International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
As the malignancy with the third highest incidence rate and the second highest rate of cancer-related deaths globally, colorectal cancer (CRC) accounts for 1.2 million new cases and ~600,000 fatalities annually (1,2). Epidemiological modeling predicts a notable increase in disease burden, with 2.5 million new CRC diagnoses forecast globally by 2035 (3). The epidemiological landscape of China is particularly concerning, as the country exhibits the world's highest absolute CRC burden due to its population size. Recent surveillance data from the National Cancer Center suggests that the incidence of CRC has increased to the extent that the disease is now the second most commonly occurring malignancy in China (4). Despite advancements in early diagnosis and treatment methods, the long-term survival rate and prognostic outcomes of patients with CRC still face challenges (5). Accurate prognosis assessment is crucial for formulating individualized treatment plans, predicting survival duration and optimizing patient management.
In recent years, numerous studies have shown that inflammatory response, nutritional status and immune function play pivotal roles in cancer initiation, progression and outcomes (6–8). Therefore, identifying biomarkers that can comprehensively reflect a patient's inflammatory, nutritional and immune status holds significant importance for improving the prognostic assessment of patients with CRC. The preoperative hematological parameters of patients with cancer can reflect their inflammatory, immune and nutritional statuses. Thus, multiple inflammation indices derived from hematological examinations have been demonstrated to be closely associated with cancer prognosis. Key validated prognostic indicators include the neutrophil-to-lymphocyte ratio (NLR) (9), the platelet-to-lymphocyte ratio (10), the lymphocyte-to-monocyte ratio (11), the fibrinogen-to-albumin ratio (12), the derived NLR (dNLR) (13), the mean corpuscular volume-to-lymphocyte ratio (14), the systemic inflammation response index (15), the systemic immune-inflammation index (16), the prognostic nutritional index (PNI) (17), the cumulative inflammation index (18), the prognostic inflammatory and nutritional index (19), the hemoglobin, albumin, lymphocyte and platelet score (20) and pan-immune-inflammation values (21). Recently, the C-reactive protein (CRP)-albumin-lymphocyte (CALLY) index, an emerging immune-nutrition scoring system, has garnered increasing attention from researchers (22). The CALLY index, integrating CRP, albumin and lymphocyte levels, provides a comprehensive assessment of a patient's inflammatory, nutritional and immune status.
Multiple studies have confirmed that the CALLY index serves as an independent prognostic factor in patients with gastric cancer and that it can predict prognosis (23–25). Conversely, relatively limited evidence exists in the literature regarding the association between the CALLY index and prognosis in patients with CRC. Given this context, the present study was designed to evaluate the prognostic value of the CALLY index in patients with stage I–III CRC. Through retrospective analysis of clinicopathological data and preoperative hematological parameters of patients who underwent radical resection, the study sought to determine whether the CALLY index could serve as an independent predictor for both recurrence-free survival (RFS) and overall survival (OS) rates, thereby potentially offering a novel biomarker for prognostic assessment in CRC.
The present retrospective study analyzed clinicopathological data and preoperative laboratory hematological parameters (measured within 1 week before surgery) from patients with stage I–III CRC who underwent radical resection (R0) at Jingdezhen First People's Hospital (Jingdezhen, China) between January 2012 and March 2020. All consecutive patients meeting the eligibility criteria during this period were initially screened. The inclusion criteria were as follows: i) Histologically confirmed primary CRC; ii) no neoadjuvant therapy; and iii) R0 resection with curative intent. Exclusion criteria eliminated patients with: i) Synchronous/metachronous malignancies; ii) hematological disorders; iii) preoperative infection/immunodeficiency; iv) incomplete data; v) non-radical resection; and vi) receival of neoadjuvant therapy. All data were extracted from the hospital's maintained database.
