Therapeutic Target Developments in Pancreatic Cancer

A special issue of Pharmaceuticals (ISSN 1424-8247). This special issue belongs to the section "Pharmacology".

Deadline for manuscript submissions: 12 May 2024 | Viewed by 6179

Special Issue Editors


E-Mail Website
Guest Editor
Molecular Pharmacology at School of Chemical Engineering, Dalian University of Technology, Dalian, China
Interests: therapeutic targets; pancreatic biology; drug resistance; tumor microenvironment; cancer metabolism

E-Mail Website
Guest Editor
Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
Interests: apoptosis; cancer biomarkers; cancer cell metabolism; cell signaling; ferroptosis; mitochondrial proteases; mitochonarial disease; mitochondrial protein homeostasis; nonalcoholic fatty liver disease

Special Issue Information

Dear Colleagues,

Pancreatic ductal adenocarcinoma (PDAC) is the fourth most common cause of cancer death in the Western world. The prognosis is poor, with 1- and 5-year survival rates of about 20% and 6%, respectively. Systemic chemotherapy administered either after tumor resection surgery or in patients with metastatic disease has been shown to prolong survival; however, surgery is the only curative treatment. About 20% of patients with pancreatic cancer can be operated on with curative intent, because most have locally advanced or metastatic pancreatic cancer at the time of diagnosis. Resistance to therapy resulting from the unique tumor environment of PDAC, consisting of desmoplastic stroma, immune suppressive cells, and activated fibroblasts, as well as a lack of biomarkers, remains a clinical challenge, one that is unmet in terms of the development of novel therapeutic targets for an effective administration of the disease. In this Special Issue, we would like to solicit manuscripts covering the development of novel therapeutical drug targets and their associated mechanisms, which can provide a basis for exploring new targets and developing more advanced strategies.

Prof. Dr. Guishan Xiao
Dr. Bin Lu
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. Pharmaceuticals 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.

Keywords

  • therapeutic targets
  • pancreatic biology
  • drug resistance
  • tumor microenvironment
  • cancer metabolism

Published Papers (3 papers)

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

Research

19 pages, 3732 KiB  
Article
A Bitter Taste Receptor as a Novel Molecular Target on Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma
by Jessica Hung, Samantha M. Perez, Siva Sai Krishna Dasa, Sarah P. Hall, Danielle B. Heckert, Brian P. Murphy, Howard C. Crawford, Kimberly A. Kelly and Lindsey T. Brinton
Pharmaceuticals 2023, 16(3), 389; https://doi.org/10.3390/ph16030389 - 03 Mar 2023
Cited by 2 | Viewed by 2502
Abstract
Cancer-associated fibroblasts (CAFs) execute diverse and complex functions in cancer progression. While reprogramming the crosstalk between CAFs and cancer epithelial cells is a promising avenue to evade the adverse effects of stromal depletion, drugs are limited by their suboptimal pharmacokinetics and off-target effects. [...] Read more.
Cancer-associated fibroblasts (CAFs) execute diverse and complex functions in cancer progression. While reprogramming the crosstalk between CAFs and cancer epithelial cells is a promising avenue to evade the adverse effects of stromal depletion, drugs are limited by their suboptimal pharmacokinetics and off-target effects. Thus, there is a need to elucidate CAF-selective cell surface markers that can improve drug delivery and efficacy. Here, functional proteomic pulldown with mass spectrometry was used to identify taste receptor type 2 member 9 (TAS2R9) as a CAF target. TAS2R9 target characterization included binding assays, immunofluorescence, flow cytometry, and database mining. Liposomes conjugated to a TAS2R9-specific peptide were generated, characterized, and compared to naked liposomes in a murine pancreatic xenograft model. Proof-of-concept drug delivery experiments demonstrate that TAS2R9-targeted liposomes bind with high specificity to TAS2R9 recombinant protein and exhibit stromal colocalization in a pancreatic cancer xenograft model. Furthermore, the delivery of a CXCR2 inhibitor by TAS2R9-targeted liposomes significantly reduced cancer cell proliferation and constrained tumor growth through the inhibition of the CXCL-CXCR2 axis. Taken together, TAS2R9 is a novel cell-surface CAF-selective target that can be leveraged to facilitate small-molecule drug delivery to CAFs, paving the way for new stromal therapies. Full article
(This article belongs to the Special Issue Therapeutic Target Developments in Pancreatic Cancer)
Show Figures

Graphical abstract

15 pages, 2072 KiB  
Article
LncRNA SNHG6 Upregulates KPNA5 to Overcome Gemcitabine Resistance in Pancreatic Cancer via Sponging miR-944
by Ge Gao, Xin Li, Hui Wu, Ling-li Huang, Yu-xin Lin, Zhi Huo, Zhong-yuan Xiang and Xiao Zhou
Pharmaceuticals 2023, 16(2), 184; https://doi.org/10.3390/ph16020184 - 25 Jan 2023
Cited by 4 | Viewed by 1312
Abstract
Gemcitabine (GEM) is the gold-standard therapeutic regimen for patients with pancreatic cancer (PC); however, patients may receive limited benefits due to the drug resistance of GEM. LncRNA SNHG6 is reported to play key roles in drug resistance, but its role and molecular mechanism [...] Read more.
Gemcitabine (GEM) is the gold-standard therapeutic regimen for patients with pancreatic cancer (PC); however, patients may receive limited benefits due to the drug resistance of GEM. LncRNA SNHG6 is reported to play key roles in drug resistance, but its role and molecular mechanism in PC remain incompletely understood. We found that LncRNA SNHG6 is drastically downregulated in GEM-resistant PC and is positively correlated with the survival of PC patients. With the help of bioinformatic analysis and molecular approaches, we show that LncRNA SNHG6 can sponge miR-944, therefore causing the upregulation of the target gene KPNA5. In vitro experiments showed that LncRNA SNHG6 and KPNA5 suppress PC cell proliferation and colony formation. The Upregulation of LncRNA SNHG6 and KPNA5 increases the response of GEM-resistant PANC-1 cells to GEM. We also show that the expression of KPNA5 is higher in patients without GEM resistance than in those who developed GEM resistance. In summary, our findings indicate that the LncRNA SNHG6/miR944/KPNA5 axis plays a pivotal role in overcoming GEM resistance, and targeting this axis may contribute to an increasing of the benefits of PC patients from GEM treatment. Full article
(This article belongs to the Special Issue Therapeutic Target Developments in Pancreatic Cancer)
Show Figures

