{"title":"Etiology-based Molecular Characterization of Hepatocellular Carcinoma Reveals SQLE's Contribution to Immunosuppressive Microenvironment.","authors":"Kangkang Yu, Qisheng Zhong, Jubo Zhang, Chong Huang","doi":"10.2174/0115680096370558250514063841","DOIUrl":"https://doi.org/10.2174/0115680096370558250514063841","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is a kind of fatal cancer with a variety of risk factors. However, a pan-etiology molecular characterization of HCC has not been explored.</p><p><strong>Objective: </strong>The objective of this study is to explore etiology-specific features of HCC and im-prove our understanding of tumorigenesis, thereby revealing potential therapeutic targets.</p><p><strong>Methods: </strong>RNA-seq, genetic alteration, copy number alteration, and clinicopathological data of The Cancer Genome Atlas -Liver Hepatocellular Carcinoma cohort were downloaded and explored. Immune-related features and single-cell and spatial transcriptomic data were adopted and analyzed.</p><p><strong>Results: </strong>Etiology-based analyses revealed that HCC with different etiologies showed different clinical features, including gender composition, ethnic composition, clinical stage distribution, and survival. In addition, distinct genetic alterations, copy number alterations, and tumor microenvironment were observed in HCC patients with different etiologies. Significantly enhanced expression of squalene epoxidase (SQLE) was observed in viral hepatitis-related HCC and was associated with poor tumor grade and overall survival. Correlation analysis revealed a negative relationship between SQLE expression and anti-tumor immunity. Single-cell and spatial transcriptomics demonstrated that SQLE contributed to reduced T cell and NK cell infiltration while increasing macrophage and monocyte infiltration.</p><p><strong>Conclusion: </strong>The current study demonstrated that HCC has etiology-specific clinical features, genetic alteration, copy number alteration, and tumor microenvironment. Overexpression of SQLE in viral hepatitis-related HCC correlate with poor clinical outcome and may contribute to reduce T cell and NK cell infiltration while increased macrophage and monocyte infiltration, which lead to immunosuppressive microenvironment and can be an actionable target.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Genetic Distinctiveness in Lung Adenocarcinoma and Lung Squamous Cell Carcinoma Cancer: Identifying Key Genomic Signatures through Differentially Expressed Gene Analysis.","authors":"Patrick Shi, Wenqiang Chen","doi":"10.2174/0115680096372957250322174718","DOIUrl":"https://doi.org/10.2174/0115680096372957250322174718","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is the most commonly diagnosed cancer type and the leading cause of cancer death worldwide. Non-small cell lung cancer (NSCLC) accounts for more than 80% of all lung cancer cases and includes two main subtypes: lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). Understanding the differences in genes causing the proliferation of LUAD and LUSC is key to advancing the diagnosis and targeted treatment development.</p><p><strong>Aims: </strong>The aim of this study was to identify candidate genes and potential tumorigenesis mechanisms distinguishing LUAD and LUSC.</p><p><strong>Methods: </strong>Three pooled transcriptomic datasets (GSE10245, GSE37745, and GSE43580) were analyzed from the Gene Expression Omnibus (GEO) database, with each dataset statistically tested for differentially expressed genes (DEGs). DEGs between lung LUAD and LUSC of the three datasets were analyzed with Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. A protein-protein interaction (PPI) network was constructed to screen candidate genes.</p><p><strong>Results: </strong>This study identified 138 shared DEGs among three patient-level gene expression datasets, containing 39 upregulated genes and 99 downregulated genes. The GO and KEGG enrichment analysis results showed the functions of DEGs to be mainly associated with epidermis development, cornified envelope, structural constituent of epidermis, and estrogen signaling pathway. Finally, through the PPI network, eight core genes were identified, including KRT14, KRT5, KRT6A, KRT16, SPRR1A, SPRR1B, SPRR3, and KRT6B.</p><p><strong>Conclusion: </strong>We have elucidated key genes and molecular mechanisms linked to NSCLC subtypes. These findings have the potential to facilitate improved diagnostic and therapeutic targets for LUAD and LUSC biomarkers.