Back to Journals » Journal of Inflammation Research » Volume 18

Robust Predictive Performance of MLPAS and CCMLP for Clinical Outcome and Risk Stratification in Patients with Colorectal Cancer

Authors Ye QY, Wang YY, Wang ZJ, Lu M, Peng HX, Wang X, Cheng XX , Ying HQ 

Received 4 October 2024

Accepted for publication 10 February 2025

Published 15 March 2025 Volume 2025:18 Pages 3889—3900

DOI https://doi.org/10.2147/JIR.S498028

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Ning Quan



Qiu-Ying Ye,1– 3,* Yuan-Yuan Wang,1,* Zhi-Jie Wang,1,* Min Lu,4 Hong-Xin Peng,5 Xin Wang,6 Xue-Xin Cheng,1 Hou-Qun Ying1,3,7

1Department of Clinical Laboratory, Immunity and Inflammation Key Laboratory of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China; 2Department of Medical Technology, Jiangxi Medical College, Shangrao, 334000, People’s Republic of China; 3Department of Laboratory Medicine, Central Hospital of Shangrao City, Shangrao, 334000, People’s Republic of China; 4Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China; 5Department of Clinical Laboratory, Nanjing First Hospital, Nanjing, 210006, People’s Republic of China; 6Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, People’s Republic of China; 7Shangrao Medical Center, The Second Affiliated Hospital of Nanchang University, Shangrao, 334000, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Hou-Qun Ying, Department of Clinical Laboratory, Immunity and Inflammation Key Laboratory of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, No. 1 of Minde Road, Nanchang, 330006, People’s Republic of China, Tel/Fax +86 0791 86297662, Email [email protected]

Background: There is no recognized biomarker is recommended to monitor or predict the prognosis of colorectal cancer (CRC) patients with negative detection of carcinoembryonic antigen (CEA) or carbohydrate antigen 19– 9 (CA19-9) and to classify high recurrence-risk cases.
Methods: Discovery and two-stage validation cohorts, which included 2111 radically resected patients with stage II–III CRC, were enrolled in this study. We detected preoperative peripheral monocyte, platelet, albumin (Alb), pre-albumin (pAlb), CEA, and CA19-9 and investigated the prognostic and risk-stratified roles of twelve new inflammatory biomarkers in the three cohorts.
Results: In our study, monocyte-to-pAlb ratio (MPAR), monocyte-to-lymphocyte -to-Alb ratio (MLAR), monocyte-to-lymphocyte-to-pAlb ratio (MLPAR), monocyte- to-pAlb score (MPAS), lymphocyte-to-monocyte-Alb score (MLAS), lymphocyte-to monocyte-pAlb score (MLPAS), and platelet-to-lymphocyte-Alb score (PLAS) were significantly associated with both RFS and OS in three cohorts. MLPAS showed the best performance in predicting RFS and OS, and it was related to right-tumor location and significant cancer burden (≥ 5cm) in the overall population. Moreover, MLPAS is a robust prognostic biomarker in subgroups stratified by CEA or CA19-9. Patients with scores zero and two of the CEA-CA19-9-MLPAS score (CCMLP) showed the lowest and highest recurrence and death rates, respectively, and significant survival differences were observed between them.
Conclusion: MLPAS is an optimal, independent, and robust prognostic biomarker in the stage II–III CRC population, especially with negative CEA or CA19-9. The CCMLP could effectively classify high recurrence-risk patients who require more focus, monitoring, and treatment for the clinic.

Keywords: colorectal cancer, inflammation, prognosis, lymphocyte to monocyte-pre-albumin score, CEA-CA19-9-MLPAS score

Introduction

Primary radical resection is the way to cure patients with early-stage colorectal cancer (CRC).1 However, approximately 22.24% and 43.09% of stage II and stage III CRC patients can be observed to have recurrence or distal metastasis in three years’ follow-up.2 So, accurately identifying patients with high-recurrence risk and efficient stratification of those who harbor unsatisfactory outcomes is essential for clinical treatment and decision-making.

