Current Cancer Drug Targets - Volume 25, Issue 12, 2025
Volume 25, Issue 12, 2025
-
-
Enhanced Detection of Gastrointestinal Malignancies using Machine Learning-Optimized Liquid Biopsy: A Mini Review
More LessAuthors: Shankar Ganesh Muthusamy and Venkateswaramurthy NallasamyBackgroundGastrointestinal (GI) cancers represent some of the most common and lethal malignancies globally, underscoring the urgent need for improved diagnostic strategies. Traditional diagnostic methods, while effective to some degree, are often invasive and unsuitable for regular screenings.
ObjectiveThis review article explores integrating machine learning (ML) with liquid biopsy techniques as a revolutionary approach to enhance the detection and monitoring of GI cancers. Liquid biopsies offer a non-invasive alternative for cancer detection through the analysis of circulating tumor DNA (ctDNA) and other biomarkers, which when combined with ML, can significantly improve diagnostic accuracy and patient outcomes.
MethodsWe conducted a comprehensive review of recent advancements in liquid biopsy and ML, focusing on their synergistic potential in the early detection of GI cancers. The review addresses the application of next-generation sequencing and digital droplet PCR in enhancing the sensitivity and specificity of liquid biopsies.
ResultsMachine learning algorithms have demonstrated remarkable ability in navigating complex datasets and identifying diagnostically significant patterns in ctDNA and other circulating biomarkers. Innovations such as machine learning-enhanced “fragmentomics” and tomographic phase imaging flow cytometry illustrate significant strides in non-invasive cancer diagnostics, offering enhanced detection capabilities with high accuracy.
ConclusionThe integration of ML in liquid biopsy represents a transformative step in the early detection and personalized treatment of GI cancers. Future research should focus on overcoming current limitations, such as the heterogeneity of tumor-derived genetic materials and the standardization of liquid biopsy protocols, to fully realize the potential of this technology in clinical settings.
-
-
-
Perspectives on Ergodic Cancer Therapy Derived from Cloning Genome Chaos via In Vivo Rhabdomyosarcoma RA-2 Models: a Narrative Review
More LessAuthors: Sergey Shityakov, Natalia Lubinets and Viacheslav KravtsovThis review explores articles concerning the experimental research cycle on genome instability in cell populations of highly malignant recurrent organotropic rhabdomyosarcoma RA-2 in rats. Clonal analysis and cloning were pivotal components of this research, which relies on the frequency of cells with micronuclei and internuclear bridges to gauge the intensity of chromothripsis and break-fusion-bridge cycles. The efficacy of cloning, determined by these indicators, stemmed from the deliberate isolation of tumor stem cells, yielding clones in which chromothripsis activity and breakage-fusion-bridge cycles were sustained. Notably, it is plausible that the stem cells themselves, progenitors of these clones, harbor dicentric chromosomes and chromosomal fragments, contributing to the formation of “fatal micronuclei” in their karyotype. Cloning based on bridges and micronuclei has proven effective up to a certain threshold (15%-18%), reaffirming the predicted reproductive extinction of malignant cell populations under mutational pressure and genome chaos, as posited by the genetic theory of cell populations. Furthermore, this review highlights the potential of ergodic cancer therapy as a novel therapeutic strategy. Ergodic therapy offers promising prospects for late-stage and terminal malignant tumors, where conventional treatments may fall short due to advanced progression. Furthermore, by “enhancing chromothripsis” through the induction of additional micronuclei and bridges, ergodic cancer therapy seeks to increase genome chaos to a critical threshold, potentially halting malignant progression. This innovative approach presents opportunities to explore new drugs and targets for chromothripsis-based oncotherapy, addressing the pressing need for effective treatments in advanced stages of malignancy.
