Review Article

Inflammatory Response Markers as Predictors of Colorectal Cancer Prognosis

Minsung Kim1https://orcid.org/0000-0002-1984-5084, Il Tae Son1https://orcid.org/0000-0003-1781-9189, Bo Young Oh1,*https://orcid.org/0000-0002-1255-0961
Author Information & Copyright
1Department of Surgery, Hallym Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
*Corresponding author: Bo Young Oh, Department of Surgery, Hallym Sacred Heart Hospital, Hallym University College of Medicine, 22 Gwanpyeong-ro 170 Beon-gil, Pyeongan-dong, Dongan-gu, Anyang 14068, Korea, Tel: 82-31-380-3772, Fax: 82-31-380-1619, E-mail: obbyy@hanmail.net

© Copyright 2023 Ewha Womans University College of Medicine and Ewha Medical Research Institute. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Sep 23, 2023; Revised: Oct 15, 2023; Accepted: Oct 25, 2023

Published Online: Dec 31, 2023

Abstract

Colorectal cancer (CRC) is a globally prevalent and challenging malignancy. Accurate prognosis prediction is essential for optimizing patient care. This comprehensive review discusses the intricate relationships between inflammatory response markers and CRC prognosis. Inflammatory response markers have gained prominence as a prognostic tool. Elevations in the preoperative neutrophil-lymphocyte ratio, platelet-lymphocyte ratio, and C-reactive protein-albumin ratio predict a poor prognosis for patients with CRC. A decreased lymphocyte-monocyte ratio is also a poor prognostic factor. A high Glasgow prognostic score and a high modified Glasgow prognostic score are associated with adverse outcomes, including reduced survival. While significant progress has been made, challenges remain in standardizing the clinical application of these inflammatory response markers. Prospective research and further investigations are warranted to refine the prognostic models. Enhanced understanding and utilization of these inflammatory response markers will help advance personalized treatment strategies, refine surveillance protocols, and improve the management of CRC.

Keywords: Colorectal neoplasms; Inflammatory response marker; Prognosis

Introduction

Colorectal cancer (CRC) is one of the most commonly diagnosed malignancies and a leading cause of cancer-related death worldwide [1,2]. Advances in screening and treatment modalities have improved the survival of patients with CRC; however, the mortality rate remains high in cases of metastasis or recurrence [1,3]. The complexity of tumor progression—including, for instance, tumor heterogeneity, resistance mechanisms, genetic alterations, and micromolecular biology—contributes to the difficulty of achieving improvements in prognosis; therefore, more sophisticated and tailored treatment strategies are needed [4-7]. It is thus important to identify factors associated with a poor prognosis in patients with CRC. The conventional prognostic model for CRC is the TNM staging system, which was proposed by the American Joint Committee on Cancer. Clinical characteristics and additional pathologic features are also used to predict the patient’s prognosis [5-6,8,9]. However, patients with a similar clinicopathologic status and staging may have different prognoses.

Biomarkers are quantifiable and measurable indicators that reflect normal biological processes, pathological conditions, or responses to therapeutic interventions. Biomarkers serve as diagnostic tools that provide early disease detection and act as prognostic markers to offer insights into disease progression and potential outcomes [9-12]. Inflammatory response markers, which are among the most easily measurable biomarkers, reflect the body’s immune response and provide insights into the tumor microenvironment and its impact on the prognosis of CRC. Numerous previous studies have reported that inflammatory response markers, such as the neutrophil-lymphocyte ratio (NLR) and the platelet-lymphocyte ratio (PLR), can be useful for predicting the CRC prognosis [3-10,13,14]. The purpose of this study was to review inflammatory response markers that, according to current research, exhibit potential for predicting the prognosis of patients with CRC. This review summarizes the inflammatory response markers that can be obtained from routinely performed blood tests before CRC treatment, with the aim of offering an understanding of how inflammatory response markers may predict the prognosis of CRC and contribute to advances in the field of precision medicine in CRC [7].

