Skip to main content

EDITORIAL article

Front. Oncol., 28 January 2022
Sec. Gastrointestinal Cancers
This article is part of the Research Topic Chemo-resistance in Gastrointestinal Cancers View all 12 articles

Editorial: Chemo-Resistance in Gastrointestinal Cancers

  • 1Research Center for Biomedical Information Technology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
  • 2College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
  • 3Centre for Human Genetics (CHG), Bengaluru, India

Editorial on the Research Topic
Chemo-Resistance in Gastrointestinal Cancers

Gastrointestinal (GI) cancers are a group of diseases originating from different organs of the gastro-intestinal tract, with very high ranking of cancer incidence and mortality (1). Chemotherapy is widely used in GI cancer treatment, and is the preferred choice of treatment for patients who cannot undergo surgical resection or in patients with advanced metastases (2). Patients with GI cancer can benefit from chemotherapy including these commonly used chemotherapeutic agents, adriamycin, platinum drugs, 5-fluorouracil, vincristine, and paclitaxel. However, it is often observed that there is poor or no response to chemotherapy, and even in patients who respond well during primary treatment, the long-term results are disappointing. The development of chemo-resistance is a major obstacle in management of GI cancers. Various causes of drug resistance have been identified, which include inactivation of apoptosis signaling pathways, loss of cell cycle checkpoint control, accelerated cell proliferation and autophagy flux, enhanced DNA damage repair capacity, cancer stem cells, as well as epithelial-mesenchymal transition (35). Unfortunately, the precise mechanism of adaptive changes during development of chemo-resistance in GI cancers is still unclear. An insight into the mechanisms of chemo-resistance in GI cancers may help to devise better and personalized treatment strategies.

This Research Topic was aimed at bringing together clinical and basic scientists and to address the problem of chemo-resistance from multiple perspectives. True to its aim, this collection has a variety of articles including original research, review of literature and case reports. They cover areas such as signaling pathways, non-coding RNAs, cancer stem cells, and biomarkers to predict tumor outcome.

The Mitogen Activated Protein Kinase (MAPK) pathway is one of the most important signaling pathways involved in normal cellular processes, and plays a key role in the development and progression of cancer (6). Once activated, MAPK exerts an important role in converting extracellular stimuli into a wide range of cellular responses. Increasing evidences support its role in response to chemotherapeutic agents (7, 8). Upon stimulation, ERK1/2 signaling could decrease the sensitivity to sorafenib in hepatocellular carcinoma cells (9). Understanding the main effector genes along with downstream pathways can identify potential therapeutic targets. In this special guest edition, Li B. et al. discovered that phosphorylation of endogenous ERK1/2 could be stimulated by GCDA (Glycochenodeoxycholate) in hepatocellular carcinoma cells. Disruption of the effect of GCDA by blocking phosphorylation and nuclear accumulation of ERK1/2 could be potentially a mode of managing GCDA-related liver cancer and chemo-resistance. In addition, Gao et al. demonstrated that CIDEA expression promoted the chemosensitivity of esophageal cancer cells to cisplatin by activation of JNK. This study further suggests a tumor suppressor role for CIDEA as well and opens a possibility of using this as a target for combinatorial therapy. A study by Li Y. et al. reported a case of advanced colorectal cancer who was successfully treated using anti-EGFR drugs (cetuximab) in combination with anti-VEGF agents (fruquintinib) after development of resistance to chemotherapy. The mechanism underlying the success of this combinatorial therapy however needs further investigation.

