Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Surg. Apr 27, 2025; 17(4): 101066
Published online Apr 27, 2025. doi: 10.4240/wjgs.v17.i4.101066
Pan-immune-inflammation in colon cancer: A prognostic biomarker and the role of tumor location in personalized care
Gaya Spolverato, Department of Surgery, University of Padova, Padua 35122, Italy
Giulia Capelli, Department of Surgery, ASST Bergamo Est, Bergamo 24068, Lombardy, Italy
Floriane Noel, Michele Steindler, Department of Research, Sibylone, Paris 75002, France
Andrew Alexander Gumbs, Department of Surgery, University of Magdeburg, Magdeburg 39130, Saxony-Anhalt, Germany
Andrew Alexander Gumbs, Department of Surgery, Service de Chirurgie Digestive Minimale Invasive, Hôpital Antoine Béclère, Assistance Publique-Hôpitaux de Paris, Clamart 92140, France
ORCID number: Andrew Alexander Gumbs (0000-0002-7044-5318).
Author contributions: Spolverato G, Capelli G, Noel F, Steindler M, and Gumbs AA contributed to this paper; Gumbs AA designed the overall concept and outline of the manuscript; Spolverato G contributed to the discussion and design of the manuscript; Noel F and Steindler M drafted the manuscript; Spolverato G and Capelli G contributed to manuscript writing, editing, and review of the literature. All authors have read and approved the final manuscript.
Conflict-of-interest statement: No conflicts of interest to report.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Andrew Alexander Gumbs, FACS, MD, MSc, Chief, Full Professor, Department of Surgery, Service de Chirurgie Digestive Minimale Invasive, Hôpital Antoine Béclère, Assistance Publique-Hôpitaux de Paris, 157 Rue de la Porte de Trivaux, Clamart 92140, France. aagumbs@gmail.com
Received: September 3, 2024
Revised: February 14, 2025
Accepted: February 25, 2025
Published online: April 27, 2025
Processing time: 206 Days and 20.6 Hours

Abstract

Despite advances in surgery, chemotherapy, and radiotherapy, the treatment of colorectal cancer (CRC) requires more personalized approaches based on tumor biology and molecular profiling. While some relevant mutations have been associated with differential response to immunotherapy, such as RAS and BRAF mutations limiting response to anti-epithelial growth factor receptor drugs or microsatellite instability predisposing susceptibility to immune checkpoint inhibitors, the role of inflammation in dictating tumor progression and treatment response is still under investigation. Several inflammatory biomarkers have been identified to guide patient prognosis. These include the neutrophil-lymphocyte ratio, Glasgow prognostic score (GPS) and its modified version, lymphocyte-C-reactive protein ratio, and platelet-lymphocyte ratio. However, these markers are not yet included in the standard clinical management of patients with CRC, and further research is needed to evaluate their efficacy in different patient populations. A recent study by Wang et al, published in the World Journal of Gastroenterology, sheds light on the prognostic significance of pan-immune-inflammation value (PIV) in CRC, particularly concerning primary tumor location. Specifically, the authors found that a high PIV was strongly correlated with worse disease-free survival in patients with left-sided colon cancer, whereas no such association was observed in patients with right-sided colon cancer. Integrating tumor location into the prognostic assessment of CRC may improve our ability to more accurately identify high-risk patients and develop personalized treatment plans that are more likely to improve patient outcomes.

Key Words: Colorectal cancer; Inflammatory biomarkers; Tumor location; Targeted therapy

Core Tip: A recent study by Wang et al highlights that a high pan-immune inflammation value is associated with worse disease-free survival in left-sided colon cancer, but not in right-sided colon cancer. This suggests that considering tumor location in colorectal cancer prognosis may help identify high-risk patients and more effectively tailor treatment plans.



INTRODUCTION

Colorectal cancer (CRC) is the third most common cancer worldwide and a leading cause of cancer-related deaths[1]. Despite advances in surgery, chemotherapy, and radiotherapy, the limitations of these treatments underscore the urgent need for more personalized approaches[2]. Biomarkers, biological molecules that hold the key to unlocking a new era of precision in CRC care, are emerging as critical tools in this regard[3,4].

