Skip to main content

EDITORIAL article

Front. Oncol., 19 December 2022
Sec. Cancer Genetics
This article is part of the Research Topic Advances in Genetics and Molecular Diagnosis in Colorectal, Stomach, and Pancreatic Cancer. View all 11 articles

Editorial: Advances in genetics and molecular diagnosis in colorectal, stomach, and pancreatic cancer

  • 1Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB) Hospital Italiano de Buenos Aires, Instituto Universitario Hospital Italiano de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (HIBA-IUHI-CONICET), Ciudad Autónoma de Buenos Aires (CABA), Argentina
  • 2Programa de Cáncer Hereditario (Pro.Can.He.), Hospital Italiano de Buenos Aires (HIBA), Ciudad Autónoma de Buenos Aires (CABA), Argentina
  • 3Department of Tumor Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo, Norway

Gastrointestinal (GI) cancer accounts for over one-quarter (26%) of all cancer cases and over one-third (35%) of cancer deaths globally (1). According to worldwide incidence and mortality rates, the most common cancers include colorectal, stomach, liver, esophageal, and pancreatic cancers (1). Although their incidence has decreased, this group of malignancies continues to pose major challenges to public health. Driven by population growth and aging worldwide, it is estimated that the number of new cases and deaths will increase by 58% and 73%, respectively, by 2040 (1).

Cancer is a complex, heterogeneous, and multifactorial disease, in which both genetic and non-genetic factors are involved. A complete identification of the risk factors, and an understanding of their role at different stages of tumor development, is essential for achieving high-quality comprehensive cancer care. In recent years, the overwhelming revolution in omics technologies, from genomics to transcriptomics, has expanded our understanding of the molecular pathogenesis of GI cancers (2). Despite the advances made in this field, extensive work is still needed to fully understand the molecular and genetic nature that leads to the development of such cancers.

This Research Topic focuses attention on advances in genetic predisposition and novel biomarkers linked to GI cancers, seeking to bring some light to this research field.

Colorectal cancer (CRC) is the most frequently diagnosed GI malignancy, and it is estimated that up to ~35% of all cases are associated with an inherited form attributable to moderate and high penetrance germline genetic alterations (2, 3). Lynch syndrome and familial adenomatous polyposis (FAP) syndrome are the two most common disorders, accounting for 2-4% and 1% of all CRCs, respectively (4). Although new predisposition genes for FAP have been identified recently, to date, up to 20% of patients with a clear FAP phenotype and as much as 80% with the attenuated form remain genetically unexplained (Olkinuora et al.). Olkinuora et al. confirmed the importance of extending the coverage of genomic studies by NGS to different populations to achieve the goal of identifying new genes that predispose to hereditary CRC. A recurrent variant in the DNA glycosylase gene NEIL1 was observed in two non-related families from Finland and Argentina, suggesting that pathogenic germline variants in this gene can explain polyposis in FAP cases without APC pathogenic alterations. To date, for certain genes, such as BRCA1, the existing data to prove their role in CRC development are contradictory. In that regard, the article by Freire et al. reported three non-related CRC cases (families) carrying a heterozygous pathogenic germline variant in the BRCA1 gene, with a clear family history of cancer. These new data provide more insight to support the role of BRCA1 in CRC development.

Despite pancreatic cancer ranking fifth among five major GI cancers, its mortality and incidence rates are almost equal, making it an extremely deadly malignancy. Heritability is estimated to account for 21.5% of pancreatic cancer cases, thus it is important to identify the genetic and molecular background behind this population at risk. By performing a GWAS analysis and a recessive genetic model, Lu et al. identified a set of SNPs showing an association with pancreatic cancer risk (specific recessive effect; OR, 0.75-1.42, p<10-5). This study also emphasized the importance of choosing the right bioinformatic tool to improve the definition of polygenic risk scores.

For the treatment of most cancers, in particular for GI malignancies such as CRC, a combination of surgery with adjuvant therapy is widely used. However, the prognostic and risk factors for these patients have not yet been clarified. Furthermore, owing to a lack of effective markers, it is also not possible to infer their prognosis by only having data on genetic alterations in primary tumor tissue. To solve these problems, Yu et al. investigated the expression and protein levels of matrix metalloproteinase (MMP) gene family members. They found that a subgroup of MMP genes could have the potential to work as a target for precision therapies in CRC patients. More precisely, modifying their expression levels could significantly improve both progression-free survival and relapse-free time. In the same line of prognostic research, but for pancreatic adenocarcinomas (PAAD), Xu et al. investigated invasive-related genes with the aim of identifying different molecular subtypes of PAAD. Based on a molecular signatures analysis, a three-gene model was constructed and validated as a marker for assessing the prognostic risk of such patients.

