molecules-logo

Journal Browser

Journal Browser

Design, Synthesis, and Evaluation of Anticancer Drugs

A special issue of Molecules (ISSN 1420-3049). This special issue belongs to the section "Medicinal Chemistry".

Deadline for manuscript submissions: closed (31 October 2023) | Viewed by 6396

Special Issue Editors


E-Mail Website
Guest Editor
Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud Post, Poojappura, Thiruvananthapuram 695014, Kerala, India
Interests: bioprospecting; chemosensitization; chemoprevention; chemotherapeutics; cancer immunology; anticancer drugs; molecular signalling

E-Mail Website
Guest Editor
Department of Biotechnology and Microbiology Kannur University, Thalassery Campus Palayad P.O., Kannur 670661, Kerala, India
Interests: computational and structural biology: studies on protein–ligand interactions; identification of enzyme inhibitors; structure-based drug design using molecular modelling and docking

E-Mail Website
Guest Editor
Department of Biotechnology, University of Calicut, Malappuram 673635, Kerala, India
Interests: bioprospecting; chemosensitization; chemoprevention; chemotherapeutics; anticancer drugs; molecular signalling

E-Mail Website
Guest Editor
Rajiv Gandhi Centre for Biotechnology (RGCB), Thycaud Post, Poojappura, Thiruvananthapuram 695014, Kerala, India
Interests: cancer biology; molecular biology

Special Issue Information

Dear Colleagues,

Natural compounds and their secondary metabolites form an integral part of anti-cancer drugs. Hence, the design, synthesis and evaluation of these molecules have a major role in drug discovery programs. The currently available therapeutic options for cancer have a number of drawbacks, the most common being multi-drug resistance followed by undesirable off-target effects and uncertainty in terms of their efficacies. The death rate that is directly attributable to cancer is still quite high. Hence, in order to successfully treat a wide variety of lethal malignancies, the discovery of innovative anticancer medicines is an absolute requirement. Elucidating the molecular targets and developing and designing drugs, which target these signaling pathways, make it possible to establish novel therapeutic strategies. Recently, new methods have been developed for screening anti-cancer drugs, such as chemical-based drugs, plant-based natural drugs, antibodies, peptides, aptamers, etc. This Special Issue in “Molecules” aims to further explore the current developments in all fields that are connected with the design, synthesis and evaluation of anticancer drug research.

Dr. Ruby John Anto
Dr. C. Sadasivan
Dr. Smitha Vadakkeveettil Bava
Dr. Kalishwaralal Kalimuthu
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Molecules is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • cancer drug synthesis
  • plant-derived natural drugs
  • structure- based drug designing
  • in silico drug discovery
  • developing new assays for drug discovery
  • screening of specific target molecules
  • peptide drug conjugation
  • antibody drug conjugation
  • aptamer drug conjugation
  • small molecules
  • apoptosis
  • ferroptosis
  • cuproptosis
  • autophagy
  • necroptosis

Published Papers (3 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

22 pages, 5060 KiB  
Article
Activity and Selectivity of Novel Chemical Metallic Complexes with Potential Anticancer Effects on Melanoma Cells
by Maria Camilla Ciardulli, Annaluisa Mariconda, Marco Sirignano, Erwin Pavel Lamparelli, Raffaele Longo, Pasqualina Scala, Raffaella D’Auria, Antonietta Santoro, Liberata Guadagno, Giovanna Della Porta and Pasquale Longo
Molecules 2023, 28(12), 4851; https://doi.org/10.3390/molecules28124851 - 19 Jun 2023
Cited by 1 | Viewed by 1575
Abstract
Human malignant melanoma cells from lymph node metastatic site (MeWo) were selected for testing several synthesized and purified silver(I) and gold(I) complexes stabilized by unsymmetrically substituted N-heterocyclic carbene (NHC) ligands, called L20 (N-methyl, N′-[2-hydroxy ethylphenyl]imidazol-2-ylide) and M1 (4,5-dichloro, N-methyl, N′-[2-hydroxy ethylphenyl]imidazol-2-ylide), having halogenide [...] Read more.
Human malignant melanoma cells from lymph node metastatic site (MeWo) were selected for testing several synthesized and purified silver(I) and gold(I) complexes stabilized by unsymmetrically substituted N-heterocyclic carbene (NHC) ligands, called L20 (N-methyl, N′-[2-hydroxy ethylphenyl]imidazol-2-ylide) and M1 (4,5-dichloro, N-methyl, N′-[2-hydroxy ethylphenyl]imidazol-2-ylide), having halogenide (Cl or I) or aminoacyl (Gly=N-(tert-Butoxycarbonyl)glycinate or Phe=(S)-N-(tert-Butoxycarbonyl)phenylalaninate) counterion. For AgL20, AuL20, AgM1 and AuM1, the Half-Maximal Inhibitory Concentration (IC50) values were measured, and all complexes seemed to reduce cell viability more effectively than Cisplatin, selected as control. The complex named AuM1 was the most active just after 8 h of treatment at 5 μM, identified as effective growth inhibition concentration. AuM1 also showed a linear dose and time-dependent effect. Moreover, AuM1 and AgM1 modified the phosphorylation levels of proteins associated with DNA lesions (H2AX) and cell cycle progression (ERK). Further screening of complex aminoacyl derivatives indicated that the most powerful were those indicated with the acronyms: GlyAg, PheAg, AgL20Gly, AgM1Gly, AuM1Gly, AgL20Phe, AgM1Phe, AuM1Phe. Indeed, the presence of Boc-Glycine (Gly) and Boc-L-Phenylalanine (Phe) showed an improved efficacy of Ag main complexes, as well as that of AuM1 derivatives. Selectivity was further checked on a non-cancerous cell line, a spontaneously transformed aneuploid immortal keratinocyte from adult human skin (HaCaT). In such a case, AuM1 and PheAg complexes resulted as the most selective allowing HaCaT viability at 70 and 40%, respectively, after 48 h of treatment at 5 μM. The same complexes tested on 3D MeWo static culture induced partial spheroid disaggregation after 24 h of culture, with almost half of the cells dead. Full article
(This article belongs to the Special Issue Design, Synthesis, and Evaluation of Anticancer Drugs)
Show Figures

