapse
|
13
|
Moore J, Gkantalis J, Guix I, Chou W, Yuen K, Lazar AA, Spitzer M, Combes A, Barcellos-Hoff MH. Identification of a conserved subset of cold tumors responsive to immune checkpoint blockade. J Immunother Cancer 2025; 13:e010528. [PMID: 40050047 PMCID: PMC11887281 DOI: 10.1136/jitc-2024-010528] [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: 09/06/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The efficacy of immune checkpoint blockade (ICB) depends on restoring immune recognition of cancer cells that have evaded immune surveillance. Transforming growth factor-beta (TGFβ) is associated with immune-poor, so-called cold tumors whereas loss of its signaling promotes DNA misrepair that could stimulate immune response. METHODS We analyzed transcriptomic data from IMvigor210, The Cancer Genome Atlas, and Tumor Immune Syngeneic MOuse data sets to evaluate the predictive value of high βAlt, a score representing low expression of a signature consisting of TGFβ targets and high expression of genes involved in error-prone DNA repair. The immune context of βAlt was assessed by evaluating tumor-educated immune signatures. An ICB-resistant, high βAlt preclinical tumor model was treated with a TGFβ inhibitor, radiation, and/or ICB and assessed for immune composition and tumor control. RESULTS We found that a high βAlt score predicts ICB response yet is paradoxically associated with an immune-poor tumor microenvironmentcancer in both human and mouse tumors. We postulated that high βAlt cancers consist of cancer cells in which loss of TGFβ signaling generates a TGFβ rich, immunosuppressive tumor microenvironment. Accordingly, preclinical modeling showed that TGFβ inhibition followed by radiotherapy could convert an immune-poor, high βAlt tumor to an immune-rich, ICB-responsive tumor. Mechanistically, TGFβ inhibition increased activated natural killer (NK) cells, which were required to recruit lymphocytes to respond to ICB in irradiated tumors. NK cell activation signatures were also increased in high βAlt, cold mouse and human tumors that responded to ICB. CONCLUSIONS These studies indicate that loss of TGFβ signaling competency and gain of error-prone DNA repair identifies a subset of cold tumors that are responsive to ICB. Our mechanistic studies show that inhibiting TGFβ activity can convert a high βAlt, cold tumor into ICB-responsive tumors via NK cells. A biomarker consisting of combined TGFβ, DNA repair, and immune context signatures is a means to prospectively identify patients whose cancers may be converted from cold to hot with appropriate therapy.
Collapse
Affiliation(s)
- Jade Moore
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Jim Gkantalis
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Ines Guix
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - William Chou
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Kobe Yuen
- Oncology Biomarker Development, Genentech, South San Francisco, California, USA
| | - Ann A Lazar
- Division of Oral Epidemiology and Division of Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Matthew Spitzer
- Depts of Otolaryngology-Head and Neck Surgery and of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | - Alexis Combes
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Hickey J, Harris RA, Meola SD, Jennings S, Moore PF, Vernau W, Harding K, Thamm DH, Schlein LJ. Mixed histiocytic sarcoma in a Bernese Mountain Dog. J Vet Diagn Invest 2025; 37:317-323. [PMID: 39866033 PMCID: PMC11773501 DOI: 10.1177/10406387241312308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025] Open
Abstract
An 8-y-old, spayed female Bernese Mountain Dog was presented to a referral center for evaluation of right thoracic limb lameness and previously suspected Evans syndrome that had been poorly responsive to immunosuppressive therapy. Based on review of examination findings and laboratory data, Evans syndrome was deemed unlikely and hemophagocytic histiocytic sarcoma (HHS) was strongly suspected. On blood smear evaluation, atypical, histiocytic cells were noted, some of which exhibited siderophagia. Considering that circulating cells are not typically observed in dogs with HHS, additional diagnostic investigation was performed. Autopsy and histopathology revealed that the dog had a mixed form of HS (dendritic-cell origin HS in the lung, and HHS in the spleen, liver, and bone marrow), and immunocytochemical characterization of cultured cells derived from blood suggested that the cells were of dendritic HS origin, rather than HHS origin, as originally suspected. Whole-exome sequencing revealed genetic similarity between cell lines derived from lung tissue and blood, providing additional evidence of the relatedness of these 2 cell populations. Our case highlights the rare entity of mixed HS and typifies the inherent challenges in classifying rare, atypical, circulating neoplastic cells.
Collapse
Affiliation(s)
| | - R. Adam Harris
- Veterinary Diagnostic Laboratory–Microbiology, Immunology, Pathology, Colorado State University, Fort Collins, CO, USA
| | | | | | - Peter F. Moore
- Department of Pathology, Microbiology and Immunology, University of California–Davis, Davis, CA, USA
| | - William Vernau
- Department of Pathology, Microbiology and Immunology, University of California–Davis, Davis, CA, USA
| | | | - Douglas H. Thamm
- Clinical Sciences Department, Colorado State University, Fort Collins, CO, USA
| | - Lisa J. Schlein
- Veterinary Diagnostic Laboratory–Microbiology, Immunology, Pathology, Colorado State University, Fort Collins, CO, USA
- Zoetis Reference Laboratories, Florham Park, NJ, USA
| |
Collapse
|
15
|
Liu X, To KK, Zeng Q, Fu L. Effect of Extracellular Vesicles Derived From Tumor Cells on Immune Evasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417357. [PMID: 39899680 PMCID: PMC11948033 DOI: 10.1002/advs.202417357] [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: 12/21/2024] [Indexed: 02/05/2025]
Abstract
The crosstalk between immunity and cancer in the regulation of tumor growth is considered a hallmark of cancer. Antitumor immunity refers to the innate and adaptive immune responses that regulate cancer development and proliferation. Tumor immune evasion represents a major hindrance to effective anticancer treatment. Extracellular vesicles (EVs) are nano-sized and lipid-bilayer-enclosed particles that are secreted to the extracellular space by all cell types. They are critically involved in numerous biological functions including intercellular communication. Tumor-derived extracellular vesicles (TEVs) can transport a variety of cargo to modulate immune cells in the tumor microenvironment (TME). This review provides the latest update about how tumor cells evade immune surveillance by exploiting TEVs. First, the biogenesis of EVs and the cargo-sorting machinery are discussed. Second, how tumor cells modulate immune cell differentiation, activation, and function via TEVs to evade immune surveillance is illustrated. Last but not least, the novel antitumor strategies that can reverse immune escape are summarized.
Collapse
Affiliation(s)
- Xuanfan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Kenneth K.W. To
- School of PharmacyThe Chinese University of Hong KongHong Kong999077P. R. China
| | - Qinsong Zeng
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
- Guangxi Hospital Division of The First Affiliated HospitalSun Yat‐sen UniversityNanning530025P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
16
|
Zarzuela L, Durán RV, Tomé M. Metabolism and signaling crosstalk in glioblastoma progression and therapy resistance. Mol Oncol 2025; 19:592-613. [PMID: 38105543 PMCID: PMC11887670 DOI: 10.1002/1878-0261.13571] [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: 08/07/2023] [Revised: 11/09/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023] Open
Abstract
Glioblastoma is the most common form of primary malignant brain tumor in adults and one of the most lethal human cancers, with high recurrence and therapy resistance. Glioblastoma cells display extensive genetic and cellular heterogeneity, which precludes a unique and common therapeutic approach. The standard of care in glioblastoma patients includes surgery followed by radiotherapy plus concomitant temozolomide. As in many other cancers, cell signaling is deeply affected due to mutations or alterations in the so-called molecular drivers. Moreover, glioblastoma cells undergo metabolic adaptations to meet the new demands in terms of energy and building blocks, with an increasing amount of evidence connecting metabolic transformation and cell signaling deregulation in this type of aggressive brain tumor. In this review, we summarize some of the most common alterations both in cell signaling and metabolism in glioblastoma, presenting an integrative discussion about how they contribute to therapy resistance. Furthermore, this review aims at providing a comprehensive overview of the state-of-the-art of therapeutic approaches and clinical trials exploiting signaling and metabolism in glioblastoma.
Collapse
Affiliation(s)
- Laura Zarzuela
- Centro Andaluz de Biología Molecular y Medicina Regenerativa – CABIMER, Consejo Superior de Investigaciones CientíficasUniversidad de Sevilla, Universidad Pablo de OlavideSevilleSpain
| | - Raúl V. Durán
- Centro Andaluz de Biología Molecular y Medicina Regenerativa – CABIMER, Consejo Superior de Investigaciones CientíficasUniversidad de Sevilla, Universidad Pablo de OlavideSevilleSpain
| | - Mercedes Tomé
- Centro Andaluz de Biología Molecular y Medicina Regenerativa – CABIMER, Consejo Superior de Investigaciones CientíficasUniversidad de Sevilla, Universidad Pablo de OlavideSevilleSpain
| |
Collapse
|
17
|
Wang H, Syed AA, Krijgsveld J, Sigismondo G. Isolation of Proteins on Chromatin Reveals Signaling Pathway-Dependent Alterations in the DNA-Bound Proteome. Mol Cell Proteomics 2025; 24:100908. [PMID: 39842777 PMCID: PMC11889358 DOI: 10.1016/j.mcpro.2025.100908] [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: 04/08/2024] [Revised: 01/03/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
Signaling pathways often convergence on transcription factors and other DNA-binding proteins that regulate chromatin structure and gene expression, thereby governing a broad range of essential cellular functions. However, the repertoire of DNA-binding proteins is incompletely understood even for the best-characterized pathways. Here, we optimized a strategy for the isolation of Proteins on Chromatin (iPOC) exploiting tagged nucleoside analogs to label the DNA and capture associated proteins, thus enabling the comprehensive, sensitive, and unbiased characterization of the DNA-bound proteome. We then applied iPOC to investigate chromatome changes upon perturbation of the cancer-relevant PI3K-AKT-mTOR pathway. Our results show distinct dynamics of the DNA-bound proteome upon selective inhibition of PI3K, AKT, or mTOR, and we provide evidence how this signaling cascade regulates the DNA-bound status of SUZ12, thereby modulating H3K27me3 levels. Collectively, iPOC is a powerful approach to study the composition of the DNA-bound proteome operating downstream of signaling cascades, thereby both expanding our knowledge of the mechanism of action of the pathway and unveiling novel chromatin modulators that can potentially be targeted pharmacologically.
