Topic Editors

Department of Biological Sciences, The Border Biomedical Research Center (BBRC), The University of Texas at El Paso, El Paso, TX 79968, USA
Department of Biological Sciences, The Border Biomedical Research Center (BBRC), The University of Texas At El Paso, El Paso, TX 79968, USA
Dr. Blanca E. Ruiz-Medina
Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
Dr. Patricia Talamás-Rohana
Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Instituto Politecnico Nacional, Mexico City, Mexico
Laboratory of Medicinal Chemistry, Department of Biology, University of Massachusetts, Amherst, MA 01003-9297, USA

Advances in Anti-Cancer Drugs: 2nd Edition

Abstract submission deadline
closed (31 May 2025)
Manuscript submission deadline
closed (31 August 2025)
Viewed by
6350

Topic Information

Dear Colleagues,

This Topic is the second edition of the collection “Advances in Anti-Cancer Drugs”, available at https://www.mdpi.com/topics/AIACD.

We hope this message finds you well. As large quantities of novel synthetic and natural molecules continue to be generated or discovered, it is challenging to identify and characterize therapeutic agents with effective anti-cancer activity.

The aim of this particular Topic, "Advances in Anti-Cancer Drugs”, is to collect a group of publications focused on novel chemical compounds with cytotoxic activity against cancer cells, whether in vitro, in vivo, or both, particularly including articles that present novel biomarkers and target proteins with potential therapeutic properties. In addition, submissions are encouraged for studies on drug repurposing, including approved, discontinued, and shelved drugs with anti-cancer activity. Moreover, immunotherapy, electrochemotherapy, gene therapy, and phytomedicine studies are highly welcome.

The submitted manuscripts should include the partial mechanism used for novel compounds and therapeutic strategies. Information on the potential mechanism of action should be included. This Topic provides a suitable platform to disseminate anti-cancer discoveries at the bench and the bedside. Thus, we are delighted to invite you to participate in this excellent opportunity to publish your manuscript in our journal.

Dr. Armando Varela-Ramirez
Dr. Elisa Robles-Escajeda
Dr. Blanca E. Ruiz-Medina
Dr. Patricia Talamás-Rohana
Dr. Rachid Skouta
Topic Editors

Keywords

  • anticancer
  • antiproliferation
  • apoptosis
  • biomarkers
  • cell cycle
  • cell signaling
  • drug discovery
  • immunotherapy
  • gene therapy
  • phytomedicine

Participating Journals

Journal Name Impact Factor CiteScore Launched Year First Decision (median) APC
Cancers
cancers
4.4 8.8 2009 20.3 Days CHF 2900
Medicines
medicines
- - 2014 45 Days CHF 1400
Medical Sciences
medsci
4.4 8.7 2013 24.3 Days CHF 1600
Cells
cells
5.2 10.5 2012 16 Days CHF 2700
Pharmaceuticals
pharmaceuticals
4.8 7.7 2004 14 Days CHF 2900
Biology
biology
3.5 7.4 2012 17.4 Days CHF 2700
Biologics
biologics
- 7.2 2021 23.5 Days CHF 1200

Preprints.org is a multidisciplinary platform offering a preprint service designed to facilitate the early sharing of your research. It supports and empowers your research journey from the very beginning.

MDPI Topics is collaborating with Preprints.org and has established a direct connection between MDPI journals and the platform. Authors are encouraged to take advantage of this opportunity by posting their preprints at Preprints.org prior to publication:

  1. Share your research immediately: disseminate your ideas prior to publication and establish priority for your work.
  2. Safeguard your intellectual contribution: Protect your ideas with a time-stamped preprint that serves as proof of your research timeline.
  3. Boost visibility and impact: Increase the reach and influence of your research by making it accessible to a global audience.
  4. Gain early feedback: Receive valuable input and insights from peers before submitting to a journal.
  5. Ensure broad indexing: Web of Science (Preprint Citation Index), Google Scholar, Crossref, SHARE, PrePubMed, Scilit and Europe PMC.

