Indexed In : Science Citation Index Expanded(SCIE), MEDLINE,
Pubmed/Pubmed Central, Elsevier Bibliographic, Google Scholar,
Databases(Scopus & Embase), KCI, KoreaMed, DOAJ
Gut and Liver is an international journal of gastroenterology, focusing on the gastrointestinal tract, liver, biliary tree, pancreas, motility, and neurogastroenterology. Gut atnd Liver delivers up-to-date, authoritative papers on both clinical and research-based topics in gastroenterology. The Journal publishes original articles, case reports, brief communications, letters to the editor and invited review articles in the field of gastroenterology. The Journal is operated by internationally renowned editorial boards and designed to provide a global opportunity to promote academic developments in the field of gastroenterology and hepatology. +MORE
Yong Chan Lee |
Professor of Medicine Director, Gastrointestinal Research Laboratory Veterans Affairs Medical Center, Univ. California San Francisco San Francisco, USA |
Jong Pil Im | Seoul National University College of Medicine, Seoul, Korea |
Robert S. Bresalier | University of Texas M. D. Anderson Cancer Center, Houston, USA |
Steven H. Itzkowitz | Mount Sinai Medical Center, NY, USA |
All papers submitted to Gut and Liver are reviewed by the editorial team before being sent out for an external peer review to rule out papers that have low priority, insufficient originality, scientific flaws, or the absence of a message of importance to the readers of the Journal. A decision about these papers will usually be made within two or three weeks.
The remaining articles are usually sent to two reviewers. It would be very helpful if you could suggest a selection of reviewers and include their contact details. We may not always use the reviewers you recommend, but suggesting reviewers will make our reviewer database much richer; in the end, everyone will benefit. We reserve the right to return manuscripts in which no reviewers are suggested.
The final responsibility for the decision to accept or reject lies with the editors. In many cases, papers may be rejected despite favorable reviews because of editorial policy or a lack of space. The editor retains the right to determine publication priorities, the style of the paper, and to request, if necessary, that the material submitted be shortened for publication.
Dan Li1,2 , Can Liu3
, Renwang Hu1,2
Correspondence to: Can Liu
ORCID https://orcid.org/0009-0007-3289-6920
E-mail canliu520@sina.com
Renwang Hu
ORCID https://orcid.org/0009-0009-8569-1122
E-mail huhu1566@sina.com
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.
Gut Liver.
Published online March 5, 2025
Copyright © Gut and Liver.
Background/Aims: To construct a new model based on folate receptor-positive circulating tumor cells (FR+-CTC) for the preoperative prediction of peritoneal metastasis in gastrointestinal malignancies and to apply this model in clinical practice.
Methods: Patients with gastrointestinal malignancies who had undergone preoperative FR+-CTC counts were retrospectively collected. Risk factors for peritoneal metastasis in patients with gastrointestinal malignancies were identified using a logistic regression model. The “pROC” package in R software was employed to plot the receiver operating characteristic curve for predicting peritoneal metastasis in these patients based on identified risk factors. Spearman correlation analysis was performed to assess the relationship between FR+-CTC counts and risk factors.
Results: A total of 396 patients meeting the inclusion criteria were finally included in the study. The number of FR+-CTC, albumin level, total protein level, and cancer antigen 125 (CA-125) level were identified as risk factors affecting peritoneal metastasis in gastrointestinal malignancies. The number of FR+-CTC was significantly negatively correlated with albumin (R=–0.21, p<0.001), and total protein levels (R=–0.10, p=0.047), and a positively correlated with CA-125 level (R=0.15, p=0.004). The number of FR+-CTCs was significantly higher in patients with peritoneal metastasis, lymph node metastasis, vascular invasion, neural invasion, and in those with stage T3-4 and III-IV gastrointestinal malignancies (p<0.05 for all). The model demonstrated stable predictive capacity, as validated through 10-fold cross-validation.
Conclusions: FR+-CTCs can serve as a novel biomarker for gastrointestinal malignancies. A new model based on FR+-CTCs demonstrated strong predictive capabilities for the preoperative assessment of peritoneal metastasis in gastrointestinal cancers.
Keywords: Stomach neoplasms, Colorectal neoplasms, Folate receptor-positive circulating tumor cells, Peritoneal metastasis, Gastrointestinal malignancies
Gastrointestinal malignancies are among the most common cancers globally,1,2 with their prognosis often hinging on the occurrence of peritoneal metastasis. Once peritoneal metastasis occurs, there is a significant reduction in patient survival rates.3-5 Accurately predicting the likelihood of peritoneal metastasis preoperatively is therefore crucial in developing personalized treatment plans. Circulating tumor cells (CTC) have gained increasing attention in cancer research in recent years.6-9 They are tumor cells that have detached from the primary tumor or metastatic sites and entered the bloodstream. Considered as a form of “real-time liquid biopsy,” CTC are seen as a powerful tool for predicting cancer progression and response to treatment. However, the detection and identification of CTC remain challenging due to their extreme rarity in the blood and high heterogeneity. Folate receptor (FR), a cell surface high-affinity folate-binding protein, is overexpressed in many types of tumors, including ovarian, breast, and lung cancers.10-12 The presence of FR-positive CTC (FR+-CTC) in gastrointestinal malignancies has been confirmed, with studies indicating that the quantity of FR+-CTC is closely associated with the clinical-pathological characteristics and prognosis of tumors.13-15 Compared to other biomarkers, such as EpCAM+ CTC or traditional tumor markers like carcinoembryonic antigen, cancer antigen 19-9, and cancer antigen 125 (CA-125), FR+-CTC demonstrate unique advantages in gastrointestinal malignancies due to their higher specificity and relevance in tumor aggressiveness and metastasis.16-18 For instance, while EpCAM-based detection is commonly employed, it has limitations in capturing mesenchymal-like CTC, which are critical in metastasis. FR+-CTC provide a broader detection spectrum that includes epithelial, mesenchymal, and hybrid phenotypes, offering a more comprehensive understanding of metastatic potential. To ensure the accuracy and sensitivity of detecting FR+-CTC, rigorous controls were implemented in our immunofluorescence and flow cytometry techniques. These controls included the use of established positive and negative cell lines to validate antibody specificity, as well as spike-in experiments to evaluate the efficiency of CTC recovery. Additionally, strict gating strategies were applied to distinguish FR+-CTC from other circulating cells, minimizing the risk of false positives. However, up to now, there is still a relative scarcity of research on the role of FR+-CTC in predicting peritoneal metastasis in gastrointestinal cancers. Most existing studies are preliminary observational studies based on small samples, lacking large-scale validation and in-depth mechanistic research. Moreover, despite their predictive value, how to accurately integrate FR+-CTC into clinical decision-making remains an unresolved issue. In this context, we have designed a new predictive model based on the detection of FR+-CTC, aimed at preoperatively predicting the risk of peritoneal metastasis in gastrointestinal malignancies, thereby guiding clinical practice.
This retrospective study collected data from patients diagnosed with gastric and colorectal cancer through surgery at Henan Provincial People's Hospital from July 2021 to May 2023. Patients who underwent preoperative testing for FR+-CTC were selected. This study was approved by the Institutional Review Board of Henan Provincial People's Hospital (approval number: 202284), and it adhered to the principles of the 1975 Declaration of Helsinki and its subsequent amendments. Written informed consent was waived.
Inclusion criteria were: (1) patients confirmed to have gastric or colorectal cancer through preoperative gastroscopy or colonoscopy; (2) preoperative testing for FR+-CTC; (3) no other related antitumor treatment before surgery; and (4) patients underwent surgical treatment. Exclusion criteria included: (1) non-gastric or colorectal cancer patients; (2) patients who received related antitumor treatments preoperatively, such as radiotherapy, chemotherapy, neoadjuvant therapy, traditional Chinese medicine, etc.; (3) patients with a history of other malignancies; and (4) patients who did not undergo surgery.
This method involves using the FR as a marker to detect and quantify CTC from blood samples. Initially, preoperative blood samples are collected from patients for detecting FR+-CTC. These cells are then separated from the blood samples using techniques such as immunomagnetic separation. Subsequently, folate (folic acid) is used as a probe to detect and quantify the expression of FR in these separated cells using techniques like immunofluorescence staining and flow cytometry. To ensure the accuracy and sensitivity of detecting FR+-CTC, rigorous and well-defined controls were employed. Positive control cell lines with known FR expression were utilized to validate antibody specificity, while negative control cell lines lacking FR expression minimized the risk of nonspecific binding. Additionally, spike-in experiments were conducted by introducing known quantities of FR+ tumor cells into blood samples from healthy donors to evaluate CTC recovery efficiency and sensitivity. For flow cytometry, strict gating strategies were applied, leveraging fluorescence intensity, cell size, and morphological parameters to differentiate FR+-CTC from other circulating cells, such as leukocytes and platelets. Furthermore, appropriate isotype controls and repeated measurements ensured reproducibility and minimized false positives. These measures collectively enhanced the reliability of FR+-CTC detection.