In GI cancers, multiple deregulated noncoding RNAs (ncRNAs), including miRNAs and lncRNAs, play pivotal roles in the development of chemo-resistance (10, 11). In this regard, Qian et al. found that miR-454-3p was significantly up-regulated in oxaliplatin-resistant colorectal cancer cells and miR-454-3p promoted oxaliplatin resistance by targeting PTEN and activating the AKT signaling pathway. PI3K/AKT/mTOR pathway is also one of the most important signaling pathways involved in chemo-resistance in many human cancers (12). The contribution by Chen et al. demonstrated that HOXA13 overexpression increased 5-fluorouracil resistance in gastric cancer cells, and the expression of HOXA13 was directly suppressed by miR-139-5p. Bai et al. demonstrated the role of lncRNA AC007639.1 in chemo-resistance of hepatocellular carcinoma using a combination of bioinformatics and experimental approaches. These findings highlight targeting ncRNAs may act as a potential therapeutic strategy for reducing resistance to chemotherapy. Given the importance of ncRNAs in cancer, systematic exploration of the crosstalk with other molecular players should aid in a better understanding of their roles during the process of development of drug resistance of GI cancer patients.

A study by Liu et al. demonstrated the role of cancer stem cells as well as epithelial-mesenchymal transition (EMT) responsible for the development of chemo-resistance in esophageal cancer. Cancer stem cells have been reported in different GI cancers and are thought to be responsible for tumor initiation, metastasis, and drug resistance (13). The role of EMT in cancer drug resistance has long been suggested. It is worth noting that tumor microenvironment (TME) is also a factor mediating EMT-driven drug resistance, and the interactions of cancer cells with TME are crucial in EMT and drug resistance (14). Therefore it would be necessary to delineate detailed relationships of cancer cells and TME during chemo-resistance, such as the spatial locations of cancer and immune cells in the tumor tissues and specific ligand-receptor interactions between them.

Identification of biomarkers could improve diagnosis, prognosis, prediction of recurrence and treatment response. Many studies have contributed to the discovery of prognostic biomarkers, but the clinical need lies much more in the predictive biomarkers which would aid in deciding therapeutic approaches to improve patient outcomes. In this context, Sun et al. comprehensively reviewed progresses on the sensitivity prediction of neoadjuvant chemoradiotherapy for GI cancers in the aspects of microRNAs, metabolic enzymes, exosomes, other biomarkers, inflammatory indicators, and imageological assessments. Notably, a recent review has a special focus on the immune markers from TME and discusses their predictive roles on response to cytotoxic chemotherapy in colorectal cancer (15). The emerging move to the discovery and establishment of biomarkers from TME has the potential to develop more robust biomarkers for therapy benefit and resistance of chemotherapy. Alternatively, prediction of drug sensitivity through bioinformatics and computational biology could represent one efficient way to manage large and complex data sets, and provide prior information in the theoretical guidance. Innovative and advanced biomarker prediction approaches, together with large cohorts validation, should be more extensively designed and conducted in the near future.

Altogether, the articles collected in this Research Topic provide a series of insightful sets of data to better understand molecular events involved in the development of chemo-resistance in GI cancers. These findings offer implications in potential therapeutic targets identification, and provide insight on further drug-resistance research. It is our hope that this effort would pave the way for more inter-disciplinary work with the goal of better management of GI cancers.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

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.

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. Thrift AP, El-Serag HB. Burden of Gastric Cancer. Clin Gastroenterol Hepatol (2020) 18(3):534–42. doi: 10.1016/j.cgh.2019.07.045

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Wagner AD, Syn NL, Moehler M, Grothe W, Yong WP, Tai BC, et al. Chemotherapy for Advanced Gastric Cancer. Cochrane Database System Rev (2017) 8:CD004064. doi: 10.1002/14651858.CD004064.pub4

CrossRef Full Text | Google Scholar

3. Nies AT, Magdy T, Schwab M, Zanger UM. Role of ABC Transporters in Fluoropyrimidine-Based Chemotherapy Response. Adv Cancer Res (2015) 125:217–43. doi: 10.1016/bs.acr.2014.10.007

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Hammond WA, Swaika A, Mody K. Pharmacologic Resistance in Colorectal Cancer: A Review. Ther Adv Med Oncol (2016) 8(1):57–84. doi: 10.1177/1758834015614530

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Czabotar PE, Lessene G, Strasser A, Adams JM. Control of Apoptosis by the BCL-2 Protein Family: Implications for Physiology and Therapy. Nat Rev Mol Cell Biol (2014) 15(1):49–63. doi: 10.1038/nrm3722

