Skip to main content

EDITORIAL article

Front. Oncol., 29 August 2024
Sec. Gastrointestinal Cancers: Colorectal Cancer
This article is part of the Research Topic Advances in Molecular Biology Knowledge of Rectal Cancer and Forthcoming Role of Liquid Biopsy View all 10 articles

Editorial: Advances in molecular biology knowledge of rectal cancer and forthcoming role of liquid biopsy

  • 1Gastroenterology and Endoscopy Unit, University Hospital of Parma, Parma, Italy
  • 2Department of Pathology, University Hospital of Parma, Parma, Italy
  • 3Medical Oncology Unit, Department of Oncology, Ospedale Sant’Andrea, La Spezia, Italy

Advances in molecular biology may significantly impact our understanding of rectal cancer and inform the development and positioning of novel therapies (1). Total Neoadjuvant Treatment (TNT) for locally advanced rectal cancer is gaining consensus and has evolved, emphasizing the need of a multidisciplinary strategy. TNT has increased complete response rates, disease-free survival, control of distant metastases and has become a new standard of care (2, 3). Also, a raising amount of reports indicates that a non-operative management (NOM), entailing close surveillance of patients with clinical complete response (cCR) after neoadjuvant therapy, could be an appropriate alternative option to rectal surgery (4). Recently, the International Watch & Wait Database (IWWD) reported superiority in terms of quality of life and a small risk of local inoperable disease recurrence without compromising survival in patients with rectal cancer managed nonoperatively after achieving a cCR following neoadjuvant therapy (5).

Selecting predictive molecular markers is thus becoming even more crucial. The tumor microenvironment plays a critical role in colorectal tumor development, progression and immune escape (6). Stromal cells (i.e. adipocytes, vasculature, lymphocytes) interact with cancer cells and may affect therapy response. As reported in the review by Mirza et al., tumor microenvironment evaluation during treatment may inform on new therapies, uncover responses and tumor resistance. The intratumoral immune contexture is a main factor of clinical outcome in both early- and advanced-stage colorectal cancer (7, 8). Specifically, in rectal cancer a local hot immune signature in the tumor before neoadjuvant therapy is correlated with improved response and prolonged disease-free survival (9). Besides, the 100% complete clinical response rate after programmed cell death protein-1 (PD-1) blockade soundly suggests that the in situ innate immune response released and enhanced by immune checkpoint inhibitors treatment can fully eradicate cancer cells precluding recurrences (10). Tumor immune infiltrate has also been described as an independent prognostic marker in a large international cohort of rectal cancer patients with cCR managed nonoperatively and could pave the way for prospective therapeutic trials guided by immunoscore to adapt follow up and/or therapy of NOM patients (11).

Colorectal cancers show genetic variations and clonal evolution, which proffer noteworthy difficulties in selecting appropriate therapies (12). In the traditional approach, the identification and choice of therapy have mainly depended on the employment of invasive tissue biopsies and imaging assays. Currently, core tumor biopsy specimens represent the gold standard biological tissue to identify and analyze predictive biomarkers. However, anatomical feasibility, tumor heterogeneity and cancer progression are major limitations of this single-snapshot approach. Liquid biopsy is increasingly gaining attention as a complementary and potentially alternative non-invasive tool to bypass these limitations.

Liquid biopsy assessment of circulating tumor DNA (ctDNA) is useful for risk stratification and detection of minimal residual disease (MRD) in early colorectal cancer (13). ctDNA can also outline the tumor mutational profile, detect mutations not identified in the tissue biopsies and offer a comprehensive and dynamic evaluation of tumor genetics, classify specific therapeutic targets thus allowing clinicians to monitor disease progression and the efficacy of treatments (13). The introduction of liquid biopsies has endorsed a noteworthy move towards precision medicine in colorectal cancer; the presence of ctDNA in high-risk stage II (T4) and stage III colorectal cancer patients correlates with adverse prognosis both post-surgery and post-adjuvant treatment independent of other conventional clinical-pathological risk factors (13). More recently, Tie and co-authors demonstrated that patients with liver-only metastases undergoing surgical resection had a lower relapse-free survival in the case of ctDNA positivity (14), thus confirming the potential role of serial ctDNA analysis as an immediate marker of therapy activity. As reported by Choi et al., liquid biopsies using extracellular vesicle DNA (evDNA) secreted by tumor cells may be a different source for the detection of cancer driver mutations and a complementary tool for the diagnosis and surveillance of colon cancer patients. Their results showed that evDNA isolated from the plasma of colon cancer patients harboring KRAS G12D and G13D mutations was significantly associated with both CEA level and survival. Unlike fragmented pieces of ‘cell-free’ DNA (cfDNA) shed from apoptotic or necrotic cells, extracellular vesicles arise from viable tumor cells. Therefore, evDNA might reveal the underlying biology of living cancer cells (15, 16) and reflect cancer driver mutations even in the early stages of cancer development (17).