The “caret” package in R software (R Foundation for Statistical Computing, Vienna, Austria) is used for random grouping of patients. All patients are randomly divided into a training group and a validation group in a 1:1 ratio.
The predictive model is constructed using a logistic regression model with the “glmnet” package in R software. The receiver operating characteristic (ROC) curve was plotted using the “pROC” package, and the area under the curve (AUC) is calculated to assess the predictive capability of the model. The model's validation is performed using a 10-fold cross-validation method, iterating 1,000 cycles to build predictive models and calculate the mean AUC values for validation. The accuracy of the predictive model is verified using the “riskRegression” and “prodlim” packages in R software to plot calibration curves.
Correlation analysis is performed using the Spearman correlation method, with the “ggplot2,” “ggpubr,” and “ggExtra” packages in R software used to plot correlation curves. Non-parametric testing (Wilcox test) is used to analyze statistical differences in FR+-CTC values and clinical relevant indicators among different gastrointestinal cancer patients, visualized using the “ggplot2” and “reshape2” packages in R software to create difference box plots. The chi-square test is used to compare baseline data between the training and validation groups. All analyses in this study are conducted in R software (version 4.0.3), with an alpha level of 0.05, considering p<0.05 as statistically significant.
A total of 396 patients meeting the criteria were included after screening. The baseline clinical characteristics, tumor pathological staging, levels of albumin, total protein, hemoglobin, FR+-CTC values, and tumor marker expressions of the included patients are summarized in Table 1.
Table 1. Baseline Clinical Information of Patients Included in This Study
Characteristic | Value (n=396) |
---|---|
Age, median (IQR), yr | 62 (55–71) |
Sex, No. (%) | |
Male | 250 (63.13) |
Female | 146 (36.87) |
Vessel invasion, No. (%) | |
No | 176 (44.44) |
Yes | 220 (55.56) |
Nerve invasion, No. (%) | |
No | 221 (55.81) |
Yes | 175 (44.19) |
T stage, No. (%) | |
T1 | 58 (14.66) |
T2 | 81 (20.45) |
T3 | 172 (43.43) |
T4 | 85 (21.46) |
N stage, No. (%) | |
N0 | 212 (53.53) |
N1 | 50 (12.63) |
N2 | 94 (23.74) |
N3 | 40 (10.10) |
M stage, No. (%) | |
M0 | 362 (91.41) |
M1 | 34 (8.59) |
Stage, No. (%) | |
I | 109 (27.52) |
II | 118 (29.80) |
III | 135 (34.09) |
IV | 34 (8.59) |
Tumor, No. (%) | |
Gastric cancer | 123 (31.06) |
Intestinal cancer | 273 (68.94) |
Peritoneal metastasis, No. (%) | |
No | 363 (91.67) |
Yes | 33 (8.33) |
Gastric cancer | 19 (57.58) |
Intestinal cancer | 14 (42.42) |
FR+-CTC, median (IQR), FU/3 mL | 10.80 (8.88–13.73) |
Albumin, median (IQR), g/L | 39.35 (34.98–41.10) |
Total protein, median (IQR), g/L | 66.75 (60.48–71.83) |
Hemoglobin, median (IQR), g/L | 122.0 (109.0–136.0) |
CEA, median (IQR), ng/mL | 1.85 (1.11–3.43) |
CA19-9, median (IQR), U/mL | 9.94 (5.83–17.54) |
CA-125, median (IQR), U/mL | 9.92 (6.66–15.32) |
IQR, interquartile range; FR+-CTC, folate receptor-positive circulating tumor cells; CEA, carcinoembryonic antigen; CA19-9, cancer antigen 19-9; CA-125, cancer antigen 125.
A logistic regression model was constructed using the “glmnet” package in R software, incorporating all risk factors into the model. As shown in Table 2, in this predictive model, FR+-CTC, albumin, total protein, and CA-125 were found to be statistically significant (p<0.05), while hemoglobin, cancer antigen 19-9, and carcinoembryonic antigen were not statistically significant (p>0.05). Therefore, the indicators ultimately included in the model are: FR+-CTC, albumin, total protein, and CA-125.
Table 2. Logistic Regression Analysis of Peritoneal Metastasis Variable in Patients with Gastrointestinal Malignancies
SE | Z-value | OR | 95% CI | p-value | |
---|---|---|---|---|---|
Intercept | 2.039 | –2.569 | 0.01 | 0.00–0.29 | 0.010 |
FR+-CTC | 0.026 | 4.553 | 1.13 | 1.07–1.19 | <0.001 |
Albumin | 0.060 | –3.127 | 0.83 | 0.74–0.93 | 0.002 |
Total protein | 0.036 | 2.729 | 1.10 | 1.03–1.18 | 0.006 |
Hemoglobin | 0.012 | 0.874 | 1.01 | 0.99–1.03 | 0.382 |
CA-125 | 0.008 | 2.335 | 1.02 | 1.00–1.04 | 0.019 |
CA19-9 | 0.002 | –0.128 | 1.00 | 0.98–1.01 | 0.899 |
CEA | 0.003 | –0.183 | 1.00 | 0.99–1.01 | 0.855 |
SE, standard error; OR, odds ratio; CI, confidence interval; FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125; CA19-9, cancer antigen 19-9; CEA, carcinoembryonic antigen.
The ROC curve was used to evaluate the accuracy of FR+-CTC and conventional clinical indicators in diagnosing peritoneal metastasis in patients with gastrointestinal malignancies. As shown in Fig. 1, the AUC values for diagnosing peritoneal metastasis with FR+-CTC, albumin, total protein, and CA-125 were 0.774, 0.686, 0.531, and 0.750, respectively. The optimal cutoff values for FR+-CTC, albumin, total protein, and CA-125 were 15.05 FU/3 mL, 37.45 g/L, 58.65 g/L, and 17.91 U/mL, respectively. This indicates that FR+-CTC had the highest predictive accuracy for peritoneal metastasis in patients with gastrointestinal malignancies.
Spearman correlation analysis was conducted to study the relationships between FR+-CTC and albumin, total protein, and CA-125 levels. As shown in Fig. 2A, there was a significant negative correlation between FR+-CTC and albumin levels (correlation coefficient R=–0.21, p<0.001). Fig. 2B showed a significant negative correlation between FR+-CTC and total protein levels (correlation coefficient R=–0.10, p=0.047). Fig. 2C indicated a significant positive correlation between FR+-CTC and CA-125 levels (correlation coefficient R=0.15, p=0.004). Although the FR+-CTC value has a certain correlation with albumin level, total protein level, and CA-125 level, the correlation coefficients are all relatively low.
As shown in Fig. 3A, patients with peritoneal metastasis had significantly higher levels of FR+-CTC and CA-125, and lower levels of albumin compared to those without peritoneal metastasis. Fig. 3B illustrated that patients with lymph node metastasis had significantly higher levels of FR+-CTC and CA-125, and lower albumin levels than those without lymph node metastasis. In Fig. 3C, patients with vascular invasion showed significantly higher FR+-CTC count and lower albumin levels than those without vascular invasion. Fig. 3D showed that patients with neural invasion had significantly higher levels of FR+-CTC and CA-125, and lower albumin levels than those without neural invasion. As per Fig. 3E, patients with T3-4 stage tumors had significantly higher FR+-CTC count compared to those in T1-2 stage. Fig. 3F indicated that patients with stage III-IV pathology had significantly higher levels of FR+-CTC and CA-125, and lower albumin levels compared to those in stage I-II. The detailed statistical data are shown in Table 3.