BIOMARKERS: A NEW ERA OF PRECISION IN CRC
Molecular profiling: Unmasking the tumor's identity

Genetic and epigenetic alterations drive the development and progression of CRC. Unraveling the unique molecular profile of the tumor through biomarker analysis is critical for guiding treatment decisions, particularly in advanced or metastatic disease[3]. In particular, the mutational profile of the tumor can predict the response to epithelial growth factor receptor (EGFR)-targeting monoclonal antibodies, such as cetuximab and panitumumab, which have been validated over the past 20 years as powerful tools for prolonging survival in patients with metastatic CRC[5].

Key players in this molecular landscape include RAS mutations (KRAS, NRAS) and BRAF mutations, particularly the V600E mutation, which confer resistance to anti-EGFR therapies and are associated with poorer prognosis[3,6,7]. Similarly, human epidermal growth factor receptor 2 (HER2) amplification, although relatively rare in CRC, can drive tumor growth and predict resistance to anti-EGFR therapies. However, anti-HER2 therapies, initially successful in treating HER2-positive breast cancer, are now showing promise in HER2-amplified metastatic CRC[3,8]. Finally, microsatellite instability-high (MSI-H) tumors have a distinct molecular profile characterized by a high number of mutations and neoantigens, making them particularly susceptible to immune checkpoint inhibitors (ICIs), a major breakthrough in CRC treatment[9]. These drugs, which block inhibitory signals that suppress the immune system, have demonstrated remarkable efficacy in patients with MSI-H/dMMR CRC, resulting in durable responses and improved survival[9,10]. Unfortunately, the majority of CRC tumors are microsatellite stable or mismatch repair proficient, rendering them less responsive to ICIs[11]. This is largely due to their lower tumor mutational burden and weaker immunogenicity, making them adept at evading immune surveillance[9,12]. Overcoming this immune evasion is a major focus of ongoing research, with strategies exploring combinations of ICIs with other treatment modalities, such as chemotherapy, targeted therapies, and radiotherapy[9,12].

Inflammation: A silent partner in CRC progression

Chronic inflammation plays a critical role in the development and progression of CRC, influencing tumor growth, angiogenesis, and metastasis[2]. In recent years, several studies have linked inflammation to postoperative recurrence in patients undergoing curative surgery for CRC. For example, Matsubara et al[13] found that high post-resection C-reactive protein (CRP) levels were linked to significantly worse recurrence-free survival in 467 patients with stage I–III CRC who underwent curative surgery. Similarly, Di Caro et al[14] discovered that high preoperative serum levels of inflammatory cytokines and the acute-phase protein Pentraxin-3 were associated with an increased risk of recurrence independently of TNM staging in 69 patients. Consequently, several inflammation-related biomarkers, readily available through routine blood tests, have emerged as valuable prognostic indicators[2]. Elevated neutrophil-lymphocyte ratio, reflecting a higher proportion of neutrophils to lymphocytes, is consistently associated with poorer prognosis in CRC[2,4,15]. The Glasgow prognostic score and its modified version incorporate serum CRP and albumin levels, reflecting both systemic inflammation and nutritional status, with higher scores linked to worse outcomes[2,4,16]. A low lymphocyte-CRP ratio, indicating a lower proportion of lymphocytes to CRP, is associated with poorer prognosis and increased risk of postoperative complications[2,17]. Elevated platelet-lymphocyte ratio, reflecting a higher proportion of platelets to lymphocytes, is linked to poorer prognosis in CRC, particularly in advanced stages[2,18]. While these biomarkers hold significant promise, their optimal cutoff values and clinical application remain areas of active investigation. Multicenter prospective studies are crucial to standardize their use and maximize their clinical utility[2].

The crucial role of inflammation in CRC should also be viewed as a driver of oncogenic mutations. For example, KRAS mutations are more commonly present in CRC arising from chronic inflammatory disease. Conversely, these mutations have been shown to create an immunosuppressive tumor microenvironment (TME), suppressing the expression of pro-inflammatory cytokines. Therefore, there is a complex and yet unclear link between chronic inflammation, the development[19] of oncogenic mutations, and the immunomodulation of the TME. Understanding this intricate relationship may lead to many interesting developments in personalized therapy in the next few years.