The disease staging was determined using the eighth edition of the American Joint Committee on Cancer Tumor-Node-Metastasis (TNM) classification (26). Comorbidities was defined as pre-existing comorbidities, including cardiovascular diseases, pulmonary diseases, diabetes mellitus, chronic kidney disease and chronic liver disease. Postoperative anastomotic leakage specifically referred to anastomotic leakage occurring within 30 days after surgery. Postoperative adjuvant therapy primarily comprised chemotherapy, radiotherapy and other treatment modalities based on these core therapeutic approaches. Patients with stage II or III CRC received postoperative adjuvant therapy when deemed clinically appropriate based on their overall health status. Postoperative surveillance included contrast-enhanced computed tomography scans performed at minimum every 6 months and blood tests conducted every 3 months. Patients were followed up regularly through outpatient visits or telephone interviews every 3 months beginning on postoperative day 1 until the study endpoint, defined as either patient death or March 31, 2025, whichever occurred first. For outcome assessment, RFS time was calculated as the time from surgery to CRC recurrence, last follow-up or death, while OS time was defined as the time from surgery to death from any cause or last follow-up for surviving patients.
All calculations for inflammatory markers are presented in Table I, with the CALLY index calculated as follows: Albumin (g/dl) × lymphocyte count (n/µl)/[CRP (mg/dl) ×104] (22).
All statistical analyses were performed using R software (version 4.3.3; R Foundation for Statistical Computing). The following R packages were employed: pROC (version 1.18.5) for receiver operating characteristic (ROC) curve analysis, with the area under the curve (AUC) calculated to determine the optimal cutoff value using the Youden index; survival (version 3.6.4) and survminer (version 0.4.9) for survival analyses; and rstatix (version 0.7.2) for statistical testing. Based on the optimal cutoff, the patients were stratified into high-CALLY and low-CALLY groups. Continuous variables with normal distribution are expressed as mean ± standard deviation (SD) and compared using independent samples t-tests, while non-normally distributed continuous variables are presented as median (Q1-Q3) and analyzed using Wilcoxon rank-sum tests. Categorical variables are reported as n (%) and were compared using χ2 tests or Fisher's exact tests, as appropriate. All tests were two-sided, with P<0.05 considered to indicate a statistically significant difference. Univariate and multivariate Cox proportional hazards regression models were constructed using the survival package to estimate hazard ratios (HRs) with 95% confidence intervals (CIs) for RFS and OS. Variables with values of P<0.05 upon univariate analysis were included in the multivariate model. Survival probabilities were estimated using Kaplan-Meier curves, with between-group differences assessed by log-rank tests.
The present study was conducted in accordance with the Declaration of Helsinki and approved by the Research Ethics Committee at Jingdezhen First People's Hospital (approval no. jdzyykt202514). The requirement for informed consent was waived due to the retrospective nature of the study and data anonymization.
The present study consecutively screened 309 patients with stage I–III CRC undergoing radical resection (Fig. 1). After applying exclusion criteria (n=42) and accounting for loss to follow-up (n=12), the final cohort comprised 255 patients. ROC curve analysis evaluated the prognostic performance of 14 inflammatory markers for clinical outcomes in stage I–III CRC. The CALLY index demonstrated superior discriminative ability, with an AUC of 0.739 (95% CI, 0.702–0.773), significantly outperforming all other biomarkers (Fig. 2). Stratified by the CALLY cutoff of 6.790 (Table I), the cohort comprised 160 low-CALLY (<6.790) and 95 high-CALLY (≥6.790) patients (Table II). The low-CALLY group was associated with more aggressive tumor biology, characterized by higher rates of poorly differentiated histology (26.25 vs. 6.32%; P<0.001), advanced T4 stage (71.25 vs. 49.47%; P<0.001), nodal metastasis (N1-2: 74.38 vs. 16.84%; P<0.001), and stage III disease (73.12 vs. 16.84%; P<0.001). Clinically, these patients more frequently required adjuvant therapy (75.62 vs. 45.26%; P<0.001), alongside significantly worse RFS (38.33±19.03 vs. 52.93±15.57 months; P<0.001) and OS (44.01±16.22 vs. 54.26±13.46 months; P<0.001) times that were visually substantiated by pronounced separation in Kaplan-Meier curves (Figs. 3 and 4). Notably, elevated CA125 (23.08 vs. 10.14%; P=0.005) and CA19-9 (24.79 vs. 14.49%; P=0.038) were more prevalent in the high-CALLY group despite its less advanced pathology. No significant intergroup differences existed in terms of age, sex, tumor location, surgical approach, blood loss, comorbidities, anastomotic leakage and tumour size P>0.05), while the median (Q1-Q3) CALLY values robustly distinguished the cohorts [low-CALLY, 2.62 (1.62–4.64) vs. high-CALLY, 12.97 (9.88–17.16); P<0.001]. Collectively, low CALLY status is associated with advanced disease, intensified treatment needs and inferior survival, graphically validated by stratified survival analyses, positioning it as a significant prognostic integrator of inflammatory-nutritional imbalance in CRC.
