Your new experience awaits. Try the new design now and help us make it even better

EDITORIAL article

Front. Oncol., 10 October 2025

Sec. Pharmacology of Anti-Cancer Drugs

Volume 15 - 2025 | https://doi.org/10.3389/fonc.2025.1705678

This article is part of the Research TopicDrug Development for Pancreatic Cancer: Novel Targets, Drug Candidates and MechanismsView all 5 articles

Editorial: Drug development for pancreatic cancer: novel targets, drug candidates and mechanisms

  • 1Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
  • 2School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China

Pancreatic ductal adenocarcinoma (PDAC), the most prevalent subtype of pancreatic cancer, constitutes approximately 90% of all pancreatic cancer cases. It is recognized as one of the most aggressive malignancies globally, with a five-year survival rate persistently around 12% (1). The absence of early specific symptoms and effective screening methods results in over 80% of patients being diagnosed at an advanced stage, frequently with local invasion or distant metastasis, thereby significantly limiting treatment options and contributing to a poor prognosis (2). This challenging scenario highlights the urgent necessity to enhance our understanding of the biological mechanisms underlying PDAC and to develop innovative therapeutic strategies.

While engineered T cell therapy has demonstrated significant success in treating hematologic malignancies, its application in PDAC remains fraught with substantial challenges (3). These include an immunosuppressive microenvironment, tumor antigen heterogeneity, and inadequate T cell infiltration coupled with functional exhaustion. Moreover, PDAC treatment is hindered by several factors: a highly heterogeneous genomic landscape, a complex tumor microenvironment (TME), a persistently immunosuppressive state, and metabolic reprogramming mechanisms, all of which contribute to therapy resistance. Addressing these challenges necessitates interdisciplinary collaboration and the development of integrated, multi-dimensional interventional strategies—this is the primary research focus that this Research Topic seeks to advance.

This collection of studies presents four pioneering investigations that illuminate the biological characteristics and therapeutic strategies of PDAC from diverse perspectives. These studies not only advance the understanding of disease mechanisms but also investigate novel interventions with translational potential, including immune cell engineering, natural product screening, and epigenetic regulation. Notably, the research conducted by Zhu et al. transcends traditional αβ T cell approaches by concentrating on the application of γδ T cells and natural killer T (NKT) cells in PDAC immunotherapy. Utilizing a mesothelin/CD3 bispecific antibody (BsAb), the authors demonstrate that these cells exhibit significantly enhanced tumor-killing activity both in vitro and in vivo, while substantially reducing the release of inflammatory cytokines such as IL-6 and IL-1β. This reduction in cytokine release mitigates the toxicities commonly associated with conventional αβ T cell-based therapies. Consequently, this study broadens the spectrum of cellular immunotherapy options and provides new pathways for enhancing treatment safety.

Chen et al. tackle the issue of CAR-T cell exhaustion in PDAC through a novel strategy involving the co-expression of the IL-15/IL-15Rα complex (IL15C). This approach activates the JAK3/STAT5 signaling pathway, thereby significantly enhancing the proliferative capacity and anti-apoptotic characteristics of NKG2D-CAR T cells. The engineered cells exhibit a marked reduction in the expression of exhaustion markers, such as PD-1 and TIM-3, and preferentially differentiate into central memory T cells, leading to more sustained tumor control in vivo. This innovative strategy offers a valuable framework for optimizing CAR-T cell therapy in the context of solid tumors.

In parallel, although Shao et al. conducted their investigation within a lung cancer model, their comprehensive research methodology offers substantial insights for PDAC drug development. By integrating network pharmacology, molecular docking, and functional validation, the researchers elucidate the anti-tumor mechanisms of cyclomorusin, an active compound derived from the traditional Chinese medicine Cortex Mori. This compound exerts its effects by inhibiting the PI3K/AKT/mTOR signaling pathway. The study highlights the contemporary scientific relevance of compounds derived from traditional medicine and provides a theoretical basis for the development of natural products targeting pancreatic ductal adenocarcinoma.

Wang et al. explore the role of protein SUMOylation in PDAC and develop a prognostic scoring system (Sscore) based on SUMO substrate-encoding genes, including CDK1, AHNAK2, and SAFB. This model demonstrates efficacy in predicting patient survival and shows associations with genomic instability, immune infiltration, and drug sensitivity. Notably, through functional experiments, the authors identify SAFB2 as a novel tumor suppressor that impedes PDAC progression by inhibiting the Wnt/β-catenin pathway. This study offers a dual theoretical foundation for SUMOylation-targeted therapy and personalized prognostic evaluation.

The studies featured in this topic collectively encompass multiple dimensions, ranging from fundamental mechanisms to translational applications, underscoring the significance of integrated multi-strategy interventions in PDAC treatment. Each article not only presents novel experimental evidence but also expands therapeutic perspectives. Whether through the diversification of immune cell types, the application of cytokine engineering, multi-target regulation via natural compounds, or mechanistic insights at the epigenetic level, these contributions reflect the field’s diversity and innovative vitality.

The studies featured in this Topic collectively cover multiple dimensions—from fundamental mechanisms to translational applications—highlighting the importance of combined multi-strategy interventions in PDAC treatment. Each article not only presents novel experimental evidence but also broadens therapeutic perspectives. Whether through the expansion of immune cell types, the application of cytokine engineering, multi-target regulation via natural compounds, or mechanistic insights at the epigenetic level, these contributions reflect the diversity and innovative vitality of the field. However, determining the most effective treatment strategy for patients, systematically evaluating the advantages and disadvantages of various therapeutic approaches, and ultimately developing a personalized precision treatment framework remain critical challenges that demand urgent attention in future research.

Author contributions

XS: Conceptualization, Writing – original draft, Writing – review & editing. HX: Data curation, Writing – original draft, Writing – review & editing.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Halbrook CJ, Lyssiotis CA, Magliano MP, and Maitra A. Pancreatic cancer: Advances and challenges. Cell. (2023) 186:1729–54. doi: 10.1016/j.cell.2023.02.014

PubMed Abstract | Crossref Full Text | Google Scholar

2. Blackford AL, Canto MI, Klein AP, Hruban RH, and Goggins M. Recent trends in the incidence and survival of stage 1A pancreatic cancer: A surveillance, epidemiology, and end results analysis. J Natl Cancer Inst. (2020) 112:1162–9. doi: 10.1093/jnci/djaa004

PubMed Abstract | Crossref Full Text | Google Scholar

3. Simnica D and Kobold S. Neoantigen T-cell receptor gene therapy in pancreatic cancer. N Engl J Med. (2022) 387:573. doi: 10.1056/NEJMc2208623

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: pancreatic cancer, pancreatic ductal adenocarcinoma, CAR-T cells, natural products, SUMOylation

Citation: Sun X and Xu H (2025) Editorial: Drug development for pancreatic cancer: novel targets, drug candidates and mechanisms. Front. Oncol. 15:1705678. doi: 10.3389/fonc.2025.1705678

Received: 15 September 2025; Accepted: 29 September 2025;
Published: 10 October 2025.

Edited and reviewed by:

Olivier Feron, Université Catholique de Louvain, Belgium

Copyright © 2025 Sun and Xu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: HongDe Xu, eGhkMTIyMEB6enUuZWR1LmNu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.