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Trimethylamine N-oxide Impairs Oocyte Maturation and Embryogenesis via NF-κB/NLRP3 Pathway Modulation.","authors":"Fengping He, Yongmei Zhang, Yanle Guo, Tizhen Yan, Jiwu Lou","doi":"10.2174/0115680096364675250419135908","DOIUrl":"https://doi.org/10.2174/0115680096364675250419135908","url":null,"abstract":"<p><strong>Background: </strong>The role of Trimethylamine N-oxide (TMAO) in oocyte maturation and embryogenesis remains unclear, particularly its impact on ovarian granulosa cells (OGCs) and its underlying mechanisms.</p><p><strong>Methods: </strong>This study examined the effects of TMAO (100-400 μmol/L) on oocyte maturation, cumulus cell expansion, mitochondrial distribution, and embryonic development in vitro and in a BALB/c mouse model. The involvement of the NF-κB/NLRP3 signaling pathway in TMAO-induced ovarian dysfunction was assessed using Western blotting and gene expression analyses. The potential therapeutic effect of miRNA-146, an NF-κB inhibitor, was also explored.</p><p><strong>Results: </strong>Western blotting confirmed that TMAO activates the NF-κB signaling pathway and induces the synthesis of caspase 3 and NLRP3 complexes. However, pretreatment with miRNA-146, an NF-κB inhibitor, significantly reduced inflammation and inflammatory gene expression during TMAO therapy. Additionally, miRNA-146 pretreatment promoted oocyte maturation by suppressing NF-κB/NLRP3 activation, OGCs apoptotic inflammatory factor expression, and the gene expression of NF-κB, caspase 3, and NLRP3.</p><p><strong>Conclusion: </strong>Findings demonstrate that TMAO disrupts oocyte development through NF- κB/NLRP3 activation, contributing to ovarian dysfunction. Notably, targeting TMAO and its downstream signaling could serve as a novel therapeutic strategy for premature ovarian insufficiency (POI).</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144141784","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Extraordinary Benefit of Regorafenib in Metastatic Cholangiocarcinoma: A Case Report.","authors":"Ayberk Bayramgil, Tugce Kubra Gunes, Fatma Gulcicek Ayranci, Duygu Yoruk Atik, Busra Nigdelioglu, Melike Ozcelik, Ozgecan Dulgar Kaya","doi":"10.2174/0115680096378189250422100120","DOIUrl":"https://doi.org/10.2174/0115680096378189250422100120","url":null,"abstract":"<p><strong>Introduction/background: </strong>Cholangiocarcinoma (CC) is a rare, aggressive cancer of the bile ducts with limited treatment advancements over recent decades. The five-year survival rate for CC remains low, around 10%, and even lower for advanced cases.</p><p><strong>Case presentation: </strong>This case report discusses a 46-year-old woman with metastatic CC who achieved remarkable progression-free survival with regorafenib, a multikinase inhibitor typi-cally used in other cancers. After multiple lines of treatment, regorafenib was given as a 7th-line therapy. Despite initial intolerance requiring dose reduction, the patient achieved 22 months of progression-free survival (PFS) with stable disease and partial regression in some areas. Her response to regorafenib significantly exceeded typical outcomes in the literature, where PFS generally ranges from 3 to 4 months. This improvement may be attributed to an FGFR2 mutation identified via next-generation sequencing, which regorafenib may effec-tively target.</p><p><strong>Conclusion: </strong>This case suggests that patients with FGFR2 mutations might benefit from regorafenib, warranting further studies to explore this associationAs can be applied as a promising target gene to develop drug resistance and remedial efficacy.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144101502","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ibrahim Karaduman, Hatice Terzi, Seyma Tastemur, Mehmet Şencan