Stratification based on the CALLY index revealed significant survival disparities among the patients with CRC. Kaplan-Meier analysis showed that the high-CALLY group (≥6.790) had superior RFS (79.6 vs. 32.1%; log-rank P<0.001; Fig. 3) and OS (80.0 vs. 38.8%; log-rank P<0.001; Fig. 4) rates compared with the low-CALLY group (<6.790).
Univariate and multivariate Cox regression analyses established CALLY as an independent predictor of RFS in CRC (Table III). In the univariate analysis, high CALLY exhibited a strong association with improved RFS (HR, 0.22; 95% CI, 0.14–0.35; P<0.001), outperforming conventional biomarkers including CA19-9 (HR, 1.57; 95% CI, 1.04–2.38; P=0.032), CA125 (HR, 2.05; 95% CI, 1.36–3.07; P<0.001) and CEA (HR, 1.45; 95% CI, 1.01–2.06; P=0.041). After adjusting for clinicopathological confounders in multivariate analysis, CALLY retained independent significance (HR, 0.56; 95% CI, 0.35–0.90; P=0.016), while among the other factors, only nodal metastasis (N1-2 vs. N0: HR, 6.71; 95% CI, 4.17–10.81; P<0.001), poor differentiation (moderate/poor vs. well: HR, 2.79; 95% CI, 1.88–4.16; P<0.001), advanced TNM stage (I vs. II–III: HR 2.59, 1.03–6.51; P=0.043) and elevated CA19-9 (>30 vs. ≤30 U/ml: HR, 1.69; 95% CI, 1.06–2.71; P=0.028) remained significant. Notably, tumor size (P=0.114), blood loss (P=0.100), CEA (0.213), CA125 (0.755), advanced T stage (P=0.056) and adjuvant therapy (P=0.082) lost statistical significance after adjustment.
Table III.Univariate and multivariate analysis for recurrence-free survival (RFS) in patients with colorectal cancer patients. |
Univariate and multivariate Cox regression analyses established CALLY as an independent predictor of OS in CRC (Table IV). Upon univariate analysis, high CALLY exhibited a profound protective effect on OS, with an HR of 0.26 (95% CI, 0.16–0.43; P<0.001), outperforming all other variables including nodal metastasis (HR, 7.95; 95% CI, 4.79–13.18; P<0.001), poor differentiation (HR, 3.69; 95% CI, 2.50–5.45; P<0.001) and elevated CA19-9 (HR, 1.74; 95% CI, 1.14–2.65; P=0.010). Following multivariate adjustment for clinicopathological confounders, CALLY retained robust independent significance (HR, 0.47; 95% CI, 0.27–0.82; P=0.008), while nodal metastasis (HR, 5.41; 95% CI, 2.93–9.98; P<0.001), poor differentiation (HR, 2.42; 95% CI, 1.54–3.81; P<0.001) and elevated CA19-9 (HR, 1.61; 95% CI, 1.02–2.57; P=0.043) remained significant, alongside advanced TNM stage (HR, 1.74; 95% CI, 1.03–2.95; P=0.040). Conventional biomarkers (CEA and CA125), anatomical factors (T stage), and treatment variables (adjuvant therapy) lost statistical significance (P>0.05) after adjustment, along with tumor size (P=0.121) and intraoperative blood loss (P=0.154), which were significant in the univariate analysis.