-
-
-
Understanding Tankyrase Inhibitors and Their Role in the Management of Different Cancer
More LessAuthors: Sinjini Das, Gowramma Byran, Kaushik Biswas and Kalirajan RajagopalCancer manifests as uncontrolled cell proliferation. Tankyrase, a poly(ADP-ribose) polymerase member, is vital in Wnt signal transmission, making it a promising cancer therapy target. The Wnt/β-catenin pathway regulates critical biological processes like genomic stability, gene expression, energy utilization, and apoptosis. Its dysregulation contributes to cancer development. Targeting tankyrase within this pathway holds the potential for inhibiting aberrant cell growth and promoting programmed cell death, offering a promising avenue for cancer treatment. ADP-ribosylation, a reversible process, modifies proteins post-synthesis, regulating diverse cellular signaling pathways. Transferase enzymes like mono and poly(ADP- ribosyl) transferases transfer ADP-ribose from NAD+ to specific amino acid side chains or ADP-ribose units on target proteins. Blocking tankyrase has emerged as a promising strategy in cancer treatment. This article reviews recent advancements in developing novel tankyrase inhibitors. It delves into structure-activity relationships, molecular docking, polypharmacology profiles, and binding mechanisms at the active site. Insights into lead structure development aid in designing potent anti-cancer medications, shedding light on promising avenues in cancer therapy.
-
-
-
Exploring the Impact of Chitosan-Based Nanoparticles in Cancer Targeted Drug Delivery: A Bibliometric Review
More LessOver the last two decades, there has been a considerable increase in the usage of chitosan nanoparticles (CSNPs) for drug delivery applications in cancer treatment. However, a comprehensive bibliometric analysis of this emerging field is lacking. The current analysis presents a detailed bibliometric assessment of the research evolution and trends in CSNPs for cancer targeted-drug delivery from 2000 to 2022. A total of 626 publications from the Web of Science Core Collection (WoSCC) database were evaluated using VOSviewer, Bibliometrix, and MS Excel. An in-depth exploration was carried out to find publication trends, the most productive authors, affiliations, countries, and journals as well as the highly cited publications within this particular research domain. Ick Chan Kwon was the most prominent author who made significant contributions to this field. China was the leading country in terms of research on CSNPs for cancer-targeted drug delivery, followed by India and Iran. Concerning the total number of publications, The International Journal of Biological Macromolecules excelled as the leading journal in this research area. Furthermore, the research trends and hotspots were also identified through the analysis of the most commonly used keywords by the authors. Our analysis highlights the growing research interest in CSNPs for cancer drug delivery, emphasizing the need for further exploration to unlock their full potential.
-
-
-
Beyond CTLA-4 and PD-1: Novel Insights into MAO-A as a Next Generation Immune Checkpoint Modulator for Cancer Immunotherapy
More LessAuthors: Prajna Maity and Debjeet SurImmune checkpoint blockade (ICB) has fundamentally transformed cancer treatment by unlocking the potency of CD8+ T cells by targeting the suppression of the CTLA-4 and PD-1/PD-L1 pathways. Nevertheless, ICBs are associated with the risk of severe side effects and resistance in certain patients, driving the search for novel and safer immune checkpoint modulators. Monoamine Oxidase A (MAO-A) plays an unexpected role in the field of cancer. Recent research has unveiled its dual nature: in the brain, it breaks down neurotransmitters, impacting mood and behaviour, while in tumours, its role takes a more complex turn. The involvement of MAO-A in the tumour microenvironment is crucial, affecting oxidative stress, hypoxia, and immune cell behaviour. Specifically, the regulation of tumour-associated macrophages (TAMs) and their polarisation underscores their potential as targets for cancer immunotherapy. MAO-A inhibition reprograms TAMs, shifting them from an immunosuppressive to an immunostimulatory state, thereby enhancing the activity of tumour-infiltrating CD8+ T cells. Interestingly, the presence of MAO-A in CD8+ T cells suggests its potential as a target for novel ICB therapy. This review lays the groundwork for a deeper understanding of the therapeutic potential of MAO-A by examining the intricate landscape of its role in cancer immunology. We envision a future in which harnessing the power of CD8+ T cells through optimised MAO-A targeting might lead to safer and more effective cancer treatments, offering hope for countless patients.