In rectal cancer liquid biopsy could be important in several steps: at the time of diagnosis, for the evaluation of MRD, treatment response and possible acquired resistance and also to modulate treatment during TNT (18). The up-to-date trimodality approach for locally advanced rectal cancer comprises chemotherapy, radiotherapy and surgery and may cause considerable morbidities; moreover, it might not be mandatory for some patients, and fails to prevent disease relapses in others. The main drawback in the present managing of rectal cancer is the absence of consistent and accurate techniques of predicting responsiveness to neoadjuvant therapies without surgical resection and subsequent pathological evaluations. For instance, among patients candidate to sphinter-sparing surgery who demonstrate adequate clinical responses to induction systemic chemotherapy, omitting radiotherapy and its associated toxicities might be a valuable therapeutic option (19). Compared to standard pathological evaluation criteria, ctDNA or modifications in ctDNA could be useful in directing therapeutic decision in this setting of patients. Additionally, a tool to improve the degree of concordance between clinical and pathological complete response could assist a NOM strategy.

In conclusion, the knowledge of tumor microenvironment and immune changes together with introduction of liquid biopsies could offer a measure of these dynamic interfaces, thus enabling the development of immunotherapies and tailored therapies. The inclusion of liquid biopsies in the design of clinical trials, in addition to other analytical modalities such as conventional tissue biopsies, is a crucial component of this development. The Research Topic gives an overview of liquid biopsy and other new technologies and methods as well as emphasizing the clinical usefulness of liquid biopsy, particularly investigating its implication as an analytic, predictive, or MRD marker.

Author contributions

FN: Writing – original draft, Writing – review & editing. LG: Writing – original draft, Writing – review & editing. CA: 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.

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. Li M, Xiao Q, Venkatachalam N, Hofheinz RD, Veldwijk MR, Herskind C, et al. : Predicting response to neoadjuvant chemoradiotherapy in rectal cancer: from biomarkers to tumor models. Ther Adv Med Oncol. (2022) 14:17588359221077972. doi: 10.1177/17588359221077972

PubMed Abstract | Crossref Full Text | Google Scholar

2. Bahadoer RR, Dijkstra EA, van Etten B, Marijnen CAM, Putter H, Kranenbarg EM, et al. Short-course radiotherapy followed by chemotherapy before total mesorectal excision (TME) versus preoperative chemoradiotherapy, TME, and optional adjuvant chemotherapy in locally advanced rectal cancer (RAPIDO): A randomised, openlabel, phase 3 trial. Lancet Oncol. (2021) 22:29–42. doi: 10.1016/S1470-2045(20)30555-6

PubMed Abstract | Crossref Full Text | Google Scholar

3. Conroy T, Bosset JF, Etienne PL, Rio E, François É, Mesgouez-Nebout N, et al. Neoadjuvant chemotherapy with FOLFIRINOX and preoperative chemoradiotherapy for patients with locally advanced rectal cancer (UNICANCER-PRODIGE 23): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. (2021) 22:702–15. doi: 10.1016/S1470-2045(21)00079-6

PubMed Abstract | Crossref Full Text | Google Scholar

4. Smith JJ, Paty PB, Garcia-Aguilar J. Watch and wait in rectal cancer or more wait and see? JAMA Surg. (2020) 155:657–58. doi: 10.1001/jamasurg.2020.0226

PubMed Abstract | Crossref Full Text | Google Scholar

5. van der Valk MJM, Hilling DE, Bastiaannet E, Meershoek-Klein Kranenbarg E, Beets GL, Figueiredo NL, et al. Long-term outcomes of clinical complete responders after neoadjuvant treatment for rectal cancer in the International Watch & Wait Database (IWWD): An international multicentre registry study. Lancet. (2018) 391:2537–45. doi: 10.1016/S0140-6736(18)31078-X

PubMed Abstract | Crossref Full Text | Google Scholar

6. Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet. (2015) 47:320–29. doi: 10.1038/ng.3225

PubMed Abstract | Crossref Full Text | Google Scholar

7. Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. (2006) 313:1960–64. doi: 10.1126/science.1129139