Inflammatory Response Markers

1. Inflammation and cancer

Chronic inflammation has been recognized as an important factor in cancer initiation and progression [15]. It induces tissue damage, in response to which cell proliferation is activated as a part of the healing process. When chronic inflammation persists, there is a repeated cycle of tissue damage and regeneration, leading to the occurrence of genetic mutations. Inflammatory cells, such as macrophages and T cells, secrete cytokines and chemokines in response to tissue damage [16]. These signaling molecules (such as tumor necrosis factor-alpha and CXCL8) can affect tumor biology, including growth, migration, and differentiation, by releasing growth factors, promoting angiogenesis, and causing DNA damage [17]. Table 1 summarizes the inflammatory response markers and the prognosis of CRC.

Table 1. Inflammatory response markers associated with the prognosis of CRC
Author Year Population Patients (n) Main outcome HR (95% CI) P-value Cut-off
Neutrophil–lymphocyte ratio
Halazun [30] 2008 CRLM following curative-intent resection 440 DFS 2.26 (1.65−3.13) <0.001 5
Ding [28] 2010 CC following curative resection (stage IIA) 141 RFS 4.88 (1.73−13.75) 0.003 4
Hung [29] 2011 CC following curative resection (stage II) 1,040 OS 1.29 (1.07−1.80) 0.012 5
Chiang [18] 2012 CRC following curative resection (stage I-III) 3,857 DFS 1.31 (1.09−1.57) (especially CC) 0.013 3
Guthrie [19] 2013 CRC following curative resection (stage I-III) 206 CSS 3.07 (1.23−7.63) <0.05 5
Malietzis [20] 2014 CRC following curative resection (stage I-III) 506 DFS 2.41 (1.12−5.15) 0.024 3
Nagasaki [25] 2015 RC following nCRT and curative resection (stage I-III) 201 OS 3.38 0.012 3
Li [21] 2016 CRC following curative resection (stage I-III) 5,336 DFS OS 1.20 (1.05−1.37) 1.23 (1.01−1.50) 0.009 0.047 2.72
Song [23] 2017 CRC following resection (stage I-IV) 1,744 CSS OS 0.74 (0.57−0.95) 0.76 (0.60−0.96) (reference: NLR≥2) 0.018 0.021 2
Pedrazzani [24] 2017 CRC following curative resection (stage I-IV) 603 CSS OS 1.22 (0.77−1.93) 1.15 (0.86−1.54) 0.40 0.003 3.5
Mao [31] 2019 CRLM following nCT and resection 183 RFS OS 1.53 (1.08−2.18) 2.43 (1.49−3.94) 0.017 <0.001 2.3
Casadei-Gardini [33] 2019 CRC following CT (stage IV) 276 PFS OS 2.27 (1.59−3.23) 14.4 (11.4−17.1) <0.001 <0.001 3
Inamoto [22] 2019 CRC following curative resection (stage I-III) 448 DFS CSS OS 1.71 (1.12−2.66) 2.11 (0.96−5.05) 2.04 (1.11−3.96) 0.01 0.06 0.02 2.05
Erstad [32] 2020 CRLM following curative-intent resection 151 OS 2.46 (1.08−5.60) 0.032 5
Yosida [27] 2020 RC following curative resection (T1-2) 151 DFS 5.11 (1.84−16.4) 0.002 2.58
Zhang [26] 2020 RC following nCRT and curative resection (stage II-III) 472 DFS OS 1.71 (1.02−2.87) 1.80 (1.01−3.20) 0.044 0.046 2.3
Platelet–lymphocyte ratio
Pedrazzani [24] 2017 CRC following curative resection (stage I-IV) 603 CSS OS 1.64 (0.74−3.62) 1.86 (1.05−3.32) 0.22 0.034 350
Erstad [32] 2020 CRLM following curative-intent resection 151 OS 2.10 (1.04−4.23) 0.037 220
An [14] 2022 RC following nCRT and curative resection (stage I-III) 168 OS 1.79 (1.01−3.17) 0.047 170
Lymphocyte–monocyte ratio
Li [21] 2016 CRC following curative resection (stage I-III) 5,336 DFS OS 0.77 (0.67−0.88) 0.76 (0.62−0.93) <0.001 0.008 2.83
Chan [35] 2017 CRC following curative resection (stage I-III) 1,623 OS 0.57 (0.48−0.68) <0.001 2.38
Chen [37] 2019 Obstructive CRC with stent insertion following resection 128 DFS OS 0.42 (0.17−1.07) 0.40 (0.18−0.92) 0.068 0.031 1.67
Glasgow prognostic score
Choi [39] 2014 CRC following resection (stage I-IV) 105 CSS 5.17 (1.76−15.18) 0.003
Inamoto [22] 2019 CRC following curative resection (stage I-III) 448 DFS CSS OS 1.68 (1.03−2.67) 2.17 (1.03−4.49) 1.73 (0.97−3.02) 0.04 0.04 0.06
Lee [40] 2020 CRC following curative-intent resection (stage I-IV) 1,590 DFS OS 1.71 (1.23−2.38) 2.34 (1.62−3.39) 0.001 0.001
Modified Glasgow prognostic score
Leitch [43] 2007 CRC following curative-intent resection (stage I-IV) 149 CSS 1.44 (1.01−2.04) 0.043
Roxburgh [41] 2009 CRC following curative resection (stage I-III) 287 CSS 2.65 (1.66−4.25) <0.001
Park [44] 2016 CRC following curative-intent resection (stage I-IV) 1,000 CSS OS 1.28 (1.09−1.52) 1.28 (1.13−1.45) 0.003 <0.001
Tokunaga [42] 2017 CRC following curative resection (stage I-III) 468 RFS OS 2.14 (1.40−3.24) 2.45 (1.53−3.88) <0.001 <0.001
Suzuki [45] 2018 CRC following curative-intent resection (stage I-IV) 727 OS 2.01 0.005

