Current Cancer Drug Targets - Volume 25, Issue 8, 2025
Volume 25, Issue 8, 2025
-
-
The Potential of Nano Pharmaceuticals to Change the Paradigm of Brain Tumor Therapy: A State-of-the-art Review
Central nervous system tumors are abnormal proliferations of neuronal cells within the brain and spinal cord. They can be primary or secondary and place a heavy financial, psychological, and physical burden on individuals. The highly selective blood-brain barrier, which only permits specific molecules to flow into the brain parenchyma, inhibits the efficacy of pharmacological medicines. Treatment options include surgery, chemoradiotherapy, and targeted therapy. Despite advances in therapy over the past few decades, the overall morbidity and mortality rates are still high, emphasizing the need for improved therapeutic choices to improve survival and quality of life further. Nano pharmaceuticals have demonstrated encouraging outcomes in in vivo trials using microscopic particles to enhance bioavailability and selectivity. The most successful clinical results to date have been achieved by liposomes, extracellular vesicles, and biomimetic nanoparticles; nevertheless, clinical trials are required to confirm their safety, efficacy, affordability, long-term impact, and success in patients from various demographics. Nano pharmaceuticals have the potential to change the paradigm of therapy for brain tumors, allowing better outcomes as primary and adjunctive therapy.
-
-
-
Smart Nanobiomaterials for Gene Delivery in Localized Cancer Therapy: An Overview from Emerging Materials and Devices to Clinical Applications
The research provides an in-depth exploration of gene therapy, covering fundamental principles, diverse implementation strategies, and innovative gene delivery vectors. Further implementations of gene therapy, such as apoptosis induction, anti-angiogenesis, and nucleic acid therapy, have been described. Gene delivery vectors, encompassing viral methods such as adenovirus, retroviral, foamy viral, adeno-associated viral, herpes simplex virus (HSV), and vaccinia virus vectors and nonviral methods, such as physical and chemical approaches have been extensively discussed. Further, a significant focus is placed on novel drug carriers, including nanoparticles, such as iron oxide, calcium carbonate, gold, carbon nanotubes, graphene oxide, quantum dots, nanogels, ceramic nanoparticles, calcium phosphate, and metal-organic frameworks. Additionally, lipids, peptides, and polymeric materials, featuring liposomes, exosomes, polymeric micelles, hydrogels, polymersomes, and dendrimers are explored as promising avenues for gene delivery. Finally, the key findings and insights underlining the dynamic landscape of gene therapy research have been summarized, which may offer a comprehensive understanding of current methodologies and potential future directions in the field of gene therapy.
-
-
-
The Role of Epigenetic Mechanisms in the Treatment of GI Cancers
Epigenetic mechanisms have been shown to play a critical role in the development and progression of gastrointestinal [GI] cancers. These mechanisms involve modifications to DNA and histones that can alter gene expression patterns and may contribute to the initiation and progression of cancers. In recent years, epigenetic therapies have emerged as a promising approach to treating GI cancers. These therapies target specific epigenetic modifications, such as DNA methylation and histone acetylation, to restore normal gene expression patterns and inhibit cancer cell growth. Several epigenetic drugs have been approved for the treatment of GI cancers. Moreover, the use of epigenetic therapies in combination with other treatments, such as chemotherapeutic agents, is being studied to improve treatment outcomes.
We have provided an overview of the role of epigenetic mechanisms in GI cancer treatment aimed to focus on recent evidence of the use of epigenetic agents in clinical and preclinical GI cancer studies, including gastric, esophageal, hepatic, pancreatic, and colorectal cancers. Overall, the role of epigenetic mechanisms in GI cancer treatments is an active area of research with the potential to improve patients' treatment outcomes and advance cancer treatment strategies.
-
-
-
Targeting the Tumor Epigenetic Regulator SETDB1 for Tumor Therapy
Authors: Cheng Peng, Nini Zhou, Tengjiang Chen, Jie Lei, Changwen Chen and Shunqin ZhuEpigenetic alterations are implicated in the early stages of tumorigenesis and are widely recognized as a ubiquitous phenomenon in cancer development. Aberrant epigenetic modifications can alter the expression of target genes, induce heterochromatin formation, and gradually drive normal cells towards immortalized tumor cells with significant consequences. SETDB1 (SET domain bifurcated histone lysine methyltransferase 1), a typical histone methyltransferase, promotes the formation of heterochromatin and inhibits the transcription of genes by modifying the methylation of lysine 9 of histone 3. SETDB1 is usually highly expressed in tumors with high copy numbers, accompanied by poor prognosis and low patient survival rates, which is a typical case of abnormal epigenetic modification. We discuss the mechanism of SETDB1 in a variety of cancers and review the epigenetic inhibitors that have been reported in recent years, along with their anti-tumor effects. In addition, we summarize the role of SETDB1 in a variety of diseases and cell functions.