-
-
-
Molecular Targets and Nano-Technological Approaches in the Treatment of Hepatic Carcinoma
More LessAuthors: Trilochan Satapathy and Maneesh Kumar DiwakarLiver cancer is a leading cause of cancer-related mortality, with about one million people losing their lives each year. The disease becomes even more dangerous when tumors cannot be removed through surgery. Globally, hepatocellular carcinoma (HCC) ranks third in terms of fatality rates among liver cancers. It is also the most frequent type of liver cancer. Due to the high mortality rate associated with this malignancy, it is a hotspot for researchers looking to improve treatment methods. Nanotechnology plays an important part in these attempts. Various types of nanoparticles (NPs) have been investigated for their ability to fight liver cancer. NPs are a vast class of materials. The article details the efforts made to include inorganic NPs, such as silver, gold, metal oxide, platinum, calcium, selenium, and other uncommon materials into drug delivery systems (DDS) for therapeutic, carrier, or imaging purposes. This review discusses the function of carbon-based NPs in DDS for the treatment of liver cancer, including polymeric, polysaccharide, lipid, and carbon dot NPs, all of which have been extensively researched for this purpose. The purpose of this review is to provide a concise overview of recent developments in the field of HCC based on current research and clinical diagnosis and treatment guidelines. Further goals include elucidating the current state of nanomaterials research, its limitations, and the potential for future advancements in the field, as well as the use of nanotechnology in the detection and treatment of HCC.
-
-
-
Variants in CYP2J2 and CYP2C9 Contribute to Susceptibility of Lung Cancer
More LessAuthors: Chan Zhang, Qi Li, Yujing Cheng, Xin Yang, Wanlu Chen, Kunhua He and Mingwei ChenBackgroundCytochrome P450 (CYP) enzymes are involved in the metabolism of xenobiotic and carcinogen. In the study, we evaluated the association of CYP2J2 and CYP2C9 variants with lung cancer.
MethodsFive polymorphisms in CYP2J2 and four polymorphisms in CYP2C9 were genotyped in 507 lung cancer patients and 505 controls with Agena MassARRAY platform. The linkage of variants with lung cancer risk was evaluated by odds ratio (OR) and 95% confidence interval (CI) in genetic models and haplotype analyses.
ResultsWe found that CYP2C9 rs1934967 alleles were associated with lung cancer risk (P < 0.05). In stratified analysis, rs2280274 (women, non-smoker, non-drinker and lymphatic metastasis), rs11207535 (non-smoker and non-drinker), rs10889159 (non-smoker and non-drinker) of CYP2J2, whereas rs1934967 (age ≤ 60, BMI > 24, squamous carcinoma) of CYP2C9 decreased lung cancer risk (P < 0.05). In addition, the results of linkage disequilibrium (LD) analysis showed that rs2280274|rs4388726 - TG (with adjustment: P = 0.042) of CYP2J2 and rs10509679|rs1934967| rs1934968|rs9332220 – GTGG (without adjustment: P = 0.044) of CYP2C9 were linked with a significantly decreased lung cancer risk.
ConclusionOur results indicated genetic variants in CYP2J2 and CYP2C9 might contribute to the susceptibility of lung cancer in Chinese population.
-
-
-
Clinical Efficacy and Safety of Anlotinib Combined with Immune Checkpoint Inhibitors in the Treatment of Advanced Squamous Cell Lung Cancer: A Retrospective Single-Center Study in China
More LessAuthors: Bin Xu, Chunhua Xu, Li Li and Yuchao WangBackgroundThere is a relative lack of real-world data regarding the treatment of advanced squamous cell lung cancer (SqCLC) with anlotinib.
Aims and ObjectiveThis study aimed to investigate the clinical efficacy and safety of combining anlotinib with ICIs for the treatment of advanced SqCLC.