CRC, colorectal cancer; HR, hazard ratio; CRLM, colorectal cancer with liver metastasis; DFS, disease-free survival; CC, colon cancer; RFS, recurrence-free survival; OS, overall survival; CSS, cancer-specific survival; RC, rectal cancer; nCRT, neoadjuvant chemoradiotherapy; NLR, neutrophil-lymphocyte ratio; PFS, progression-free survival.

Download Excel Table
2. Neutrophil-lymphocyte ratio

The NLR is a widely used biomarker to predict prognosis in CRC; it is defined as the absolute neutrophil count divided by the absolute lymphocyte count. Several studies have shown that a high preoperative NLR is associated with a poor prognosis in patients with stage I–III CRC who underwent curative resection. The cutoff values were different depending on the study and ranged from 2.05–5.00 [18-23]. Chiang et al. analyzed 3,857 patients with stage I–III CRC who underwent curative resection and found that a preoperative NLR>3 was a significant predictor of disease-free survival (DFS) [18]. Li et al. reported that a preoperative NLR>2.72 was associated with significantly lower DFS and overall survival (OS) rates in 5,336 patients with stage I–III CRC who underwent curative resection [21]. Some studies have concluded that a high preoperative NLR predicted a poor prognosis in patients with CRC who underwent curative-intent resection, including stage IV patients [23,24]. Song et al. reported that a preoperative NLR>2 was associated with lower cancer-specific survival (CSS) and OS rates in patients with stage I–IV CRC who underwent resection [23]. Several studies have focused on high NLR values and the prognosis of rectal cancer [25-28]. Zhang et al. analyzed 472 patients with advanced rectal cancer who underwent preoperative chemoradiotherapy followed by curative resection. A high NLR before chemoradiotherapy was significantly associated with worse DFS and OS [26]. Yosida et al. reported that a preoperative NLR>2.58 was associated with a lower DFS in patients with stage I–II rectal cancer who underwent curative resection [27]. Other studies have focused on high NLR values and the prognosis of colon cancer [28,29]. Hung et al. analyzed 1,040 patients with stage II colon cancer who underwent curative resection and found that a preoperative NLR>5 was associated with a lower OS rate [29].

Several studies have investigated the role of pretreatment NLR in colon cancer with distant metastasis [30-33]. A study by Halazun et al. demonstrated that a preoperative NLR>5 had a poor prognostic impact in patients with concurrent CRC liver metastasis who underwent curative-intent resection [30]. Mao et al. analyzed 183 patients who were diagnosed with CRC with liver metastasis and performed neoadjuvant chemotherapy followed by surgery. An NLR>2.3 before chemotherapy was associated with poor recurrence-free survival and OS [31]. Casadei-Gardini et al. performed a randomized-controlled trial in patients with Stage IV CRC who underwent chemotherapy and reported that a pretreatment NLR>3 was associated with poor progression-free survival and OS [33] .