Currently, carcinoembryonic antigen (CEA) is one of the most common biomarkers used to monitor recurrence and predict the survival of postoperative patients.3 Its sensitivity ranged from 41% to 97% and specificity from 52% to 100% for CRC.4 Unfortunately, a small number of patients with negative CEA detection and CEA neither classify the patients with high-recurrence risk nor predict the survival of these patients.5,6 Carbohydrate antigen 19–9 (CA19-9) is the second most common biomarker to predict the survival of these postoperative patients. High and low CA19-9 detections can stratify patients with different outcomes.6,7 However, the expected efficacy is unsatisfactory, and significant survival heterogeneity was observed in patients with either high or low CA19-9.8 Hence, it is urgent to discover the ideal biomarker to monitor and predict CRC patients, especially for patients with negative CEA or CA19-9.

It is well known that cancer-elicited inflammation is a hallmark of malignancy, including CRC.9 Systematic chronic inflammation can regulate the expression of albumin (Alb), and pre-albumin (pAlb). High circulating, monocyte (Mon) and platelet (Plt) counting, and low lymphocyte (Lym) counting are commonly detected in peripheral samples of CRC patients. Our previous study showed that new combined inflammatory ratios could help select patients who could benefit from adjuvant chemotherapy and bevacizumab-based target therapy and were superior to the single biomarker in predicting the prognosis of patients with early or advanced CRC.1013 Several inflammatory scores, such as the inflammation-Immunity-Nutrition score, Glasgow prognostic score, and Naples prognostic score, were also considered as independent prognostic biomarkers for CRC.14–16 However, there is no study to report the prognostic role of Mon to Alb ratio (MAR), Mon to pAlb ratio (MPAR), ratio of Mon to Lym to Alb (MLAR), ratio of Mon to Lym to pAlb (MLPAR), Plt to Alb ratio (PAR), ratio of Plt to Lym to Alb (PLAR) in stage II–III CRC. The predicting efficacy of Mon-Alb score (MAS), Mon-pAlb score (MPAS), Lym to Mon-Alb score (MLAS), Lym to Mon-pAlb score (MLPAS), Plt-Alb score (PAS), and Plt to Lym-Alb score (PLAS) in the prognosis of these patients remains unknown.

In this study, we formed twelve inflammatory biomarkers, and three cohorts, including 2111 patients, were enrolled to investigate: 1) the prognostic role of these biomarkers in CRC; 2) the optimal prognostic biomarker between them, especially in subgroups with negative CEA or CA19-9; 3) the stratification role of the optimal prognostic biomarker in identifying the patients with high-recurrence risk and unsatisfactory outcome.

Materials and Methods

Population

In our study, 1413 patients recruited from the Second Affiliated Hospital of Nanchang University were randomly divided into discovery and internal validation cohorts in a 6:4 ratio following previous research.17 An external validation cohort consisted of patients from Nanjing First Hospital, and the Second Hospital of Shandong First Medical University, respectively. All enrolled patients shall meet the following inclusion criteria: 1) clinical and pathological detection confirmed patients with stage II–III CRC according to the Chinese Protocol of Diagnosis and Treatment of Colorectal Cancer;18 2) the enrolled patients received radical resection; 3) all the patients participated voluntarily and signed the informed consent form. The patients who meet the following criteria shall be excluded: 1) combined with other malignancies, hematological or autoimmune diseases, as well as recent infections or injuries; 2) combined with benign chronic inflammatory intestinal disease; 3) age under 18 years old; 3) non-firstly clinical confirmation or performed clinical intervention ahead of clinical confirmation. The Ethics Committee of each hospital approves this study, and all eligible patients have signed the informed consent form.

Data Collection

The following clinical characteristics: gender, age, life history (smoking and drinking), and chronic diseases (diabetes, hypertension) were collected from the electronic medical record system in each hospital. Two senior pathological experts examined tumor pathological characteristics, such as tumor invasion, lymph node status or distal metastasis, cell differentiation, and tumor size.