Graphical abstract

19 pages, 3231 KiB  
Article
Exploitation and Verification of a Stroma- and Metastasis-Associated Risk Prognostic Signature in Pancreatic Adenocarcinoma
by Jia-Hao Zheng, Hong-Fei Yao, Zong-Hao Duan, Pei-Xuan Ji, Jian Yang, Yu-Heng Zhu, Qin-Yuan Jia, Jian-Yu Yang, De-Jun Liu, Yong-Wei Sun, Peng-Cheng Chen, Pei-Dong Shi and Li Chen
Pharmaceuticals 2022, 15(11), 1336; https://doi.org/10.3390/ph15111336 - 28 Oct 2022
Viewed by 1368
Abstract
Pancreatic adenocarcinoma (PAAD), one of the most malignant tumors, not only has abundant mesenchymal components, but is also characterized by an extremely high metastatic risk. The purpose of this study was to construct a model of stroma- and metastasis-associated prognostic signature, aiming to [...] Read more.
Pancreatic adenocarcinoma (PAAD), one of the most malignant tumors, not only has abundant mesenchymal components, but is also characterized by an extremely high metastatic risk. The purpose of this study was to construct a model of stroma- and metastasis-associated prognostic signature, aiming to benefit the existing clinical staging system and predict the prognosis of patients. First, stroma-associated genes were screened from the TCGA database with the ESTIMATE algorithm. Subsequently, transcriptomic data from clinical tissues in the RenJi cohort were screened for metastasis-associated genes. Integrating the two sets of genes, we constructed a risk prognostic signature by Cox and LASSO regression analysis. We then obtained a risk score by a quantitative formula and divided all samples into high- and low-risk groups based on the scores. The results demonstrated that patients with high-risk scores have a worse prognosis than those with low-risk scores, both in the TCGA database and in the RenJi cohort. In addition, tumor mutation burden, chemotherapeutic drug sensitivity and immune infiltration analysis also exhibited significant differences between the two groups. In exploring the potential mechanisms of how stromal components affect tumor metastasis, we simulated different matrix stiffness in vitro to explore its effect on EMT key genes in PAAD cells. We found that cancer cells stimulated by high matrix stiffness may trigger EMT and promote PAAD metastasis. Full article
(This article belongs to the Special Issue Therapeutic Target Developments in Pancreatic Cancer)
Show Figures

Figure 1

Planned Papers

The below list represents only planned manuscripts. Some of these manuscripts have not been received by the Editorial Office yet. Papers submitted to MDPI journals are subject to peer-review.

Tentative outline:

Endoscopic Ultrasound Tissue Acquisition in the era of precision medicine for pancreatic cancer

 

Alberto Fantin, Martina Tessari, Ottavia De Simoni, Giulia Peserico, Monica Franco, Annalisa Masier, Sabina Grillo, Cingolani Linda, Chiara Cristofori, Tiziana Morbin, Attilio Pirillo, Mario Gruppo

 

Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal malignancies and it has been estimated that pancreatic cancer will surpass breast cancer as the third leading cause of cancer death by 2025. Regarding the diagnosis of pancreatic cancer, Endoscopic Ultrasound (EUS) is more sensitive, specific and accurate in the detection of pancreatic lesions than high-quality cross-sectional imaging, furthermore it provides the histopathological diagnosis performing EUS-guided tissue acquisition (EUS-TA). Unfortunately, the majority of PDAC patients are diagnosed at late tumor stages with locally advanced or metastatic  cancer and the overall survival at 5 years is about 5%. To improve survival in patients with PDAC, current research aims at identifying molecular targets and subgroups of PDAC that may benefit from personalized treatment, opening a new landscape for the so-called ‘precision oncology’ as well as finding biomarkers predicting a better treatment response. Recent insights from modern next-generation sequencing (NGS) technologies shed light on the biological rationale of the results achieved so far and showed the chance to improve the molecular characteristics of this cancer in order to better define a treatment.

The concept of precision medicine for PDAC has become popular in recent years and many work groups have made important efforts to describe its genetic and molecular alterations in order to stratify subclasses of patients with prognostic relevance and possible molecular based-treatment opportunity. Genomic profiling has revealed several actionable alterations and therapies targeting aberrations such as BRCA1/2 mutations, mismatch repair (MMR) deficiencies or NTRK1–3 fusions are now available in this subset of patients. The real-world implication of molecular classification remains to be established and numerous clinical trials are currenting under development to enforce precision medicine in clinical practice.

Other studies were able to create pancreatic cancer organoids that mimic the original characteristic of primitive neoplasm by means of EUS-TA to enable testing targeted drugs.

In this scenario,  thanks to the ability to obtain tissue samples with high adequacy, EUS-TA allows to deepen the knowledge of cancer by expanding the offer of effective drugs against it.

Back to TopTop