Table 3. Statistical Analysis of FR+-CTC Count, Albumin Level, Total Protein Level, and CA-125 Level in Various Types of Gastrointestinal Malignancies
Covariates | Type | FR+-CT, FU/3 mL | Albumin, g/L | Total protein, g/L | CA-125, U/mL | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
median (IQR) | p-value | median (IQR) | p-value | median (IQR) | p-value | median (IQR) | p-value | |||||
Peritoneal metastasis | PM (–) | 10.5 (8.8–13.3) | <0.001 | 39.9 (35.3–43.2) | <0.001 | 66.8 (60.6–71.9) | 0.551 | 9.05 (6.59–14.16) | <0.001 | |||
PM (+) | 18.8 (12.2–25.8) | 35.0 (31.1–38.4) | 65.6 (57.4–70.8) | 21.79 (9.67–78.92) | ||||||||
Lymph node metastasis | N (–) | 10.4 (8.9–12.7) | 0.028 | 40.5 (35.9–43.5) | 0.003 | 66.3 (60.8–71.9) | 0.726 | 8.76 (6.46–12.81) | <0.001 | |||
N (+) | 11.6 (8.8–15.5) | 38.0 (34.7–42.3) | 67.4 (60.0–71.8) | 10.43 (6.82–19.41) | ||||||||
Vascular invasion | Vessel (–) | 10.2 (8.8–12.5) | 0.005 | 40.5 (36.0–43.4) | 0.013 | 66.6 (61.4–71.5) | 0.906 | 9.09 (6.59–13.58) | 0.074 | |||
Vessel (+) | 11.6 (9.0–15.5) | 38.2 (34.4–42.9) | 67.0 (59.8–71.8) | 9.46 (6.69–17.58) | ||||||||
Neural invasion | Nerve (–) | 10.4 (8.8–12.7) | 0.039 | 40.3 (35.2–43.5) | 0.014 | 66.6 (60.6–72.0) | 0.892 | 9.05 (6.58–13.77) | 0.036 | |||
Nerve (+) | 11.2 (8.9–15.9) | 38.1 (34.9–42.3) | 67.1 (59.9–71.5) | 9.80 (6.79–18.77) | ||||||||
T stage | T1–2 | 10.2 (8.2–12.0) | <0.001 | 40.2 (35.2–43.3) | 0.536 | 66.2 (59.8–70.9) | 0.331 | 9.18 (6.58–13.97) | 0.290 | |||
T3–4 | 11.4 (9.1–15.3) | 39.2 (34.9–43.0) | 67.1 (60.8–71.9) | 9.26 (6.81–16.14) | ||||||||
Stage | Stage I–II | 10.3 (8.9–12.4) | 0.004 | 40.5 (35.6–43.6) | 0.005 | 66.2 (60.0–71.9) | 0.452 | 8.81 (6.55–13.65) | 0.005 | |||
Stage III–IV | 12.0 (8.8–16.1) | 38.0 (34.8–42.1) | 67.4 (60.8–71.8) | 10.0 (6.82–19.97) |
FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125; PM, peritoneal metastasis; lQR, interquartile range.
Patients were randomly divided into a trial group and a validation group in a 1:1 ratio. Detailed clinical information of patients in both groups is shown in Table 4. There were no significant differences between the training and validation groups in terms of age, sex, vascular invasion, neural invasion, T stage, N stage, M stage, stage, tumor location, and presence of peritoneal metastasis (p>0.05), indicating comparability between the two groups.
Table 4. Clinical Data of Patients Included in the Training and Validation Groups
Covariates | Type | Train set (n=198) | Validation set (n=198) | p-value |
---|---|---|---|---|
Age | ≤62 yr | 100 (50.51) | 102 (51.52) | 0.920 |
>62 yr | 98 (49.49) | 96 (48.48) | ||
Sex | Male | 124 (62.63) | 126 (63.64) | 0.917 |
Female | 74 (37.37) | 72 (36.36) | ||
Vessel invasion | No | 85 (42.93) | 91 (45.96) | 0.613 |
Yes | 113 (57.07) | 107 (54.04) | ||
Nerve invasion | No | 111 (56.06) | 110 (55.56) | 0.998 |
Yes | 87 (43.94) | 88 (44.44) | ||
T stage | T1 | 28 (14.14) | 30 (15.15) | 0.854 |
T2 | 39 (19.70) | 42 (21.21) | ||
T3 | 85 (42.93) | 87 (43.94) | ||
T4 | 46 (23.23) | 39 (19.70) | ||
N stage | N0 | 96 (48.48) | 116 (58.58) | 0.094 |
N1 | 30 (15.15) | 20 (10.10) | ||
N2 | 47 (23.74) | 47 (23.74) | ||
N3 | 25 (12.63) | 15 (7.58) | ||
M stage | M0 | 178 (89.90) | 184 (92.93) | 0.369 |
M1 | 20 (10.10) | 14 (7.07) | ||
Stage | I | 47 (23.74) | 62 (31.31) | 0.293 |
II | 59 (29.80) | 59 (29.80) | ||
III | 72 (36.36) | 63 (31.82) | ||
IV | 20 (10.10) | 14 (7.07) | ||
Tumor | Gastric cancer | 66 (33.33) | 57 (28.79) | 0.385 |
Intestinal cancer | 132 (66.67) | 141 (71.21) | ||
Peritoneal metastasis | No | 178 (89.90) | 185 (93.43) | 0.275 |
Yes | 20 (10.10) | 13 (6.57) |
Data are presented as number (%).
As shown in Fig. 4A-C, the AUC values for the training group, validation group, and the entire patient cohort were 0.813, 0.874, and 0.837, respectively, indicating that the model has a strong predictive capability. Fig. 4D illustrates a nomogram, which visualizes the predictive model, enhancing the ability to predict the probability of peritoneal metastasis in patients with gastrointestinal malignancies, thus applying this predictive model in clinical settings. By determining the scores for each indicator and calculating the total score, the probability of a patient having peritoneal metastasis can be estimated. The specific parameters of this predictive model, as shown in Table 5, indicate that indicators such as FR+-CTC, albumin, total protein, and CA-125 are statistically significant (p<0.05).
Table 5. Basic Parameters of the Predictive Model for Determining Peritoneal Metastasis in Patients with Gastrointestinal Malignancies
SE | Z-value | OR | 95% CI | p-value | |
---|---|---|---|---|---|
Intercept | 1.895 | –2.443 | 0.01 | 0.00–0.40 | 0.014 |
FR+-CTC | 0.026 | 4.593 | 1.13 | 1.07–1.18 | <0.001 |
Albumin | 0.057 | –3.036 | 0.84 | 0.75–0.94 | 0.002 |
Total protein | 0.036 | 2.780 | 1.10 | 1.03–1.18 | 0.005 |
CA-125 | 0.005 | 3.468 | 1.02 | 1.01–1.03 | 0.001 |
SE, standard error; OR, odds ratio; CI, confidence interval; FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125.
The accuracy of the predictive model was validated using a 10-fold cross-validation method, iterating 1,000 cycles to build predictive models and calculate the mean AUC values for validation. As depicted in Fig. 5A-C, the cross-validation AUC values for the training group, validation group, and the entire patient cohort were 0.813, 0.886, and 0.844, respectively. These AUC values, all greater than 0.8, indicate a stable predictive capability of the model. The calibration curve, shown in Fig. 5D, demonstrates that the red curve closely aligns with the 45° diagonal line, suggesting that the probability of peritoneal metastasis predicted by the model closely matches the actual occurrence of peritoneal metastasis in patients, confirming the model's strong predictive accuracy.
Gastrointestinal malignancies, such as gastric and colorectal cancers, are among the most common malignant tumors globally.19-22 The risk associated with these cancers stems from their high invasiveness and tendency to metastasize, especially to the peritoneum. Peritoneal metastasis is a common late-stage phenomenon in gastrointestinal malignancies, significantly impacting patient quality of life and overall survival rates.23,24 Traditional imaging methods have limitations in early detection of peritoneal metastasis, hence developing new, more sensitive, and specific methods for early prediction and diagnosis of peritoneal metastasis in gastrointestinal malignancies is of paramount importance.25-27 FR+-CTC have received extensive attention in recent cancer research.28-30 The FR is a glycoprotein associated with the cell membrane that efficiently mediates cellular uptake of folate, an indispensable element for cell growth and division. In certain types of tumor cells, the expression of the FR is significantly upregulated, making it a marker distinguishing tumor cells from normal cells. CTC are tumor cells that have shed from the primary tumor into the peripheral blood circulation. Due to their high specificity and ease of detection, FR+-CTC are recognized as powerful biomarkers for cancer diagnosis and prognosis. Studies have indicated that FR+-CTC are not only involved in tumor progression but also actively contribute to the metastatic cascade. Overexpression of FR has been linked to enhanced tumor cell proliferation, migration, invasion, and immune evasion, all of which are critical steps in metastatic development. Recent evidence suggests that FR+-CTC facilitates hematogenous dissemination by interacting with the microenvironment and promoting the establishment of pre-metastatic niches. For example, FR-mediated signaling pathways have been implicated in epithelial-mesenchymal transition, a process that endows tumor cells with increased motility and invasiveness, further enhancing their ability to metastasize.31-33 These findings provide mechanistic insights into the role of FR+-CTC in peritoneal metastasis, highlighting their potential as both a predictive biomarker and a therapeutic target. Furthermore, the unique biology of FR+-CTC, including their ability to survive in circulation and evade anoikis, underscores their clinical relevance in predicting metastatic risk and guiding treatment strategies. Recent studies suggest that detecting FR+-CTC can predict the presence and metastasis of tumors at asymptomatic or early stages.