PubMed Abstract | Crossref Full Text | Google Scholar

8. Pagès F, Mlecnik B, Marliot F, Bindea G, Ou FS, Bifulco C, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. (2018) 391:2128–39. doi: 10.1016/S0140-6736(18)30789-X

PubMed Abstract | Crossref Full Text | Google Scholar

9. Chatila WK, Kim JK, Walch H, Marco MR, Chen CT, Wu F, et al. Genomic and transcriptomic determinants of response to neoadjuvant therapy in rectal cancer. Nat Med. (2022) 28:1646–55. doi: 10.1038/s41591-022-01930-z

PubMed Abstract | Crossref Full Text | Google Scholar

10. Cercek A, Lumish M, Sinopoli J, Weiss J, Shia J, Lamendola-Essel M, et al. PD-1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N Engl J Med. (2022) 386:2363–76. doi: 10.1056/NEJMoa2201445

PubMed Abstract | Crossref Full Text | Google Scholar

11. El Sissy C, Kirilovsky A, Lagorce Pagès C, Marliot F, Custers PA, Dizdarevic E, et al. International validation of the immunoscore biopsy in patients with rectal cancer managed by a watch-and-wait strategy. J Clin Oncol. (2024) 42:70–80. doi: 10.1200/JCO.23.00586

PubMed Abstract | Crossref Full Text | Google Scholar

12. Saoudi González N, Salvà F, Ros J, Baraibar I, Rodríguez-Castells M, García A, et al. Unravelling the complexity of colorectal cancer: heterogeneity, clonal evolution, and clinical implications. Cancers (Basel). (2023) 15:4020. doi: 10.3390/cancers15164020

PubMed Abstract | Crossref Full Text | Google Scholar

13. Dasari A, Morris VK, Allegra CJ, Atreya C, Benson AB 3rd, Boland P, et al. ctDNA applications and integration in colorectal cancer: an NCI Colon and Rectal-Anal Task Forces whitepaper. Nat Rev Clin Oncol. (2020) 17:757–70. doi: 10.1038/s41571-020-0392-0

PubMed Abstract | Crossref Full Text | Google Scholar

14. Tie J, Wang Y, Cohen J, Li L, Hong W, Christie M, et al. Circulating tumor DNA dynamics and recurrence risk in patients undergoing curative intent resection of colorectal cancer liver metastases: A prospective cohort study. PloS Med. (2021) 18:e1003620. doi: 10.1371/journal.pmed.1003620

PubMed Abstract | Crossref Full Text | Google Scholar

15. Allenson K, Castillo J, San Lucas FA, Scelo G, Kim DU, Bernard V, et al. High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann Oncol. (2017) 28:741–47. doi: 10.1093/annonc/mdx004

PubMed Abstract | Crossref Full Text | Google Scholar

16. Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. (2015) 523:177–82. doi: 10.1038/nature14581

PubMed Abstract | Crossref Full Text | Google Scholar

17. Möhrmann L, Huang HJ, Hong DS, Tsimberidou AM, Fu S, Piha-Paul SA, et al. Liquid biopsies using plasma exosomal nucleic acids and plasma cell-free DNA compared with clinical outcomes of patients with advanced cancers. Clin Cancer Res. (2018) 24:181–88. doi: 10.1158/1078-0432.CCR-17-2007

PubMed Abstract | Crossref Full Text | Google Scholar

18. Massihnia D, Pizzutilo EG, Amatu A, Tosi F, Ghezzi S, Bencardino K, et al. Liquid biopsy for rectal cancer: A systematic review. Cancer Treat Rev. (2019) 79:101893. doi: 10.1016/j.ctrv.2019.101893

PubMed Abstract | Crossref Full Text | Google Scholar

19. Schrag D, Shi Q, Weiser MR, Gollub MJ, Saltz LB, Musher BL, et al. Preoperative treatment of locally advanced rectal cancer. N Engl J Med. (2023) 389:322–34. doi: 10.1056/NEJMoa2303269

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: rectal cancer, molecular biology, liquid biopsy, prognosis, biomarker

Citation: Negri F, Gnetti L and Aschele C (2024) Editorial: Advances in molecular biology knowledge of rectal cancer and forthcoming role of liquid biopsy. Front. Oncol. 14:1476174. doi: 10.3389/fonc.2024.1476174

Received: 05 August 2024; Accepted: 16 August 2024;
Published: 29 August 2024.

Edited and Reviewed by:

Liang Qiao, The University of Sydney, Australia

Copyright © 2024 Negri, Gnetti and Aschele. 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: Francesca Negri, fnegri@ao.pr.it

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.