{"title":"Evaluation of the Antitumoral Effect of PI3K/AKT/mTOR Pathway Inhibition and Antioxidant Combination on Burkitt Lymphoma Cell Line.","authors":"Ibrahim Karaduman, Hatice Terzi, Seyma Tastemur, Mehmet Şencan","doi":"10.2174/0115680096366040250414114030","DOIUrl":"https://doi.org/10.2174/0115680096366040250414114030","url":null,"abstract":"<p><strong>Background and aim: </strong>Burkitt lymphoma is a highly aggressive type of B-cell non-Hodgkin lymphoma. While it is more commonly seen in children and adolescents, it accounts for about 1-2% of adult lymphomas. Since it has high Ki-67 ratios, both its progression rate and sensitivity to chemotherapy are high. However, treatment success decreases in chemotherapy-resistant relapsed/refractory groups. Therefore, new and less toxic therapeutic agents targeting biological pathways related to Burkitt lymphoma pathogenesis are needed. In our study, we aimed to determine the antitumoral efficacy of PI3K/AKT/mTOR pathway inhibitor ETP 45658 and antioxidant Resveratrol, which play a role in Burkitt lymphoma pathogenesis on a Burkitt lymphoma cell line.</p><p><strong>Methods: </strong>Burkitt lymphoma Raji cell line was used in our study. ETP 45658 was applied to the cell line at concentrations of 50, 25, 12.5, 1, 0.1, and 0.01 μM, both alone and in combination with 100 μM resveratrol. The cytotoxic effects of ETP 45658 and the combination treatment on Raji cells were then evaluated using an in vitro XTT cell viability test. IC50 value was calculated according to the results obtained in the XTT test. Based on the IC50 value, the apoptosis assay was studied using the flow cytometry method.</p><p><strong>Results: </strong>Our research showed that increasing doses of ETP 45658, individually and in combination, had a statistically significant cytotoxic effect on Burkitt lymphoma Raji cells. This cytotoxic effect was found to be higher in combination treatment. In apoptosis experiments with the calculated IC50 value, it induced apoptosis weakly.</p><p><strong>Conclusion: </strong>Our study showed that ETP 45668 alone and in combination with resveratrol has antitumoral activity on the Burkitt lymphoma cell line. PI3K/Akt/mTOR pathway inhibitors may be an alternative treatment option in patients with relapsed/refractory Burkitt lymphoma. However, more preclinical and clinical studies are needed.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144076561","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Promotion of Melanoma Progression through MCM4-Induced Immune Suppression and Polarization of Macrophages by Carcinogenic Exosomes.","authors":"Xuewei Zhang, Sirong Liu, Deni Kang, Ronghua Yang","doi":"10.2174/0115680096357750250507073948","DOIUrl":"https://doi.org/10.2174/0115680096357750250507073948","url":null,"abstract":"<p><strong>Objective: </strong>This study aims to provide a comprehensive understanding of the crucial role of MCM4 in melanoma progression regarding the regulatory communication between macrophages and cancer cells mediated by extracellular vesicles.</p><p><strong>Methods: </strong>Initially, a preliminary analysis was conducted using the Tumor Immune Estimation Resource (TIMER) database. Subsequently, the role of MCM4 knockdown on the polarization of THP- 1 and RAW264.7 macrophages was observed. Finally, the biological functionalities of exosomes derived from A375 cells overexpressing MCM4 on normal melanocytes (HEM-L) were explored.</p><p><strong>Results: </strong>On the one hand, MCM4 knockdown resulted in the upregulation of M1 macrophage markers and downregulation of M2 macrophage markers, indicating that MCM4 could facilitate polarization of macrophages toward the M2 phenotype and suggesting its oncogenic potential. On the other hand, MCM4 overexpression in melanocytes increased the secretion of exosomes, enhancing the proliferation, clonogenic, and DNA synthesis abilities of normal melanocytes. In addition, MCM4 overexpression-induced secretion of exosomes promoted the migration and invasion capabilities of normal melanocytes.</p><p><strong>Conclusion: </strong>Exosomes secreted by MCM4-overexpressed melanocytes could stimulate their proliferation, migration, and invasion abilities. MCM4 promoted M2 polarization of macrophages, indicating its crucial role in tumor microenvironment formation and thereby facilitating tumor development.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144076562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advances and Future Prospects of Lipid-Based Nanocarriers in Targeted Cancer Therapy: A Comprehensive Review.","authors":"Suraj Mandal","doi":"10.2174/0115680096351325250412164059","DOIUrl":"https://doi.org/10.2174/0115680096351325250412164059","url":null,"abstract":"<p><strong>Introduction: </strong>Targeted cancer therapy has significantly transformed the field of oncology by improving treatment precision and reducing the occurrence of unintended side effects. Lipid-based nanocarriers have become a viable platform for delivering chemothera-peutic drugs due to their biocompatibility, flexibility, and ability to be functionalised.