Figure 1

18 pages, 4241 KiB  
Article
Characterization of Thymoquinone-Sulfobutylether-β-Cyclodextrin Inclusion Complex for Anticancer Applications
by Eltayeb E. M. Eid, Amer A. Almaiman, Salah Abdalrazak Alshehade, Wardah Alsalemi, Sareh Kamran, FakhrEldin O. Suliman and Mohammed Abdullah Alshawsh
Molecules 2023, 28(10), 4096; https://doi.org/10.3390/molecules28104096 - 15 May 2023
Cited by 10 | Viewed by 2278
Abstract
Thymoquinone (TQ) is a quinone derived from the black seed Nigella sativa and has been extensively studied in pharmaceutical and nutraceutical research due to its therapeutic potential and pharmacological properties. Although the chemopreventive and potential anticancer effects of TQ have been reported, its [...] Read more.
Thymoquinone (TQ) is a quinone derived from the black seed Nigella sativa and has been extensively studied in pharmaceutical and nutraceutical research due to its therapeutic potential and pharmacological properties. Although the chemopreventive and potential anticancer effects of TQ have been reported, its limited solubility and poor delivery remain the major limitations. In this study, we aimed to characterize the inclusion complexes of TQ with Sulfobutylether-β-cyclodextrin (SBE-β-CD) at four different temperatures (293–318 K). Additionally, we compared the antiproliferative activity of TQ alone to TQ complexed with SBE-β-CD on six different cancer cell lines, including colon, breast, and liver cancer cells (HCT-116, HT-29, MDA-MB-231, MCF-7, SK-BR-3, and HepG2), using an MTT assay. We calculated the thermodynamic parameters (ΔH, ΔS, and ΔG) using the van’t Holf equation. The inclusion complexes were characterized by X-ray diffraction (XRD), Fourier transforms infrared (FT-IR), and molecular dynamics using the PM6 model. Our findings revealed that the solubility of TQ was improved by ≥60 folds, allowing TQ to penetrate completely into the cavity of SBE-β-CD. The IC50 values of TQ/SBE-β-CD ranged from 0.1 ± 0.01 µg/mL against SK-BR-3 human breast cancer cells to 1.2 ± 0.16 µg/mL against HCT-116 human colorectal cancer cells, depending on the cell line. In comparison, the IC50 values of TQ alone ranged from 0.2 ± 0.01 µg/mL to 4.7 ± 0.21 µg/mL. Overall, our results suggest that SBE-β-CD can enhance the anticancer effect of TQ by increasing its solubility and bioavailability and cellular uptake. However, further studies are necessary to fully understand the underlying mechanisms and potential side effects of using SBE-β-CD as a drug delivery system for TQ. Full article
(This article belongs to the Special Issue Design, Synthesis, and Evaluation of Anticancer Drugs)
Show Figures

Figure 1

12 pages, 6620 KiB  
Article
Enhanced Intracellular Photosensitizer Uptake and Retention by Targeting Viral Oncoproteins in Human Papillomavirus Infected Cancer Cells and Cancer Stem Cells
by Elvin Peter Chizenga and Heidi Abrahamse
Molecules 2023, 28(2), 647; https://doi.org/10.3390/molecules28020647 - 8 Jan 2023
Viewed by 1891
Abstract
Immunogenic proteins in cancer are relevant targets for drug delivery. In Photodynamic Therapy (PDT), surface antigens have previously been used to deliver the photosensitizer (PS) to the tumor microenvironment for specific targeting. However, can we target intracellular antigens to achieve more than surface [...] Read more.
Immunogenic proteins in cancer are relevant targets for drug delivery. In Photodynamic Therapy (PDT), surface antigens have previously been used to deliver the photosensitizer (PS) to the tumor microenvironment for specific targeting. However, can we target intracellular antigens to achieve more than surface recognition? Can we possibly increase PS intracellular localization and prevent drug efflux at the same time? In this study, these questions were addressed by using a compound that can not only specifically recognize and bind to intracellular E6 oncoproteins in Human Papillomavirus (HPV)-Transformed cancer cells, but is also capable of enhancing transmembrane uptake using the cells’ own active transport mechanisms. HPV-transformed SiHa cells were cultured in vitro, and the resistant subpopulation was isolated using Magnetic Activated Cell Sorting (MACS). PDT was performed on four different cell types with varying physiognomies in terms of HPV oncoprotein expression and physiological form. Results demonstrated that tagging PSs on a carrier molecule that specifically delivers the PS inside the cells that express the target proteins enhanced both cellular uptake and retention of the PS even in the presence of drug efflux proteins on resistant subpopulations. These findings provide insight into the possibility of preventing cell-mediated resistance to PDT. Full article
(This article belongs to the Special Issue Design, Synthesis, and Evaluation of Anticancer Drugs)
Show Figures

Figure 1

Back to TopTop