Published Papers (8 papers)

Order results
Result details
Journals
Select all
Export citation of selected articles as:
18 pages, 23288 KB  
Article
Ginseng Polysaccharides Inhibit Aspergillus sydowii-Driven Lung Adenocarcinoma via Modulating Gut Microbiota–Bile Acid Metabolism Axis
by Jinlian He, Xiao Shu, Hudan Pan, Mingming Wang, Yuanyuan Song, Feng Zhou, Lirong Lian, Liqing Chen, Gangyuan Ma, Yicheng Zhao, Runze Li and Liang Liu
Cancers 2025, 17(19), 3134; https://doi.org/10.3390/cancers17193134 - 26 Sep 2025
Viewed by 255
Abstract
Background: Lung cancer is the leading cause of cancer-related mortality globally, with lung adenocarcinoma (LUAD) as the most common subtype. Dysbiotic intratumoral mycobiomes drive LUAD pathogenesis, and Aspergillus sydowii (A. sydowii) acts as a key oncogenic fungal species. Ginseng polysaccharides (GPs), [...] Read more.
Background: Lung cancer is the leading cause of cancer-related mortality globally, with lung adenocarcinoma (LUAD) as the most common subtype. Dysbiotic intratumoral mycobiomes drive LUAD pathogenesis, and Aspergillus sydowii (A. sydowii) acts as a key oncogenic fungal species. Ginseng polysaccharides (GPs), bioactive phytochemicals with immunomodulatory and oncostatic properties, counteract fungal infections and restore immunosurveillance in LUAD. Methods: Subcutaneous and orthotopic LUAD murine models were established by implanting Lewis lung carcinoma (LLC) cells. Subcutaneous tumors were infected intratumorally and orthotopic models via nasal inoculation. GPs (200 mg/kg/day) were orally administered to evaluate tumor growth. Metagenomic and targeted bile acid metabolomic profiling of fecal and tumor tissues was performed, with Spearman correlations analyzed using R packages. Results: GPs significantly inhibited A. sydowii-induced tumor growth in both models. In subcutaneous tumors; GPs reduced volume (p < 0.05) and weight vs. infected controls. In orthotopic models, GPs decreased pathological nodules and lung weight, with micro-CT/H&E confirming attenuated hyperplasia. Metagenomics showed GPs restored gut homeostasis by enriching Lactobacillus/Muribaculum intestinale and suppressing pro-inflammatory Alistipes. Targeted metabolomics revealed reduced β-Hyodeoxycholic Acid (3β-HDCA), Chenodeoxycholic acid 24-acyl-b-D-glucuronide (CDCA-24G) and 3β-hydroxychol-5-en-24-oic acid (5-isoLCA) after GP treatment. Network analysis confirmed significant microbe–bile acid interactions. Conclusions: GPs exert antitumor effects against A. sydowii-induced LUAD by modulating gut microbiota and bile acid metabolism. This identifies GPs as a promising therapy for mycobiome-influenced cancers, with dual targeting of fungal infection and metabolic reprogramming. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Graphical abstract