Collapse
Affiliation(s)
- Huiyu Wang
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Azmal Ali Syed
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany.
| | - Gianluca Sigismondo
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
18
|
Biswas S, Lee JE, Xie G, Masclef L, Ren Z, Côté J, Affar EB, Ge K, Kutateladze TG. Colorectal cancer hot spot mutations attenuate the ASXL-MLL4 interaction. J Biol Chem 2025; 301:108333. [PMID: 39984049 PMCID: PMC11957774 DOI: 10.1016/j.jbc.2025.108333] [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: 12/18/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025] Open
Abstract
Human additional sex combs like (ASXL) proteins are involved in the maintenance of both transcriptional activation and repression through their ability to bind multiple chromatin regulators, including two tumor suppressors: deubiquitinase BAP1 and methyltransferase MLL4 (KMT2D). The ASXL genes are often altered in colorectal cancer (CRC), and ASXL1 is one of the four hub genes related to the pathogenesis of CRC. Here, we show that MLL4 and BAP1 interdependently target specific genomic regions and positively or negatively regulate expression of a subset of genes in the human colon carcinoma HCT116 cells. MLL4 and BAP1 colocalize on a subset of enhancers and promoters in an interdependent manner. Genomic distribution of BAP1 in CRC cells differs from that in ESCs, with substantially more BAP1 binding sites identified on enhancers and promoters in HCT116 cells. MLL4 occupancy on MLL4+ BAP1+ genomic regions depends on functional ASXLs that interact with both MLL4 and BAP1, and CRC-relevant mutations attenuate the formation of the MLL4-ASXL complex. Mutational analysis and binding assays identified CRC hot spot mutations in ASXLs. Our findings suggest that alterations in the genomic distribution of the MLL4-ASXL-BAP1 axis and CRC hot spot mutations in ASXLs perturb normal transcriptional programs and may trigger pathogenic events in colon cancer.
Collapse
Affiliation(s)
- Soumi Biswas
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ji-Eun Lee
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Guojia Xie
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Louis Masclef
- Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada
| | - Zhizhong Ren
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Jacques Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Quebec City, Québec, Canada
| | - El Bachir Affar
- Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada; Department of Medicine, University of Montréal, Montréal, Québec, Canada
| | - Kai Ge
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA.
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado, USA.
| |
Collapse
|
19
|
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
|
20
|
Zou JX, Chang MR, Kuznetsov NA, Kee JX, Babak MV, Ang WH. Metal-based immunogenic cell death inducers for cancer immunotherapy. Chem Sci 2025:d4sc08495k. [PMID: 40160356 PMCID: PMC11949249 DOI: 10.1039/d4sc08495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunogenic cell death (ICD) has attracted enormous attention over the past decade due to its unique characteristics in cancer cell death and its role in activating innate and adaptive immune responses against tumours. Many efforts have been dedicated to screening, identifying and discovering ICD inducers, resulting in the validation of several based on metal complexes. In this review, we provide a comprehensive summary of current metal-based ICD inducers, their molecular mechanisms for triggering ICD initiation and subsequent protective antitumour immune responses, along with considerations for validating ICD both in vitro and in vivo. We also aim to offer insights into the future development of metal complexes with enhanced ICD-inducing properties and their applications in potentiating antitumour immunity.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Meng Rui Chang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Nikita A Kuznetsov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Jia Xuan Kee
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- NUS Graduate School - Integrative Science and Engineering Programme (ISEP), National University of Singapore 21 Lower Kent Ridge Rd Singapore 119077 Singapore
| |
Collapse
|
21
|
Pal J, Riester M, Ganner A, Ghosh A, Dhamija S, Mookherjee D, Voss C, Frew IJ, Kotsis F, Neumann-Haefelin E, Spang A, Diederichs S. Nonstop mutations cause loss of renal tumor suppressor proteins VHL and BAP1 and affect multiple stages of protein translation. SCIENCE ADVANCES 2025; 11:eadr6375. [PMID: 39937911 PMCID: PMC11817944 DOI: 10.1126/sciadv.adr6375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
Nonstop extension or stop-loss mutations lead to the extension of a protein at its carboxyl terminus. Recently, nonstop mutations in the tumor suppressor SMAD Family Member 4 (SMAD4) have been discovered to lead to proteasomal SMAD4 degradation. However, this mutation type has not been studied in other cancer genes. Here, we explore somatic nonstop mutations in the tumor suppressor genes BRCA1 Associated Protein 1 (BAP1) and Von Hippel-Lindau (VHL) enriched in renal cell carcinoma. For BAP1, nonstop mutations generate an extremely long extension. Instead of proteasomal degradation, the extension decreases translation and depletes BAP1 messenger RNA from heavy polysomes. For VHL, the short extension leads to proteasomal degradation. Unexpectedly, the mutation alters the selection of the translational start site shifting VHL isoforms. We identify germline VHL nonstop mutations in patients leading to the early onset of severe disease manifestations. In summary, nonstop extension mutations inhibit the expression of renal tumor suppressor genes with pleiotropic effects on translation and protein stability.
Collapse
Affiliation(s)
- Jagriti Pal
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marisa Riester
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Athina Ganner
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Avantika Ghosh
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center, Freiburg, Germany
| | - Sonam Dhamija
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center, Freiburg, Germany
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | | | - Christian Voss
- Department of Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ian J. Frew
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fruzsina Kotsis
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elke Neumann-Haefelin
- Renal Division, Department of Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anne Spang
- Biozentrum, University of Basel, Basel, Switzerland
| | - Sven Diederichs
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center, Freiburg, Germany
| |
Collapse
|
22
|
Ganesan IP, Kiyokawa H. A Perspective on Therapeutic Targeting Against Ubiquitin Ligases to Stabilize Tumor Suppressor Proteins. Cancers (Basel) 2025; 17:626. [PMID: 40002221 PMCID: PMC11853300 DOI: 10.3390/cancers17040626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/26/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
The loss of functions of tumor suppressor (TS) genes plays a key role in not only tumor initiation but also tumor progression leading to poor prognosis. While therapeutic inhibition of oncogene-encoded kinases has shown clinical success, restoring TS functions remains challenging due to conceptual and technical limitations. E3 ubiquitin ligases that ubiquitinate TS proteins for accelerated degradation in cancers emerge as promising therapeutic targets. Unlike proteasomal inhibitors with a broad spectrum, inhibitors of an E3 ligase would offer superior selectivity and efficacy in enhancing expression of its substrate TS proteins as far as the TS proteins retain wild-type structures. Recent advances in developing E3 inhibitors, including MDM2 inhibitors, highlight their potential and ultimately guide the framework to establish E3 inhibition as effective strategies to treat specific types of cancers. This review explores E3 ligases that negatively regulate bona fide TS proteins, the developmental status of E3 inhibitors, and their promise and pitfalls as therapeutic agents for anti-cancer precision medicine.
Collapse
Affiliation(s)
| | - Hiroaki Kiyokawa
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| |
Collapse
|
23
|
Aliev TI, Yudkin DV. AAV-based vectors for human diseases modeling in laboratory animals. Front Med (Lausanne) 2025; 11:1499605. [PMID: 40007819 PMCID: PMC11859266 DOI: 10.3389/fmed.2024.1499605] [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: 09/26/2024] [Accepted: 11/26/2024] [Indexed: 02/27/2025] Open
Abstract
The development of therapeutic drugs and vaccines requires the availability of appropriate model animals that replicate the pathogenesis of human diseases. Both native and transgenic animals can be utilized as models. The advantage of transgenic animals lies in their ability to simulate specific properties desired by researchers. However, there is often a need for the rapid production of transgenic animal models, especially in situations like a pandemic, as was evident during COVID-19. An important tool for transgenesis is the adeno-associated virus. The genome of adeno-associated virus serves as a convenient expression cassette for delivering various DNA constructs into cells, and this method has proven effective in practice. This review analyzes the features of the adeno-associated virus genome that make it an advantageous vector for transgenesis. Additionally, examples of utilizing adeno-associated viral vectors to create animal models for hereditary, oncological, and viral human diseases are provided.
Collapse
Affiliation(s)
- Timur I. Aliev
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Dmitry V. Yudkin
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
24
|
Poon G, Vedi A, Sanders M, Laurenti E, Valk P, Blundell JR. Single-cell DNA sequencing reveals pervasive positive selection throughout preleukemic evolution. CELL GENOMICS 2025; 5:100744. [PMID: 39842433 PMCID: PMC11872528 DOI: 10.1016/j.xgen.2024.100744] [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: 05/22/2024] [Revised: 08/03/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
The representation of driver mutations in preleukemic hematopoietic stem cells (pHSCs) provides a window into the somatic evolution that precedes acute myeloid leukemia (AML). Here, we isolate pHSCs from the bone marrow of 16 patients diagnosed with AML and perform single-cell DNA sequencing on thousands of cells to reconstruct phylogenetic trees of the major driver clones in each patient. We develop a computational framework that can infer levels of positive selection operating during preleukemic evolution from the statistical properties of these phylogenetic trees. Combining these data with 67 previously published phylogenetic trees, we find that the highly variable structures of preleukemic trees emerge naturally from a simple model of somatic evolution with pervasive positive selection typically in the range of 9%-24% per year. At these levels of positive selection, we show that the identification of early multiple-mutant clones could be used to identify individuals at risk of future AML.