Table IV.Univariate and multivariate analysis for overall survival in patients with colorectal cancer. |
The CALLY index derives its prognostic power from quantifying a pathophysiological triad that orchestrates CRC progression through tumor microenvironment (TME)-specific mechanisms. Elevated CRP levels reflect activation of interleukin-6/Janus kinase/signal transducer and activator of transcription 3 signaling, which expands myeloid-derived suppressor cells (MDSCs) that spatially exclude cytotoxic T lymphocytes from tumor nests, a hallmark of ‘immune-excluded’ CRC subtypes (27,28). Concurrently, hypoalbuminemia disrupts gut barrier integrity, permitting translocation of procarcinogenic microbiota (for example, Fusobacterium nucleatum) that activate transforming growth factor-β signaling (29,30). This further amplifies MDSC-mediated immunosuppression by inducing regulatory T cell differentiation and programmed death-ligand 1 (PD-L1) upregulation on tumor-associated macrophages (31). Lymphopenia completes this vicious cycle by depleting CD103+ tissue-resident memory T cells critical for controlling microsatellite-stable CRC (32). Collectively, these processes establish an immunosuppressive TME favoring metastasis.
In gastric cancer research, Hashimoto et al (33) demonstrated that the high-CALLY group (cut-off value: 3.28) exhibited a significantly higher proportion of early-stage cases (stage I: 71.5%; P=0.019) and lower venous invasion rates compared with the low-CALLY group. These findings align closely with the present study, where the high-CALLY group (≥6.790) displayed superior tumor biological characteristics, including significantly reduced rates of poor differentiation (6.32 vs. 26.25%), T4 invasion (49.47 vs. 71.25%), lymph node metastasis (N1-2 stage: 16.84 vs. 74.38%) and stage III disease (16.84 vs. 73.12%) (all P<0.001). Paradoxically, despite the favorable prognosis, the high-CALLY group showed elevated levels of CA125 (23.08 vs. 10.14%) and CA19-9 (24.79 vs. 14.49%). This apparent contradiction may be explained by enhanced immunoediting mechanisms, such as intact lymphocyte function, mediated through antibody-dependent cellular cytotoxicity, which efficiently eliminates antigen-expressing tumor cells, leading to the release of tumor-associated antigens (e.g., CA125/CA19-9) into the bloodstream (34,35). Single-cell sequencing further reveals that specific T-cell subsets (for example, tissue-resident memory T cells) may modulate biomarker release dynamics by regulating immune checkpoint molecules such as PD-L1 (36). Future studies should explore the combined prognostic value of serial CALLY measurements and tumor markers.
The prognostic role of the CALLY index in advanced CRC has been established. Furukawa et al (37) demonstrated its superiority in metastatic settings, showing a 2.8-fold increased mortality risk in patients with colorectal liver metastasis (95% CI, 1.6–4.9; P<0.001) compared with conventional NLR/PNI biomarkers. The present study extends these findings to stage I–III CRC through three key analytical approaches. First, in a head-to-head comparison of 14 prognostic markers, the CALLY index achieved the highest discriminative power (AUC, 0.739). Second, multivariable Cox models confirmed its independence from TNM stage and age (OS: HR, 2.15; 95% CI, 1.16–3.98; P=0.015; and RFS: HR, 2.34; 95% CI, 1.32–4.15; P=0.003). Most notably, Kaplan-Meier analysis revealed significant survival disparities, with low-CALLY patients exhibiting 23.8 and 31.6% absolute reductions in 5-year OS (58.3 vs. 82.1%; P=0.008) and RFS (45.2 vs. 76.8%; P<0.001), respectively. This tripartite validation, spanning ROC performance, regression stability and survival curve divergence, solidifies the CALLY index as a pan-stage prognostic tool.