</p><p><strong>Aim and objectives: </strong>The objective of this research is to investigate the present tactics and future prospects of lipid-based nanocarriers in the field of targeted cancer treatment. The goals include the examination of recent progress in the development of lipid nanocarriers, evaluating their effectiveness in clinical uses, and pinpointing opportunities for enhancement and novelty.</p><p><strong>Material and method: </strong>An extensive literature analysis was performed, including examining current research on the use of lipid-based nanocarriers in cancer treatment. An analysis was conducted on data pertaining to nanocarrier formulations, targeting mechanisms, treatment results, and clinical trials in order to detect trends and new technologies.</p><p><strong>Results: </strong>Recent progress in lipid-based nanocarriers, including liposomes, solid lipid nano-particles, and nanoemulsions, has shown enhanced drug delivery and decreased toxicity. Targeting methods, such as the utilisation of ligand-receptor interactions and stimuli-responsive systems, have significantly improved the precision and effectiveness of therapeu-tics.</p><p><strong>Conclusion: </strong>Lipid-based nanocarriers have great potential for advancing targeted cancer treatment in the future. Although there has been significant progress, it is essential to con-tinue developing formulation processes and targeting tactics to overcome existing con-straints and improve clinical results. Ongoing cooperation across several fields of study will be crucial in effectively applying these nanotechnologies from the laboratory to practical use in healthcare.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144076516","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Exploring the Role of Long Non-Coding RNAs in Mediating Cisplatin Resistance in Colorectal Cancer.","authors":"Shirin Azizidoost, Omid Anbiyaee, Atefeh Bahmani, Maryam Khombi Shooshtari, Pezhman Alavinejad, Sara Zarasvandnia, Maryam Farzaneh","doi":"10.2174/0115680096376480250408070103","DOIUrl":"https://doi.org/10.2174/0115680096376480250408070103","url":null,"abstract":"<p><p>Colorectal cancer (CRC) usually begins as adenomatous polyps in the colorectal or rectal epithelial cells. Currently, there are no reliable biomarkers for early CRC screening or prognostic prediction, leading to late-stage diagnoses when surgical options may no longer be viable. The disease is driven by mutations in oncogenes, tumor suppressor genes, and DNA repair genes, with rapid growth and metastasis contributing to treatment failure. Over the past two decades, research on non-coding RNAs (ncRNAs), particularly long ncRNAs (lncRNAs), has expanded significantly, revealing their critical roles in cancer biology. LncRNAs are involved in numerous biological processes such as cell proliferation, apoptosis, metabolism, and drug resistance, and they are often abnormally expressed in various cancers, including hepatocellular carcinoma, pancreatic cancer, and bladder cancer. In CRC, lncRNAs play a regulatory role by influencing cell cycle, proliferation, apoptosis, and epithelial-mesenchymal transition, and some have been shown to affect CRC cell proliferation, invasion, and resistance to cisplatin, highlighting their potential as therapeutic targets and biomarkers in cancer treatment. This review highlights current investigations on the functions and mechanisms of lncRNAs in cisplatin resistance in CRC. Such overview is anticipated to contribute to figuring out that lncRNAs can be applied as a promising target gene to develop drug resistance and remedial efficacy.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143972300","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Promising Response to Pyrotinib in Non-Small-Cell Lung Cancer with the Rare HER2 R456C Mutation: A Case Report.","authors":"Yajie Wang, Jiaqi Hu, Runze Liu, Pei Li, Luokun Wang, Guangjian Yang","doi":"10.2174/0115680096371951250409093625","DOIUrl":"https://doi.org/10.2174/0115680096371951250409093625","url":null,"abstract":"<p><strong>Background: </strong>HER2 exon 20 insertions exhibit relative resistance to chemotherapy and covalent HER2-targeted tyrosine kinase inhibitors (TKIs) in non-small-cell lung cancer (NSCLC). Furthermore, specific missense mutations in the extracellular domain of the HER2 protein have been identified as oncogenic drivers in NSCLC. However, their structural properties and clinical response to HER2-targeted inhibitors remain poorly understood, warranting further investigation. R456C represents a rare exon 12 missense mutation in the HER2 extracellular domain, with limited documentation in NSCLC.