Collapse
Affiliation(s)
- Gladys Poon
- Early Cancer Institute, University of Cambridge, Cambridge, UK.
| | - Aditi Vedi
- Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Cambridge University Hospital NHS Foundation Trust, Cambridge, UK
| | - Mathijs Sanders
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Elisa Laurenti
- Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Peter Valk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | |
Collapse
|
25
|
Huang M, Huang Z, Miao S, Chen X, Tan Y, Zhou Y, Wang S, Shi J. Bioinformatics Analysis of coagulation-related genes in lung adenocarcinoma: unveiling prognostic indicators and treatment pathways. Sci Rep 2025; 15:4972. [PMID: 39929884 PMCID: PMC11811422 DOI: 10.1038/s41598-025-87669-2] [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: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
Lung adenocarcinoma (LUAD) frequently precipitates a hypercoagulable state, resulting in venous thromboembolism and associated hemostatic complications. Furthermore, the coagulation cascade holds a pivotal role within the tumor microenvironment (TME) of LUAD. Utilizing unsupervised clustering of coagulation-related genes (CRG) and integrating clinical attributes, distinctions and correlations in clustering across various groups were assessed. Principal component analysis (PCA) was employed to derive the CRGscore for LUAD patients. Subsequently, a prognostic signature was established to contrast the impacts of immunological and pharmacological treatments across groups. The expression of PIK3CA, posited as a potential biomarker, was corroborated via immunohistochemistry(IHC) and Western blotting. This research delineated pronounced variances in immune signatures and prognostic categorizations among four coagulation-related subtypes, and delved into their associations with three gene cluster subtypes. A prognostic model based on coagulation-related scores was formulated for risk stratification and prognosis estimation. Disparities in immune infiltration, treatment modalities, and drug responsiveness among risk cohorts were discerned. Notably, an augmented expression of the coagulation-associated gene PIK3CA was observed in tumor samples. Coagulatory function is intimately linked with LUAD and its immune microenvironment, offering predictive potential for LUAD survival prognosis. Specifically, subgroups manifesting elevated PIK3CA expression might serve as foundational indicators for optimal treatment selection.
Collapse
Affiliation(s)
- Minliang Huang
- Department of Thoracic Surgery, Medical School, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China
- Medical School of Nantong University, Nantong, 226001, China
| | - Zhanghao Huang
- Department of Thoracic Surgery, Medical School, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China
- Medical School of Nantong University, Nantong, 226001, China
| | - Shichen Miao
- Medical School of Nantong University, Nantong, 226001, China
| | - Xingyou Chen
- Medical School of Nantong University, Nantong, 226001, China
| | - Yue Tan
- Medical School of Nantong University, Nantong, 226001, China
| | - Youlang Zhou
- Hand Surgery Research Center, Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Shuo Wang
- Department of Thoracic Surgery, Medical School, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China.
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.
| | - Jiahai Shi
- Department of Thoracic Surgery, Medical School, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China.
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, NO.20, Xisi Road, Nantong, 226001, Jiangsu, China.
- Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
26
|
Josephides JM, Chen CL. Unravelling single-cell DNA replication timing dynamics using machine learning reveals heterogeneity in cancer progression. Nat Commun 2025; 16:1472. [PMID: 39922809 PMCID: PMC11807193 DOI: 10.1038/s41467-025-56783-0] [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: 02/15/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
Genomic heterogeneity has largely been overlooked in single-cell replication timing (scRT) studies. Here, we develop MnM, an efficient machine learning-based tool that allows disentangling scRT profiles from heterogenous samples. We use single-cell copy number data to accurately perform missing value imputation, identify cell replication states, and detect genomic heterogeneity. This allows us to separate somatic copy number alterations from copy number changes resulting from DNA replication. Our methodology brings critical insights into chromosomal aberrations and highlights the ubiquitous aneuploidy process during tumorigenesis. The copy number and scRT profiles obtained by analysing >119,000 high-quality human single cells from different cell lines, patient tumours and patient-derived xenograft samples leads to a multi-sample heterogeneity-resolved scRT atlas. This atlas is an important resource for cancer research and demonstrates that scRT profiles can be used to study replication timing heterogeneity in cancer. Our findings also highlight the importance of studying cancer tissue samples to comprehensively grasp the complexities of DNA replication because cell lines, although convenient, lack dynamic environmental factors. These results facilitate future research at the interface of genomic instability and replication stress during cancer progression.
Collapse
Affiliation(s)
- Joseph M Josephides
- Institut Curie, PSL Research University, CNRS UMR3244, Dynamics of Genetic Information, Sorbonne Université, Paris, France
| | - Chun-Long Chen
- Institut Curie, PSL Research University, CNRS UMR3244, Dynamics of Genetic Information, Sorbonne Université, Paris, France.
| |
Collapse
|
27
|
Fan XP, Wang JR, Chen SY, Li XR, Cao JL, Wang HB, Ding LY, Che TJ, Yang L. Mechanistic insights into PROS1 inhibition of bladder cancer progression and angiogenesis via the AKT/GSK3β/β-catenin pathway. Sci Rep 2025; 15:4748. [PMID: 39922934 PMCID: PMC11807197 DOI: 10.1038/s41598-025-89217-4] [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: 08/26/2024] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
Bladder cancer (BLCA) is one of the ten most common cancers worldwide. However, the deregulation of PROS1 and its specific function in BLCA is not well understood. By combining proteomic and transcriptomic datasets, we discovered PROS1 expression was significantly reduced in BLCA tissues and revealed the clinical relevance of PROS1 with BLCA. Analysis of multiple BLCA datasets consistently showed the group with reduced PROS1 expression was linked to cancer-promoting pathways, more aggressive characteristics, and a greater chance of responding positively to immunotherapy. Next, various functional experiments were performed and the results revealed PROS1 overexpression inhibited the proliferation, cell cycle progression, migration, invasion, and angiogenesis of BLCA. In recovery trials, the AKT activator SC79 offered additional proof that PROS1 may influence BLCA cells via the AKT/GSK3β/β-catenin pathway. In conclusion, as an angiogenesis-related gene, PROS1 may play an inhibitory role in the biological functions of bladder cancer.
Collapse
Affiliation(s)
- Xin-Peng Fan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, China
| | - Ji-Rong Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, China
| | - Si-Yu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, China
| | - Xiao-Ran Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, China
| | - Jin-Long Cao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, China
| | - Hua-Bin Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, China
| | - Li-Yun Ding
- School of Physical Science and Technology, Lanzhou University, Lanzhou, China
| | - Tuan-Jie Che
- Baiyuan Company for Gene Technology, Lanzhou, China
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, China.
| |
Collapse
|
28
|
Ma R, Yang D, Wang P, Zhang Z, Zhang X, Song J, Liu H, Liu S, Zhang Y, Zou L. Oncogenic RIT1 mutations confer ferroptosis vulnerability in lung adenocarcinoma. Biol Direct 2025; 20:19. [PMID: 39920793 PMCID: PMC11804091 DOI: 10.1186/s13062-025-00613-2] [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: 07/28/2024] [Accepted: 01/28/2025] [Indexed: 02/09/2025] Open
Abstract
Members from the RAS GTPase superfamily have been closely implicated in the tumorigenesis of various human cancers. Recent sequencing analysis of lung adenocarcinoma has revealed the prevalence of alterations in the RIT1 gene that is a close RAS paralog. However, relative to RAS subfamily members KRAS, NRAS, and HRAS, our characterization of RIT1 oncogenic properties remains incomplete. Therefore, further investigation on RIT1 will facilitate future development of targeted therapies. Our bioinformatic analysis revealed that RIT1 alterations in lung cancer predicted poor survivals but differed from its RAS paralogs by showing largely amplification and mutation. Through biochemical characterization of RIT1 hotspot mutations, we propose that RIT1 alterations were associated with increased protein abundance that promoted cell growth. Transcriptomic profiling indicated that oncogenic RIT1 mutant expression influenced common tumorigenic RAS/MAPK, PI3K/AKT, and E2F1 pathways, in addition to altered NFE2L2 target expression. Importantly, RIT1 mutants markedly sensitized cells to ferroptosis induction, and RIT1 knockdown suppressed ferroptotic cell death. Lung adenocarcinoma NCI-H2110 cells containing endogenous RIT1 M90I mutation were susceptible to ferroptosis induction both in vitro and in vivo within xenograft models. Hence, our study unravels a novel aspect of RIT1 mutations in lung cancer and suggests ferroptosis induction as a potential therapeutic strategy to treat lung cancer patients carrying RIT1 mutations.
Collapse
Affiliation(s)
- Ruilan Ma
- Department of Radiation Oncology, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Dian Yang
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Peng Wang
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Ziyi Zhang
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xuehong Zhang
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jialiang Song
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Han Liu
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shuyan Liu
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yingqiu Zhang
- The Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Lijuan Zou
- Department of Radiation Oncology, Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
29
|
Ye J, Chen Z, Zhang C, Xie R, Chen H, Ren P. PPIH is a novel diagnostic biomarker associated with immune infiltration in cholangiocarcinoma. BMC Cancer 2025; 25:218. [PMID: 39920663 PMCID: PMC11806719 DOI: 10.1186/s12885-025-13607-0] [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: 09/09/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Cholangiocarcinoma (CHOL) is the second most common primary liver malignancy, characterized by high aggressiveness and heterogeneity. It is typically diagnosed at an advanced stage, leading to a poor prognosis. Although Peptidyl Proline Isomerase H (PPIH) has been implicated in various tumors, its role in CHOL remains unexplored. This study aims to investigate the diagnostic value and potential function of PPIH in CHOL. METHODS We analyzed the expression levels, prognostic significance, and diagnostic efficiency of PPIH in CHOL using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets, coupled with gene enrichment analyses. The CIBERSORT database was employed to assess the correlation between PPIH expression and immune cell infiltration in CHOL. Additionally, immunohistochemical experiments were conducted to validate PPIH expression levels in CHOL tissues and to explore its correlation with TP53 gene mutations. RESULTS Our findings indicate that overexpression of PPIH mRNA in CHOL is associated with poor prognosis, with increased PPIH protein levels observed in CHOL tissues. Furthermore, PPIH expression showed a positive correlation with TP53 mutations. PPIH demonstrated strong diagnostic value for CHOL. Moreover, PPIH may influence tumor progression through its involvement in cell cycle regulation and spliceosome pathways, and is associated with immune cell infiltration levels. CONCLUSION The results of this study suggest that PPIH is a potential novel biomarker with significant diagnostic value for patients with CHOL. PPIH may also play a role in modulating the immune microenvironment, contributing to poor prognosis.