In our study, we have developed a predictive model based on FR+-CTC for predicting peritoneal metastasis in gastrointestinal malignancies. The establishment of this model not only provides a new perspective for understanding and predicting the clinical progression of gastrointestinal malignancies but also may offer substantial support for clinical decision-making. Firstly, we found that FR+-CTC, albumin, total protein, and CA-125 are significant factors in predicting peritoneal metastasis in gastrointestinal malignancies. FR+-CTC shows a negative correlation with albumin and total protein, and a positive correlation with CA-125. The observed negative correlation between FR+-CTC and albumin or total protein may reflect the systemic impact of advanced malignancies. Hypoalbuminemia and decreased total protein, often observed in cancer patients, are markers of malnutrition, systemic inflammation, and poor prognosis. Systemic inflammation, characterized by elevated levels of pro-inflammatory cytokines such as interleukin-6 and tumor necrosis factor-α, suppresses hepatic albumin synthesis and enhances protein catabolism, weakening the host’s immune defenses. These systemic changes create a permissive environment for the survival and dissemination of CTC by allowing them to evade immune surveillance and establish metastatic colonies. Moreover, hypoalbuminemia may reduce antioxidant capacity, further enhancing the resilience of tumor cells in circulation against oxidative stress. The decline in albumin and total protein also reflects the metabolic reprogramming associated with cancer cachexia, wherein resources are diverted to sustain tumor growth and progression. Conversely, the positive correlation between FR+-CTC and CA-125 underscores CA-125’s role as a biomarker of tumor burden and peritoneal involvement. CA-125, secreted by mesothelial cells lining the peritoneum, is frequently elevated in response to peritoneal irritation or tumor invasion. Elevated CA-125 levels may reflect a pro-inflammatory state in the peritoneal cavity, driven by tumor cell-secreted cytokines and growth factors. This inflammatory environment could facilitate FR+-CTC adhesion and implantation by promoting extracellular matrix remodeling via matrix metalloproteinases.34-37 Additionally, CA-125 is involved in mediating cell adhesion and aggregation through its interactions with MUC16 and integrins, thereby supporting FR+-CTC driven metastatic cascades. These interactions between FR+-CTC and CA-125 highlight their complementary roles in local and systemic aspects of tumor progression and underscore their combined value as predictive biomarkers for peritoneal metastasis. These findings suggest that monitoring the dynamics of FR+-CTC in conjunction with albumin, total protein, and CA-125 could provide a more comprehensive assessment of tumor progression and metastatic potential. Previous studies have extensively researched the role of the FR in the growth and metastasis of tumor cells, with increased expression often correlating with poor prognosis in malignant tumor patients.38,39 Our study not only quantitatively confirms this observation but also finds that the expression of FR+-CTC is significantly related to the risk of the tumor peritoneal metastasis. This finding further strengthens the importance of FR+-CTC as a potential biomarker, potentially offering a more accurate, biology-based predictive tool. Secondly, our predictive model demonstrates high predictive accuracy. While the AUC values reported for our model exceed 0.8 in the training group, validation group, and across all patient groups, additional metrics provide a fuller picture of the model’s performance. Specifically, our model demonstrated sensitivity, specificity, and overall accuracy exceeding 85%. This balance between sensitivity and specificity ensures the model’s capacity to accurately identify patients at risk while minimizing false positives, offering a robust tool for clinical application. These supplementary metrics, alongside the high AUC value, emphasize the model’s reliability and practicality. This indicates that our model can effectively differentiate patients at risk of peritoneal metastasis and provide accurate predictive results for clinical decision-making. More importantly, the model's advantage lies in using clinical indicators available from routine examinations, making it more feasible for practical application. This model aids understanding and prediction of the progression of gastrointestinal malignancies and potentially improving treatment strategies for patients.
However, despite the high predictive accuracy and clinical value of our predictive model, we must acknowledge some limitations in our study. Firstly, this study is based on a patient group from a single center, which may introduce sample selection bias. Future studies should be conducted across multiple centers to enhance the generalizability of the results. Secondly, although our predictive model performs well in the training and validation groups, its applicability to gastrointestinal malignancies in other countries needs further validation. One key limitation of these validation groups is the lack of subgroup analyses to assess the model’s performance across diverse clinical and demographic patient populations. Without this, the robustness and consistency of the model in varied settings remain unclear. Future studies should address these gaps by including larger, multi-center cohorts that better represent different populations and clinical contexts. Such analyses would help determine whether the predictive performance observed in the current study holds across different healthcare systems and patient demographics. Moreover, incorporating external validation datasets could further confirm the model’s reliability and minimize potential biases. These steps are crucial to enhance the clinical applicability of our findings. Future research should focus on elucidating the mechanisms linking FR+-CTC, albumin, total protein, and CA-125. Investigating whether the inflammatory state or specific cytokine profiles mediate these relationships could uncover new therapeutic targets. Exploring the dynamic interplay between nutritional status, systemic inflammation, and tumor biology will also be critical. Furthermore, incorporating additional biomarkers such as circulating tumor DNA or exosomal proteins could complement FR+-CTC analysis, enhancing the predictive model’s robustness. These efforts may not only refine the model but also provide a foundation for personalized therapeutic strategies. Current cancer research is focusing on the discovery of more precise biomarkers and personalized treatment strategies. In this context, the discovery of FR+-CTC and its application in predicting tumor metastasis is of significant importance. Future research could explore combining FR+-CTC with other emerging biomarkers to enhance the accuracy and clinical applicability of the predictive model.40,41 Moreover, with the development of precision medicine and personalized treatment strategies, our model can be further refined to meet the specific needs of different patient groups.
In summary, this discovery provides new insights for the early diagnosis and treatment of gastrointestinal malignancies. Particularly in predicting peritoneal metastasis preoperatively, the detection of FR+-CTC may become an essential tool. The development of a preoperative predictive model based on FR+-CTC is a key step in translating these research findings into clinical practice. This model integrates patients' biomarker data and clinical characteristics, offering clinicians a quantitative assessment of the risk of peritoneal metastasis. This is crucial for formulating individualized treatment strategies, such as deciding on adjuvant chemotherapy and selecting surgical methods. We anticipate that future research will further expand and refine this model, enabling it to play a more significant role in the personalized treatment of cancer.
In conclusion, FR+-CTC can serve as a novel biomarker for gastrointestinal malignancies. A new model based on FR+-CTC demonstrates strong predictive capabilities for preoperative assessment of peritoneal metastasis in gastrointestinal cancers.
No potential conflict of interest relevant to this article was reported.
Study concept and design: R.H. Data acquisition: D.L. Data analysis and interpretation: D.L. Drafting of the manuscript: D.L. Critical revision of the manuscript for important intellectual content: C.L. Statistical analysis: C.L. dministrative, technical, or material support; study supervision: R.H. Approval of final manuscript: all authors.
The data of this study can be obtained from the corresponding author according to reasonable requirements.
Gut and Liver
Published online March 5, 2025
Copyright © Gut and Liver.
Dan Li1,2 , Can Liu3
, Renwang Hu1,2
1Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, Zhengzhou, China; 2Department of Gastrointestinal Surgery, Zhengzhou University People’s Hospital, Zhengzhou, China; 3Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, China
Correspondence to:Can Liu
ORCID https://orcid.org/0009-0007-3289-6920
E-mail canliu520@sina.com
Renwang Hu
ORCID https://orcid.org/0009-0009-8569-1122
E-mail huhu1566@sina.com
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.
Background/Aims: To construct a new model based on folate receptor-positive circulating tumor cells (FR+-CTC) for the preoperative prediction of peritoneal metastasis in gastrointestinal malignancies and to apply this model in clinical practice.
Methods: Patients with gastrointestinal malignancies who had undergone preoperative FR+-CTC counts were retrospectively collected. Risk factors for peritoneal metastasis in patients with gastrointestinal malignancies were identified using a logistic regression model. The “pROC” package in R software was employed to plot the receiver operating characteristic curve for predicting peritoneal metastasis in these patients based on identified risk factors. Spearman correlation analysis was performed to assess the relationship between FR+-CTC counts and risk factors.
Results: A total of 396 patients meeting the inclusion criteria were finally included in the study. The number of FR+-CTC, albumin level, total protein level, and cancer antigen 125 (CA-125) level were identified as risk factors affecting peritoneal metastasis in gastrointestinal malignancies. The number of FR+-CTC was significantly negatively correlated with albumin (R=–0.21, p<0.001), and total protein levels (R=–0.10, p=0.047), and a positively correlated with CA-125 level (R=0.15, p=0.004). The number of FR+-CTCs was significantly higher in patients with peritoneal metastasis, lymph node metastasis, vascular invasion, neural invasion, and in those with stage T3-4 and III-IV gastrointestinal malignancies (p<0.05 for all). The model demonstrated stable predictive capacity, as validated through 10-fold cross-validation.