PAN-IMMUNE-INFLAMMATION VALUE AS A LOCATION-SPECIFIC BIOMARKER

A recent study by Wang et al[20], published in the World Journal of Gastroenterology, sheds light on the prognostic significance of the pan-immune-inflammation value (PIV) in colon cancer, particularly in relation to primary tumor location. This research is a pivotal contribution to our understanding of the complex interactions between systemic inflammation and tumor biology in CRC.

The distinction between left-sided colon cancer (LCC) and right-sided colon cancer (RCC) has been well established, with each subtype exhibiting distinct clinical, pathological, and molecular characteristics[21]. The study by Wang et al[20] adds to this narrative by demonstrating that the prognostic utility of PIV is significantly influenced by tumor location within the colon. Specifically, the authors found that a high PIV was strongly correlated with poorer disease-free survival in patients with LCC, whereas no such association was observed in patients with RCC.

The implications of these results are profound, suggesting that the prognostic value of systemic inflammatory response markers, such as PIV, may not be universally applicable across all colon cancer subtypes. Instead, the efficacy of these biomarkers may vary depending on tumor location, necessitating a more nuanced approach to their clinical application. In patients with LCC, a high PIV could serve as a critical indicator of poor prognosis and guide more aggressive and tailored treatment strategies. Conversely, the lack of a significant correlation between PIV and disease-free survival in patients with RCC highlights the need for further research to identify more relevant prognostic markers for this subgroup.

The uncertainty surrounding the prognostic value of PIV in RCC may be attributed to the distinct characteristics of the TME inherent to tumor location. RCCs exhibit a higher prevalence of KRAS and BRAF mutations. In contrast, TP53 mutations are more frequently observed in LCC. These genetic alterations contribute to an immunosuppressive TME, influencing tumor behavior and patient outcomes.

Furthermore, MSI-H tumors, which are more prevalent in RCC, exhibit an augmented inflammatory response and an enhanced recognition by the immune system, rendering them more responsive to immunotherapy. Consequently, elevated circulating inflammatory biomarkers may potentially predict a patient’s response to treatment. In rectal cancer, increased neutrophil-lymphocyte ratio and platelet-lymphocyte ratio before neoadjuvant chemoradiotherapy have been linked to poorer prognosis in patients with MSI-H. While this correlation has yet to be demonstrated in patients with colon cancer, it presents promising avenues for future research. Therefore, genetic, molecular, and inflammation-related characteristics should be considered when designing personalized therapies for patients with CRC. Further investigation is warranted to elucidate the relationship between inflammation and TME characteristics across different tumor locations and to understand the potential prognostic roles of inflammatory markers in CRC[22-24].

The large sample size and rigorous methodology of the study by Wang et al[20] lend credibility to its conclusions and strongly support integrating tumor location into the prognostic assessment of colon cancer. This integration could improve our ability to more accurately identify high-risk patients and develop personalized treatment plans that are more likely to improve patient outcomes. However, the study also raises important questions about the underlying mechanisms driving the observed differences in the prognostic value of PIV between LCC and RCC. The role of various immune and inflammatory cells in the TME, as well as their interaction with tumor cells, may differ significantly between these subtypes, influencing the overall prognostic landscape. The role of TME in influencing tumor behavior has already been studied in rectal cancer, particularly in relation to response to neoadjuvant treatment, and immunological and molecular differences in different populations have been associated with different prognoses[25,26]. Further research is needed to elucidate these mechanisms and to explore whether other systemic inflammatory response markers may also have site-specific prognostic value.

Furthermore, it should be noted that the study by Wang et al[20] is not without limitations, as acknowledged by the authors themselves. Notably, the absence of data on molecular mutations—specifically RAS mutations—and the lack of information regarding adjuvant therapy limit the applicability of their results. These gaps highlight the need for further research to validate and expand upon their findings.