Collapse
Affiliation(s)
- Jun Ye
- Precision Medical Laboratory Center, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, 556000, China
| | - Zhitao Chen
- Department of Pathology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, 556000, China
| | - Chuan Zhang
- Department of Pathology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, 556000, China
| | - Rui Xie
- Chengdu Gaoxin -Daan Medical Laboratory Co., Ltd, Chengdu, Sichuan, 610000, China
| | - Haini Chen
- Precision Medical Laboratory Center, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, 556000, China.
- The Second Affiliated Hospital of Guizhou Medical University, Kangfu Road, Kaili, 556000, China.
| | - Peng Ren
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, 556000, China.
- The Second Affiliated Hospital of Guizhou Medical University, Kangfu Road, Kaili, 556000, China.
| |
Collapse
|
30
|
Qi J, Zhou M, Yang N, Ma H, He M, Wu G, Ge C, Jin L, Cheng L, Liao W, Ren H, Lei C. TUBA1B as a novel prognostic biomarker correlated with immunosuppressive tumor microenvironment and immunotherapy response. Front Pharmacol 2025; 16:1517887. [PMID: 39968182 PMCID: PMC11832512 DOI: 10.3389/fphar.2025.1517887] [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: 10/27/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Background: Tubulin alpha 1b (TUBA1B) is a key microtubule protein essential for maintaining cellular structure and function. This protein contributes significantly to cytoskeletal formation and is implicated in various diseases. Despite its fundamental roles, TUBA1B's impact on tumor prognosis and the tumor immune microenvironment across cancer types remains inadequately understood. Methods To elucidate TUBA1B's role in cancer prognosis and immune response, we conducted a comprehensive analysis, integrating data from established databases such as The Cancer Genome Atlas, Genotype Tissue Expression, Cancer Cell Lineage Encyclopedia, Human Protein Atlas, Kaplan-Meier Plotter, cBioPortal, TIMER, and ImmuCellAI, along with a large-scale clinical study and immunotherapy cohort. We also conducted in vitro functional assays to assess TUBA1B's functional role in tumor cells, allowing for a detailed examination of its relationship with cancer prognosis and immune modulation. Results: Our findings indicate that TUBA1B expression is dysregulated across multiple cancers, correlating strongly with poor survival outcomes and advanced pathological stages. Functional enrichment analyses further revealed that TUBA1B regulates key cell cycle processes, driving tumor proliferation, migration, and invasion. It also influences immune functions within both the innate and adaptive immune systems, affecting immune-related signaling pathways. These insights underscore TUBA1B's multifaceted role in cancer progression and immune response. Conclusion: This study highlights TUBA1B's potential as a human oncogene with substantial roles in tumorigenesis and immune regulation. Elevated TUBA1B levels are associated with an immunosuppressive tumor microenvironment, impacting cancer progression and treatment outcomes. Targeting TUBA1B may offer promising therapeutic avenues for enhancing cancer treatment, offering new perspectives for innovative anti-tumor strategies with high clinical impact.
Collapse
Affiliation(s)
- Juntao Qi
- Rehabilitation Medicine Department, Hunan Aerospace Hospital, Hunan Normal University, Changsha, Hunan, China
- Research Center of Clinical Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, China
| | - Mingming Zhou
- Department of Critical Care Medicine, Chongqing University Affiliated Cancer Hospital, Chongqing, China
| | - Na Yang
- Laboratory of Oncology and Immunology, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Min He
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Gujie Wu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Chang Ge
- School of Medicine, Wuhan University, Wuhan, China
| | - Liuyin Jin
- School of Medicine, Wuhan University, Wuhan, China
| | - Lin Cheng
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Liao
- Department of Otolaryngology and Head and Neck Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Hefei Ren
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Caiyun Lei
- Rehabilitation Medicine Department, Hunan Aerospace Hospital, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
31
|
Hebert JD, Tang YJ, Szamecz M, Andrejka L, Lopez SS, Petrov DA, Boross G, Winslow MM. Combinatorial In Vivo Genome Editing Identifies Widespread Epistasis and an Accessible Fitness Landscape During Lung Tumorigenesis. Mol Biol Evol 2025; 42:msaf023. [PMID: 39907430 PMCID: PMC11824425 DOI: 10.1093/molbev/msaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/15/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Lung adenocarcinoma, the most common subtype of lung cancer, is genomically complex, with tumors containing tens to hundreds of non-synonymous mutations. However, little is understood about how genes interact with each other to enable the evolution of cancer in vivo, largely due to a lack of methods for investigating genetic interactions in a high-throughput and quantitative manner. Here, we employed a novel platform to generate tumors with inactivation of pairs of ten diverse tumor suppressor genes within an autochthonous mouse model of oncogenic KRAS-driven lung cancer. By quantifying the fitness of tumors with every single and double mutant genotype, we show that most tumor suppressor genetic interactions exhibited negative epistasis, with diminishing returns on tumor fitness. In contrast, Apc inactivation showed positive epistasis with the inactivation of several other genes, including synergistic effects on tumor fitness in combination with Lkb1 or Nf1 inactivation. Sign epistasis was extremely rare, suggesting a surprisingly accessible fitness landscape during lung tumorigenesis. These findings expand our understanding of the interactions that drive tumorigenesis in vivo.
Collapse
Affiliation(s)
- Jess D Hebert
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuning J Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Márton Szamecz
- Faculty of Informatics, Eötvös Loránd University, Budapest, Hungary
- National Laboratory for Health Security, Centre for Eco-Epidemiology, Budapest, Hungary
- Institute of Evolution, HUN-REN Centre for Ecological Research, Budapest, Hungary
| | - Laura Andrejka
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven S Lopez
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dmitri A Petrov
- Department of Biology, Stanford University, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Gábor Boross
- National Laboratory for Health Security, Centre for Eco-Epidemiology, Budapest, Hungary
- Institute of Evolution, HUN-REN Centre for Ecological Research, Budapest, Hungary
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
32
|
Harris JC, Schubert J, Lockhart B, Olson R, Paessler ME, Margolskee E, Pillai V, Wu J, Golenberg N, Chen J, Denenberg EH, Luke T, Luo M, Zhong Y, Li MM, Wertheim GB. Genetically Distinct Acute Megakaryoblastic Leukemia following Low Hypodiploid B-Lymphoblastic Leukemia linked by TP53 Mutation. Pediatr Dev Pathol 2025:10935266251316150. [PMID: 39898462 DOI: 10.1177/10935266251316150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
We report a case of acute myeloid leukemia with megakaryoblastic differentiation (AMKL) that developed after an initial B-lymphoblastic leukemia (B-ALL) with low hypodiploidy. Although the AMKL was initially thought either to be a phenotypic change from the original B-ALL or to have arisen as a result of treatment (acute myeloid leukemia, post cytotoxic therapy, AML-pCT [WHO]; AML, therapy related [ICC]), genetic evaluation of both the AMKL and the B-ALL suggest that neither of these considerations was correct. Rather, the AMKL did not harbor the most common genetic hallmark of AML-pCT-rearrangement of KMT2- and was genetically distinct from the B-ALL. Both the B-ALL and the AMKL, however, showed an identical TP53 mutation by next generation sequencing (NGS), while germline testing was negative for this mutant allele. Hence, either the patient had a tissue restricted constitutional TP53 mutation or had a somatic mutation in a multipotent hematopoietic precursor. This case highlights the necessity for close monitoring of patients with TP53-mutant tumors, as they may develop multiple lesions despite negative germline testing.
Collapse
Affiliation(s)
- Jaryse C Harris
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey Schubert
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brian Lockhart
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rachel Olson
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michele E Paessler
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth Margolskee
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Vinodh Pillai
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jinhua Wu
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Netta Golenberg
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jiani Chen
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth H Denenberg
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tammy Luke
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Minjie Luo
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yiming Zhong
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marilyn M Li
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gerald B Wertheim
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
33
|
Yan Y, Gong Y, Liang X, Xiong Q, Lin J, Wu Y, Zhang L, Chen H, Jin J, Luan X. Decoding β-catenin associated protein-protein interactions: Emerging cancer therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2025; 1880:189232. [PMID: 39643250 DOI: 10.1016/j.bbcan.2024.189232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
The hyperactive Wnt/β-catenin signaling circuit has been proven to be closely related to the progression of various cancers, with β-catenin serving as a central regulator of pro-tumorigenic processes. Preclinical evidences strongly support β-catenin as a promising therapeutic target. However, it has long been considered "undruggable" due to challenges such as the lack of crystal structures for its N- and C-terminal domains, high mutation rates, and limited availability of inhibitors. Notably, the network of β-catenin-associated protein-protein interactions (PPIs) is vital in the progression of multiple diseases. These interactions form a cancer-specific network that participates in all phases of oncogenesis, from cell metastasis to immunosuppressive microenvironment formation. Thus, researches on these PPIs are essential for unraveling the molecular mechanisms behind tumors with aberrant β-catenin activation, as well as for developing new targeted therapies. In this review, we delve into how β-catenin's PPIs orchestrate cancer progression and highlight biological and clinical dilemmas, proposing frontier technologies and potential challenges in targeting β-catenin for cancer therapy.