3. Platelet-lymphocyte ratio

The PLR, which is defined as the ratio of the platelet count to the lymphocyte count, has also been suggested as a prognostic marker for CRC. Several studies have reported that a high preoperative PLR was associated with a poor prognosis in patients with CRC [24-32,34]. Pedrazzani et al. analyzed 603 patients with CRC who underwent curative resection and found that a preoperative PLR>350 was a significant predictor of CSS and OS [24]. Erstad et al. reported that a preoperative PLR>220 in patients with concurrent CRC liver metastasis who underwent curative-intent resection was associated with a worse OS [32].

4. Lymphocyte-monocyte ratio

The lymphocyte-monocyte ratio (LMR), which is defined as the ratio of the lymphocyte count to the monocyte count, can predict the prognosis of CRC. Several studies have shown that a high preoperative LMR is associated with a poor prognosis in patients with CRC who underwent curative resection [21-35,36]. Chan et al. analyzed 1,623 patients with stage I–III CRC and the prognostic impact of LMR. A preoperative LMR<2.38 was an independent prognostic factor and was superior to other biomarkers, such as the NLR and PLR [35]. Li et al. also reported that a preoperative LMR<2.83 was associated with lower DFS and OS in patients with stage I–III CRC who underwent curative resection [21]. A study by Chen et al. focused on obstructive CRC and the prognostic impact of the pretreatment LMR. An LMR<1.67 before endoscopic stenting was associated with poor DFS and OS [37].

5. Glasgow prognostic score and modified Glasgow prognostic score

The combination of a higher CRP value and hypoalbuminemia can be a sensitive biomarker for prognosis of CRC. The Glasgow prognostic score (GPS) is a useful scoring system for predicting the prognosis of patients with CRC, as well as other malignant tumors [38]. The GPS is based on the combination of hypoalbuminemia (<3.5 g/dL) and elevated CRP (>10 mg/L); if both are abnormal, the score is 2; if one or the other is abnormal, the score is 1; if neither is abnormal, the score is 0. Multiple studies have shown that a high GPS before surgery was associated with a poor prognosis for patients with stage I–III CRC who have undergone curative resection [22,38]. Choi et al. reported that a preoperative GPS of 2 was associated with a worse CSS in patients with stage I–IV CRC who underwent resection [39]. A study by Lee et al. evaluated 1,590 patients with CRC, including stage IV, who underwent curative-intent resection and revealed that a GPS of 1 or 2 was associated with DFS and OS rates [40]. The modified GPS (mGPS) is defined as follows: patients with a CRP level ≤10 mg/L and an albumin level ≥3.5 g/dL are scored as 0; those with a CRP level >10 mg/L are scored as 1; and those with a CRP level >10 mg/L and an albumin level <3.5 g/dL are scored as 2. Several studies have demonstrated that the preoperative mGPS was associated with the prognosis in patients with stage I–III CRC who underwent curative resection [41,42]. Roxburgh et al. reported that a preoperative mGPS of 1 or 2 was associated with lower CSS rates in patients with stage I–III CRC who underwent curative resection [41]. Other studies have evaluated the association between the prognosis of patients with CRC (including stage IV) who underwent curative-intent resection and had a preoperative mGPS [43-45]. Leitch et al. reported that a preoperative mGPS of 1 or 2 was associated with lower CSS rates in patients with stage I–IV CRC who underwent curative-intent resection [43]. A study by Park et al. analyzed 1,000 patients with stage I–IV CRC who underwent curative-intent resection and reported that a preoperative GPS of 1 or 2 was associated with poor CSS and OS [44]. A study by Suzuki et al. evaluated 737 patients with stage I–IV CRC who underwent curative-intent resection and concluded that a preoperative mGPS of 1 or 2 was associated with poor CSS [45].

Conclusion

In summary, our comprehensive review has shed light on the complex interplay between the prognosis of CRC and the roles of inflammatory response markers. These non-invasive biomarkers are easily accessible both before and after surgery. The findings discussed herein collectively highlight the critical significance of considering these inflammatory response markers when assessing the clinical prognosis of patients with CRC.