MethodsSixty patients with advanced SqCLC from December, 2017 to December, 2022, were retrospectively recruited in the final analysis. The overall survival (OS), adverse events (AEs), and short-term clinical effectiveness, including complete remission (CR), partial remission (PR), stable disease (SD), and progression disease (PD) of the study populations, were observed. Objective response rate (ORR) was defined as the combined percentage of CR and PR in all patients. Disease control rate (DCR) was defined as the combined percentage of CR, PR, and SD in all patients. Subgroups were classified based on age (<65 or ≥65), sex (female or male), and the eastern cooperative oncology group (ECOG) score (0-1 or 1-2).
ResultsThe median age of the study population was 65 years, and 41 (68.3%) of the participants were male. ORR was 21.67%, and DCR was 83.33%. The median PFS and OS were 7.5 and 19.5 months, respectively. Furthermore, the Kaplan-Meier curves demonstrated no significant difference (P >0.05) in PFS and OS between different age groups, sex, and ECOG scores. Bone marrow suppression was the most common AE, followed by immune pneumonia.
ConclusionThe findings of this study confirmed the effectiveness and safety of anlotinib and ICIs for advanced SqCLC.
-
-
-
Complement Factor H Promotes the Growth of Lung Adenocarcinoma Cells through the JAK2/STAT3 Signaling Pathway
More LessAuthors: Kaili Wang, Xitan Wang, Jiang Li, Yangyang Wang and Peng GaoBackgroundOver 50% of lung adenocarcinoma patients have high levels of complement factor H (CFH) expression. Previous studies have reported that CFH inhibits the migration of endothelial cells. In this study, we investigated the mechanism by which CFH affects lung adenocarcinoma development via phosphorylation of STAT3.
MethodsAdenovirus expressing mice Cfh gene was used to infect C57 mice for two weeks, and then Lewis Lung Carcinoma (LLC) was injected to develop a subcutaneous tumor. The effect of CFH on human A549 cells was also detected. Moreover, we collected CFH over-expressed conditional medium from HepG-2 cells infected with adenovirus expressing human CFH gene. A549 cells were incubated with the conditional medium, and the effect of the CFH-conditional medium on cell proliferation and migration was detected.
ResultsIt was found that CFH promoted lung adenocarcinoma growth in vivo, and CFH-conditional medium treatment significantly increased the viability and migration area of A549 cells. CFH-conditional medium increased the phosphorylation of JAK2 and STAT3 in A549 cells. While using STATTIC to block STAT3 phosphorylation, CFH-conditional medium treatment did not affect A549 cell viability or migration compared to the control group.
ConclusionThese data suggested that CFH promoted the proliferation and migration of A549 cells by increasing the phosphorylation level of the JAK2/STAT3 signaling pathway. Furthermore, CFH has the potential to be a target for antitumor therapy.
-
-
-
Illuminating the Role of Wound Healing-Related Hub Genes in Colorectal Adenocarcinoma: Molecular Mechanisms, Prognostic Implications and Therapeutic Potential
More LessAuthors: Yichao Yan, Hailiang Liang, Yongbai Li, Dongbo Chen, Bo Li and Abduh MurshedBackgroundColorectal adenocarcinoma (COAD) is a prevalent and lethal form of cancer. Understanding the molecular mechanisms underlying COAD progression is crucial for developing effective diagnostic and therapeutic strategies.
MethodsThis study aims to explore wound healing-related genes in COAD and their potential roles in tumorigenesis and prognosis using in silico and in vitro methodology.