Sample Collection and Laboratory Detection

We collected clinical samples, a 2mL EDTA anti-coagulation peripheral blood sample, a 2mL sodium citrate anti-coagulation plasma sample, and a 4mL serum sample at 7:00~9:00 am from each patient before clinical surgery. Peripheral blood cell counting, such as circulating neutrophil, monocyte, lymphocyte, and platelet counting, was measured by Sysmex HST-302 analyzer (Sysmex, Tokyo, Japan). AU5400 analyzer (Beckman Coulter, Tokyo, Japan) was used to detect serum Alb and pAlb using bromocresol green colorimetry and immunoturbidimetric assay, respectively. The cancer biomarkers CEA and CA19-9 were detected by chemiluminescent immunoassay using the SIEMENS ADVIA Centaur XP machine (Siemens, Erlangen, Germany). We measured peripheral blood counting within two hours after sample collection, and the other detections were completed within six hours. The coefficients of variation within intra- and inter-assay of the detection was less than 10%.

According to the computational formula, we formed and calculated six inflammatory biomarkers, MAR, MPAR, MLAR, MLPAR, PAR, and PLAR, based on the detection results. We also established six inflammatory scores, MAS, MPAS, MLAS, MLPAS, PAS, and PLAS, and each of them was defined as score zero, one, and two according to the cut-off value of each parameter. The detailed formulas and defined scores are displayed in Table S1.

Follow-Up

We performed three years of follow-up in each eligible patient, three months in the first and second years and six months in the third year. The follow-up was conducted through clinical follow-up, telephone, email, and consultation of clinical records. Recurrence and distal metastasis were diagnosed by clinical imaging detection. The deadline for follow-up was Dec 31, 2023. The follow-up endpoints are recurrence-free survival (RFS) and overall survival (OS), and the times from surgery to clinical recurrence/distal metastasis or death and follow-up deadline were defined as RFS and OS, respectively.

Statistics

The optimal cut-off values of each inflammatory biomarker and six inflammatory ratios were defined using X-tile software (Yale University, New Haven, Connecticut) relying on RFS (Table S1). Binary variables were displayed as numbers and percentages, and the differences in each group were compared using the chi-square test and Fisher’s exact test. Continuous variables were shown as average ± standard deviation (SD) and were compared using the Kruskal–Wallis H-test in intergroup analysis. We conducted the Kaplan–Meier curve (Log rank test) and the univariate and multivariate Cox regression model to identify independent factors in influencing RFS and OS for the patients, determining the hazard ratios (HR) and 95% confidence intervals (CI). We used time-dependent receiver operating characteristic (ROC) curves to assess and compare the effectiveness of these indicators in predicting prognosis. Statistical analysis was performed using SPSS. 27.0 (IBM Corp, Armonk, NY, USA), R4.3.3 (Institute for Statistics and Mathematics, Vienna, Austria), along with GraphPad Prism 10 (GraphPad Software Inc., San Diego, USA).

Results

According to inclusion and exclusion criteria, 2111 eligible patients were enrolled in this study. The discovery, internal, and external validation cohorts included 847, 566, and 698 cases. The detailed baseline characteristics are displayed in Table 1. In three cohorts, it shows significant differences in MAR, MAS, MPAR, MPAS, MLAS, PAR, PAS, PLAR, recurrence rate, and death rate. More than 75% of the patients received postoperative adjuvant chemotherapy in each cohort. The postoperative recurrence rates were 27.20%, 26.30%, and 36.80% in discovery, internal, and external validation cohorts. However, the death rates were similar in the three cohorts.