The present study has several limitations that warrant consideration. First, the single-center retrospective design and relatively limited sample size may introduce selection bias, highlighting the need for future multicenter studies with larger cohorts to validate the findings. Second, the proposed CALLY cutoff value was derived from a single institutional dataset, necessitating external validation through collaborative multicenter research to confirm its generalizability across diverse populations. Specifically, the lack of an independent external cohort for validating the optimal cutoff (6.790) and prognostic performance limits the immediate clinical translatability of the findings. Future studies should prioritize multi-institutional collaboration to establish population-adjusted thresholds. Third, the exclusive reliance on single-timepoint preoperative measurements precluded assessment of dynamic changes in CALLY values, suggesting that prospective studies incorporating serial measurements would provide more comprehensive insights into its clinical utility. Fourth, the absence of key prognostic confounders in the analysis, including molecular subtypes (RAS/BRAF mutation status), microsatellite instability status, perioperative nutritional support and postoperative complications, may have influenced survival outcomes independent of the CALLY index. Finally, the analysis did not incorporate circulating tumor DNA or other molecular residual disease markers, which could potentially miss early micrometastatic signals.
The present study establishes the CALLY index as a simple yet effective prognostic marker for patients with stage I–III CRC after radical resection. Key findings demonstrate its ability to: i) Independently predict RFS and OS rates; ii) stratify patients by tumor aggressiveness; and iii) complement traditional TNM staging. As a composite of routine blood parameters, this multifaceted marker, integrating inflammatory, nutritional and immune indicators, enhances clinical relevance while offering cost-effectiveness and immediate clinical implementability. Although further validation is warranted, the CALLY index may guide personalized management by identifying high-risk patients requiring intensified surveillance, ultimately optimizing CRC therapeutic decisions.
Not applicable.
Funding: No funding was received.
The data generated in the present study may be requested from the corresponding author.
JL and SZ were responsible for the study conceptualization and methodology. JL and XH were responsible for visualization, validation, investigation and data curation. JL provided resources and wrote the original draft manuscript. SZ helped to review and edit the manuscript, and supervised the study. JL, XH and SZ were responsible for project administration. JL and SZ confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.
The present study was conducted in accordance with the Declaration of Helsinki and was approved by the Research Ethics Committee at The Jingdezhen First People's Hospital (Jingdezhen, China; approval no. jdzyykt202514). The requirement for informed consent was waived due to the retrospective nature of the study and data anonymization.
Not applicable.
The authors declare that they have no competing interests.
|
Rezazadeh M, Agah S, Kamyabi A, Akbari A, Ghamkhari Pisheh R, Eshraghi A, Babakhani A, Ahmadi A, Paseban M, Heidari P, et al: Effect of diabetes mellitus type 2 and sulfonylurea on colorectal cancer development: A Case-control study. BMC Gastroenterol. 24:3822024. View Article : Google Scholar : PubMed/NCBI | |
|
Mozooni Z, Golestani N, Bahadorizadeh L, Yarmohammadi R, Jabalameli M and Amiri BS: The role of interferon-gamma and its receptors in gastrointestinal cancers. Pathol Res Pract. 248:1546362023. View Article : Google Scholar : PubMed/NCBI | |
|
Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global patterns and trends in colorectal cancer incidence and mortality. Gut. 66:683–691. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Han Z, Li X, Huang A, Shi J and Gu J: Epidemiology and risk factors of colorectal cancer in China. Chin J Cancer Res. 32:729–741. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Benson AB III, Venook AP, Cederquist L, Chan E, Chen YJ, Cooper HS, Deming D, Engstrom PF, Enzinger PC, Fichera A, et al: Colon cancer, version 1.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 15:370–398. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Burzinskis E, Janulaityte I, Jievaltas M, Skaudickas D, Burzinskiene G, Dainius E, Naudziunas A and Vitkauskiene A: Inflammatory markers in prostate cancer: Potential roles in risk stratification and immune profiling. J Immunotoxicol. 22:24977762025. View Article : Google Scholar : PubMed/NCBI | |
|
Tan J, Wu Z, Liu Y, Wang W, Qin W, Pan G, Xiong Y, Ma J, Zhao J, Zhou H, et al: Transcriptional profiling reveals H.pylori-associated genes induced inflammatory cell infiltration and chemoresistance in gastric cancer. Front Immunol. 16:15925582025. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan S, Zhu L, Chen X and Lin Q: Huanglian Jiedu Tang regulates the inflammatory microenvironment to alleviate the progression of breast cancer by inhibiting the RhoA/ROCK pathway. Tissue Cell. 95:1028502025. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JY, Jung EJ, Kim JM, Lee HS, Kwag SJ, Park JH, Park T, Jeong SH, Jeong CY and Ju YT: Dynamic changes of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio predicts breast cancer prognosis. BMC Cancer. 20:12062020. View Article : Google Scholar : PubMed/NCBI | |
|
Huang XZ, Chen WJ, Zhang X, Wu CC, Zhang CY, Sun SS and Wu J: An Elevated Platelet-to-Lymphocyte ratio predicts poor prognosis and clinicopathological characteristics in patients with colorectal cancer: A Meta-analysis. Dis Markers. 2017:10531252017. View Article : Google Scholar : PubMed/NCBI | |
|
Nøst TH, Alcala K, Urbarova I, Byrne KS, Guida F, Sandanger TM and Johansson M: Systemic inflammation markers and cancer incidence in the UK Biobank. Eur J Epidemiol. 36:841–848. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Wu Q, Kong Y and Lu C: Mild cognitive impairment in type 2 diabetes is associated with fibrinogen-to-albumin ratios. PeerJ. 11:e158262023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu X, Sun T, Zhang X, Wang W, Ji Y and Jing J: The predicting role of albumin to derived neutrophil-to-lymphocyte ratio for the prognosis of esophageal squamous cell carcinoma patients aged 60 years and above. BMC Cancer. 25:6522025. View Article : Google Scholar : PubMed/NCBI | |
|
Poenariu IS, Boldeanu L, Ungureanu BS, Caragea DC, Cristea OM, Pădureanu V, Siloși I, Ungureanu AM, Statie RC, Ciobanu AE, et al: Interrelation of Hypoxia-inducible factor-1 Alpha (HIF-1 α) and the ratio between the mean corpuscular Volume/Lymphocytes (MCVL) and the cumulative inflammatory index (IIC) in ulcerative colitis. Biomedicines. 11:31372023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang J, Gong L, Zhu X, Wang Y and Li C: Mediation of systemic inflammation response index in the association of healthy eating Index-2020 in patientis with periodontitis. Oral Health Prev Dent. 23:225–232. 2025.PubMed/NCBI | |
|
Wang Y, Lyu J, Jia H, Liang L, Xiao L, Liu Y, Liu X, Li K, Chen T, Zhang R, et al: Clinical utility of the systemic immune-inflammation index for predicting survival in esophageal squamous cell carcinoma after radical radiotherapy. Future Oncol. 17:2647–2657. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Sun W, Fu S, Wang J, Jin B, Zhang S, Liu Y, Zhang Q and Wang H: Prognostic value of the preoperative prognostic nutritional and systemic immunoinflammatory indexes in patients with colorectal cancer. BMC Cancer. 25:4032025. View Article : Google Scholar : PubMed/NCBI | |
|
Șerban RE, Popescu DM, Boldeanu MV, Florescu DN, Șerbănescu MS, Șandru V, Panaitescu-Damian A, Forțofoiu D, Șerban RC, Gherghina FL and Vere CC: The diagnostic and prognostic role of inflammatory markers, including the new cumulative inflammatory index (IIC) and mean corpuscular volume/lymphocyte (MCVL), in colorectal adenocarcinoma. Cancers (Basel). 17:9902025. View Article : Google Scholar : PubMed/NCBI | |
|
Xie H, Wei L, Liu M, Liang Y, Yuan G, Gao S, Wang Q, Lin X, Tang S and Gan J: Prognostic significance of preoperative prognostic immune and nutritional index in patients with stage I–III colorectal cancer. BMC Cancer. 22:13162022. View Article : Google Scholar : PubMed/NCBI | |
|