17 pages, 5123 KB  
Article
Bioinformatics-Based Analysis of the Screening and Evaluation of Potential Targets of FTY720 for the Treatment of Non-Small Cell Lung Cancer
by Mengyuan Han, Sendaer Hailati, Dilihuma Dilimulati, Alhar Baishan, Alifeiye Aikebaier and Wenting Zhou
Biology 2025, 14(10), 1311; https://doi.org/10.3390/biology14101311 - 23 Sep 2025
Viewed by 355
Abstract
Background: A range of cancer cells are significantly inhibited by FTY720. It is unknown, nevertheless, how FTY720 influences the onset of non-small cell lung cancer (NSCLC). Using bioinformatics techniques, we analyzed and the possible molecular mechanisms and targets of FTY720 for the treatment [...] Read more.
Background: A range of cancer cells are significantly inhibited by FTY720. It is unknown, nevertheless, how FTY720 influences the onset of non-small cell lung cancer (NSCLC). Using bioinformatics techniques, we analyzed and the possible molecular mechanisms and targets of FTY720 for the treatment of NSCLC. Methods: DEGs (Differentially expressed genes) were acquired by differential analysis of the dataset GSE10072. Obtained FTY720 target genes and NSCLC disease genes from databases such as Swiss-TargetPrediction and GeneCard. Subsequently, target and disease genes, as well as DEGs, were merged for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, gene ontology (GO), and protein interaction analysis. The overlapping genes of DEGs and target genes, and disease genes were also obtained separately and subjected to survival as well as expression analyses. We constructed the regulatory network of miRNAs and transcription factors (TFs) on hub genes. Finally, the immune cell association of hub genes was evaluated using the ssGSEA method, molecular docking of FTY720 to hub genes was carried out utilizing Autodock, and molecular dynamics simulations were conducted. Results: In this study, 444 DEGs, 232 target genes of FTY720, and 466 disease genes were obtained. Moreover, a total of 1062 genes were obtained by removing duplicate values after merging, among which PIK3R1, Akt1, and S1PR1 had the highest DEGREE values in the protein interactions network, and these genes were primarily enriched in MAPK, PI3K-Akt signaling pathways, with the PI3K-Akt signaling pathway being the most prominent. Among the overlapping genes, three potential targets of FTY720 for NSCLC treatment were found: S1PR1, ZEB2, and HBEGF. ZEB2 and S1PR1 were determined to be hub genes and to significantly affect NSCLC prognosis by survival analysis. Furthermore, hsa-miR-132-3p, hsa-miR-192-5p, and hsa-miR-6845-3p were strongly associated with FTY720 for the treatment of NSCLC; CTBP1 (carboxy-terminal binding protein 1), EZH2 (protein lysine N-methyltransferase), and ZNF610 (zinc-finger protein 610) may all influence the expression of ZEB2 and S1PR1. Hub genes had a substantial negative link with memory B cells and a significant positive correlation with memory CD8 T cells and Th17 helper T cells. The molecular docking and kinetic simulation results of FTY720 with the two hub genes indicate that the protein-ligand complex has good stability. Conclusion: Our research indicates that FTY720 may inhibit NSCLC via possible targets ZEB2 and S1PR1, further laying the theoretical foundation for the utilization of FTY720 in NSCLC treatment. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Figure 1

29 pages, 7913 KB  
Article
Synthesis, Characterization, and Screening Anticancer—Antibiofilm Activities of Theophylline Derivatives Containing CF3/OCF3 Moiety
by Serpil Demir Düşünceli, Kübra Açıkalın Coşkun, Murat Kaloğlu, Elvan Üstün, Reyhan Çalışkan and Yusuf Tutar
Biology 2025, 14(9), 1180; https://doi.org/10.3390/biology14091180 - 2 Sep 2025
Viewed by 663
Abstract
Background: Theophylline, which is biologically important and found in tea, coffee, and cocoa beans, can be synthesized chemically or by direct extraction and concentration from natural sources. Theophylline derivatives have garnered attention in recent years for their potential therapeutic effects on Mycobacterium tuberculosis [...] Read more.
Background: Theophylline, which is biologically important and found in tea, coffee, and cocoa beans, can be synthesized chemically or by direct extraction and concentration from natural sources. Theophylline derivatives have garnered attention in recent years for their potential therapeutic effects on Mycobacterium tuberculosis, antihistaminic, anti-inflammatory, and anticancer. Also, trifluoromethyl (CF3) group has also been widely used in drug and agrochemical design. Methods: In this study, a series of new theophylline derivatives containing substituted trifluoromethyl and trifluoromethoxy groups were synthesized. The structures of these new compounds were confirmed by NMR, FT-IR, and elemental analyses. Additionally, the anticancer activities of the molecules were analyzed against VEGFR-2, CYP P450, and estrogen receptor by molecular docking method. Furthermore, in vitro biological effects of the compounds were comprehensively evaluated in cancer (A549 and HeLa) and normal (BEAS-2B) cells. Cell viability was assessed by MTT assay, and selectivity index (SI) values were calculated to determine tumor-specific toxicity. Results: N(7)-substituted theophyllines were prepared by the reaction of 1,3-dimethyl-3,7-dihydro-1H-purine-2,6-dione (theophylline) and trifluoromethyl substituted benzyl halide compounds. The synthesized N(7)-substituted theophyllines were obtained as white powder in high yield. The structure of synthesized compounds was confirmed by various spectroscopic techniques such as 1H, 13C, 19F NMR, and FT-IR spectroscopy, and elemental analysis. The highest interaction was recorded as −5.69 kcal/mol for 3-CF3 substituted against VEGFR-2 structure while the best binding affinity was determined for 4-OCF3 substituted with −6.69 kcal/mol against Human Cytochrome P450 with in silico analysis. The in vitro anticancer activities of the molecules were also evaluated against A549 and HeLa cells, and displayed considerably higher cytotoxicity with 2-CF3, 3-CF3, and 4-CF3 substituted molecules in Hela and A549 cell line. To elucidate the molecular mechanism, apoptosis-related gene expression changes were analyzed by RT-qPCR in A549 and HeLa cells treated with compound 2-CF3. Significant upregulation of pro-apoptotic markers and downregulation of anti-apoptotic genes were observed. Consistently, ELISA-based quantification confirmed increased protein levels of Caspase-3, BAX, and Cytochrome C, and decreased BCL-2, validating the apoptotic mechanism at the protein level. Also, the antibacterial and antibiofilm activity details of the molecules were evaluated against DNA Gyrase, and SarA crystal structures by molecular docking method. The highest interaction was recorded as −5.56 kcal/mol for 2-CF3 substituted with H-bonds with Asn46, Val71, Asp73, and Thr165 against DNA Gyrase crystal structure while 3-CF3 substituted has the best binding affinity against SarA. The in vitro antimicrobial effects of the molecules were also evaluated. Conclusions: The synthesized molecules may provide insight into the development of potential therapeutic agents to the increasing antimicrobial resistance and biofilm-forming capacity of microorganisms. Additionally, compound 2-CF3 substituted exhibited promising and selective anticancer activity through apoptosis induction, supported by gene and protein level evidence. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Graphical abstract