Table 1 Baseline Clinical Characteristics of the Eligible Patients in Three Cohorts

Table 2 Kaplan–Meier Curve and Cox Regression Analysis in Discovery Cohort

In the discovery cohort, significant survival differences were observed in high- and low-groups stratified by cell differentiation, lymph node status (LN), CEA, CA19-9, MAR, MAS, MPAR, MPAS, MLAR, MLAS, MLPAR, MLPAS, PAR, PAS, PLAR, and PLAS in terms of RFS and OS in the analysis of Kaplan–Meier curve, respectively (Figure 1A and B). Moreover, LN, CEA, CA19-9, MAR, MAS, MPAR, MPAS, MLAR, MLAS, MLPAR, MLPAS, PAR, PAS and PLAS were still significantly associated with clinical outcomes of the patients with stage II–III CRC, respectively (Table 2).

Figure 1 Kaplan–Meier curve of MLPAS in the three cohorts. (A and B) recurrence-free survival and overall survival in the discovery cohort, respectively; (C and D) recurrence-free survival and overall survival in the internal validation cohort, respectively; (E and F) recurrence-free survival and overall survival in the external validation cohort, respectively.

Notes: *p < 0.001.

In internal validation cohort, LN, CA19-9, MAS, MPAR, MPAS, MLAR, MLAS, MLPAR, MLPAS, PAR, PAS, PLAR, and PLAS were validated to be significantly associated with survival of the patients, respectively (Figure 1C and D, 2A and B). Moreover, the significant associations of CEA, CA19-9, MPAR, MPAS, MLAR, MLAS, MLPAR, MLPAS, and PLAS with the prognosis were still observed in the external validation cohort (Figures 1E and F, 2C and D).

Figure 2 Forest plot of LN status, CEA, CA19-9, and the twelve inflammatory biomarkers in the internal and external validation cohorts. (A) recurrence-free survival in the internal validation cohort; (B) overall survival in the internal validation cohort; (C) recurrence-free survival in the external validation cohort; (D) overall survival in the external validation cohort.

Time-dependent ROC showed that MLPAS harbored the highest areas under the curve (AUCs) to predict 12 (AUC = 0.65 for RFS, AUC = 0.67 for OS), 24 (AUC = 0.64 for RFS, AUC = 0.64 for OS), and 36 (AUC = 0.63 for RFS, AUC = 0.64 for OS) months’ survival in the overall population (Figure 3A and B). The AUCs of MLPAS are higher than CEA (p < 0.01) or CA19-9 (p < 0.01) in predicting one, two, and three years’ of RFS and OS of the patients (Table S2). Furthermore, the distribution of left primary tumor locations is significantly higher in patients with low-MLPAS than in high-MLPAS patients (p < 0.01). Patients with high MLPAS harbored a significantly enormous cancer burden (≥5cm) compared to patients with low MLPAS (p < 0.01) (Figure 3C and D). However, no distribution difference between high- and low-MLPAS was observed in the subgroups stratified by TNM stage, cell differentiation, or LN.

Figure 3 Prognostic area under time-dependent ROC (AUROC) of seven new inflammatory biomarkers and association of MLPAS with primary tumor location and cancer burden in the overall population. (A) recurrence-free survival; (B) overall survival; (C) MLPAS and primary tumor location; (D) MLPAS and cancer burden.

Notes: *p < 0.001.

In patients with negative CEA, the recurrence rate was 24.03%, and the rate within the high-MLPAS cases was significantly higher than in the low-MLPAS patients (p < 0.01 for 42.01% vs 17.77%). Recurrence rates were 26.46%, 43.48%, and 19.63% in CA19-9-negative patients and patients with high- or low-MLPAS, respectively (Table S3). The survival of patients with low-MLPAS was superior to that of high-MLPAS patients in subgroups with either negative CEA or CA19-9 or both negative CEA and CA19-9 (Figure 4A–F). Moreover, significant survival differences were also observed in high- and low-MLPAS patients with positive CEA or CA19-9.

Figure 4 Kaplan–Meier curve of CEA, CA19-9, MLPAS, CCMLP in the overall population. (A and C) MLPAS in negative CEA subgroup; (B and D) MLPAS in negative CA19-9 subgroup; (E and F) MLPAS in both negative CEA and CA19-9 subgroup (G) RFS comparison in subgroups with negative CEA, CA19-9, MLPAS, both negative CEA and CA19-9, and CCMLP (score zero); (H) RFS comparison in subgroups with positive CEA, CA19-9, and MLPAS (score two), and CCMLP (score two); (I and J) clinical outcome comparison in subgroups stratified by CCMLP score.