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Khavari TA, Rinn J. Ras/Erk MAPK Signaling in Epidermal Homeostasis and Neoplasia. Cell Cycle (2007) 6(23):2928–31. doi: 10.4161/cc.6.23.4998

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Deacon K, Mistry P, Chernoff J, Blank JL, Patel R. P38 Mitogen-Activated Protein Kinase Mediates Cell Death and P21-Activated Kinase Mediates Cell Survival During Chemotherapeutic Drug-Induced Mitotic Arrest. Mol Biol Cell (2003) 14(5):2071–87. doi: 10.1091/mbc.e02-10-0653

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Lou X, Zhou Q, Yin Y, Zhou C, Shen Y. Inhibition of the Met Receptor Tyrosine Kinase Signaling Enhances the Chemosensitivity of Glioma Cell Lines to CDDP Through Activation of P38 MAPK Pathway. Mol Cancer Ther (2009) 8(5):1126–36. doi: 10.1158/1535-7163.MCT-08-0904

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Zhong J, Yu X, Dong X, Lu H, Zhou W, Li L, et al. Downregulation of Secreted Clusterin Potentiates the Lethality of Sorafenib in Hepatocellular Carcinoma in Association With the Inhibition of ERK1/2 Signals. Int J Mol Med (2018) 41(5):2893–900. doi: 10.3892/ijmm.2018.3463

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Zhang F, Li Y, Xu W, He L, Tan Y, Xu H. Long non-Coding RNA ZFAS1 Regulates the Malignant Progression of Gastric Cancer via the microRNA-200b-3p/Wnt1 Axis. Biosci Biotechnol Biochem (2019) 83(7):1289–99. doi: 10.1080/09168451.2019.1606697

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Xi Z, Si J, Nan J. LncRNA MALAT1 Potentiates Autophagyassociated Cisplatin Resistance by Regulating the Microrna30b/Autophagyrelated Gene 5 Axis in Gastric Cancer. Int J Oncol (2019) 54(1):239–48. doi: 10.3892/ijo.2018.4609

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Fattahi S, Amjadi-Moheb F, Tabaripour R, Ashrafi GH, Akhavan-Niaki H. PI3K/AKT/mTOR Signaling in Gastric Cancer: Epigenetics and Beyond. Life Sci (2020) 262:118513. doi: 10.1016/j.lfs.2020.118513

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Taniguchi H, Moriya C, Igarashi H, Saitoh A, Yamamoto H, Adachi Y, et al. Cancer Stem Cells in Human Gastrointestinal Cancer. Cancer Sci (2016) 107(11):1556–62. doi: 10.1111/cas.13069

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Erin N, Grahovac J, Brozovic A, Efferth T. Tumor Microenvironment and Epithelial Mesenchymal Transition as Targets to Overcome Tumor Multidrug Resistance. Drug Resist Updates: Rev Commentaries Antimicrob Anticancer Chemother (2020) 53:100715. doi: 10.1016/j.drup.2020.100715

CrossRef Full Text | Google Scholar

15. Wilkinson K, Ng W, Roberts TL, Becker TM, Lim SH, Chua W, et al. Tumour Immune Microenvironment Biomarkers Predicting Cytotoxic Chemotherapy Efficacy in Colorectal Cancer. J Clin Pathol (2021) 74(10):625–34. doi: 10.1136/jclinpath-2020-207309

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: gastric cancer, drug-resistance, mechanism, targets, biomarker

Citation: Li X, Yang D-H and Ranganathan P (2022) Editorial: Chemo-Resistance in Gastrointestinal Cancers. Front. Oncol. 12:821212. doi: 10.3389/fonc.2022.821212

Received: 24 November 2021; Accepted: 05 January 2022;
Published: 28 January 2022.

Edited and reviewed by:

Liang Qiao, Westmead Institute for Medical Research, Australia

Copyright © 2022 Li, Yang and Ranganathan. 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: Xia Li, xia.li@siat.ac.cn; bioinflix@126.com

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.