19 pages, 12866 KB  
Article
Solanum lyratum-Derived Solalyraine A1 Suppresses Non-Small Cell Lung Cancer Through Regulation of Exosome Secretion and Related Protein Biomarkers
by Pu Jiang, Liangyu Liu, Lixian Chen, Bing Han and Xiao Du
Pharmaceuticals 2025, 18(9), 1280; https://doi.org/10.3390/ph18091280 - 27 Aug 2025
Viewed by 553
Abstract
Background: Lung cancer is a prevalent malignancy globally, with non-small cell lung cancer (NSCLC) accounting for 80–85% of cases. Solalyraine A1 (SA1) is a steroidal glycoalkaloid derived from Solanum lyratum. However, the effect and mechanism of SA1 on NSCLC remain unclear. [...] Read more.
Background: Lung cancer is a prevalent malignancy globally, with non-small cell lung cancer (NSCLC) accounting for 80–85% of cases. Solalyraine A1 (SA1) is a steroidal glycoalkaloid derived from Solanum lyratum. However, the effect and mechanism of SA1 on NSCLC remain unclear. Methods: The exosomes from SA1-treated A549 cells were prepared and administered to A549 xenograft mice. Proteomics analysis of SA1-treated A549 cells and their exosomes was conducted to assess the mechanism. Bioinformatics analysis was utilized to identify differentially expressed proteins (DEPs) and key signaling pathways. Western blot analysis confirmed the expression of potential targets. Results: SA1 effectively suppressed tumor growth in A549 xenografts, demonstrating a remarkable inhibition rate of 70.48%. A total of 1154 DEPs were identified in A549 cells, primarily associated with the ribosome pathway. Additionally, 746 DEPs were identified in exosomes, mainly involved in the spliceosome pathway. Five highly regulated DEPs were selected for verification. SA1 was found to suppress MUC5B and elevate APOB expression in A549 cells, while inhibiting MFGM, ANGL4 and increasing GCN1 expression in exosomes. Conclusions: This study demonstrates that SA1 exhibits anti-NSCLC effects by regulating exosome function and related protein expression, providing novel insights for NSCLC treatment. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Figure 1