Notes: *p < 0.001.

According to the cut-off values of CEA, CA19-9, and MLPAS, we established a new prognostic score named CCMLP. Among them, 43.58%, 55.25%, 1.17% of the patients were defined as score zero, one, and two of CCMLP, respectively. The recurrence rates of the patients with CCMLP scores zero, one, and two were 16.03%, 38.62%, and 71.43%, respectively, and the rate in score zero and two patients is low and significantly high compared to CEA, CA19-9, or MLPAS as well as combined CEA or CA19-9 and MLPAS scores (Table S3). The death rates were 8.08%, 25.78%, and 47.26% in each subgroup stratified by CCMLP (Table S3). The Kaplan–Meier curve showed that the patients with scores zero and two harbored the best and worst outcomes (Figure 4G–J), and significant clinical outcome differences were observed in comparison of the score one vs score 0 [adjusted HR (95% CI) = 2.56 (2.03–3.22) for RFS; adjusted HR (95% CI) = 3.41 (2.44–4.77) for OS] and score two vs score 0 [adjusted HR (95% CI) = 10.86 (5.89–20.00) for RFS; adjusted HR (95% CI) = 9.53 (4.35–20.92) for OS] (Table 3).

Table 3 Kaplan–Meier Curve and Cox Regression Analysis of CCMLP in Overall Population

Discussion

It is unknown what the prognostic and stratifying role of MLPAS is in patients of stage II–III CRC, particularly in cases with negative CEA or CA19-9. Our study observed that three new inflammatory ratios (MPAR, MLAR, MLPAR) and four new inflammatory scores (MPAS, MLAS, MLPAS, PLAS) were independent predictive biomarkers for the prognosis of patients in the three cohorts. MLPAS was the best factor in predicting the clinical outcome of the patients, especially in the CEA or CA19-9 negative subgroup. Moreover, CCMLP could effectively identify postoperative patients with the lowest and the highest risk of recurrence and death.

As we know, CRC is a kind of metabolic wasting disease accompanied by metabolic reprogramming, contributing to malnutrition, hypoalbuminemia, sarcopenia, or even cachexia.19,20 Moreover, high systematic chronic inflammation, which CRC elicits, is commonly observed in middle- or advanced-stage disease, showing elevated counting of peripheral neutrophils, monocytes, platelets, and lymphocytopenia.2,12,21 Our previous studies showed that the Neu to Lym ratio and fibrinogen to pAlb ratio were independent prognostic biomarkers for CRC.22,23 Xie also reported that combined neutrophil/lymphocyte ratio and C-reaction protein index could serve as an effective biomarker to predict the survival of cancer.24 In this study, we found that three new ratios (MPAR, MLAR, MLPAR) and four scores (MPAS, MLAS, MLPAS, PLAS) were significantly associated with the prognosis of stage II–III CRC patients in discovery, internal, and external validation cohorts, demonstrating that these new inflammatory biomarkers were robust and independent biomarkers to predict the outcome of the patients. MLPAS harbored the highest predicted efficacy among these biomarkers for predicting the clinical outcome of the patients, implying that it is an optimal and ideal prognostic factor. Significant associations were observed not only in patients with positive CEA or CA19-9 but also in the negative patients, revealing that MLPAS was superior to CEA and CA19-9, and it could be recognized as a supplementary biomarker for cancer biomarkers to predict the outcome, particular for the patients with negative results.