Conclusions: FR+-CTCs can serve as a novel biomarker for gastrointestinal malignancies. A new model based on FR+-CTCs demonstrated strong predictive capabilities for the preoperative assessment of peritoneal metastasis in gastrointestinal cancers.
Keywords: Stomach neoplasms, Colorectal neoplasms, Folate receptor-positive circulating tumor cells, Peritoneal metastasis, Gastrointestinal malignancies
Gastrointestinal malignancies are among the most common cancers globally,1,2 with their prognosis often hinging on the occurrence of peritoneal metastasis. Once peritoneal metastasis occurs, there is a significant reduction in patient survival rates.3-5 Accurately predicting the likelihood of peritoneal metastasis preoperatively is therefore crucial in developing personalized treatment plans. Circulating tumor cells (CTC) have gained increasing attention in cancer research in recent years.6-9 They are tumor cells that have detached from the primary tumor or metastatic sites and entered the bloodstream. Considered as a form of “real-time liquid biopsy,” CTC are seen as a powerful tool for predicting cancer progression and response to treatment. However, the detection and identification of CTC remain challenging due to their extreme rarity in the blood and high heterogeneity. Folate receptor (FR), a cell surface high-affinity folate-binding protein, is overexpressed in many types of tumors, including ovarian, breast, and lung cancers.10-12 The presence of FR-positive CTC (FR+-CTC) in gastrointestinal malignancies has been confirmed, with studies indicating that the quantity of FR+-CTC is closely associated with the clinical-pathological characteristics and prognosis of tumors.13-15 Compared to other biomarkers, such as EpCAM+ CTC or traditional tumor markers like carcinoembryonic antigen, cancer antigen 19-9, and cancer antigen 125 (CA-125), FR+-CTC demonstrate unique advantages in gastrointestinal malignancies due to their higher specificity and relevance in tumor aggressiveness and metastasis.16-18 For instance, while EpCAM-based detection is commonly employed, it has limitations in capturing mesenchymal-like CTC, which are critical in metastasis. FR+-CTC provide a broader detection spectrum that includes epithelial, mesenchymal, and hybrid phenotypes, offering a more comprehensive understanding of metastatic potential. To ensure the accuracy and sensitivity of detecting FR+-CTC, rigorous controls were implemented in our immunofluorescence and flow cytometry techniques. These controls included the use of established positive and negative cell lines to validate antibody specificity, as well as spike-in experiments to evaluate the efficiency of CTC recovery. Additionally, strict gating strategies were applied to distinguish FR+-CTC from other circulating cells, minimizing the risk of false positives. However, up to now, there is still a relative scarcity of research on the role of FR+-CTC in predicting peritoneal metastasis in gastrointestinal cancers. Most existing studies are preliminary observational studies based on small samples, lacking large-scale validation and in-depth mechanistic research. Moreover, despite their predictive value, how to accurately integrate FR+-CTC into clinical decision-making remains an unresolved issue. In this context, we have designed a new predictive model based on the detection of FR+-CTC, aimed at preoperatively predicting the risk of peritoneal metastasis in gastrointestinal malignancies, thereby guiding clinical practice.
This retrospective study collected data from patients diagnosed with gastric and colorectal cancer through surgery at Henan Provincial People's Hospital from July 2021 to May 2023. Patients who underwent preoperative testing for FR+-CTC were selected. This study was approved by the Institutional Review Board of Henan Provincial People's Hospital (approval number: 202284), and it adhered to the principles of the 1975 Declaration of Helsinki and its subsequent amendments. Written informed consent was waived.
Inclusion criteria were: (1) patients confirmed to have gastric or colorectal cancer through preoperative gastroscopy or colonoscopy; (2) preoperative testing for FR+-CTC; (3) no other related antitumor treatment before surgery; and (4) patients underwent surgical treatment. Exclusion criteria included: (1) non-gastric or colorectal cancer patients; (2) patients who received related antitumor treatments preoperatively, such as radiotherapy, chemotherapy, neoadjuvant therapy, traditional Chinese medicine, etc.; (3) patients with a history of other malignancies; and (4) patients who did not undergo surgery.
This method involves using the FR as a marker to detect and quantify CTC from blood samples. Initially, preoperative blood samples are collected from patients for detecting FR+-CTC. These cells are then separated from the blood samples using techniques such as immunomagnetic separation. Subsequently, folate (folic acid) is used as a probe to detect and quantify the expression of FR in these separated cells using techniques like immunofluorescence staining and flow cytometry. To ensure the accuracy and sensitivity of detecting FR+-CTC, rigorous and well-defined controls were employed. Positive control cell lines with known FR expression were utilized to validate antibody specificity, while negative control cell lines lacking FR expression minimized the risk of nonspecific binding. Additionally, spike-in experiments were conducted by introducing known quantities of FR+ tumor cells into blood samples from healthy donors to evaluate CTC recovery efficiency and sensitivity. For flow cytometry, strict gating strategies were applied, leveraging fluorescence intensity, cell size, and morphological parameters to differentiate FR+-CTC from other circulating cells, such as leukocytes and platelets. Furthermore, appropriate isotype controls and repeated measurements ensured reproducibility and minimized false positives. These measures collectively enhanced the reliability of FR+-CTC detection.
The “caret” package in R software (R Foundation for Statistical Computing, Vienna, Austria) is used for random grouping of patients. All patients are randomly divided into a training group and a validation group in a 1:1 ratio.
The predictive model is constructed using a logistic regression model with the “glmnet” package in R software. The receiver operating characteristic (ROC) curve was plotted using the “pROC” package, and the area under the curve (AUC) is calculated to assess the predictive capability of the model. The model's validation is performed using a 10-fold cross-validation method, iterating 1,000 cycles to build predictive models and calculate the mean AUC values for validation. The accuracy of the predictive model is verified using the “riskRegression” and “prodlim” packages in R software to plot calibration curves.
Correlation analysis is performed using the Spearman correlation method, with the “ggplot2,” “ggpubr,” and “ggExtra” packages in R software used to plot correlation curves. Non-parametric testing (Wilcox test) is used to analyze statistical differences in FR+-CTC values and clinical relevant indicators among different gastrointestinal cancer patients, visualized using the “ggplot2” and “reshape2” packages in R software to create difference box plots. The chi-square test is used to compare baseline data between the training and validation groups. All analyses in this study are conducted in R software (version 4.0.3), with an alpha level of 0.05, considering p<0.05 as statistically significant.
A total of 396 patients meeting the criteria were included after screening. The baseline clinical characteristics, tumor pathological staging, levels of albumin, total protein, hemoglobin, FR+-CTC values, and tumor marker expressions of the included patients are summarized in Table 1.
Table 1 . Baseline Clinical Information of Patients Included in This Study.
Characteristic | Value (n=396) |
---|---|
Age, median (IQR), yr | 62 (55–71) |
Sex, No. (%) | |
Male | 250 (63.13) |
Female | 146 (36.87) |
Vessel invasion, No. (%) | |
No | 176 (44.44) |
Yes | 220 (55.56) |
Nerve invasion, No. (%) | |
No | 221 (55.81) |
Yes | 175 (44.19) |
T stage, No. (%) | |
T1 | 58 (14.66) |
T2 | 81 (20.45) |
T3 | 172 (43.43) |
T4 | 85 (21.46) |
N stage, No. (%) | |
N0 | 212 (53.53) |
N1 | 50 (12.63) |
N2 | 94 (23.74) |
N3 | 40 (10.10) |
M stage, No. (%) | |
M0 | 362 (91.41) |
M1 | 34 (8.59) |
Stage, No. (%) | |
I | 109 (27.52) |
II | 118 (29.80) |
III | 135 (34.09) |
IV | 34 (8.59) |
Tumor, No. (%) | |
Gastric cancer | 123 (31.06) |
Intestinal cancer | 273 (68.94) |
Peritoneal metastasis, No. (%) | |
No | 363 (91.67) |
Yes | 33 (8.33) |
Gastric cancer | 19 (57.58) |
Intestinal cancer | 14 (42.42) |
FR+-CTC, median (IQR), FU/3 mL | 10.80 (8.88–13.73) |
Albumin, median (IQR), g/L | 39.35 (34.98–41.10) |
Total protein, median (IQR), g/L | 66.75 (60.48–71.83) |
Hemoglobin, median (IQR), g/L | 122.0 (109.0–136.0) |
CEA, median (IQR), ng/mL | 1.85 (1.11–3.43) |
CA19-9, median (IQR), U/mL | 9.94 (5.83–17.54) |
CA-125, median (IQR), U/mL | 9.92 (6.66–15.32) |
IQR, interquartile range; FR+-CTC, folate receptor-positive circulating tumor cells; CEA, carcinoembryonic antigen; CA19-9, cancer antigen 19-9; CA-125, cancer antigen 125..