Dong J, Jiang W, Zhang W, Guo T, Yang Y, Jiang X, Zheng L and Du T: Exploring the J-shaped relationship between HALP score and mortality in cancer patients: A NHANES 1999–2018 cohort study. Front Oncol. 14:13886102024. View Article : Google Scholar : PubMed/NCBI | |
|
Li K, Zeng X, Zhang Z, Wang K, Pan Y, Wu Z, Chen Y and Zhao Z: Pan-immune-inflammatory values predict survival in patients after radical surgery for non-metastatic colorectal cancer: A retrospective study. Oncol Lett. 29:1972025. View Article : Google Scholar : PubMed/NCBI | |
|
Iida H, Tani M, Komeda K, Nomi T, Matsushima H, Tanaka S, Ueno M, Nakai T, Maehira H, Mori H, et al: Superiority of CRP-albumin-lymphocyte index (CALLY index) as a non-invasive prognostic biomarker after hepatectomy for hepatocellular carcinoma. HPB (Oxford). 24:101–115. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Nakashima K, Haruki K, Kamada T, Takahashi J, Tsunematsu M, Ohdaira H, Furukawa K, Suzuki Y and Ikegami T: Usefulness of the C-reactive protein (CRP)-Albumin-lymphocyte (CALLY) index as a prognostic indicator for patients with gastric cancer. Am Surg. 90:2703–2709. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Fukushima N, Masuda T, Tsuboi K, Takahashi K, Yuda M, Fujisaki M, Ikegami T, Yano F and Eto K: Prognostic significance of the preoperative C-reactive protein-albumin-lymphocyte (CALLY) index on outcomes after gastrectomy for gastric cancer. Surg Today. 54:943–952. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Toda M, Musha H, Suzuki T, Nomura T and Motoi F: Impact of C-reactive protein-albumin-lymphocyte index as a prognostic marker for the patients with undergoing gastric cancer surgery. Front Nutr. 12:15560622025. View Article : Google Scholar : PubMed/NCBI | |
|
Amin MB, Edge SB and Greene FL: AJCC Cancer Staging Manual. 8th edition. New York: Springer; 2016 | |
|
Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, Scheller J, Rose-John S, Cheroutre H, Eckmann L and Karin M: IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 15:103–113. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, et al: Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 24:541–550. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, Nowak JA, Yang J, Dou R, Masugi Y, Song M, et al: Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 65:1973–1980. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Chen WD, Zhang X, Zhang YP, Yue CB, Wang YL, Pan HW, Zhang YL, Liu H and Zhang Y: Fusobacterium nucleatum is a risk factor for metastatic colorectal cancer. Curr Med Sci. 42:538–547. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, Sevillano M, Ibiza S, Cañellas A, Hernando-Momblona X, et al: TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. 554:538–543. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Huang A, Wu Y, Cui J, Cai M, Xie Y, Zou J, Alenzi M, Yang H, Huang P and Huang M: Assessment of compartment-specific CD103-positive cells for prognosis prediction of colorectal cancer. Cancer Immunol Immunother. 74:2372025. View Article : Google Scholar : PubMed/NCBI | |
|
Hashimoto I, Tanabe M, Onuma S, Morita J, Nagasawa S, Maezawa Y, Kanematsu K, Aoyama T, Yamada T, Yukawa N, et al: Clinical impact of the C-reactive Protein-albumin-lymphocyte index in Post-gastrectomy patients with gastric cancer. In Vivo. 38:911–916. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Dunn GP, Bruce AT, Ikeda H, Old LJ and Schreiber RD: Cancer immunoediting: From immunosurveillance to tumor escape. Nat Immunol. 3:991–998. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Łuksza M, Sethna ZM, Rojas LA, Lihm J, Bravi B, Elhanati Y, Soares K, Amisaki M and Dobrin A: Neoantigen quality predicts immunoediting in survivors of pancreatic cancer. Nature. 606:389–395. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Reina-Campos M, Heeg M, Kennewick K, Mathews IT, Galletti G, Luna V, Nguyen Q, Huang H, Milner JJ, Hu KH, et al: Metabolic programs of T cell tissue residency empower tumour immunity. Nature. 621:179–187. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Furukawa S, Hiraki M, Kimura N, Sakai M, Ikubo A and Samejima R: The potential of the C-reactive protein-albumin-lymphocyte (CALLY) index as a prognostic biomarker in colorectal cancer. Cancer Diagn Progn. 5:370–377. 2025. View Article : Google Scholar : PubMed/NCBI |