</p><p><strong>Case presentation: </strong>This study presents an atypical case of NSCLC with a HER2 R456C mutation, where the patient experienced a favorable response and substantial survival benefit from the HER2-targeted inhibitor pyrotinib. A patient, a 65-year-old man diagnosed with stage IIIB lung adenocarcinoma, initially underwent radical concurrent chemoradiotherapy. Upon disease recurrence, polymerase chain reaction assay detected no oncogenic alterations, and programmed cell death ligand 1 (PD-L1) expression was negative. Chemotherapy in combination with bevacizumab resulted in stable disease, providing a progression-free survival (PFS) benefit of 6 months. However, anlotinib proved ineffective against brain metastasis, necessitating brain radiotherapy. A subsequent lung biopsy confirmed adenocarcinoma and next-generation sequencing identified a somatic HER2 exon 12 missense mutation, p.R456C. Following pyrotinib administration, the patient's pulmonary metastases significantly diminished, and the brain metastasis regressed, resulting in a partial response and a PFS benefit of 13 months.</p><p><strong>Conclusion: </strong>To the best of our knowledge, this study represents the first reported case demonstrating the promising efficacy of pyrotinib in HER2-altered NSCLC harboring the ra-re exon 12 R456C mutation. Heterogeneous alterations in the HER2 extracellular segment, such as R456C, may be targetable and could confer survival benefits with HER2-targeted inhibitors.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143995271","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hany A Al-Hussaniy, Amjad I Oraibi, Zahraa Salam Al-Tameemi, Ali Hikmat Alburghaif, Meena Akeel Naji, Fatima Akeel Naji
{"title":"The Association of KRAS and P53 Gene Mutations and MDM2 Expression with the Occurrence of Colorectal Cancer.","authors":"Hany A Al-Hussaniy, Amjad I Oraibi, Zahraa Salam Al-Tameemi, Ali Hikmat Alburghaif, Meena Akeel Naji, Fatima Akeel Naji","doi":"10.2174/0115680096349198250407100724","DOIUrl":"https://doi.org/10.2174/0115680096349198250407100724","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC) encompasses various cancers located in the rectosigmoid junction, rectum, and anus, as well as parts of the colon. Globally, CRC is the second leading cause of cancer-related mortality and the third most prevalent malignan-cy. The KRAS oncogene was found to be mutated in 30 to 50% of CRC cases, leading to dysregulated cellular functions. Furthermore, the connection between KRAS and P53 gene mutations and the incidence of both colorectal and breast cancer remains an area of interest.</p><p><strong>Method: </strong>This comprehensive narrative review was carried out by mining data from recog-nized databases, such as PubMed, Google Scholar, and ResearchGate. The purpose was to extensively explore and understand the association between the KRAS and P53 gene muta-tions and the prevalence of colorectal and breast cancers.</p><p><strong>Results: </strong>The mutation in the KRAS oncogene has been identified as a key player in cellular signaling pathways, including MAPK, PI3K, and PLD. Despite extensive research, gene therapies targeting these mutations have seen limited success, especially in codons 12, 13, 61, and 143.</p><p><strong>Conclusion: </strong>Mutations in the KRAS and P53 genes, along with aberrant MDM2 expression, play pivotal roles in the onset and progression of colorectal cancer by disrupting key cellular signaling pathways, such as MAPK, PI3K, and PLD. Despite advancements in understand-ing these mechanisms, current gene therapy approaches have shown limited success, particu-larly in targeting KRAS codon mutations. This underscores the urgent need for innovative therapeutic strategies and further research to develop effective treatments for colorectal can-cer and its potential links to other malignancies, such as breast cancer.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":" ","pages":""},"PeriodicalIF":2.3,"publicationDate":"2025-05-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143972710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}