A logistic regression model was constructed using the “glmnet” package in R software, incorporating all risk factors into the model. As shown in Table 2, in this predictive model, FR+-CTC, albumin, total protein, and CA-125 were found to be statistically significant (p<0.05), while hemoglobin, cancer antigen 19-9, and carcinoembryonic antigen were not statistically significant (p>0.05). Therefore, the indicators ultimately included in the model are: FR+-CTC, albumin, total protein, and CA-125.
Table 2 . Logistic Regression Analysis of Peritoneal Metastasis Variable in Patients with Gastrointestinal Malignancies.
SE | Z-value | OR | 95% CI | p-value | |
---|---|---|---|---|---|
Intercept | 2.039 | –2.569 | 0.01 | 0.00–0.29 | 0.010 |
FR+-CTC | 0.026 | 4.553 | 1.13 | 1.07–1.19 | <0.001 |
Albumin | 0.060 | –3.127 | 0.83 | 0.74–0.93 | 0.002 |
Total protein | 0.036 | 2.729 | 1.10 | 1.03–1.18 | 0.006 |
Hemoglobin | 0.012 | 0.874 | 1.01 | 0.99–1.03 | 0.382 |
CA-125 | 0.008 | 2.335 | 1.02 | 1.00–1.04 | 0.019 |
CA19-9 | 0.002 | –0.128 | 1.00 | 0.98–1.01 | 0.899 |
CEA | 0.003 | –0.183 | 1.00 | 0.99–1.01 | 0.855 |
SE, standard error; OR, odds ratio; CI, confidence interval; FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125; CA19-9, cancer antigen 19-9; CEA, carcinoembryonic antigen..
The ROC curve was used to evaluate the accuracy of FR+-CTC and conventional clinical indicators in diagnosing peritoneal metastasis in patients with gastrointestinal malignancies. As shown in Fig. 1, the AUC values for diagnosing peritoneal metastasis with FR+-CTC, albumin, total protein, and CA-125 were 0.774, 0.686, 0.531, and 0.750, respectively. The optimal cutoff values for FR+-CTC, albumin, total protein, and CA-125 were 15.05 FU/3 mL, 37.45 g/L, 58.65 g/L, and 17.91 U/mL, respectively. This indicates that FR+-CTC had the highest predictive accuracy for peritoneal metastasis in patients with gastrointestinal malignancies.
Spearman correlation analysis was conducted to study the relationships between FR+-CTC and albumin, total protein, and CA-125 levels. As shown in Fig. 2A, there was a significant negative correlation between FR+-CTC and albumin levels (correlation coefficient R=–0.21, p<0.001). Fig. 2B showed a significant negative correlation between FR+-CTC and total protein levels (correlation coefficient R=–0.10, p=0.047). Fig. 2C indicated a significant positive correlation between FR+-CTC and CA-125 levels (correlation coefficient R=0.15, p=0.004). Although the FR+-CTC value has a certain correlation with albumin level, total protein level, and CA-125 level, the correlation coefficients are all relatively low.
As shown in Fig. 3A, patients with peritoneal metastasis had significantly higher levels of FR+-CTC and CA-125, and lower levels of albumin compared to those without peritoneal metastasis. Fig. 3B illustrated that patients with lymph node metastasis had significantly higher levels of FR+-CTC and CA-125, and lower albumin levels than those without lymph node metastasis. In Fig. 3C, patients with vascular invasion showed significantly higher FR+-CTC count and lower albumin levels than those without vascular invasion. Fig. 3D showed that patients with neural invasion had significantly higher levels of FR+-CTC and CA-125, and lower albumin levels than those without neural invasion. As per Fig. 3E, patients with T3-4 stage tumors had significantly higher FR+-CTC count compared to those in T1-2 stage. Fig. 3F indicated that patients with stage III-IV pathology had significantly higher levels of FR+-CTC and CA-125, and lower albumin levels compared to those in stage I-II. The detailed statistical data are shown in Table 3.
Table 3 . Statistical Analysis of FR+-CTC Count, Albumin Level, Total Protein Level, and CA-125 Level in Various Types of Gastrointestinal Malignancies.
Covariates | Type | FR+-CT, FU/3 mL | Albumin, g/L | Total protein, g/L | CA-125, U/mL | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
median (IQR) | p-value | median (IQR) | p-value | median (IQR) | p-value | median (IQR) | p-value | |||||
Peritoneal metastasis | PM (–) | 10.5 (8.8–13.3) | <0.001 | 39.9 (35.3–43.2) | <0.001 | 66.8 (60.6–71.9) | 0.551 | 9.05 (6.59–14.16) | <0.001 | |||
PM (+) | 18.8 (12.2–25.8) | 35.0 (31.1–38.4) | 65.6 (57.4–70.8) | 21.79 (9.67–78.92) | ||||||||
Lymph node metastasis | N (–) | 10.4 (8.9–12.7) | 0.028 | 40.5 (35.9–43.5) | 0.003 | 66.3 (60.8–71.9) | 0.726 | 8.76 (6.46–12.81) | <0.001 | |||
N (+) | 11.6 (8.8–15.5) | 38.0 (34.7–42.3) | 67.4 (60.0–71.8) | 10.43 (6.82–19.41) | ||||||||
Vascular invasion | Vessel (–) | 10.2 (8.8–12.5) | 0.005 | 40.5 (36.0–43.4) | 0.013 | 66.6 (61.4–71.5) | 0.906 | 9.09 (6.59–13.58) | 0.074 | |||
Vessel (+) | 11.6 (9.0–15.5) | 38.2 (34.4–42.9) | 67.0 (59.8–71.8) | 9.46 (6.69–17.58) | ||||||||
Neural invasion | Nerve (–) | 10.4 (8.8–12.7) | 0.039 | 40.3 (35.2–43.5) | 0.014 | 66.6 (60.6–72.0) | 0.892 | 9.05 (6.58–13.77) | 0.036 | |||
Nerve (+) | 11.2 (8.9–15.9) | 38.1 (34.9–42.3) | 67.1 (59.9–71.5) | 9.80 (6.79–18.77) | ||||||||
T stage | T1–2 | 10.2 (8.2–12.0) | <0.001 | 40.2 (35.2–43.3) | 0.536 | 66.2 (59.8–70.9) | 0.331 | 9.18 (6.58–13.97) | 0.290 | |||
T3–4 | 11.4 (9.1–15.3) | 39.2 (34.9–43.0) | 67.1 (60.8–71.9) | 9.26 (6.81–16.14) | ||||||||
Stage | Stage I–II | 10.3 (8.9–12.4) | 0.004 | 40.5 (35.6–43.6) | 0.005 | 66.2 (60.0–71.9) | 0.452 | 8.81 (6.55–13.65) | 0.005 | |||
Stage III–IV | 12.0 (8.8–16.1) | 38.0 (34.8–42.1) | 67.4 (60.8–71.8) | 10.0 (6.82–19.97) |
FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125; PM, peritoneal metastasis; lQR, interquartile range..
Patients were randomly divided into a trial group and a validation group in a 1:1 ratio. Detailed clinical information of patients in both groups is shown in Table 4. There were no significant differences between the training and validation groups in terms of age, sex, vascular invasion, neural invasion, T stage, N stage, M stage, stage, tumor location, and presence of peritoneal metastasis (p>0.05), indicating comparability between the two groups.
Table 4 . Clinical Data of Patients Included in the Training and Validation Groups.