-
-
-
Groundbreaking mRNA Lung Cancer Vaccine Trials: A New Dawn in Cancer Treatment
Authors: Md Sadique Hussain, Ayesha Sultana, Ajay Singh Bisht and Gaurav GuptaThe advent of mRNA vaccines has heralded a transformative era in oncology, exemplified by the BNT116 mRNA lung cancer vaccine. Leveraging the same groundbreaking technology as COVID-19 vaccines, BNT116 delivers tumor-specific genetic instructions to the immune system, targeting non-small cell lung cancer (NSCLC), the most prevalent lung cancer subtype. This approach contrasts with conventional therapies that lack precision and often damage healthy tissues. By encoding tumor antigens, BNT116 educates cytotoxic T cells to recognize and eradicate malignant cells, aligning with the principles of precision medicine. Early-phase clinical trials (e.g., NCT05142189) have demonstrated a favorable safety profile and promising antitumor activity, with ongoing research exploring its use in combination therapies, such as checkpoint inhibitors. Despite logistical challenges, such as mRNA instability and cold chain requirements, advances in lipid nanoparticle delivery systems are enhancing vaccine stability and efficacy. The adaptability of mRNA technology positions it as a cornerstone for personalized oncology, with potential applications extending to other cancers. Success in the BNT116 trials could redefine NSCLC treatment paradigms, offering a targeted, less cytotoxic alternative. This innovation can not only improve therapeutic outcomes, but also pave the way for preventive cancer vaccines, signaling a new dawn in cancer treatment.
-
-
-
Exosomal CircRNAs: A Future Star in Colorectal Cancer
Authors: Yuanzhi Zhou, Chengyan Wei, Yuqi Xu, Jingjing Wang, Chunwei Zhang and Yong JinColorectal cancer (CRC) is currently the third most common malignancy worldwide, with an increasing mortality rate and treatment resistance. Due to the lack of effective biomarkers and therapeutic targets, the early diagnosis and treatment of colorectal cancer remain suboptimal. Circular RNAs (circRNAs) are a novel class of non-coding RNAs with covalent closed-loop structures that are well stabilized and conserved and are involved in multiple pathological conditions in humans. CircRNAs have been identified to be enriched and stable in exosomes. In addition, there is growing proof that exosomal circRNAs that have been identified as oncogenes or tumor suppressors regulate CRC growth, migration, and sensitivity to radiotherapy and chemotherapy. Exosomal circRNAs represent promising candidates as diagnostic biomarkers and anti-tumor targets. In this article, we explore recent studies on exosomal circRNAs in CRC and describe their biological functions in colorectal cancer development, illustrating their potential as biomarkers and targeted therapeutic capabilities.
-
-
-
Bioinformatics Strategy for Investigating the Mechanism of Hispolon in the Treatment of Triple-Negative Breast Cancer s with In vitro Experiments
Authors: Junfeng Li, Jingfei Bao, Lichao Wu, Tengfei Sun, Junhui Zhao, Fei Luo, Fangfang Tao and Wenhong LiuBackgroundHispolon, a phenolic compound isolated from the medicinal yellow fungal mulberry, exhibits a strong anti-triple-negative breast cancer (TNBC) effect. However, the antitumor mechanisms of Hispolon have not been fully explored.
ObjectiveIn this study, we systematically investigated the mechanism of Hispolon against TNBC based on bioinformatics and in vitro experiments.
MethodsThe Hispolon-related targets were first collected from the SwissTarget database. Differential Expression Genes (DEG) were screened between TNBC and normal breast tissue using the Gene Expression Comprehensive (GEO) dataset. The overlapping targets between Hispolon and DEG were analyzed by plotting Venn maps. Protein-protein interaction (PPI) network was constructed to analyze the interactions among these targets. The focus was on mining the core targets of anti-TNBC effects of Hispolon via the Cytohubba and MCODE plugin of Cytoscape 3.7.2 software. We performed survival analysis on these core targets to screen the best-matched targets, including EGFR, KIT, and PLAU. This correlated strongly with our validation of Hispolon by molecular docking. In addition, Gene Ontology (GO) analysis and KEGG pathway analysis were performed using R software (ClusterProfiler package). Finally, in vitro experiments were performed to assess the accuracy of predicted target genes.