The evidence presented suggests that elevated levels of inflammatory response markers are associated with a poor prognosis in patients with CRC. These markers reflect the systemic inflammation that often accompanies malignancies, as well as the intricate relationship between the tumor microenvironment and the host immune response. Incorporating these markers into clinical practice could enhance the precision of prognosis prediction and inform treatment decisions. When used in combination with clinical assessments, these markers offer valuable insight into the management of patients with CRC.

Despite significant progress in understanding the relationship between these inflammatory response markers and the prognosis of CRC, challenges remain. The heterogeneity of CRC and the influence of various factors on inflammatory response marker levels underscore the need for continued research. Prospective studies, multi-center trials, and the exploration of emerging inflammatory response markers hold promise for refining prognostic models and improving patient outcomes. Ultimately, the integration of these inflammatory response markers into the clinical evaluation of patients with CRC is a promising way to improve personalized treatment strategies, optimize surveillance protocols, and advance the field of CRC management. As our understanding of these inflammatory response markers continues to evolve, so will our ability to predict, prevent, and effectively treat malignancies.

Acknowledgements

Not applicable.

Conflict of Interest

Bo Young Oh serves as the editorial board members of the Ewha Medical Journal, but had no role in the decision to publish this article. No other potential conflict of interest relevant to this article was reported.

Author Contribution

Conceptualization: Oh BY

Formal Analysis: Kim M

Investigation: Kim M

Methodology: Kim M

Project Administration: Oh BY

Writing – Original Draft: Kim M

Writing – Review & Editing: Kim M, Son IT, Oh BY

Ethics Approval and Consent to Participate

Not applicable.

References

1.

Kim MH, Park S, Yi N, Kang B, Park IJ. Colorectal cancer mortality trends in the era of cancer survivorship in Korea: 2000–2020. Ann Coloproctol. 2022; 38(5):343-352

2.

Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021; 71(1):7-33

3.

An S, Shim H, Kim K, Kim B, Bang HJ, Do H, et al. Pretreatment inflammatory markers predicting treatment outcomes in colorectal cancer. Ann Coloproctol. 2022; 38(2):97-108

4.

Kim JC, Bodmer WF. Genotypic and phenotypic characteristics of hereditary colorectal cancer. Ann Coloproctol. 2021; 37(6):368-381

5.

Lim Y, Gang DY, Lee WY, Yun SH, Cho YB, Huh JW, et al. Proteomic identification of arginine-methylated proteins in colon cancer cells and comparison of messenger RNA expression between colorectal cancer and adjacent normal tissues. Ann Coloproctol. 2022; 38(1):60-68

6.

Kim YH, Kim KJ. Upregulation of prostaglandin E2 by inducible microsomal prostaglandin E synthase-1 in colon cancer. Ann Coloproctol. 2022; 38(2):153-159

7.

Park IJ. Precision medicine for primary rectal cancer will become a reality. Ann Coloproctol. 2022; 38(4):281-282

8.

Eom T, Lee Y, Kim J, Park I, Gwak G, Cho H, et al. Prognostic factors affecting disease-free survival and overall survival in T4 colon cancer. Ann Coloproctol. 2021; 37(4):259-265

9.

Duffy MJ, Lamerz R, Haglund C, Nicolini A, Kalousová , Holubec L, et al. Tumor markers in colorectal cancer, gastric cancer and gastrointestinal stromal cancers: European group on tumor markers 2014 guidelines update. Int J Cancer. 2014; 134(11):2513-2522

10.

Lech G, Słotwiński R, Słodkowski M, Krasnodębski IW. Colorectal cancer tumour markers and biomarkers: recent therapeutic advances. World J Gastroenterol. 2016; 22(5):1745-1755

11.

Joo JI, Lim SW, Oh BY. Prognostic impact of carcinoembryonic antigen levels in rectal cancer patients who had received neoadjuvant chemoradiotherapy. Ann Coloproctol. 2021; 37(3):179-185

12.

Kang S. Carcinoembryonic antigen, the most accessible test for predicting colorectal cancer prognosis: exploring alternative roles. Ann Coloproctol. 2021; 37(3):129-130

13.

Guo J, Chok AY, Lim HJ, Tay WX, Lye WK, Samarakoon LB, et al. Prognostic value of neutrophil-to-lymphocyte ratio in obstructing colorectal cancer treated by endoscopic stenting as a bridge to surgery. Ann Coloproctol. 2021; 37(3):159-165

14.