Covariates | Type | Train set (n=198) | Validation set (n=198) | p-value |
---|---|---|---|---|
Age | ≤62 yr | 100 (50.51) | 102 (51.52) | 0.920 |
>62 yr | 98 (49.49) | 96 (48.48) | ||
Sex | Male | 124 (62.63) | 126 (63.64) | 0.917 |
Female | 74 (37.37) | 72 (36.36) | ||
Vessel invasion | No | 85 (42.93) | 91 (45.96) | 0.613 |
Yes | 113 (57.07) | 107 (54.04) | ||
Nerve invasion | No | 111 (56.06) | 110 (55.56) | 0.998 |
Yes | 87 (43.94) | 88 (44.44) | ||
T stage | T1 | 28 (14.14) | 30 (15.15) | 0.854 |
T2 | 39 (19.70) | 42 (21.21) | ||
T3 | 85 (42.93) | 87 (43.94) | ||
T4 | 46 (23.23) | 39 (19.70) | ||
N stage | N0 | 96 (48.48) | 116 (58.58) | 0.094 |
N1 | 30 (15.15) | 20 (10.10) | ||
N2 | 47 (23.74) | 47 (23.74) | ||
N3 | 25 (12.63) | 15 (7.58) | ||
M stage | M0 | 178 (89.90) | 184 (92.93) | 0.369 |
M1 | 20 (10.10) | 14 (7.07) | ||
Stage | I | 47 (23.74) | 62 (31.31) | 0.293 |
II | 59 (29.80) | 59 (29.80) | ||
III | 72 (36.36) | 63 (31.82) | ||
IV | 20 (10.10) | 14 (7.07) | ||
Tumor | Gastric cancer | 66 (33.33) | 57 (28.79) | 0.385 |
Intestinal cancer | 132 (66.67) | 141 (71.21) | ||
Peritoneal metastasis | No | 178 (89.90) | 185 (93.43) | 0.275 |
Yes | 20 (10.10) | 13 (6.57) |
Data are presented as number (%)..
As shown in Fig. 4A-C, the AUC values for the training group, validation group, and the entire patient cohort were 0.813, 0.874, and 0.837, respectively, indicating that the model has a strong predictive capability. Fig. 4D illustrates a nomogram, which visualizes the predictive model, enhancing the ability to predict the probability of peritoneal metastasis in patients with gastrointestinal malignancies, thus applying this predictive model in clinical settings. By determining the scores for each indicator and calculating the total score, the probability of a patient having peritoneal metastasis can be estimated. The specific parameters of this predictive model, as shown in Table 5, indicate that indicators such as FR+-CTC, albumin, total protein, and CA-125 are statistically significant (p<0.05).
Table 5 . Basic Parameters of the Predictive Model for Determining Peritoneal Metastasis in Patients with Gastrointestinal Malignancies.
SE | Z-value | OR | 95% CI | p-value | |
---|---|---|---|---|---|
Intercept | 1.895 | –2.443 | 0.01 | 0.00–0.40 | 0.014 |
FR+-CTC | 0.026 | 4.593 | 1.13 | 1.07–1.18 | <0.001 |
Albumin | 0.057 | –3.036 | 0.84 | 0.75–0.94 | 0.002 |
Total protein | 0.036 | 2.780 | 1.10 | 1.03–1.18 | 0.005 |
CA-125 | 0.005 | 3.468 | 1.02 | 1.01–1.03 | 0.001 |
SE, standard error; OR, odds ratio; CI, confidence interval; FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125..
The accuracy of the predictive model was validated using a 10-fold cross-validation method, iterating 1,000 cycles to build predictive models and calculate the mean AUC values for validation. As depicted in Fig. 5A-C, the cross-validation AUC values for the training group, validation group, and the entire patient cohort were 0.813, 0.886, and 0.844, respectively. These AUC values, all greater than 0.8, indicate a stable predictive capability of the model. The calibration curve, shown in Fig. 5D, demonstrates that the red curve closely aligns with the 45° diagonal line, suggesting that the probability of peritoneal metastasis predicted by the model closely matches the actual occurrence of peritoneal metastasis in patients, confirming the model's strong predictive accuracy.
Gastrointestinal malignancies, such as gastric and colorectal cancers, are among the most common malignant tumors globally.19-22 The risk associated with these cancers stems from their high invasiveness and tendency to metastasize, especially to the peritoneum. Peritoneal metastasis is a common late-stage phenomenon in gastrointestinal malignancies, significantly impacting patient quality of life and overall survival rates.23,24 Traditional imaging methods have limitations in early detection of peritoneal metastasis, hence developing new, more sensitive, and specific methods for early prediction and diagnosis of peritoneal metastasis in gastrointestinal malignancies is of paramount importance.25-27 FR+-CTC have received extensive attention in recent cancer research.28-30 The FR is a glycoprotein associated with the cell membrane that efficiently mediates cellular uptake of folate, an indispensable element for cell growth and division. In certain types of tumor cells, the expression of the FR is significantly upregulated, making it a marker distinguishing tumor cells from normal cells. CTC are tumor cells that have shed from the primary tumor into the peripheral blood circulation. Due to their high specificity and ease of detection, FR+-CTC are recognized as powerful biomarkers for cancer diagnosis and prognosis. Studies have indicated that FR+-CTC are not only involved in tumor progression but also actively contribute to the metastatic cascade. Overexpression of FR has been linked to enhanced tumor cell proliferation, migration, invasion, and immune evasion, all of which are critical steps in metastatic development. Recent evidence suggests that FR+-CTC facilitates hematogenous dissemination by interacting with the microenvironment and promoting the establishment of pre-metastatic niches. For example, FR-mediated signaling pathways have been implicated in epithelial-mesenchymal transition, a process that endows tumor cells with increased motility and invasiveness, further enhancing their ability to metastasize.31-33 These findings provide mechanistic insights into the role of FR+-CTC in peritoneal metastasis, highlighting their potential as both a predictive biomarker and a therapeutic target. Furthermore, the unique biology of FR+-CTC, including their ability to survive in circulation and evade anoikis, underscores their clinical relevance in predicting metastatic risk and guiding treatment strategies. Recent studies suggest that detecting FR+-CTC can predict the presence and metastasis of tumors at asymptomatic or early stages.
In our study, we have developed a predictive model based on FR+-CTC for predicting peritoneal metastasis in gastrointestinal malignancies. The establishment of this model not only provides a new perspective for understanding and predicting the clinical progression of gastrointestinal malignancies but also may offer substantial support for clinical decision-making. Firstly, we found that FR+-CTC, albumin, total protein, and CA-125 are significant factors in predicting peritoneal metastasis in gastrointestinal malignancies. FR+-CTC shows a negative correlation with albumin and total protein, and a positive correlation with CA-125. The observed negative correlation between FR+-CTC and albumin or total protein may reflect the systemic impact of advanced malignancies. Hypoalbuminemia and decreased total protein, often observed in cancer patients, are markers of malnutrition, systemic inflammation, and poor prognosis. Systemic inflammation, characterized by elevated levels of pro-inflammatory cytokines such as interleukin-6 and tumor necrosis factor-α, suppresses hepatic albumin synthesis and enhances protein catabolism, weakening the host’s immune defenses. These systemic changes create a permissive environment for the survival and dissemination of CTC by allowing them to evade immune surveillance and establish metastatic colonies. Moreover, hypoalbuminemia may reduce antioxidant capacity, further enhancing the resilience of tumor cells in circulation against oxidative stress. The decline in albumin and total protein also reflects the metabolic reprogramming associated with cancer cachexia, wherein resources are diverted to sustain tumor growth and progression. Conversely, the positive correlation between FR+-CTC and CA-125 underscores CA-125’s role as a biomarker of tumor burden and peritoneal involvement. CA-125, secreted by mesothelial cells lining the peritoneum, is frequently elevated in response to peritoneal irritation or tumor invasion. Elevated CA-125 levels may reflect a pro-inflammatory state in the peritoneal cavity, driven by tumor cell-secreted cytokines and growth factors. This inflammatory environment could facilitate FR+-CTC adhesion and implantation by promoting extracellular matrix remodeling via matrix metalloproteinases.34-37 Additionally, CA-125 is involved in mediating cell adhesion and aggregation through its interactions with MUC16 and integrins, thereby supporting FR+-CTC driven metastatic cascades. These interactions between FR+-CTC and CA-125 highlight their complementary roles in local and systemic aspects of tumor progression and underscore their combined value as predictive biomarkers for peritoneal metastasis. These findings suggest that monitoring the dynamics of FR+-CTC in conjunction with albumin, total protein, and CA-125 could provide a more comprehensive assessment of tumor progression and metastatic potential. Previous studies have extensively researched the role of the FR in the growth and metastasis of tumor cells, with increased expression often correlating with poor prognosis in malignant tumor patients.38,39 Our study not only quantitatively confirms this observation but also finds that the expression of FR+-CTC is significantly related to the risk of the tumor peritoneal metastasis. This finding further strengthens the importance of FR+-CTC as a potential biomarker, potentially offering a more accurate, biology-based predictive tool. Secondly, our predictive model demonstrates high predictive accuracy. While the AUC values reported for our model exceed 0.8 in the training group, validation group, and across all patient groups, additional metrics provide a fuller picture of the model’s performance. Specifically, our model demonstrated sensitivity, specificity, and overall accuracy exceeding 85%. This balance between sensitivity and specificity ensures the model’s capacity to accurately identify patients at risk while minimizing false positives, offering a robust tool for clinical application. These supplementary metrics, alongside the high AUC value, emphasize the model’s reliability and practicality. This indicates that our model can effectively differentiate patients at risk of peritoneal metastasis and provide accurate predictive results for clinical decision-making. More importantly, the model's advantage lies in using clinical indicators available from routine examinations, making it more feasible for practical application. This model aids understanding and prediction of the progression of gastrointestinal malignancies and potentially improving treatment strategies for patients.