FUTURE DIRECTIONS: TOWARDS A CONTINUUM OF PERSONALIZED CARE

Recent studies have highlighted the crucial function of inflammation in the genesis and recurrence of colon cancer, prompting a shift toward the investigation of novel targets for systemic therapy. Preclinical models have demonstrated encouraging outcomes with agents such as JAK inhibitors, chemokine receptor antagonists (e.g., CXCR2 inhibitors), antibodies targeting the interleukin-6 receptor or its signaling pathway, and cyclooxygenase-2 inhibitors[27]. The integration of these therapeutic modalities with chemotherapy and surgical interventions holds considerable potential to significantly improve the prognosis of patients with colon cancer.

The future of CRC care lies in a comprehensive, multiomics approach that integrates data from genomics, transcriptomics, proteomics, and metagenomics to create a holistic picture of the tumor and its microenvironment[3,28]. Liquid biopsies, which analyze circulating tumor DNA, offer the potential for real-time monitoring of tumor evolution and treatment response, further refining personalized treatment strategies[3,29]. The integration of artificial intelligence could enhance the clinical application of whole-genome sequencing analysis, enabling more precise therapeutic decision-making and improving patient prognosis[30].

CONCLUSION

The study by Wang et al[20] represents a significant advancement in our understanding of the role of PIV as a prognostic biomarker in CRC. By highlighting the importance of tumor location, this research paves the way for more precise and personalized approaches to CRC treatment. As we continue to explore the molecular and inflammatory landscape of CRC, the integration of such biomarkers into clinical practice will be critical to improving patient outcomes and advancing the field of precision medicine in oncology.

Biomarkers are changing the landscape of CRC care, shifting the focus beyond the limitations of traditional approaches toward a future of precision medicine. By unlocking the molecular secrets of the tumor and harnessing the power of the immune system, we can tailor treatment strategies to individual patients, maximizing their chances of survival and improving their quality of life. As the field advances, the integration of studies like Wang et al’s will be critical to refining and expanding our arsenal of prognostic tools, ultimately improving patient care and outcomes[20].