Latest epidemiological data support the notion that at least 90% of cancer deaths from solid tumors are caused by metastases (5). Cancer metastases to the liver over a course of CRC occur in more than 50% of patients (6). To date, only a small number of cases have been candidates for a multimodal treatment, for which high drug resistance and relapse rates have been observed (7). As the mechanisms linked to those events are poorly understood, Kyrochristos et al. conducted a pilot study to assess the potential translational implications of intrapatient heterogeneity as a tool for defining appropriate matched drug therapy. Although it was a proof-of-concept approach, they provided strong evidence to support further evaluation, even in future clinical trials.

To fully understand GI cancer carcinogenesis, uncovering molecular data at each stage is essential. Several authors approached this topic by submitting review and original articles. Lv et al. described that, in CRC, Fibrillin-1 gene methylation can serve as a biomarker for disease development, as its transcriptional inactivation might start from normal colonic epithelium and increase through adenoma to CRC. Using a meta-analysis, Zhang et al. revealed that the Leptin G19A polymorphism may decrease the risk for CRC and esophageal GI cancers, as well as urinary tract cancer. Furthermore, Gao et al. demonstrated that a polymorphism (rs7158663) in the maternally expressed three-lncRNAs can be used as a genetic marker for predicting the risk of GI (CRC and gastric) and breast cancers. Finally, Mulder et al., in their submitted article, underlined that the development of orphan medicinal products for PAAD treatment should be closely monitored to increase the success rate of drugs reaching the market.

All of the collected articles support the notion that a deepened understanding of the molecular and genetic factors linked to the development of GI cancers could help to improve the translation of basic scientific research into clinical services and practices, pointing towards personalized medicine, and thus, more effective treatment of GI malignancies.

Author contributions

All of the listed authors equally, substantially, and intellectually contributed to this Editorial and approved it for publication.

Funding

The authors acknowledge funding from the Agencia Nacional de Promoción Científica y Tecnológica (grant number PICT_2017 -3210 to CV and -3197 to WP). The Agencia Nacional de Promoción Científica y Tecnológica offered a paid doctoral student fellowship to JS. MD-V acknowledges funding from the Norwegian Cancer Society (contract 194751-2017).

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. Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, et al. Global burden of 5 major types of gastrointestinal cancer. Gastroenterology (2020) 159:335–49.e15. doi: 10.1053/j.gastro.2020.02.068

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Olkinuora AP, Peltomäki PT, Aaltonen LA, Rajamäki K. From APC to the genetics of hereditary and familial colon cancer syndromes. Hum Mol Genet (2021) 30:R206–24. doi: 10.1093/hmg/ddab208

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Katabathina VS, Menias CO, Khanna L, Murphy L, Dasyam AK, Lubner MG, et al. Hereditary gastrointestinal cancer syndromes: Role of imaging in screening, diagnosis, and management. RadioGraphics (2019) 39:1280–301. doi: 10.1148/rg.2019180185

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Samadder NJ, Baffy N, Giridhar KV, Couch FJ, Riegert-Johnson D. Hereditary cancer syndromes–a primer on diagnosis and management, part 2: Gastrointestinal cancer syndromes. Mayo Clin Proc (2019) 94:1099–116. doi: 10.1016/j.mayocp.2019.01.042

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Dillekås H, Rogers MS, Straume O. Are 90% of deaths from cancer caused by metastases? Cancer Med (2019) 8:5574–6. doi: 10.1002/cam4.2474

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Zarour LR, Anand S, Billingsley KG, Bisson WH, Cercek A, Clarke MF, et al. Colorectal cancer liver metastasis: Evolving paradigms and future directions. Cell Mol Gastroenterol Hepatol (2017) 3:163–73. doi: 10.1016/j.jcmgh.2017.01.006

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Nordlinger B, Sorbye H, Glimelius B, Poston GJ, Schlag PM, Rougier P, et al. Perioperative FOLFOX4 chemotherapy and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC 40983): long-term results of a randomised, controlled, phase 3 trial. Lancet Oncol (2013) 14:1208–15. doi: 10.1016/S1470-2045(13)70447-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: gastrointestinal cancer, colorectal cancer, pancreatic cancer, genetic predisposition, hereditary syndrome

Citation: Vaccaro CA, Soarez J, Dominguez-Valentin M and Pavicic WH (2022) Editorial: Advances in genetics and molecular diagnosis in colorectal, stomach, and pancreatic cancer. Front. Oncol. 12:1104671. doi: 10.3389/fonc.2022.1104671

Received: 21 November 2022; Accepted: 30 November 2022;
Published: 19 December 2022.

Edited and Reviewed by:

Heather Cunliffe, University of Otago, New Zealand

Copyright © 2022 Vaccaro, Soarez, Dominguez-Valentin and Pavicic. 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: Walter Hernán Pavicic, walter.pavicic@hospitalitaliano.org.ar

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.