An SH, Kim IY. Can pretreatment platelet-to-lymphocyte and neutrophil-to-lymphocyte ratios predict long-term oncologic outcomes after preoperative chemoradiation followed by surgery for locally advanced rectal cancer?. Ann Coloproctol. 2022; 38(3):253-261

15.

Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008; 454:436-444

16.

Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002; 420:860-867

17.

Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010; 140(6):883-899

18.

Chiang SF, Hung HY, Tang R, Changchien CR, Chen JS, You YT, et al. Can neutrophil-to-lymphocyte ratio predict the survival of colorectal cancer patients who have received curative surgery electively?. Int J Colorectal Dis. 2012; 27:1347-1357

19.

Guthrie GJK, Roxburgh CSD, Farhan-Alanie OM, Horgan PG, McMillan DC. Comparison of the prognostic value of longitudinal measurements of systemic inflammation in patients undergoing curative resection of colorectal cancer. Br J Cancer. 2013; 109(1):24-28

20.

Malietzis G, Giacometti M, Askari A, Nachiappan S, Kennedy RH, Faiz O, et al. A preoperative neutrophil to lymphocyte ratio of 3 predicts disease-free survival after curative elective colorectal cancer surgery. Ann Surg. 2014; 260(2):287-292

21.

Li Y, Jia H, Yu W, Xu Y, Li X, Li Q, et al. Nomograms for predicting prognostic value of inflammatory biomarkers in colorectal cancer patients after radical resection. Int J Cancer. 2016; 139(1):220-231

22.

Inamoto S, Kawada K, Okamura R, Hida K, Sakai Y. Prognostic impact of the combination of neutrophil-to-lymphocyte ratio and Glasgow prognostic score in colorectal cancer: a retrospective cohort study. Int J Colorectal Dis. 2019; 34(7):1303-1315

23.

Song Y, Yang Y, Gao P, Chen X, Yu D, Xu Y, et al. The preoperative neutrophil to lymphocyte ratio is a superior indicator of prognosis compared with other inflammatory biomarkers in resectable colorectal cancer. BMC Cancer. 2017; 17(1):744

24.

Pedrazzani C, Mantovani G, Fernandes E, Bagante F, Salvagno GL, Surci N, et al. Assessment of neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio and platelet count as predictors of long-term outcome after R0 resection for colorectal cancer. Sci Rep. 2017; 7(1):1494

25.

Nagasaki T, Akiyoshi T, Fujimoto Y, Konishi T, Nagayama S, Fukunaga Y, et al. Prognostic impact of neutrophil-to-lymphocyte ratio in patients with advanced low rectal cancer treated with preoperative chemoradiotherapy. Dig Surg. 2015; 32(6):496-503

26.

Zhang Y, Liu X, Xu M, Chen K, Li S, Guan G. Prognostic value of pretreatment systemic inflammatory markers in patients with locally advanced rectal cancer following neoadjuvant chemoradiotherapy. Sci Rep. 2020; 10(1):8017

27.

Yoshida D, Minami K, Sugiyama M, Ota M, Ikebe M, Morita M, et al. Prognostic impact of the neutrophil-to-lymphocyte ratio in stage I-II rectal cancer patients. J Surg Res. 2020; 245:281-287

28.

Ding PR, An X, Zhang RX, Fang YJ, Li LR, Chen G, et al. Elevated preoperative neutrophil to lymphocyte ratio predicts risk of recurrence following curative resection for stage IIA colon cancer. Int J Colorectal Dis. 2010; 25(12):1427-1433

29.

Hung HY, Chen JS, Yeh CY, Changchien CR, Tang R, Hsieh PS, et al. Effect of preoperative neutrophil–lymphocyte ratio on the surgical outcomes of stage II colon cancer patients who do not receive adjuvant chemotherapy. Int J Colorectal Dis. 2011; 26(8):1059-1065

30.

Halazun KJ, Aldoori A, Malik HZ, Al-Mukhtar A, Prasad KR, Toogood GJ, et al. Elevated preoperative neutrophil to lymphocyte ratio predicts survival following hepatic resection for colorectal liver metastases. Eur J Surg Oncol. 2008; 34(1):55-60

31.