MLPAS is a new inflammatory score for the Lym to Mon ratio and pAlb. Cytotoxic T lymphocytes and B lymphocytes are the main components consistent with tumor-infiltrating lymphocytes and play an essential role in anti-tumor characteristics, contributing to a favorite clinical outcome in CRC regardless of microsatellite instability status.25 Tumor-associated macrophages, differentiated from circulating monocytes, can accelerate CRC tumorigenesis via IL-6 and IL-8 secretion.26,27 Serum Alb, especially pre-Alb, is the optimal factor reflecting the nutritional status in patients, and inflammatory reactions can contribute to hypoalbuminemia, leading to poor survival.28 Moreover, MLPAS was significantly correlated with CRC characteristics, including primary tumor location and cancer burden. The association between these clinical features and the progression of CRC consequently affects the prognosis of patients with CRC. Thus, MLPAS is an optimal biomarker to reflect systematic inflammation and predict clinical outcomes in patients with CRC.

No widely recognized solution exists to classify the postoperative progression patients or the best survival cases.29 Exosomes, circulating tumor cells, and circulating tumor DNA detections were reported to stratify CRC patients relying on microfluidics-based methods, nanoparticles, electrochemical biosensors, respectively.30–32 However, the high detection requirements of these biomarkers restrict their popularization and application, especially in many primary medical units. In this study, we newly formed CCMLP, and we found that the recurrence and death rates among patients with CCMLP zero score were the lowest compared to negative CEA, CA19-9, MLPAS, or combined two of them. On the contrary, the highest recurrence and death rates were found in patients with triple-positive biomarkers (MLPAS score two), and the highest recurrence and death rates were approximately 4.5-fold and six-fold compared to the lowest rates, respectively. Moreover, CCMLP was an independent and robust biomarker for predicting the survival of patients in the overall population. These results illustrate that CCMLP could identify subgroups with significant differences in outcome, and it helped to distinguish a class of patients with high-recurrence risk who need clinical focus and real-time monitoring.

The new inflammatory indexes, estimated based on peripheral blood counting and circulating Alb or pAlb detection, are simple, economical, practical, and easy to popularize. In our large sample size and multiple center study, we first reported that MLPAS was a robust and independent prognostic score for patients with both negative- and positive CEA or CA19-9 patients. It is also the first time to report a new biomarker CCMLP, and we confirmed that the newly combined score could serve as a new classification system based on chronic inflammation. However, some limitations should be addressed as follows: 1) we did not obtain comprehensive treatment data, such as the detailed chemotherapy regimen and cycle, as well as clinical response data; 2) we did not detect some other important biomarkers, for example, circulating tumor cell, due to the shortage of research fund; 3) some studies showed postoperative detection was the better choice for prediction of the patients, we only detected the preoperative sample, and it is still a question for preoperative or postoperative detection; 4) cancer-elicited inflammation may vary according to its molecular subtype. However, we did not obtain the molecular characteristics, such as mutation of RAS, BRAF, and microsatellite instability status, in these patients. Thus, the prognostic roles of MLPAS and CCMLP in subgroups stratified by these molecular characteristics remain unclear.

In summary, MLPAS is a robust, independent, economical, and practical inflammatory indicators for patients with either negative or positive CEA or CA19-9. CCMLP is helpful in precise inflammatory classification and identifying a clinical focus and high-risk populations.

Data Sharing Statement

All data generated or analyzed during this study are included in this published article.

Acknowledgments

We thank all the patients for participating in the work.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This work was supported by the National Natural Science Foundation of China (grant number: 82360416 and 82460545), the Second Affiliated Hospital of Nanchang University Funding Program (2022efyA02).

Disclosure

The authors declare no conflicts of interest in this work.

