Anticancer Drug Targeting Using Novel Delivery Systems and Medical Devices

A special issue of Pharmaceutics (ISSN 1999-4923). This special issue belongs to the section "Drug Targeting and Design".

Deadline for manuscript submissions: closed (31 January 2023) | Viewed by 4630

Special Issue Editors


E-Mail Website
Guest Editor
School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
Interests: human and veterinary pharmaceuticals; advanced drug delivery; cancer; infection; 3D printing
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences (MCOPS), Manipal University, Manipal, Karnataka 576104, India
Interests: controlled drug delivery; polymeric nanoparticles; liposomes; nanoemulsions; lipid nanoparticles; chronotherapeutics; solubility enhancement
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Cancer is one of the leading causes of death worldwide and as per the cancer reports, it is expected that by the year 2040, the cases may rise by 29.5 million. A plethora of strategies are available in the market for the treatment of cancer, which has been widely used in clinics. These include chemotherapy, immunotherapy, targeted therapies, and a combination of radiation-chemotherapy. Despite these strategies, multidrug resistance remains a major obstacle in the effectiveness of cancer therapy. To combat this, novel targeted approaches encompassing organic and inorganic nanocarriers, cancer stem cell therapy, biomaterials have been explored for improving its therapeutic efficacy over the conventional approaches. These help in the preferential uptake of the therapeutic agents by the tumor cells leading to their eradication with reduced side effects and alleviating the likelihood of multidrug resistance.

This Special Issue focuses on an assemblage of the key findings and contributions focused on the area of novel drug delivery systems and medical devices research in cancer therapy, diagnostics and bioimaging. In this issue, we welcome original research, short communications, and reviews with key topics comprising of the novel, innovative therapeutic approaches involving the synthesis, targeted drug delivery for theranostics purposes, multimodal drug therapy for combating multidrug resistance, biomedical devices addressing the fields of cancer therapy and diagnostics.

Prof. Dr. Sanjay Garg
Dr. Usha Y. Nayak
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. Pharmaceutics is an international peer-reviewed open access monthly 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 2900 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.

Published Papers (2 papers)

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

Research

14 pages, 2430 KiB  
Article
Glucose-Modified Zein Nanoparticles Enhance Oral Delivery of Docetaxel
by Yabing Xing, Xiao Li, Weiwei Cui, Meng Xue, Yanan Quan and Xinhong Guo
Pharmaceutics 2022, 14(7), 1361; https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics14071361 - 27 Jun 2022
Cited by 6 | Viewed by 1632
Abstract
Based on glucose (G) transporters (GLUTs), structuring nanoparticles with G as a target are an effective strategy to enhance oral bioavailability and anti-tumor effects of drugs. A novel drug delivery system using G-modified zein (GZ) nanoparticles loaded with docetaxel (DTX) (DTX-GNPs) was prepared [...] Read more.
Based on glucose (G) transporters (GLUTs), structuring nanoparticles with G as a target are an effective strategy to enhance oral bioavailability and anti-tumor effects of drugs. A novel drug delivery system using G-modified zein (GZ) nanoparticles loaded with docetaxel (DTX) (DTX-GNPs) was prepared and characterized in vitro and in vivo via assessment of cellular uptake, absorption site, pharmacokinetics, ex vivo distribution, and anti-tumor effects. The DTX-GNPs were approximately 120 nm in size. Compared with DTX-NPs, G modification significantly enhanced cellular uptake of DTX-GNPs by 1.22 times in CaCo-2 cells, which was related to GLUT mediation and the enhancement of endocytosis pathways via clathrin, micropinocytosis, and caveolin. Compared to DTX-NPs, G modification significantly enhanced DTX-NP absorption in the jejunum and ileum, delayed plasma concentration peak time, prolonged the average residence time in vivo, and increased oral bioavailability (from 43.82% to 96.04%). Cellular uptake and oral bioavailability of DTX were significantly affected by the G modification ratio. Compared with DTX-NPs, G modification significantly reduced drug distribution in the liver, lungs, and kidneys and increased tumor distribution and tumor growth inhibition rate without obvious systemic toxicity. This study demonstrated the potential of GZ-NPs as nanocarriers for DTX to enhance oral bioavailability and anti-tumor effects. Full article
Show Figures

Graphical abstract

16 pages, 2107 KiB  
Article
Effects of a Novel Thiadiazole Derivative with High Anticancer Activity on Cancer Cell Immunogenic Markers: Mismatch Repair System, PD-L1 Expression, and Tumor Mutation Burden
by Sofia Sagredou, Panagiotis Dalezis, Eirini Papadopoulou, Maria Voura, Maria V. Deligiorgi, Michail Nikolaou, Mihalis I. Panayiotidis, George Nasioulas, Vasiliki Sarli and Dimitrios T. Trafalis
Pharmaceutics 2021, 13(6), 885; https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics13060885 - 15 Jun 2021
Cited by 1 | Viewed by 2385
Abstract
Microsatellite instability (MSI), tumor mutation burden (TMB), and programmed cell death ligand-1 (PD-L1) are particularly known as immunotherapy predictive biomarkers. MSI and TMB are closely related to DNA mismatch repair (MMR) pathway functionality, while the PD-L1 checkpoint mediates cancer cell evasion from immune [...] Read more.
Microsatellite instability (MSI), tumor mutation burden (TMB), and programmed cell death ligand-1 (PD-L1) are particularly known as immunotherapy predictive biomarkers. MSI and TMB are closely related to DNA mismatch repair (MMR) pathway functionality, while the PD-L1 checkpoint mediates cancer cell evasion from immune surveillance via the PD-L1/PD-1 axis. Among all the novel triazolo[3,4-b]thiadiazole derivatives, the compound KA39 emerged as the most potent anticancer agent. In the present study, potential alterations in MSI, TMB, and/or PD-L1 expression upon cell treatment with KA39 are explored. We tested three MMR-deficient (DLD-1, LS174T, and DU-145) and two MMR-proficient (HT-29 and PC-3) human cancer cell lines. Our findings support KA39-induced PD-L1 overexpression in all cancer cell lines, although the most outstanding increase was observed in MMR-proficient HT-29 cells. MSI analysis showed that KA39 affects the MMR system, impairing its recognition or repair activity, particularly in MMR-deficient DLD-1 and DU-145 cells, enhancing oligonucleotide production. There were no remarkable alterations in the TMB between untreated and treated cells, indicating that KA39 does not belong to mutagenic agents. Taking together the significant in vitro anticancer activity with PD-L1 upregulation and MSI increase, KA39 should be investigated further for its implication in chemo-immunotherapy of cancer. Full article
Show Figures

Figure 1

Back to TopTop