Collapse
Affiliation(s)
- Yue Yan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiting Gong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaohui Liang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qingyi Xiong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ye Wu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lijun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jinmei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
34
|
Cazzola M, Malcovati L. Genome sequencing in the management of myelodysplastic syndromes and related disorders. Haematologica 2025; 110:312-329. [PMID: 39445412 PMCID: PMC11788631 DOI: 10.3324/haematol.2023.284947] [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: 04/13/2024] [Accepted: 07/18/2024] [Indexed: 10/25/2024] Open
Abstract
Myeloid neoplasms originate from the clonal proliferation of hematopoietic stem cells, which is driven by the acquisition of somatic genetic mutations. Within these disorders, myelodysplastic syndromes (MDS) are specifically characterized by morphological abnormalities (dysplasia) and impaired maturation of myeloid precursors (ineffective hematopoiesis), resulting in peripheral blood cytopenia. Several studies have advanced the field of MDS, with a few landmark papers leading to a paradigm shift, opening new avenues of research and enabling a molecular revolution. These seminal papers include the first description of the 5q- syndrome, the identification of somatic mutations of TET2 in myeloid neoplasms, the detection of common pathway mutations in the splicing machinery, and the discovery of clonal hematopoiesis. The somatic genomic landscape of MDS is now well defined. Genes that are recurrently mutated include epigenetic regulators, as well as genes of RNA splicing machinery, transcription regulation, DNA repair control, cohesin complex, and signal transduction. Furthermore, several disorders with a germline genetic predisposition to MDS have been identified, collectively accounting for up to 15% of all MDS cases. Genomic profiling can significantly improve the diagnostic approach to MDS, allowing the identification of distinct nosological entities such as SF3B1-mutant or TP53-mutant MDS. The Molecular International Prognostic Scoring System for MDS has already proven to be a valuable tool for individualized risk assessment and treatment decisions. In addition, the recently developed molecular taxonomy of MDS will likely facilitate the implementation of precision medicine approaches for these disorders. This will necessitate the establishment of specialized infrastructures within public health systems, involving close collaboration between healthcare institutions, academia, and the life-sciences industry.
Collapse
Affiliation(s)
- Mario Cazzola
- Department of Molecular Medicine, University of Pavia, and Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia.
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, and Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia
| |
Collapse
|
35
|
Garcia-Murillas I, Cutts RJ, Walsh-Crestani G, Phillips E, Hrebien S, Dunne K, Sidhu K, Daber R, Hubert B, Graybill C, DeFord PM, Wooten DJ, Zhao J, Ellsworth RE, Johnston SRD, Ring A, Russell S, Evans A, Skene A, Wheatley D, Smith IE, Korn WM, Turner NC. Longitudinal monitoring of circulating tumor DNA to detect relapse early and predict outcome in early breast cancer. Breast Cancer Res Treat 2025; 209:493-502. [PMID: 39424680 PMCID: PMC11785695 DOI: 10.1007/s10549-024-07508-2] [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: 07/09/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE Detection of molecular residual disease (MRD) allows for the identification of breast cancer patients at high-risk of recurrence, with the potential that early initiation of treatment at early stages of relapse could improve patient outcomes. The Invitae Personalized Cancer Monitoring™ assay (PCM) is a newly developed next-generation sequencing approach that utilizes up to 50 patient-specific, tumor-informed DNA variants, to detect circulating tumor DNA (ctDNA). The ability of the PCM assay to detect MRD before clinical relapse was evaluated. METHODS The cohort included 61 female patients with high-risk breast cancer who underwent neoadjuvant chemotherapy. Plasma samples were collected before and during neoadjuvant therapy, after surgery and during monitoring. PCM was used to detect ctDNA at each time point. RESULTS The sensitivity to detect ctDNA in plasma from patients who relapsed during the monitoring phase was 76.9% (10/13). Specificity and positive predictive values were both 100% with all (10/61, 16%) of the patients who had ctDNA detected during the monitoring phase subsequently relapsing. Detection of ctDNA during monitoring was associated with a high-risk of future relapse (HR 37.2, 95% CI 10.5-131.9, p < 0.0001), with a median lead-time from ctDNA detection to clinical relapse of 11.7 months. CONCLUSION PCM detected ctDNA in patients who relapsed with a long lead-time over clinical relapse, shows strong association with relapse-free survival and may be used to identify patients at high-risk for relapse, allowing for earlier intervention.
Collapse
Affiliation(s)
- Isaac Garcia-Murillas
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Rosalind J Cutts
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Giselle Walsh-Crestani
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - Sarah Hrebien
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Kathryn Dunne
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Kally Sidhu
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | - Simon Russell
- Hinchingbrooke Hospital, Hinchingbrooke Park, Huntingdon, UK
| | | | | | - Duncan Wheatley
- Department of Oncology, Royal Cornwall Hospitals NHS Trust, Truro, UK
| | - Ian E Smith
- Breast Unit, Royal Marsden Hospital, London, UK
| | - W Michael Korn
- Former Employees of Invitae Corp, San Francisco, CA, USA
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Nicholas C Turner
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
- Breast Unit, Royal Marsden Hospital, London, UK.
- The Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK.
| |
Collapse
|
36
|
Umemura S, Udagawa H, Ikeda T, Murakami H, Daga H, Toyozawa R, Kozuki T, Sakakibara-Konishi J, Ohe Y, Morise M, Kato T, Shingyoji M, Hara S, Furuya N, Teranishi S, Takata S, Miyamoto S, Nakachi I, Wakabayashi M, Nomura S, Sato A, Ishii G, Tsuchihara K, Sugiyama E, Kirita K, Sakai T, Shibata Y, Izumi H, Nosaki K, Zenke Y, Matsumoto S, Yoh K, Niho S, Goto K. Clinical Significance of a Prospective Large Genomic Screening for SCLC: The Genetic Classification and a Biomarker-Driven Phase 2 Trial of Gedatolisib. J Thorac Oncol 2025; 20:177-193. [PMID: 39395663 DOI: 10.1016/j.jtho.2024.10.004] [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: 10/16/2023] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
INTRODUCTION SCLC has been treated as a single entity resulting in limited survival improvement. Developing effective tools for guiding appropriate therapeutic strategies is crucial. METHODS A total of 1035 SCLCs were prospectively analyzed by a genomic screening platform: LC-SCRUM-Asia. Fresh frozen tumor samples were subjected to a next-generation sequencing system enabling the integrative analysis of cancer-related genes. A phase 2 trial of gedatolisib for SCLC with PI3K/AKT/mTOR pathway mutations was conducted based on this screening. RESULTS On the basis of the treatment outcomes and therapeutic targets, the following five distinct genetic subgroups were identified in SCLC: NSCLC-subgroup (genetic alterations associated with NSCLC, 8.5%); Hotspot-subgroup (targetable hotspot mutations common in tumors, 3.0%); PI3K-subgroup (PI3K/AKT/mTOR pathway mutations, 7.4%); MYC-subgroup (MYC family amplifications, 13.0%); and HME-subgroup (mutations in the histone-modifying enzymes, 17.6%). The NSCLC-subgroup (hazard ratio = 1.57; 95% confidence interval: 1.22-2.03) and MYC-subgroup (hazard ratio = 1.56; 95% confidence interval: 1.26-1.93) had significantly shorter progression-free survivals after first-line platinum-based treatment. The Hotspot-subgroup and MYC-subgroup were candidates for novel targeted therapies. The HME-subgroup had a favorable survival in patients who received programmed cell death (ligand) 1 inhibitor-based therapies (p = 0.005, log-rank test) regardless of some overlap with other subgroups. There were 15 patients enrolled into the phase 2 trial of gedatolisib in the PI3K-subgroup, and the overall response rate and the disease control rate were 6.7% and 20%, respectively. The MYC-subgroup or NSCLC-subgroup was associated with unfavorable clinical outcomes in this trial. CONCLUSIONS Molecular classification of SCLC by genetic approach is beneficial for predicting the treatment outcomes and effectively guiding the clinical choices.
Collapse
Affiliation(s)
- Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaya Ikeda
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Haruyasu Murakami
- Division of Thoracic Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Haruko Daga
- Department of Medical Oncology, Osaka City General Hospital, Osaka, Japan
| | - Ryo Toyozawa
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Toshiyuki Kozuki
- Department of Thoracic Oncology and Medicine, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Jun Sakakibara-Konishi
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masahiro Morise
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | | | - Satoshi Hara
- Department of Respiratory Medicine, Itami City Hospital, Itami, Japan
| | - Naoki Furuya
- Division of Respiratory Medicine, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Shuhei Teranishi
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Saori Takata
- Department of Respiratory Medicine, Kyorin University School of Medicine, Mitaka, Japan
| | - Shingo Miyamoto
- Department of Medical Oncology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Ichiro Nakachi
- Department of Internal Medicine, Saiseikai Utsunomiya Hospital, Utsunomiya, Japan
| | - Masashi Wakabayashi
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Shogo Nomura
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan; Department of Biostatistics and Bioinformatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Eri Sugiyama
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keisuke Kirita
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuya Sakai
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuji Shibata
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroki Izumi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kaname Nosaki
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Zenke
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Seiji Niho
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
37
|
Alshenaifi JY, Vetere G, Maddalena G, Yousef M, White MG, Shen JP, Vilar E, Parseghian C, Dasari A, Morris VK, Huey R, Overman MJ, Wolff R, Raghav KP, Willis J, Alfaro K, Futreal A, You YN, Kopetz S. Mutational and co-mutational landscape of early onset colorectal cancer. Biomarkers 2025; 30:64-76. [PMID: 39761813 PMCID: PMC11856746 DOI: 10.1080/1354750x.2024.2447089] [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: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Colorectal cancer (CRC) incidence and mortality before 50 have been rising alarmingly in the recent decades. METHODS Using a cohort of 10,000 patients, this study investigates the clinical, mutational, and co-mutational features of CRC in early-onset (EOCRC, < 50 years) compared to late-onset (LOCRC, ≥ 50 years). RESULTS EOCRC was associated with a higher prevalence of Asian and Hispanic patients, rectal or left-sided tumors (72% vs. 59%), and advanced-stage disease. Molecular analyses revealed differences in mutation patterns, with EOCRC having higher frequencies of TP53 (74% vs. 68%, p < 0.01) and SMAD4 (17% vs. 14%, p = 0.015), while BRAF (5% vs. 11%, p < 0.001) and NOTCH1 (2.7% vs. 4.1%, p = 0.01) mutations were more prevalent in LOCRC. Stratification by tumor site and MSI status highlighted significant location- and age-specific molecular differences, such as increased KRAS and CTNNB1 mutations in right-sided EOCRC and higher BRAF prevalence in MSI-H LOCRC (47% vs. 6.7%, p < 0.001). Additionally, co-occurrence analysis revealed unique mutational networks in EOCRC MSS, including significant co-occurrences of FBXW7 with NOTCH3, RB1, and PIK3R1. CONCLUSION This study highlights the significance of age-specific molecular profiling, offering insights into the unique biology of EOCRC and potential clinical applications.