22 pages, 3676 KB  
Article
Multiple Strategies Confirm the Anti Hepatocellular Carcinoma Effect of Cinnamic Acid Based on the PI3k-AKT Pathway
by Jiageng Guo, Lijiao Yan, Qi Yang, Huaying Li, Yu Tian, Jieyi Yang, Jinling Xie, Fan Zhang and Erwei Hao
Pharmaceuticals 2025, 18(8), 1205; https://doi.org/10.3390/ph18081205 - 14 Aug 2025
Viewed by 698
Abstract
Background: Hepatocellular carcinoma is one of the leading causes of cancer-related deaths worldwide. Its high recurrence rate and limited treatment options underscore the urgent need for the development of new and highly effective drugs. Methods: This study systematically explores the molecular mechanism [...] Read more.
Background: Hepatocellular carcinoma is one of the leading causes of cancer-related deaths worldwide. Its high recurrence rate and limited treatment options underscore the urgent need for the development of new and highly effective drugs. Methods: This study systematically explores the molecular mechanism of cinnamic acid against hepatocellular carcinoma through integrated machine learning prediction, network pharmacological analysis and in vitro experimental verification. Results: The prediction of anti-tumor activity based on the random forest model showed that cinnamic acid has significant anti-tumor potential (probability = 0.69). Network pharmacology screened 185 intersection targets of cinnamic acid and liver cancer, of which 39 core targets (such as PIK3R1, AKT1, MAPK1) were identified as key regulatory hubs through protein interaction network and topological analysis. Functional enrichment analysis showed that these targets were mainly enriched in the PI3K/AKT signaling pathway (p = 2.1 × 10−12), the cancer pathway (p = 3.8 × 10−10), and apoptosis-related biological processes. Molecular docking validation revealed that the binding energies of cinnamic acid with the 19 core targets were all below −5 kcal/mol, a threshold indicating strong binding affinity in molecular docking. The binding modes to PIK3R1 (−5.4 kcal/mol) and AKT1 (−5.1 kcal/mol) stabilized through hydrogen bonding. In vitro, cinnamic acid dose-dependently inhibited Hep3B proliferation/migration, induced apoptosis, downregulated PI3K, p-AKT, and Bcl-2, and upregulated Bax and Caspase-3/8. Conclusions: This study systematically reveals, for the first time, that the multi-target mechanism of cinnamic acid exerts anti-hepatic cancer effects by targeting the PI3K/AKT signaling pathway, supporting its potential as a natural anti-tumor drug. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Figure 1

13 pages, 1537 KB  
Article
Correlation of SERPINA-1 Gene Over-Expression with Inhibition of Cell Proliferation and Modulation of the Expression of IL-6, Furin, and NSD2 Genes
by Nassim Tassou, Hajar Anibat, Ahmed Tissent and Norddine Habti
Biologics 2025, 5(3), 22; https://doi.org/10.3390/biologics5030022 - 6 Aug 2025
Viewed by 667
Abstract
Background and Objectives: The cytokine IL-6, methyltransferase NSD2, pro-protein convertase Furin, and growth factor receptor IGF-1R are essential factors in the proliferation of cancer cells. These proteins are involved in the tumor process by generating several cell-signaling pathways. However, the interactions of these [...] Read more.
Background and Objectives: The cytokine IL-6, methyltransferase NSD2, pro-protein convertase Furin, and growth factor receptor IGF-1R are essential factors in the proliferation of cancer cells. These proteins are involved in the tumor process by generating several cell-signaling pathways. However, the interactions of these oncogenic biomarkers, Furin, IL-6, and NSD2, and their links with the inhibitor SERPINA-1 remain largely unknown. Materials and Methods: Cell proliferation is measured by colorimetric and enzymatic methods. The genetic expressions of SERPINA-1, Furin, IL-6, and NSD2 are measured by qRT-PCR, while the expression of IGF-1R on the cell surface is measured by flow cytometry. Results: The proliferation of cells overexpressing SERPINA-1 (JP7pSer+) is decreased by more than 90% compared to control cells (JP7pSer-). The kinetics of the gene expression ratios of Furin, IL-6, and NSD2 show an increase for 48 h, followed by a decrease after 72 h for the three biomarkers in JP7pSer+ cells compared to JP7pSer- cells. The expression of IGF-1R on the cell surface in both cell lines is low, with JP7pSer- cells expressing 1.33 times more IGF-1R than JP7pSer+ cells. Conclusions: These results suggest gene correlations of SERPINA-1 overexpression with decreased cell proliferation and modulation of gene expression of Furin, IL-6, and NSD2. This study should be complemented by molecular transcriptomic and proteomic experiments to better understand the interaction of SERPINA-1 with IL-6, Furin, and NSD2, and their effect on tumor progression. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Figure 1