ResultsThe ADME results suggested that Hispolon has great potential to develop into a drug. Twenty overlapping targets were screened by matching the 107 targets of Hispolon to the 2,013 targets of TNBC DEG. Seven core targets of Hispolon against TNBC were initially identified, including EGFR, IGFBP3, MMP9, MMP2, MMP1, PLAU, and KIT. GO enrichment analysis demonstrated that the biological process of Hispolon acting on TNBC mainly involves lymphocyte activation in immune response and phosphatidylinositol-mediated signaling. Additionally, the relaxin signaling pathway, estrogen signaling pathway, proteoglycans in cancer, and others might be the key pathways of Hispolon against TNBC. Furthermore, Hispolon inhibited the proliferation of MDA-MB-231 cells in a concentration-dependent manner and regulated the RNA and protein expression of the core targets EGFR, PLAU, and KIT for the treatment of TNBC.
ConclusionIn this study, the polygenic pharmacological mechanism of action of Hispolon against TNBC was explored through network pharmacology and in vitro experiments, providing a new insight into the mechanism of TCM monomer against TNBC.
-
-
-
Cardiovascular Mortality Risk Among Patients with Liver Cancer: A SEER Database Study
Authors: Juan Li, Junyong Zhang and Song HeCrucial for understanding liver cancer patients overall health outcomes. This research aimed to assess the CVM risk of them.
Methods and MaterialsData sourced from the Surveillance, Epidemiology, and End Results (SEER) database encompassing liver cancer diagnoses from 2000 to 2017 were utilized. The standardized mortality rate (SMR) was computed using general population reference data, and multivariate competing risk models were employed for analysis.
ResultsAnalysis of 70,733 liver cancer patient records revealed 1,954 instances of CVM. The overall CVM SMR for liver cancer patients was 12.01 (95% CI: 11.48-12.55). Various demographic and clinical factors, including sex, race, age, year of diagnosis, pathological type, general stage, treatment modalities, and matrimonial status, emerged as liver cancer patients` independent predictors of CVM.
ConclusionLiver cancer patients have a notably heightened susceptibility to cardiovascular mortality (CVM) in contrast to the general populace. It is imperative to promptly recognize high-risk subcategories and execute tailored cardiovascular interventions as crucial measures to bolster survival rates within this cohort of patients.
-
-
-
Macranthoside B Suppresses the Growth of Adenocarcinoma of Esophagogastric Junction by Regulating Iron Homeostasis and Ferroptosis through NRF2 Inhibition
Authors: Lingling Wang, Guangzhao Pan, Sichao Tian, Che Zhang, Fangfang Tao and Jiang-Jiang QinBackgroundMacranthoside B (MB) is a saponin compound extracted from honeysuckle that has been reported to exhibit significant medicinal values, particularly anti-tumor activities. This study aimed to evaluate the anticancer efficacy of MB in treating adenocarcinoma of the esophagogastric junction (AEG) and elucidate its underlying mechanisms.
MethodsThree AEG cell lines and normal gastric epithelial cells were used to assess the anticancer activity of MB in vitro. A series of experiments, including RNA sequencing (RNA-seq) analysis, transmission electron microscopy (TEM), immunofluorescence, and western blot assay, were conducted to validate the molecular mechanisms by which MB may mediate these physiological changes. Finally, we used shRNA assays to silence the key gene driving these changes and examined the expression of molecules involved in the affected pathways.
ResultsMB exhibited significant anti-AEG cell activity with IC50 values ranging from 9.5 to 12.7 μM. RNA-seq results indicated that MB treatment in AEG cells significantly altered mRNA levels of autophagy- and ferroptosis-related genes. Further experiments revealed that MB treatment led to the up-regulation of lipid reactive oxygen species (Lip-ROS), oxidative stress-related pathway genes, and LC3B-labeled autophagic vesicles in AEG cells. Moreover, MB mediated NCOA4-dependent ferritinophagy, disrupting iron homeostasis and causing subsequent ferroptosis. We further confirmed that the intrinsic connection between autophagy and ferroptosis was due to the inhibition of NRF2 by MB. The inhibition of NRF2 by MB triggered transcriptional repression of its downstream effector molecules HERC2 and VAMP8, thus stabilizing NCOA4.
ConclusionThis study demonstrated MB to inhibit AEG cell growth by regulating iron homeostasis and inducing ferroptosis through the inhibition of NRF2, providing a basis for the development of novel drugs for AEG treatment.
-
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