ResultsA set of 70 genes associated with the “wound healing” term ere extracted from the Gene Ontology (GO) database (GO:0042060) and a protein-protein interaction (PPI) network was constructed using the STRING database. The PPI network was analyzed with the CytoHubba plugin in Cytoscape, identifying four major hub genes: MMP2, FN1, NF1, and PTK7. We then analyzed the expression of these hub genes across 16 COAD cell lines and nine normal colon cell lines using RT-qPCR, finding significant overexpression in COAD cell lines. ROC curve analysis confirmed the diagnostic potential of these genes, with MMP2, FN1, and NF1 showing high AUC values. Expression validation using the TCGA COAD cohort, OncoDB, and HPA databases corroborated these findings, highlighting the overexpression and high protein levels of these genes in COAD. Promoter methylation analysis indicated lower methylation levels in COAD samples, suggesting dysregulation through epigenetic mechanisms. Genetic alteration analysis via cBioPortal revealed a spectrum of mutations, with FN1 being the most frequently mutated. Prognostic analysis using a KM plotter showed that high expression of the hub genes is associated with poorer overall survival (OS) and disease-free survival (DFS). Functional state correlations via CancerSEA suggested that these genes promote cell cycle, proliferation, metastasis, and stemness in COAD. Expression analysis in immune cells and drug sensitivity analyses highlighted the roles of MMP2, FN1, and NF1 in macrophages and drug resistance. A miRNA-mRNA network constructed using miRNet identified hsa-miR-200a-3p as a central regulator. Finally, functional assays in HCT116 cells demonstrated that knockdown of MMP2 and FN1 reduced proliferation, colony formation, and wound healing, suggesting these genes as potential therapeutic targets in COAD.
ConclusionIn conclusion, our study identifies MMP2, FN1, NF1, and PTK7 as key wound healing-related hub genes in COAD.
-
-
-
Identification of PANoptosis Subtypes to Assess the Prognosis and Immune Microenvironment of Lung Adenocarcinoma Patients: A Bioinformatics Combined Machine Learning Study
More LessAuthors: Xiaofeng Zhou, Bolin Wang, Di Wu, Lu Gao, Zhihua Wan and Ruifeng WuBackgroundPANoptosis, a novelty mechanism of cell death involving crosstalk between apoptosis, pyroptosis, and necroptosis, is strongly associated with tumor cell death and immunotherapy efficacy. However, its relevance in lung adenocarcinoma (LUAD) remains to be elucidated.
MethodsIn this study, we acquired 18 PANoptosis-related differentially expressed gene (PRDEG) of LUAD. Based on these genes, LUAD samples were identified with different subtypes by unsupervised clustering. Next, we compared the differences between the subtypes, including clinical features, immune microenvironment, and potentially sensitive drugs. Furthermore, we used machine learning to identify hub prognostic PRDEGs, construct a risk score, and validate it on other external datasets. We incorporated the patient's clinical information and risk score into the proportional hazards model and lasso-cox models to find key prognostic features and constructed five prognostic models. The best model was identified via the area under the curve and validated on an external dataset.
ResultsLUAD patients were divided into two clusters named C1 and C2, respectively. The C2 cluster exhibited shorter survival time, more advanced tumor stage, higher suppressive immune cell scores, such as dendritic cells, and higher expression of inhibitory immune checkpoints, such as LAG3 and CD86. TIMP1, CAV1, and CD69 were recognized as key prognostic factors, and risk scores predicted survival with significant differences in the external validation set. Risk score and N-stage were identified as critical prognostic features. The Coxph model outperformed other machine learning clinical models. The 1-, 3-, and 5-year time-ROCs in the external validation set were 0.55, 0.59, and 0.60, respectively.
ConclusionWe demonstrated the potential of PANoptosis-based molecular clustering and prognostic features in predicting the survival of patients with LUAD as well as the tumor microenvironment.
-
Volumes & issues
-
Volume 25 (2025)
-
Volume 24 (2024)
-
Volume 23 (2023)
-
Volume 22 (2022)
-
Volume 21 (2021)
-
Volume 20 (2020)
-
Volume 19 (2019)
-
Volume 18 (2018)
-
Volume 17 (2017)
-
Volume 16 (2016)
-
Volume 15 (2015)
-
Volume 14 (2014)
-
Volume 13 (2013)
-
Volume 12 (2012)
-
Volume 11 (2011)
-
Volume 10 (2010)
-
Volume 9 (2009)
-
Volume 8 (2008)
-
Volume 7 (2007)
-
Volume 6 (2006)
-
Volume 5 (2005)
-
Volume 4 (2004)
-
Volume 3 (2003)
-
Volume 2 (2002)
-
Volume 1 (2001)
Most Read This Month