Mao R, Zhao JJ, Bi XY, Zhang YF, Li ZY, Huang Z, et al. A low neutrophil to lymphocyte ratio before preoperative chemotherapy predicts good outcomes after the resection of colorectal liver metastases. J Gastrointest Surg. 2019; 23(3):563-570

32.

Erstad DJ, Taylor MS, Qadan M, Tanabe KK, Chang DC, Ferrone CR, et al. Platelet and neutrophil to lymphocyte ratios predict survival in patients with resectable colorectal liver metastases. Am J Surg. 2020; 220(6):1579-1585

33.

Casadei-Gardini A, Scarpi E, Ulivi P, Palladino MA, Accettura C, Bernardini I, et al. Prognostic role of a new inflammatory index with neutrophil-to-lymphocyte ratio and lactate dehydrogenase (CII: colon inflammatory index) in patients with metastatic colorectal cancer: results from the randomized Italian Trial in Advanced Colorectal Cancer (ITACa) study. Cancer Manag Res. 2019; 11:4357-4369

34.

Ozawa T, Ishihara S, Nishikawa T, Tanaka T, Tanaka J, Kiyomatsu T, et al. The preoperative platelet to lymphocyte ratio is a prognostic marker in patients with stage II colorectal cancer. Int J Colorectal Dis. 2015; 30(9):1165-1171

35.

Chan JC, Chan DL, Diakos CI, Engel A, Pavlakis N, Gill A, et al. The lymphocyte-to-monocyte ratio is a superior predictor of overall survival in comparison to established biomarkers of resectable colorectal cancer. Ann Surg. 2017; 265(3):539-546

36.

Dolan RD, McSorley ST, Park JH, Watt DG, Roxburgh CS, Horgan PG, et al. The prognostic value of systemic inflammation in patients undergoing surgery for colon cancer: comparison of composite ratios and cumulative scores. Br J Cancer. 2018; 119(1):40-51

37.

Chen XQ, Xue CR, Hou P, Lin BQ, Zhang JR. Lymphocyte-to-monocyte ratio effectively predicts survival outcome of patients with obstructive colorectal cancer. World J Gastroenterol. 2019; 25(33):4970-4984

38.

Lu X, Guo W, Xu W, Zhang X, Shi Z, Zheng L, et al. Prognostic value of the Glasgow prognostic score in colorectal cancer: a meta-analysis of 9,839 patients. Cancer Manag Res. 2019; 11:229-249

39.

Choi KW, Hong SW, Chang YG, Lee WY, Lee B, Paik IW, et al. Inflammation-based score (Glasgow prognostic score) as an independent prognostic factor in colorectal cancer patients. Ann Surg Treat Res. 2014; 86(6):309-313

40.

Lee SC, Huh JW, Lee WY, Yun SH, Kim HC, Cho YB, et al. Prognostic value of serum inflammatory markers in colorectal cancer. Int J Colorectal Dis. 2020; 35(7):1211-1219

41.

Roxburgh CS, Salmond JM, Horgan PG, Oien K, McMillan DC. Comparison of the prognostic value of inflammation-based pathologic and biochemical criteria in patients undergoing potentially curative resection for colorectal cancer. Ann Surg. 2009; 249(5):788-793

42.

Tokunaga R, Sakamoto Y, Nakagawa S, Izumi D, Kosumi K, Taki K, et al. Comparison of systemic inflammatory and nutritional scores in colorectal cancer patients who underwent potentially curative resection. Int J Clin Oncol. 2017; 22(4):740-748

43.

Leitch EF, Chakrabarti M, Crozier JEM, McKee RF, Anderson JH, Horgan PG, et al. Comparison of the prognostic value of selected markers of the systemic inflammatory response in patients with colorectal cancer. Br J Cancer. 2007; 97(9):1266-1270

44.

Park JH, Watt DG, Roxburgh CS, Horgan PG, McMillan DC. Colorectal cancer, systemic inflammation, and outcome: staging the tumor and staging the host. Ann Surg. 2016; 263(2):326-336

45.

Suzuki Y, Okabayashi K, Hasegawa H, Tsuruta M, Shigeta K, Kondo T, et al. Comparison of preoperative inflammation-based prognostic scores in patients with colorectal cancer. Ann Surg. 2018; 267(3):527-531