Collapse
Affiliation(s)
- Jumanah Yousef Alshenaifi
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guglielmo Vetere
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Giulia Maddalena
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael G. White
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eduardo Vilar
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine Parseghian
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Van Karlyle Morris
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Wolff
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kanwal P. Raghav
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andy Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y. Nancy You
- Department of Colon & Rectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
38
|
Siegenthaler F, Imboden S, Büchi C, Christe L, Solass W, Saner F, Rauh C, Hofer S, Schlatter B, Wampfler J, Mohr S, Papadia A, Anokhina M, Göring W, Rau TT, Mueller MD. Added prognostic value of sentinel lymph node mapping in endometrial cancer to molecular subgroups. Gynecol Oncol 2025; 193:12-19. [PMID: 39764854 DOI: 10.1016/j.ygyno.2024.12.014] [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: 10/02/2024] [Accepted: 12/22/2024] [Indexed: 03/03/2025]
Abstract
OBJECTIVE Treatment approaches for endometrial cancer became more personalized in the last decade, mainly due to two key advancements - sentinel lymph node (SLN) mapping and molecular classification. However, their prognostic interaction remains relatively unexplored. METHODS This retrospective cohort study included patients with endometrial cancer, who underwent surgical treatment including SLN mapping at the Bern University Hospital, Switzerland. Ultrastaging of the SLNs and a molecular analysis on the primary tumor was performed. RESULTS The study cohort included 206 patients, of which 197 tumor samples underwent molecular classification. 11.2 % were classified as POLEmut, 25.9 % as MMRd, 46.2 % as NSMP, and 16.8 % as p53abn. Overall, 834 SLN were removed. SLN macrometastasis were most prevalent in patients with p53abn tumors (24.2 %), followed by MMRd (13.7 %), NSMP (5.5 %), and POLEmut (0 %) tumors (p = .006). Mean follow-up time was 70.9 months. SLN macrometastasis was significantly associated with a higher risk of recurrence in the entire study cohort (p > .001) and the NSMP subgroup (p > .001). In the MMRd subgroup, SLN macrometastasis remained a significant predictor of recurrence (p = .030) and disease-specific death (p = .047) in multivariate Cox regression analysis. For patients with p53abn endometrial cancer, there was no association between SLN macrometastasis and risk of recurrence (p = .618) or disease specific death (p = .798). CONCLUSIONS SLN macrometastasis is an independent predictor of recurrence and disease-specific death in patients with MMRd endometrial cancer. In the subgroup of p53abn endometrial cancers, SLN macrometastasis did not have an added impact on oncological outcome.
Collapse
Affiliation(s)
- Franziska Siegenthaler
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland.
| | - Sara Imboden
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Carol Büchi
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Lucine Christe
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
| | - Wiebke Solass
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland
| | - Flurina Saner
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Claudia Rauh
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Seline Hofer
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Bettina Schlatter
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Julian Wampfler
- Department of Medical Oncology, Bern University Hospital, Bern, Switzerland
| | - Stefan Mohr
- Department of Gynecology and Obstetrics, Bürgerspital, Solothurn, Switzerland
| | - Andrea Papadia
- Ospedale Regionale di Lugano, Civico, Lugano, Switzerland
| | - Maria Anokhina
- Institute of Pathology, University Hospital and Heinrich-Heine-University Düsseldorf, Germany
| | - Wolfgang Göring
- Institute of Pathology, University Hospital and Heinrich-Heine-University Düsseldorf, Germany
| | - Tilman T Rau
- Institute of Tissue Medicine and Pathology, University of Bern, Switzerland; Institute of Pathology, University Hospital and Heinrich-Heine-University Düsseldorf, Germany
| | - Michael D Mueller
- Department of Obstetrics and Gynecology, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
39
|
Du F, Wu X, He Y, Zhao S, Xia M, Zhang B, Tong J, Xia T. Identification of an Amino Acid Metabolism Reprogramming Signature for Predicting Prognosis, Immunotherapy Efficacy, and Drug Candidates in Colon Cancer. Appl Biochem Biotechnol 2025; 197:714-734. [PMID: 39222169 DOI: 10.1007/s12010-024-05049-4] [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] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Colon cancer ranked third among the most frequently diagnosed cancers worldwide. Amino acid metabolic reprogramming was related to the occurrence and development of colon cancer. We looked for the amino acid metabolism genes (AMGs) associated with amino acid metabolism from molecular signatures database as prognostic markers and constructed amino acid metabolism scoring model (AMS). According to AMS, the patients were divided into high AMS and low AMS groups, and the prognostic characteristics, molecular phenotypes, somatic cell mutation characteristics, immune cell infiltration characteristics, and immunotherapy effect of the two groups were systematically analyzed. Finally, the compounds targeting AMGs were also screened. We screen out 6 prognostic AMGs (P < 0.05) and construct an AMS model based on them. K-M curve indicated that OS in low AMS group was significantly higher than that in high group (P < 0.05), which were validated in multiple datasets. And different AMS groups had different molecular phenotypes, somatic cell mutation characteristics and immune cell infiltration characteristics. Low AMS group had a better effect for immunotherapy. In addition, we predicted potential therapeutic compounds that could bind to AMGs target proteins. AMS model can be used as a hierarchical tool to evaluate the prognosis, immune infiltration characteristics and immunotherapy response ability of colon cancer. And the compounds screened based on AMGs may become new anti-tumor drugs.
Collapse
Affiliation(s)
- Fenqi Du
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Xiangxin Wu
- Ganzhou Cancer Hospital, Ganzhou, Jiangxi Province, People's Republic of China
| | - Yibo He
- Department of Acupuncture Massage & Rehabilitation, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, Shandong Province, People's Republic of China
| | - Shihui Zhao
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Mingyu Xia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Bomiao Zhang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China
| | - Jinxue Tong
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China.
| | - Tianyi Xia
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin Medial University, Harbin, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
40
|
Zhang D, Jiang M, Li P, Laster KV, Zhao D, Zhi Y, Wei H, Nie W, Gao Y, Wu Q, Xiang P, He X, Liu K, Dong Z. CHI-KAT8i5 suppresses ESCC tumor growth by inhibiting KAT8-mediated c-Myc stability. Cell Rep 2025; 44:115135. [PMID: 39772391 DOI: 10.1016/j.celrep.2024.115135] [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: 04/25/2024] [Revised: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The integrated analysis of histone modifier enzymes in solid tumors, especially in esophageal squamous cell carcinoma (ESCC), is still inadequate. Here, we investigate the expression levels of histone modifier enzymes in ESCC tissues. Notably, KAT8 (lysine acetyltransferase 8) is identified as a prognostic and therapeutic biomarker in ESCC. Esophageal-tissue-specific deletion of KAT8 in mice led to less tumor burden after induction of tumorigenesis via 4-nitroquinoline N-oxide (4NQO) treatment compared with wild-type mice. Meanwhile, silencing KAT8 significantly suppresses tumor growth in cell-line-derived xenograft (CDX) and patient-derived xenograft (PDX) models. Mechanically, we confirm that KAT8 regulates c-Myc protein stability by directly binding it. Furthermore, we design and screen a specific KAT8 inhibitor (CHI-KAT8i5) that significantly attenuates tumor growth in vitro and in vivo, providing promising potential for clinical application. Thus, our work identifies that KAT8 could serve as a potential clinically relevant biomarker and therapeutic target in patients with ESCC and that KAT8 inhibitor is a promising lead candidate for ESCC therapy.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Ming Jiang
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Pan Li
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Kyle Vaughn Laster
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Dengyun Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Yafei Zhi
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Huifang Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Yunfeng Gao
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Qiong Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Pu Xiang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450000 Henan, China
| | - Xinyu He
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000 Henan, China.
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000 Henan, China.
