- Home
- A-Z Publications
- Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry - Anti-Cancer Agents)
- Issue Home
Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry - Anti-Cancer Agents) - Current Issue
Volume 25, Issue 10, 2025
-
-
Advances in VEGFR Inhibitors: A Comprehensive Review of Novel Anticancer Agents
Authors: Sumeet Jha, Sneha Gupta, Supriya Rani, Pinky Arora, Neeraj Choudhary and Shubham KumarCancer, characterized by aberrant cell growth, presents a formidable health challenge, impacting millions of individuals worldwide each year. Among the myriad mechanisms facilitating tumor progression, Vascular Endothelial Growth Factor receptors (VEGFR) play a pivotal role in driving angiogenesis the process by which tumors develop their own blood supply. This vascularization not only supports tumor nourishment and growth but also facilitates metastasis, enabling cancer to spread to distant sites. VEGFR inhibitors offer a strategic approach to disrupt the VEGF-VEGFR binding pathway, thereby impeding angiogenesis, metastasis, and the proliferation of cancer cells. This review elucidates the latest advancements in medicinal chemistry pertaining to VEGFR inhibitors, showcasing a variety of chemical moieties and assessing their efficacy across different cancer cell lines. The novel compounds highlighted in this review exhibit significant promise for anticancer evaluation through targeted VEGFR kinase inhibition. A robust body of in vivo, in vitro, and ex vivo studies supports these findings, demonstrating the antitumor effects of these compounds. Computational analyses further enhance our understanding by predicting compound binding affinities, pharmacokinetics, and overall drug-likeness. Despite the significant progress made in developing effective VEGFR inhibitors, challenges remain in refining these agents for optimal cancer treatment. This review not only summarizes the advancements achieved in VEGFR inhibitor development but also emphasizes the ongoing hurdles that must be addressed to enhance the efficacy of cancer therapies.
-
-
-
Bioactive Products Targeting C-Met As Potential Antitumour Drugs
Authors: Liying Zhao, Chunmei Qian, Xiaoqi Ma and Xiaoyu WangMesenchymal‒epithelial transition factor (c-Met), a receptortyrosine kinase (RTK), plays a vital role in cell proliferation, migration and invasion, and tumour metastasis.
ObjectiveWith increasing duration of treatment, many tumours gradually develop drug resistance. Therefore, novel antitumour drugs need to be developed to treat patients with tumours. Targeting c-met inhibitors may be an effective treatment strategy.
MethodsScientific databases such as ScienceDirect, PubMed, the Wiley Online Library, and Social Sciences Citation Index were used to collect information. All the relevant literature was reviewed, and the available literature was screened. The upstream and downstream pathways of c-Met and their relevance to antitumour effects were searched based on the articles' title, abstract, and full text. The c-Met-targeting drugs with antitumour effects are summarized below. A “citation within a citation” or snowballing approach was used in this screening process to identify additional papers that may have been missed in the initial literature screening process. High-quality studies published in peer-reviewed journals were summarized and prioritized for citation in the review.
ResultsIn recent years, research on small-molecule targeted drugs has developed rapidly. Many results have also been achieved in the synthesis and isolation of c-Met inhibitors from natural compounds and traditional Chinese medicines.
ConclusionThis article summarizes the developments in anti-c-Met drugs, which are synthesized and isolated from natural compounds and traditional Chinese medicine (TCM). This study provides primary resources for the development of c-Met inhibitors.
-
-
-
Schisanhenol Inhibits the Proliferation of Hepatocellular Carcinoma Cells by Targeting Programmed Cell Death-ligand 1 via the STAT3 Pathways
Authors: Zhihong Zhang, Yiwen Zhong, Xu Han, Xueyang Hu, Yuhan Wang, Lei Huang, Siying Li, Ziqing Li, Chunmei Wang, He Li, Jinghui Sun, Wenyue Zhuang, Mengyang Wang, Jianguang Chen, Wei Liu, Chang Liu, Xin Guo, Siyu Yuan and Jiping WuBackgroundProgrammed cell death-ligand 1 (PD-L1) is overexpressed in tumor cells, which promotes tumor cell survival and cell proliferation and causes tumor cells to escape T-cell killing. Schisanhenol, a biphenyl cyclooctene lignin-like compound, was extracted and isolated from the plant named Schisandra rubriflora (Franch.).
PurposeIn this work, we studied the anticancer potential of schisanhenol and explored whether schisanhenol mediated its effect by inhibiting the expression of PD-L1 in vitro and in vivo.
Materials and MethodsIn vitro, we performed western blot, immunofluorescence, immunoprecipitation, and colony formation assays to study the proteins, genes, and pathways related to the anti-tumour activity of schisanhenol. In vivo, we explored the antitumor activity of schisanhenol through orthotopic liver transplantation and subcutaneous transplantation tumor models of hepatocellular carcinoma (HCC) cells.