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single-blind

Specialty type: Gastroenterology and hepatology

Country of origin: Germany

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade C

Creativity or Innovation: Grade C

Scientific Significance: Grade C

P-Reviewer: Park SJ S-Editor: Qu XL L-Editor: Filipodia P-Editor: Zhang L

References
1.  Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2023. CA Cancer J Clin. 2023;73:233-254.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 1226]  [Reference Citation Analysis (2)]
2.  Ciardiello F, Ciardiello D, Martini G, Napolitano S, Tabernero J, Cervantes A. Clinical management of metastatic colorectal cancer in the era of precision medicine. CA Cancer J Clin. 2022;72:372-401.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in RCA: 261]  [Article Influence: 87.0]  [Reference Citation Analysis (0)]
3.  Lech G, Słotwiński R, Słodkowski M, Krasnodębski IW. Colorectal cancer tumour markers and biomarkers: Recent therapeutic advances. World J Gastroenterol. 2016;22:1745-1755.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in CrossRef: 234]  [Cited by in RCA: 276]  [Article Influence: 30.7]  [Reference Citation Analysis (8)]
4.  Yamamoto T, Kawada K, Obama K. Inflammation-Related Biomarkers for the Prediction of Prognosis in Colorectal Cancer Patients. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in RCA: 211]  [Article Influence: 52.8]  [Reference Citation Analysis (0)]
5.  Doleschal B, Petzer A, Rumpold H. Current concepts of anti-EGFR targeting in metastatic colorectal cancer. Front Oncol. 2022;12:1048166.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
6.  Kasper S, Reis H, Ziegler S, Nothdurft S, Mueller A, Goetz M, Wiesweg M, Phasue J, Ting S, Wieczorek S, Even A, Worm K, Pogorzelski M, Breitenbuecher S, Meiler J, Paul A, Trarbach T, Schmid KW, Breitenbuecher F, Schuler M. Molecular dissection of effector mechanisms of RAS-mediated resistance to anti-EGFR antibody therapy. Oncotarget. 2017;8:45898-45917.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
7.  Bahrami A, Hesari A, Khazaei M, Hassanian SM, Ferns GA, Avan A. The therapeutic potential of targeting the BRAF mutation in patients with colorectal cancer. J Cell Physiol. 2018;233:2162-2169.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
8.  Lai E, Liscia N, Donisi C, Mariani S, Tolu S, Pretta A, Persano M, Pinna G, Balconi F, Pireddu A, Impera V, Dubois M, Migliari M, Spanu D, Saba G, Camera S, Musio F, Ziranu P, Puzzoni M, Demurtas L, Pusceddu V, Dettori M, Massa E, Atzori F, Dessì M, Astara G, Madeddu C, Scartozzi M. Molecular-Biology-Driven Treatment for Metastatic Colorectal Cancer. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
9.  Johdi NA, Sukor NF. Colorectal Cancer Immunotherapy: Options and Strategies. Front Immunol. 2020;11:1624.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 235]  [Cited by in RCA: 260]  [Article Influence: 52.0]  [Reference Citation Analysis (0)]
10.  Taieb J, Svrcek M, Cohen R, Basile D, Tougeron D, Phelip JM. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur J Cancer. 2022;175:136-157.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 113]  [Reference Citation Analysis (0)]
11.  Akin Telli T, Bregni G, Vanhooren M, Saude Conde R, Hendlisz A, Sclafani F. Regorafenib in combination with immune checkpoint inhibitors for mismatch repair proficient (pMMR)/microsatellite stable (MSS) colorectal cancer. Cancer Treat Rev. 2022;110:102460.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
12.  Pecci F, Cantini L, Bittoni A, Lenci E, Lupi A, Crocetti S, Giglio E, Giampieri R, Berardi R. Beyond Microsatellite Instability: Evolving Strategies Integrating Immunotherapy for Microsatellite Stable Colorectal Cancer. Curr Treat Options Oncol. 2021;22:69.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in RCA: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
13.  Matsubara D, Arita T, Nakanishi M, Kuriu Y, Murayama Y, Kudou M, Konishi H, Komatsu S, Shiozaki A, Otsuji E. The impact of postoperative inflammation on recurrence in patients with colorectal cancer. Int J Clin Oncol. 2020;25:602-613.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 42]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
14.  Di Caro G, Carvello M, Pesce S, Erreni M, Marchesi F, Todoric J, Sacchi M, Montorsi M, Allavena P, Spinelli A. Circulating Inflammatory Mediators as Potential Prognostic Markers of Human Colorectal Cancer. PLoS One. 2016;11:e0148186.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
15.  Tohme S, Sukato D, Chalhoub D, McDonald KA, Zajko A, Amesur N, Orons P, Marsh JW, Geller DA, Tsung A. Neutrophil-lymphocyte ratio is a simple and novel biomarker for prediction of survival after radioembolization for metastatic colorectal cancer. Ann Surg Oncol. 2015;22:1701-1707.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in RCA: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
16.  He L, Li H, Cai J, Chen L, Yao J, Zhang Y, Xu W, Geng L, Yang M, Chen P, Zheng J, Yang Y, Gong S. Prognostic Value of the Glasgow Prognostic Score or Modified Glasgow Prognostic Score for Patients with Colorectal Cancer Receiving Various Treatments: a Systematic Review and Meta-Analysis. Cell Physiol Biochem. 2018;51:1237-1249.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