| |
Collapse
|
41
|
Nikanjam M, Kato S, Allen T, Sicklick JK, Kurzrock R. Novel clinical trial designs emerging from the molecular reclassification of cancer. CA Cancer J Clin 2025. [PMID: 39841128 DOI: 10.3322/caac.21880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Next-generation sequencing has revealed the disruptive reality that advanced/metastatic cancers have complex and individually distinct genomic landscapes, necessitating a rethinking of treatment strategies and clinical trial designs. Indeed, the molecular reclassification of cancer suggests that it is the molecular underpinnings of the disease, rather than the tissue of origin, that mostly drives outcomes. Consequently, oncology clinical trials have evolved from standard phase 1, 2, and 3 tissue-specific studies; to tissue-specific, biomarker-driven trials; to tissue-agnostic trials untethered from histology (all drug-centered designs); and, ultimately, to patient-centered, N-of-1 precision medicine studies in which each patient receives a personalized, biomarker-matched therapy/combination of drugs. Innovative technologies beyond genomics, including those that address transcriptomics, immunomics, proteomics, functional impact, epigenetic changes, and metabolomics, are enabling further refinement and customization of therapy. Decentralized studies have the potential to improve access to trials and precision medicine approaches for underserved minorities. Evaluation of real-world data, assessment of patient-reported outcomes, use of registry protocols, interrogation of exceptional responders, and exploitation of synthetic arms have all contributed to personalized therapeutic approaches. With greater than 1 × 1012 potential patterns of genomic alterations and greater than 4.5 million possible three-drug combinations, the deployment of artificial intelligence/machine learning may be necessary for the optimization of individual therapy and, in the near future, also may permit the discovery of new treatments in real time.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
| | - Shumei Kato
- Division of Hematology-Oncology, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
| | | | - Jason K Sicklick
- Moores Cancer Center, University of California San Diego Health, La Jolla, California, USA
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, San Diego, California, USA
- Department of Pharmacology, University of California San Diego, San Diego, California, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin, USA
- Worldwide Innovative Networking in Personalized Cancer Medicine Consortium, Paris, France
| |
Collapse
|
42
|
Rzepecka IK, Tysarowski A, Konopka B, Dansonka-Mieszkowska A, Kupryjanczyk J. High Frequency of PIK3R1 Alterations in Ovarian Cancers: Clinicopathological and Molecular Associations. Cancers (Basel) 2025; 17:269. [PMID: 39858051 PMCID: PMC11764438 DOI: 10.3390/cancers17020269] [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: 11/22/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The phosphoinositide 3-kinase (PI3K) pathway is activated in multiple cancers. However, the significance of PIK3R1 encoding the PI3K regulatory subunit, an inhibitor of the PI3K catalytic subunit encoded by PIK3CA, in ovarian cancer development is largely unknown. METHODS Here, we investigated PIK3R1 genomic alterations and gene expression by direct sequencing and qPCR methods in 197 ovarian cancers. The results were correlated with clinicopathological and molecular variables and patient outcomes. RESULTS In addition to mutations (3.5%) and allelic losses (28.4%), we observed a very high frequency of decreased PIK3R1 mRNA levels in ovarian carcinomas (95.8%). Tumors with PIK3R1 mutations mostly represented low-stage cancers of endometrioid and clear-cell type. Tumors with PIK3R1 deletion and underexpression shared similar phenotypes of high-grade carcinomas (p = 0.003 and p = 0.025, respectively). Allelic loss was also associated with advanced stages (p = 0.003) and high-grade serous histotypes (p = 0.004). The PIK3R1 copy number correlated with mRNA levels (p = 0.009). PIK3R1 mutations coexisted with PTEN mutations (p = 0.041), whereas PIK3R1 deletion and underexpression were linked to PIK3CA amplification (p = 0.038 and p = 0.033, respectively). Low PIK3R1 expression diminished the probability of a complete response (OR 0.07, p = 0.03) in patients treated with platinum-based regimens. CONCLUSIONS PIK3R1 alterations may contribute to the development of ovarian cancers with different malignant potential and molecular changes. The high frequency of PIK3R1 aberrations suggests their importance in PI3K pathway deregulation, and they may potentially serve as an alternative to PIK3CA markers for therapy with these pathway inhibitors.
Collapse
Affiliation(s)
- Iwona K. Rzepecka
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (B.K.); (A.D.-M.)
| | - Andrzej Tysarowski
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (B.K.); (A.D.-M.)
| | - Bozena Konopka
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (B.K.); (A.D.-M.)
| | - Agnieszka Dansonka-Mieszkowska
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.T.); (B.K.); (A.D.-M.)
| | - Jolanta Kupryjanczyk
- Department of Cancer Pathomorphology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| |
Collapse
|
43
|
Barakat A, El-Senduny FF, Islam MS, Al-Majid AM, Elshaier YAMM, Mazyed EA, Badria FA. Nanoformulation of Spirooxindole and Methods for Treating Hepatocellular Carcinoma. Pharmaceutics 2025; 17:93. [PMID: 39861743 PMCID: PMC11768502 DOI: 10.3390/pharmaceutics17010093] [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: 11/07/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Objectives: This in vivo study introduces a newly developed spirooxindole derivative that is deemed safe and effective as a potential targeted therapy for various cancers. Methods: Extensive in vivo investigations, including histopathology, immunohistochemistry, and molecular biology, validated its potential for further preclinical and clinical exploration, necessitating comprehensive examinations of its bioavailability, pharmacodynamics, and pharmacokinetics. Additionally, this study involves the development of a commercially viable proniosomal drug delivery system for the compound, facilitating controlled drug release. Results: The data revealed efficacy of spirooxindole derivative in halting the progression of liver cancer, metastasis, and portal vein thrombosis, with potential implications for enhancing regeneration and recovery of early-stage cancer cells in multiple organs, thereby improving recovery rates and remission among cancer patients. The proniosomes, loaded with the compound, exhibited high entrapment efficiency and prolonged drug release rates of up to 12 h in vitro. The optimized formula demonstrated superior drug release percentages and stability compared to conventional niosomes. Further analysis via FTIR and DSC confirmed the absence of chemical interactions and proper entrapment of the compound within the nanovesicles, indicating a stable and effective drug delivery system. Conclusions: This study presents a novel, safe, and effective chemical entity of spirooxindole derivatives for further preclinical and clinical studies.
Collapse
Affiliation(s)
- Assem Barakat
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.I.); (A.M.A.-M.)
| | - Fardous F. El-Senduny
- Department of Pathology & Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, FL 33136, USA;
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Mohammad Shahidul Islam
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.I.); (A.M.A.-M.)
| | - Abdullah Mohammed Al-Majid
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.S.I.); (A.M.A.-M.)
| | - Yaseen A. M. M. Elshaier
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Menoufiya 32958, Egypt;
| | - Eman A. Mazyed
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Kaferelsheikh University, Kaferelsheikh 33516, Egypt;
| | - Farid A. Badria
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
44
|
Liu M, Chen P, Wei B, Tan HL, Zhao YX, Ai L, Li N, Jiang YK, Lin J, Li SJ, Chang S. FN1 shapes the behavior of papillary thyroid carcinoma through alternative splicing of EDB region. Sci Rep 2025; 15:327. [PMID: 39747903 PMCID: PMC11695688 DOI: 10.1038/s41598-024-83369-5] [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: 06/30/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Papillary thyroid cancer (PTC) is often characterized by indolent behavior, small tumors with slow cell proliferation and a tendency to metastasize to cervical lymph node simultaneously, and the molecular mechanisms underlying that remain poorly understood. In this study, FN1 was the hottest gene of PTC and distinctive expression in PTC cells. FN1 deficiency severely inhibited the p53 signaling pathway, especially cyclin proteins, resulting in increased cell growth but hampered invasion. The alternatively splicing EDB region of FN1 was exclusively expressed in tumors, which impacted integrin β1 (ITGB1) bonding FN1 and its secretion process, resulting in completely distinct roles of two isoforms that FN1 including and skipping EDB domain. The isoform EDB(-)FN1 intracellularly inhibited tumor proliferation by upregulating p21 expression, whereas extracellular EDB(+)FN1 promoted lymph node metastasis via the VEGF signaling pathway in vitro and in vivo. Moreover, the alternative splicing EDB region of FN1 was modulated by p53-targeted protein ZMAT3 which activated cell migration and lymphoangiogenesis. Collectively, combined with p53-induced proteins, FN1 played both anti- and pro-cancer roles owing to EDB domain alternative splicing. FN1 is a potential determinant behind the characteristic behavior of PTC, which may contribute to a deeper understanding of the peculiarity of PTC and provide a promising target for regional lymph node metastasis.
Collapse
Affiliation(s)
- Mian Liu
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, China
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan, China
| | - Pei Chen
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Bo Wei
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Hai-Long Tan
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Ya-Xin Zhao
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Lei Ai
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Ning Li
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Ying-Ke Jiang
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Jing Lin
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Shi-Jin Li
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China
| | - Shi Chang
- Department of General Surgery, XiangYa Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China.
- Clinical Research Center for Thyroid Disease in Hunan Province, Xiangya Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China.
- Hunan Provincial Engineering Research Center for Thyroid and Related Diseases Treatment Technology, Xiangya Hospital Central South University, No. 87 XiangYa Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
45
|
Wang X, Bao S, Jiang M, Zou X, Yin Y. Clinical, pathological and gene expression profiling of estrogen receptor discordance in breast cancer. Clin Transl Oncol 2025; 27:233-256. [PMID: 38926258 DOI: 10.1007/s12094-024-03547-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Breast cancer (BC) is the world's largest tumor species in which hormone receptor-positive patients have relatively good prognosis. However, majority of patients will develop late resistance, one of the important factors is due to the loss of the original estrogen receptor (ER) expression. METHODS We conducted this study in 115 patients with BC who experienced second biopsy at Jiangsu Province Hospital (JSPH) and divided patients into two subgroups ER + to - and ER + to + . First, clinicopathological characteristics between two groups were evaluated. Second, we explored candidate genes related to BC ER intratumor heterogeneity by applying next-generation sequencing (NGS) in 42 patients. Multi-omics integrative analysis of tumor transcriptomic, cancer-related pathway, diagnostic and prognostic value and immune profile were conducted. Besides, preliminary assay were also used to evaluate the correlation between KMT2C and ERα (ESR1) expression. The CCK-8, 5-Ethynyl-2'-deoxyuridine (EdU) assays, Transwell assays and the wound scratch tests were applied to explore the cellular interactions between KMT2C and BC. RESULTS We find the histological type (p = 0.008) and disease-free survival (DFS) (p = 0.004) were significantly different in two subgroups. In Cox survival analysis, metastasis (Hazard ratio (HR) > 1, p = 0.007) and neo-adjuvant (HR < 1, p < 0.001) are independent prognostic factors of DFS. Besides, by analyzing NGS results, we found four genes KMT2C, FGFR19, FGF1 and FGF4 were highly mutated genes in ER + to - subgroup. Furthermore, the gene KMT2C displayed significant diagnostic value and prognostic value in BC and pan-cancer. In addition, a positive correlation between KMT2C expression and immune infiltrating levels of T cell CD4 + , macrophage and neutrophil was found. In the end, Western blot and RT-qPCR assay were used and found KMT2C and ERα (ESR1) expressions are strongly positive correlated in mRNA and protein level. Inhibition of KMT2C significantly reduced proliferation, invasion, and migration of MCF7 cells. CONCLUSION People in two cohorts from JSPH presented different clinical characteristics and prognosis. The gene KMT2C may affect the progression of BC by regulating the molecular, epigenetic activity and immune infiltration. It may also serve as a novel prognostic biomarker for BC patients who underwent ER status converted from positive to negative.