References

1. Keck J, Gaedcke J, Ghadimi M, Lorf T. Surgical therapy in patients with colorectal liver metastases. Digestion. 2022;103(4):245–252. doi:10.1159/000524022

2. Ying HQ, Sun F, Liao YC, Cai D, Yang Y, Cheng XX. The value of circulating fibrinogen-to-pre-albumin ratio in predicting survival and benefit from chemotherapy in colorectal cancer. Therapeut Adv Med Oncol. 2021;13:17588359211022886. doi:10.1177/17588359211022886

3. Fakih M, Sandhu J, Wang C, et al. Evaluation of comparative surveillance strategies of circulating tumor DNA, imaging, and carcinoembryonic antigen levels in patients with resected colorectal cancer. JAMA Network Open. 2022;5(3):e221093. doi:10.1001/jamanetworkopen.2022.1093

4. Nicholson BD, Shinkins B, Pathiraja I, et al. Blood CEA levels for detecting recurrent colorectal cancer. Cochrane Database Syst Rev. 2015;2015(12):Cd011134. doi:10.1002/14651858.CD011134.pub2

5. Shen D, Wang X, Wang H, et al. Current surveillance after treatment is not sufficient for patients with rectal cancer with negative baseline CEA. J National Compr Cancer Network. 2022;20(6):653–662.e3. doi:10.6004/jnccn.2021.7101

6. Li Z, Zhu H, Pang X, et al. Preoperative serum CA19-9 should be routinely measured in the colorectal patients with preoperative normal serum CEA: a multicenter retrospective cohort study. BMC Cancer. 2022;22(1):962. doi:10.1186/s12885-022-10051-2

7. Li C, Zhao K, Zhang D, et al. Prediction models of colorectal cancer prognosis incorporating perioperative longitudinal serum tumor markers: a retrospective longitudinal cohort study. BMC Med. 2023;21(1):63. doi:10.1186/s12916-023-02773-2

8. Sun F, Peng HX, Gao QF, et al. Preoperative circulating FPR and CCF score are promising biomarkers for predicting clinical outcome of stage II-III colorectal cancer patients. Cancer Manage Res. 2018;10:2151–2161. doi:10.2147/CMAR.S167398

9. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discovery. 2022;12(1):31–46. doi:10.1158/2159-8290.CD-21-1059

10. Ying HQ, Chen W, Xiong CF, Wang Y, Li XJ, Cheng XX. Quantification of fibrinogen-to-pre-albumin ratio provides an integrating parameter for differential diagnosis and risk stratification of early-stage colorectal cancer. Cancer Cell Int. 2022;22(1):137. doi:10.1186/s12935-022-02532-y

11. Ying HQ, You XH, Liao YC, Sun F, Cheng XX. High-grade inflammation attenuates chemosensitivity and confers to poor survival of surgical stage III CRC patients. Front Oncol. 2021;11:580455. doi:10.3389/fonc.2021.580455

12. Chen QG, Zhang L, Sun F, et al. Elevated FPR confers to radiochemoresistance and predicts clinical efficacy and outcome of metastatic colorectal cancer patients. Aging. 2019;11(6):1716–1732. doi:10.18632/aging.101864

13. Ying HQ, Liao YC, Sun F, Peng HX, Cheng XX. The role of cancer-elicited inflammatory biomarkers in predicting early recurrence within stage II-III colorectal cancer patients after curable resection. J Inflamm Res. 2021;14:115–129. doi:10.2147/JIR.S285129

14. Zhou J, Wei W, Hou H, et al. Prognostic value of C-reactive protein, Glasgow prognostic score, and C-reactive protein-to-albumin ratio in colorectal cancer. Front Cell Dev Biol. 2021;9:637650. doi:10.3389/fcell.2021.637650

15. Li XY, Yao S, He YT, et al. Inflammation-immunity-nutrition score: a novel prognostic score for patients with resectable colorectal cancer. J Inflamm Res. 2021;14:4577–4588. doi:10.2147/JIR.S322260

16. Park SH, Woo HS, Hong IK, Park EJ. Impact of postoperative Naples prognostic score to predict survival in patients with stage II-III colorectal cancer. Cancers. 2023;15(20):5098. doi:10.3390/cancers15205098

17. Geßele C, Saller T, Smolka V, Dimitriadis K, Amann U, Strobach D. Development and validation of a new drug-focused predictive risk score for postoperative delirium in orthopaedic and trauma surgery patients. BMC Geriatr. 2024;24(1):422. doi:10.1186/s12877-024-05005-1