17.  Okugawa Y, Toiyama Y, Fujikawa H, Kawamura M, Yasuda H, Yokoe T, Mochiki I, Okita Y, Ohi M, Nakatani K. Cumulative perioperative lymphocyte/C-reactive protein ratio as a predictor of the long-term outcomes of patients with colorectal cancer. Surg Today. 2021;51:1906-1917.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
18.  Chen N, Li W, Huang K, Yang W, Huang L, Cong T, Li Q, Qiu M. Increased platelet-lymphocyte ratio closely relates to inferior clinical features and worse long-term survival in both resected and metastatic colorectal cancer: an updated systematic review and meta-analysis of 24 studies. Oncotarget. 2017;8:32356-32369.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in RCA: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
19.  Pereira F, Ferreira A, Reis CA, Sousa MJ, Oliveira MJ, Preto A. KRAS as a Modulator of the Inflammatory Tumor Microenvironment: Therapeutic Implications. Cells. 2022;11.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
20.  Wang QY, Zhong WT, Xiao Y, Lin GL, Lu JY, Xu L, Zhang GN, Du JF, Wu B. Pan-immune-inflammation value as a prognostic biomarker for colon cancer and its variation by primary tumor location. World J Gastroenterol. 2024;30:3823-3836.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (1)]
21.  Boeckx N, Koukakis R, Op de Beeck K, Rolfo C, Van Camp G, Siena S, Tabernero J, Douillard JY, André T, Peeters M. Primary tumor sidedness has an impact on prognosis and treatment outcome in metastatic colorectal cancer: results from two randomized first-line panitumumab studies. Ann Oncol. 2017;28:1862-1868.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in RCA: 168]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
22.  Ciepiela I, Szczepaniak M, Ciepiela P, Hińcza-Nowak K, Kopczyński J, Macek P, Kubicka K, Chrapek M, Tyka M, Góźdź S, Kowalik A. Tumor location matters, next generation sequencing mutation profiling of left-sided, rectal, and right-sided colorectal tumors in 552 patients. Sci Rep. 2024;14:4619.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
23.  Mulet-Margalef N, Linares J, Badia-Ramentol J, Jimeno M, Sanz Monte C, Manzano Mozo JL, Calon A. Challenges and Therapeutic Opportunities in the dMMR/MSI-H Colorectal Cancer Landscape. Cancers (Basel). 2023;15.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
24.  Lee JH, Kang BH, Song C, Kang SB, Lee HS, Lee KW, Chie EK, Kim JS. Microsatellite Instability Correlated Inflammatory Markers and their Prognostic Value in the Rectal Cancer Following Neoadjuvant Chemoradiotherapy: A Hypothesis-generating Study. In Vivo. 2020;34:2119-2126.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
25.  Spolverato G, Fassan M, Capelli G, Scarpa M, Negro S, Chiminazzo V, Kotsafti A, Angriman I, Campi M, De Simoni O, Ruffolo C, Astghik S, Vignotto C, Scognamiglio F, Becherucci G, Rivella G, Marchegiani F, Facci L, Bergamo F, Brignola S, Businello G, Guzzardo V, Dal Santo L, Salmaso R, Massani M, Pozza A, Cataldo I, Stecca T, Dei Tos AP, Zagonel V, Pilati P, Franzato B, Scapinello A, Pirozzolo G, Recordare A, Merenda R, Bordignon G, Guerriero S, Romiti C, Portale G, Cipollari C, Zizzo M, Porzionato A, Agostini M, Cavallin F, Di Camillo B, Bardini R, Maretto I, Castagliuolo I, Pucciarelli S, Scarpa M. IMMUNOREACT 5: female patients with rectal cancer have better immune editing mechanisms than male patients - a cohort study. Int J Surg. 2023;109:323-332.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
26.  Stepanyan A, Fassan M, Spolverato G, Castagliuolo I, Scarpa M, Scarpa M; IMMUNOREACT Study Group. IMMUNOREACT 0: Biopsy-based immune biomarkers as predictors of response to neoadjuvant therapy for rectal cancer-A systematic review and meta-analysis. Cancer Med. 2023;12:17878-17890.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
27.  Crutcher MM, Baybutt TR, Kopenhaver JS, Snook AE, Waldman SA. Emerging drug targets for colon cancer: A preclinical assessment. Expert Opin Ther Targets. 2022;26:207-216.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
28.  Gumbs AA, Croner R, Abu-hilal M, Bannone E, Ishizawa T, Spolverato G, Frigerio I, Siriwardena A, Messaoudi N. Surgomics and the Artificial intelligence, Radiomics, Genomics, Oncopathomics and Surgomics (AiRGOS) Project. Art Int Surg. 2023;3:180-5.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
29.  Zhou H, Zhu L, Song J, Wang G, Li P, Li W, Luo P, Sun X, Wu J, Liu Y, Zhu S, Zhang Y. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. Mol Cancer. 2022;21:86.  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 126]  [Article Influence: 42.0]  [Reference Citation Analysis (0)]
30.  Alagarswamy K, Shi W, Boini A, Messaoudi N, Grasso V, Cattabiani T, Turner B, Croner R, Kahlert UD, Gumbs A. Should AI-Powered Whole-Genome Sequencing Be Used Routinely for Personalized Decision Support in Surgical Oncology-A Scoping Review. BioMedInformatics. 2024;4:1757-1772.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]