Collapse
Affiliation(s)
- Xi Wang
- Department of Radiotherapy, Affiliated Hospital 2 of Nantong University (Nantong First People's Hospital), Nantong, 226300, Jiangsu, China
| | - Shengnan Bao
- Department of Oncology, Tumor Hospital Affiliated to Nantong University, Nantong, 226300, Jiangsu, China
| | - Mengping Jiang
- Department of Radiotherapy, Affiliated Hospital 2 of Nantong University (Nantong First People's Hospital), Nantong, 226300, Jiangsu, China
| | - Xian Zou
- Clinical Medicine, School of Medicine, Nantong University, Nantong, 226001, Jiangsu, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
46
|
Campos-León K, Ferguson J, Günther T, Wood CD, Wingett SW, Pekel S, Varghese CS, Jones LS, Stockton JD, Várnai C, West MJ, Beggs A, Grundhoff A, Noyvert B, Roberts S, Parish JL. Repression of CADM1 transcription by HPV type 18 is mediated by three-dimensional rearrangement of promoter-enhancer interactions. PLoS Pathog 2025; 21:e1012506. [PMID: 39869645 PMCID: PMC11801731 DOI: 10.1371/journal.ppat.1012506] [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: 08/15/2024] [Revised: 02/06/2025] [Accepted: 12/02/2024] [Indexed: 01/29/2025] Open
Abstract
Upon infection, human papillomavirus (HPV) manipulates host cell gene expression to create an environment that is supportive of a productive and persistent infection. The virus-induced changes to the host cell's transcriptome are thought to contribute to carcinogenesis. Here, we show by RNA-sequencing that oncogenic HPV18 episome replication in primary human foreskin keratinocytes (HFKs) drives host transcriptional changes that are consistent between multiple HFK donors. We have previously shown that HPV18 recruits the host protein CTCF to viral episomes to control the differentiation-dependent viral transcriptional programme. Since CTCF is an important regulator of host cell transcription via coordination of epigenetic boundaries and long-range chromosomal interactions, we hypothesised that HPV18 may also manipulate CTCF to contribute to host transcription reprogramming. Analysis of CTCF binding in the host cell genome by ChIP-Seq revealed that while the total number of CTCF binding sites is not altered by the virus, there are a sub-set of CTCF binding sites that are either enriched or depleted of CTCF. Many of these altered sites are clustered within regulatory elements of differentially expressed genes, including the tumour suppressor gene cell adhesion molecule 1 (CADM1), which supresses epithelial cell growth and invasion. We show that HPV18 establishment results in reduced CTCF binding at the CADM1 promoter and upstream enhancer. Loss of CTCF binding is coincident with epigenetic repression of CADM1, in the absence of CpG hypermethylation, while adjacent genes including the transcriptional regulator ZBTB16 are activated. These data indicate that the CADM1 locus is subject to topological rearrangement following HPV18 establishment. We tested this hypothesis using 4C-Seq (circular chromosome confirmation capture-sequencing) and show that HPV18 establishment causes a loss of long-range chromosomal interactions between the CADM1 transcriptional start site and the upstream transcriptional enhancer. These data show that HPV18 manipulates host cell promoter-enhancer interactions to drive transcriptional reprogramming that may contribute to HPV-induced disease progression.
Collapse
Affiliation(s)
- Karen Campos-León
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Jack Ferguson
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | | | - C. David Wood
- School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Steven W. Wingett
- The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Selin Pekel
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Christy S. Varghese
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Leanne S. Jones
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Joanne D. Stockton
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Csilla Várnai
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Michelle J. West
- School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Andrew Beggs
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | | | - Boris Noyvert
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
- Birmingham CRUK Centre, University of Birmingham, Birmingham, United Kingdom
| | - Sally Roberts
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Joanna L. Parish
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
- National Institute of Health Research, Biomedical Research Centre, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
47
|
Casalini R, Romei C, Ciampi R, Ramone T, Prete A, Gambale C, Matrone A, Torregrossa L, Ugolini C, Elisei R. Minor role of TP53 and TERT promoter mutations in medullary thyroid carcinoma: report of new cases and revision of the literature. Endocrine 2025; 87:243-251. [PMID: 39179735 DOI: 10.1007/s12020-024-03990-2] [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: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE Aims of this study were to investigate the prevalence of TP53 and TERT mutations in Medullary Thyroid carcinoma (MTC) and their role in inducing aggressiveness in positive cases. METHODS We performed a literature search in PubMed to identify studies investigating the prevalence of TERT and TP53 mutations in MTC. We also included data on MTC cases (n = 193) obtained at our center and unpublished. The in-silico pathogenicity of the TP53 mutations has been evaluated by predictor tools. RESULTS We identified a total of 25 and 11 published papers: all together 1280 cases have been investigated for the presence of TP53 mutations and 974 for TERT promoter mutation. Twenty-five out of 1280 (2%) cases had a TP53 mutation while only 3/974 MTC cases (0.3%) have been found to be positive for TERT promoter mutations. Among all, we identified 19 different TP53 mutations that in 12 cases were demonstrated to have an in silico predicted high pathogenic role and a high impact on protein function. Three non-sense and 4 probably not damaging mutations were also reported. The pathogenic role of the TERT promoter mutations has been previously in vitro determined. No correlation between TP53 and/or TERT mutations and aggressiveness of MTC has been demonstrated. CONCLUSION The prevalence of TP53 and TERT promoter mutations is very low in MTC. The reported mutations are pathogenic in the majority of cases. Because of their rarity it is not possible to clarify if they play or not a role in the pathogenesis and/or aggressiveness of this specific thyroid tumor.
Collapse
Affiliation(s)
- Roberta Casalini
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Cristina Romei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Raffaele Ciampi
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Teresa Ramone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Alessandro Prete
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Carla Gambale
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Antonio Matrone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Liborio Torregrossa
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Unit of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Clara Ugolini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Unit of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Rossella Elisei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy.
| |
Collapse
|
48
|
Nakayama M, Saito H, Murakami K, Oshima H, Oshima M. Missense Mutant p53 Transactivates Wnt/β-Catenin Signaling in Neighboring p53-Destabilized Cells through the COX-2/PGE2 Pathway. CANCER RESEARCH COMMUNICATIONS 2025; 5:13-23. [PMID: 39641656 PMCID: PMC11695814 DOI: 10.1158/2767-9764.crc-24-0471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
SIGNIFICANCE There is intratumor heterogeneity in the stabilization of missense mutant p53, and it has been thought that only cells with nuclear accumulation of mutant p53 have oncogenic function. However, using mouse intestinal tumor-derived organoids, we show that mutant p53-stabilized cells transactivate Wnt/β-catenin signaling in neighboring p53-destabilized cells through activating the COX-2/PGE2 pathway. These results suggest that both p53-stabilized cells and p53-destabilized cells contribute to malignant progression through interaction within the intratumor microenvironment.
Collapse
Affiliation(s)
- Mizuho Nakayama
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Hiroshi Saito
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa, Japan
| | - Kazuhiro Murakami
- Division of Epithelial Stem Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hiroko Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
- WPI Nano-Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| |
Collapse
|
49
|
Kounatidou NE, Vitkos E, Palioura S. Ocular surface squamous neoplasia: Update on genetics, epigenetics and opportunities for targeted therapy. Ocul Surf 2025; 35:1-14. [PMID: 39608452 DOI: 10.1016/j.jtos.2024.11.006] [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: 08/02/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE The purpose of this review is to explore the molecular foundations of ocular surface squamous neoplasia (OSSN), focusing on the genetic and epigenetic aspects. While current management strategies include surgical excision and medical therapies, the understanding of OSSN's molecular basis remains limited, hindering the development of targeted treatments. METHODS A comprehensive MEDLINE search was conducted for literature published between January 1993 and October 2023. Only studies with original data on molecular, genetic, or epigenetic mechanisms, such as mutations, gene expression, and genetic predispositions were included. Articles were excluded if they focused solely on clinical management without addressing these factors, or if they were reviews, editorials, or opinion pieces. RESULTS The search yielded a total of 108 articles, out of which 39 articles met the criteria for further analysis. Investigations into OSSN have identified key DNA mutations in the TP53, HGF, EGFR, TERT, and CDKN2A genes, indicating common oncogenic pathways shared with other squamous cell carcinomas (SCCs). Significant epigenetic changes were identified, including DNA methylation, histone modifications, and altered miRNA expression patterns. Epigenetic dysregulation of critical tumor suppressors and oncoproteins, further highlight the complex genetic landscape of OSSN. CONCLUSION The molecular alterations identified in OSSN not only enhance our understanding of its biology but also have potential as novel biomarkers for early detection, prognostic evaluation, and as therapeutic targets. The identification of genetic and epigenetic markers in OSSN signifies progress towards personalized medicine approaches. Further studies and collaborative efforts are essential to validate these molecular markers and translate them into clinical practice, potentially revolutionizing OSSN management and improving patient outcomes.
Collapse
Affiliation(s)
| | - Evangelos Vitkos
- Department of Oral and Maxillofacial Surgery, Klinikum Dortmund, Dortmund, Germany
| | - Sotiria Palioura
- Department of Ophthalmology, University of Cyprus Medical School, Nicosia, Cyprus.
| |
Collapse
|
50
|
Pevzner K, Simchi N, Arad G, Seger E. Identification of Protein Kinase Drug Targets Using Activity Estimation in Clinical Phosphoproteomics. Methods Mol Biol 2025; 2905:163-169. [PMID: 40163304 DOI: 10.1007/978-1-0716-4418-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
This chapter describes protein kinase (will be termed here kinase) activity estimation methods and their application to clinical cancer phosphoproteomics datasets, proposing a novel approach for identification of protein kinases as therapeutic targets. Despite significant advances in genomics-based target identification, clinical proteomics and phosphoproteomics remain underutilized. We highlight the growing availability of proteomics data from projects like Clinical Proteomic Tumor Analysis Consortium (CPTAC) and Proteomics Identifications Database (PRIDE), and review key kinase activity estimation algorithms, including PTM-SEA, KSEA, Rokai, KStar, and Kinome Atlas. Applying these methods on clinical phosphoproteomic data, we demonstrate the identification of hyperactivated kinases in specific cancer indications and highlight HER2 and EGFR as benchmarks. Our description underscores the potential of integrating kinase activity estimation with clinical phosphoproteomics to uncover new therapeutic targets and develop precision oncology therapies.
Collapse
|