18. Hospital Authority of National Health and Family Planning Commission of the People′s Republic of China. Chinese protocol of diagnosis and treatment of colorectal cancer. Zhonghua Wai Ke Za Zhi. 2018;56(4):241–258. doi:10.3760/cma.j.issn.0529-5815.2018.E001

19. Flint TR, Janowitz T, Connell CM, et al. Tumor-induced IL-6 reprograms host metabolism to suppress anti-tumor immunity. Cell Metab. 2016;24(5):672–684. doi:10.1016/j.cmet.2016.10.010

20. Feng L, Wang X, Guo X, et al. Identification of novel target DCTPP1 for colorectal cancer therapy with the natural small-molecule inhibitors regulating metabolic reprogramming. Angew Chem. 2024;63(47):e202402543. doi:10.1002/anie.202402543

21. Cong J, Liu P, Han Z, et al. Bile acids modified by the intestinal microbiota promote colorectal cancer growth by suppressing CD8(+) T cell effector functions. Immunity. 2024;57(4):876–889.e11. doi:10.1016/j.immuni.2024.02.014

22. Ying HQ, Deng QW, He BS, et al. The prognostic value of preoperative NLR, d-NLR, PLR and LMR for predicting clinical outcome in surgical colorectal cancer patients. Med Oncol. 2014;31(12):305. doi:10.1007/s12032-014-0305-0

23. Li SQ, You XH, Sun F, et al. Albumin to fibrinogen ratio and fibrinogen to pre-albumin ratio are economical, simple and promising prognostic factors for solid malignancy. J Thoracic Dis. 2019;11(Suppl 15):S2036–S2038. doi:10.21037/jtd.2019.08.96

24. Xie H, Ruan G, Ge Y, et al. Inflammatory burden as a prognostic biomarker for cancer. Clin Nutr. 2022;41(6):1236–1243. doi:10.1016/j.clnu.2022.04.019

25. Wankhede D, Yuan T, Kloor M, Halama N, Brenner H, Hoffmeister M. Clinical significance of combined tumour-infiltrating lymphocytes and microsatellite instability status in colorectal cancer: a systematic review and network meta-analysis. Lancet Gastroenterol Hepatol. 2024;9(7):609–619. doi:10.1016/S2468-1253(24)00091-8

26. Nguyen DK, Kang MJ, Oh SJ, et al. Parvimonas micra-polarized M2-like tumor-associated macrophages accelerate colorectal cancer development via IL-8 secretion. Anim Cells Syst. 2025;29(1):24–34. doi:10.1080/19768354.2024.2442401

27. Li XM, Yang Y, Jiang FQ, et al. Histone lactylation inhibits RARγ expression in macrophages to promote colorectal tumorigenesis through activation of TRAF6-IL-6-STAT3 signaling. Cell Rep. 2024;43(2):113688. doi:10.1016/j.celrep.2024.113688

28. Almasaudi AS, Dolan RD, Edwards CA, McMillan DC. Hypoalbuminemia reflects nutritional risk, body composition and systemic inflammation and is independently associated with survival in patients with colorectal cancer. Cancers. 2020;12(7):1986. doi:10.3390/cancers12071986

29. Zhou Y, Tao L, Qiu J, et al. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther. 2024;9(1):132. doi:10.1038/s41392-024-01823-2

30. Islam MS, Gopalan V, Lam AK, Shiddiky MJA. Current advances in detecting genetic and epigenetic biomarkers of colorectal cancer. Biosens Bioelectron. 2023;239:115611.

31. Zhou H, Zhu L, Song J, et al. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. mol Cancer. 2022;21(1):86. doi:10.1186/s12943-022-01556-2

32. Raza A, Khan AQ, Inchakalody VP, et al. Dynamic liquid biopsy components as predictive and prognostic biomarkers in colorectal cancer. J Exp Clin Cancer Res. 2022;41(1):99. doi:10.1186/s13046-022-02318-0

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.