20 pages, 3781 KB  
Article
Ginsenoside Rg3 Adjunctively Increases the Efficacy of Gefitinib Against NSCLC by Regulating EGFR Copy Number
by Xinyi Lv, Yuehan Song, Tianhua Liu, Dingdan Zhang, Xinpeng Ye, Qingqing Wang, Rongrong Li, Jiayi Chen, Shujing Zhang, Xue Yu and Chunying Hou
Pharmaceuticals 2025, 18(7), 1077; https://doi.org/10.3390/ph18071077 - 21 Jul 2025
Viewed by 746
Abstract
Background: Lung cancer has the highest morbidity and mortality of all tumors, and the development of TKI drugs targeting EGFR activating mutations has brought lung cancer treatment into the targeted era. In view of their low efficacy and susceptibility to drug resistance, [...] Read more.
Background: Lung cancer has the highest morbidity and mortality of all tumors, and the development of TKI drugs targeting EGFR activating mutations has brought lung cancer treatment into the targeted era. In view of their low efficacy and susceptibility to drug resistance, there is an urgent need to find strategies to increase their efficacy and reduce the incidence of drug resistance. Methods: In this study, we examined the distribution and probability of EGFR mutations in non-small cell lung cancer patients in the cBioPortal database and compared the survival prognosis of patients with normal and abnormal EGFR, NSCLC patients treated with and without TKI, and NSCLC patients with different EGFR gene copy numbers. We established a mouse lung cancer model and examined the histomorphological characteristics of lung tissues via hematoxylin and eosin staining. Additionally, changes in the copy number of the EGFR gene and its protein expression levels were detected using RT-qPCR and Western blotting. Furthermore, we quantified the concentration of the EGFR protein using ELISA. Results: We found no significant advantage of EGFR-TKI therapy over first-line chemotherapeutic agents in patients with EGFR-abnormal NSCLC. The reason for this may be related to the abnormal EGFR gene copy number; the higher the copy number increases, the worse the survival prognosis of the patients. In molecular biology experiments, we demonstrated that ginsenoside Rg3 down-regulated the copy number of 18, 19, 20, and 21 exons and protein expression of EGFR in lung adenocarcinoma cells. The results of in vivo pharmacodynamic assays confirmed that sequential administration of ginsenoside Rg3 with TKI drugs could achieve a gainful complementary effect. Conclusions: Ginsenoside Rg3 down-regulates the copy number of EGFR important exons in EGFR-mutant cells of lung adenocarcinoma and reduces EGFR protein expression, thus providing a high gainful complementary effect in combination with EGFR-TKI. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Figure 1

21 pages, 1210 KB  
Article
Taurine-Based Hybrid Drugs as Potential Anticancer Therapeutic Agents: In Vitro, In Vivo Evaluations
by Saltanat Nakypova, Andrey Smolobochkin, Tanzilya Rizbayeva, Rakhymzhan Turmanov, Almir Gazizov, Nurgali Akylbekov, Rakhmetulla Zhapparbergenov, Roza Narmanova, Saltanat Ibadullayeva, Alena Zalaltdinova, Marat Syzdykbayev, Julia Voronina, Anna Lyubina, Alexandra Voloshina, Elena Klimanova, Tatiana Sashenkova, Denis Mishchenko and Alexander Burilov
Pharmaceuticals 2025, 18(7), 1056; https://doi.org/10.3390/ph18071056 - 18 Jul 2025
Viewed by 895
Abstract
Background/Objectives: The development of antitumor agents possessing low toxicity against non-cancerous cells is still a challenge in medicinal chemistry. In this paper, we report the antitumor activity of “hybrid structures” derived from the amino acid taurine. We have synthesized 26 compounds, structures [...] Read more.
Background/Objectives: The development of antitumor agents possessing low toxicity against non-cancerous cells is still a challenge in medicinal chemistry. In this paper, we report the antitumor activity of “hybrid structures” derived from the amino acid taurine. We have synthesized 26 compounds, structures of which were confirmed using NMR, X-ray diffractometry, and other techniques. Cytotoxicity of the obtained compounds has been evaluated using three human cancer cell lines. Pyrrolidine 4p has exhibited the strongest antiproliferative activity against HL-60 cells with an IC50 of 76.7 μM, while IC50 against normal cells was 176.3 μM. Water-soluble derivatives of taurine have been tested for antileukemia activity in mice of the BDF1 line. Compound 4p has been identified as the leading compound, which increases the mean survival time of mice from 40 to 100% as compared to the control group. Together, these results prove that taurine-based hybrid structures can be a promising scaffold for the discovery of potential antiproliferative agents. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Graphical abstract

Back to TopTop