ResultsWe found that schisanhenol decreased the viability of HCC cells. It inhibited the expression of programmed cell death ligand-1 (PD-L1), which plays a pivotal role in tumorigenesis. Subsequently, schisanhenol suppressed the expression of PD-L1 by decreasing the activation of STAT3. Furthermore, we found that schisanhenol inhibited the activation of STAT3 via JAK/STAT3 (T705), Src/STAT3 (T705), and PI3K/AKT/mTOR/STAT3 (S727) pathways. Colony formation tests showed that schisanhenol suppressed cell proliferation by inhibiting PD-L1. Schisanhenol also enhanced cytotoxic T lymphocytes (CTL) activity and regained their ability to kill tumour cells in co-culture. Finally, in vivo observation confirmed the antitumor activity of schisanhenol.
ConclusionSchisanhenol inhibits the proliferation of HCC cells by targeting PD-L1 via the STAT3 pathways. These findings prove that schisanhenol is a valuable candidate for HCC therapeutics and reveal previously unknown characteristics of schisanhenol.
-
-
-
Rhizopogon luteolus and Ganoderma adspersum Extracts Inhibit Invasion through the Crosstalk between Anti-oxidant Activity and Apoptosis Induced by pAKT/Rb
Authors: Aydın Demiray, Ege Rıza Karagür, Gülsen Tel-Çayan, Onur Tokgün, Hakan Akça and Mehmet Emin DuruObjectiveLung cancer is the primary cause of cancer-related deaths globally. Protein kinase B (AKT) protein is associated with many pathways in non-small cell lung cancer (NSCLC), such as proliferation, migration, invasion, and apoptosis. Mushrooms have a long history of being used in traditional medicine to treat various diseases. Scientists have been exploring the potential of mushrooms for their antioxidant and anticancer properties. In our study, the anti-oxidant, invasion, and apoptosis effects of mushroom extracts were investigated in NSCLC.
Materials and MethodsNon-Small Cell Lung Cancer cell lines H1299, PC-3, and PC-14 were used in our study. After obtaining the extracts of Rhizopogon luteolus and Ganoderma adspersum, IC50 value was calculated as 25.04-11.73-16.54 ng/ul for R. luteolus and 2.97-1.53-1.01 μg/ul for G. adspersum, respectively, in H1299, PC3 and PC14 cell lines. Afterward, proliferative and invasion effects, as well as apoptosis and anti-oxidant effects, were investigated using the IC50 dose. Western blotting was performed to investigate the pathways of these effects.
ResultsAccording to the results of our study, Rhizopogon luteolus and Ganoderma adspersum extracts have anti-proliferative and anti-invasive effects on non-small lung cancer cell lines and induced apoptosis, which has been found to increase the anti-oxidant effect. It was found that this effect was due to cross-talk between antioxidant activity and the AKT-Rb pathway.
ConclusionWe anticipate that Rhizopogon luteolus and Ganoderma adspersum extracts will be effective in cancer treatment by suppressing lung cancer cells via p-Akt and Rb.
-
-
-
Investigation of the Anticarcinogenic Effects of Hypericum perforatum Extract on Human Thyroid Cancer
Authors: Çiğdem Gökçek-Saraç, Gizem Altunkaya and Serdar KarakurtIntroduction/ObjectivePlants and their bioactive compounds play a crucial role in the pharmaceutical industry for treating cancer. To date, the cytotoxic and antiproliferative effects of Hypericum perforatum methanol extract on human thyroid cancer cell lines have not been thoroughly explored. The present study aimed to assess the potential anti-cancer effects of HPME on human thyroid cancer and investigate its potential therapeutic benefits.
MethodsHPME was prepared using the maceration method, and its antioxidant activity was examined. Cytotoxicity studies were then carried out, followed by an investigation of the possible effects of HPME on metastasis and colony-forming capacities of human thyroid cancer cells. Afterward, qRT-PCR, western blotting, and apoptosis assays were performed.
ResultsCytotoxicity studies revealed notable cytotoxicity of HPME against the TT cell line. Moreover, HPME significantly curtailed metastasis and invasion of TT cells in an in vitro wound healing assay. Analyses of gene expressions demonstrated an elevation in caspase-12, caspase-3, and Bax, coupled with a reduction in BcL-2, APOE, and CLU expression. Following HPME treatment, there was an increase in the protein expression levels of Bax and Caspase-12, while a decrease in the BcL-2, APOE, and CLU protein expression. Furthermore, apoptotic studies indicated an increase in early apoptosis.
ConclusionOverall results revealed that HPME demonstrates a notable antioxidant capacity in human thyroid cancer. It exerts an influence on crucial biological processes associated with cancer, indicating its potential to hinder the proliferation of human thyroid cancer cells by enhancing apoptosis through the upregulation of gene and protein expression, particularly involving caspases.
-
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)
Most Read This Month Most Read RSS feed