However, despite the high predictive accuracy and clinical value of our predictive model, we must acknowledge some limitations in our study. Firstly, this study is based on a patient group from a single center, which may introduce sample selection bias. Future studies should be conducted across multiple centers to enhance the generalizability of the results. Secondly, although our predictive model performs well in the training and validation groups, its applicability to gastrointestinal malignancies in other countries needs further validation. One key limitation of these validation groups is the lack of subgroup analyses to assess the model’s performance across diverse clinical and demographic patient populations. Without this, the robustness and consistency of the model in varied settings remain unclear. Future studies should address these gaps by including larger, multi-center cohorts that better represent different populations and clinical contexts. Such analyses would help determine whether the predictive performance observed in the current study holds across different healthcare systems and patient demographics. Moreover, incorporating external validation datasets could further confirm the model’s reliability and minimize potential biases. These steps are crucial to enhance the clinical applicability of our findings. Future research should focus on elucidating the mechanisms linking FR+-CTC, albumin, total protein, and CA-125. Investigating whether the inflammatory state or specific cytokine profiles mediate these relationships could uncover new therapeutic targets. Exploring the dynamic interplay between nutritional status, systemic inflammation, and tumor biology will also be critical. Furthermore, incorporating additional biomarkers such as circulating tumor DNA or exosomal proteins could complement FR+-CTC analysis, enhancing the predictive model’s robustness. These efforts may not only refine the model but also provide a foundation for personalized therapeutic strategies. Current cancer research is focusing on the discovery of more precise biomarkers and personalized treatment strategies. In this context, the discovery of FR+-CTC and its application in predicting tumor metastasis is of significant importance. Future research could explore combining FR+-CTC with other emerging biomarkers to enhance the accuracy and clinical applicability of the predictive model.40,41 Moreover, with the development of precision medicine and personalized treatment strategies, our model can be further refined to meet the specific needs of different patient groups.
In summary, this discovery provides new insights for the early diagnosis and treatment of gastrointestinal malignancies. Particularly in predicting peritoneal metastasis preoperatively, the detection of FR+-CTC may become an essential tool. The development of a preoperative predictive model based on FR+-CTC is a key step in translating these research findings into clinical practice. This model integrates patients' biomarker data and clinical characteristics, offering clinicians a quantitative assessment of the risk of peritoneal metastasis. This is crucial for formulating individualized treatment strategies, such as deciding on adjuvant chemotherapy and selecting surgical methods. We anticipate that future research will further expand and refine this model, enabling it to play a more significant role in the personalized treatment of cancer.
In conclusion, FR+-CTC can serve as a novel biomarker for gastrointestinal malignancies. A new model based on FR+-CTC demonstrates strong predictive capabilities for preoperative assessment of peritoneal metastasis in gastrointestinal cancers.
No potential conflict of interest relevant to this article was reported.
Study concept and design: R.H. Data acquisition: D.L. Data analysis and interpretation: D.L. Drafting of the manuscript: D.L. Critical revision of the manuscript for important intellectual content: C.L. Statistical analysis: C.L. dministrative, technical, or material support; study supervision: R.H. Approval of final manuscript: all authors.
The data of this study can be obtained from the corresponding author according to reasonable requirements.
Table 1 Baseline Clinical Information of Patients Included in This Study
Characteristic | Value (n=396) |
---|---|
Age, median (IQR), yr | 62 (55–71) |
Sex, No. (%) | |
Male | 250 (63.13) |
Female | 146 (36.87) |
Vessel invasion, No. (%) | |
No | 176 (44.44) |
Yes | 220 (55.56) |
Nerve invasion, No. (%) | |
No | 221 (55.81) |
Yes | 175 (44.19) |
T stage, No. (%) | |
T1 | 58 (14.66) |
T2 | 81 (20.45) |
T3 | 172 (43.43) |
T4 | 85 (21.46) |
N stage, No. (%) | |
N0 | 212 (53.53) |
N1 | 50 (12.63) |
N2 | 94 (23.74) |
N3 | 40 (10.10) |
M stage, No. (%) | |
M0 | 362 (91.41) |
M1 | 34 (8.59) |
Stage, No. (%) | |
I | 109 (27.52) |
II | 118 (29.80) |
III | 135 (34.09) |
IV | 34 (8.59) |
Tumor, No. (%) | |
Gastric cancer | 123 (31.06) |
Intestinal cancer | 273 (68.94) |
Peritoneal metastasis, No. (%) | |
No | 363 (91.67) |
Yes | 33 (8.33) |
Gastric cancer | 19 (57.58) |
Intestinal cancer | 14 (42.42) |
FR+-CTC, median (IQR), FU/3 mL | 10.80 (8.88–13.73) |
Albumin, median (IQR), g/L | 39.35 (34.98–41.10) |
Total protein, median (IQR), g/L | 66.75 (60.48–71.83) |
Hemoglobin, median (IQR), g/L | 122.0 (109.0–136.0) |
CEA, median (IQR), ng/mL | 1.85 (1.11–3.43) |
CA19-9, median (IQR), U/mL | 9.94 (5.83–17.54) |
CA-125, median (IQR), U/mL | 9.92 (6.66–15.32) |
IQR, interquartile range; FR+-CTC, folate receptor-positive circulating tumor cells; CEA, carcinoembryonic antigen; CA19-9, cancer antigen 19-9; CA-125, cancer antigen 125.
Table 2 Logistic Regression Analysis of Peritoneal Metastasis Variable in Patients with Gastrointestinal Malignancies
SE | Z-value | OR | 95% CI | p-value | |
---|---|---|---|---|---|
Intercept | 2.039 | –2.569 | 0.01 | 0.00–0.29 | 0.010 |
FR+-CTC | 0.026 | 4.553 | 1.13 | 1.07–1.19 | <0.001 |
Albumin | 0.060 | –3.127 | 0.83 | 0.74–0.93 | 0.002 |
Total protein | 0.036 | 2.729 | 1.10 | 1.03–1.18 | 0.006 |
Hemoglobin | 0.012 | 0.874 | 1.01 | 0.99–1.03 | 0.382 |
CA-125 | 0.008 | 2.335 | 1.02 | 1.00–1.04 | 0.019 |
CA19-9 | 0.002 | –0.128 | 1.00 | 0.98–1.01 | 0.899 |
CEA | 0.003 | –0.183 | 1.00 | 0.99–1.01 | 0.855 |
SE, standard error; OR, odds ratio; CI, confidence interval; FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125; CA19-9, cancer antigen 19-9; CEA, carcinoembryonic antigen.
Table 3 Statistical Analysis of FR+-CTC Count, Albumin Level, Total Protein Level, and CA-125 Level in Various Types of Gastrointestinal Malignancies
Covariates | Type | FR+-CT, FU/3 mL | Albumin, g/L | Total protein, g/L | CA-125, U/mL | |||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
median (IQR) | p-value | median (IQR) | p-value | median (IQR) | p-value | median (IQR) | p-value | |||||
Peritoneal metastasis | PM (–) | 10.5 (8.8–13.3) | <0.001 | 39.9 (35.3–43.2) | <0.001 | 66.8 (60.6–71.9) | 0.551 | 9.05 (6.59–14.16) | <0.001 | |||
PM (+) | 18.8 (12.2–25.8) | 35.0 (31.1–38.4) | 65.6 (57.4–70.8) | 21.79 (9.67–78.92) | ||||||||
Lymph node metastasis | N (–) | 10.4 (8.9–12.7) | 0.028 | 40.5 (35.9–43.5) | 0.003 | 66.3 (60.8–71.9) | 0.726 | 8.76 (6.46–12.81) | <0.001 | |||
N (+) | 11.6 (8.8–15.5) | 38.0 (34.7–42.3) | 67.4 (60.0–71.8) | 10.43 (6.82–19.41) | ||||||||
Vascular invasion | Vessel (–) | 10.2 (8.8–12.5) | 0.005 | 40.5 (36.0–43.4) | 0.013 | 66.6 (61.4–71.5) | 0.906 | 9.09 (6.59–13.58) | 0.074 | |||
Vessel (+) | 11.6 (9.0–15.5) | 38.2 (34.4–42.9) | 67.0 (59.8–71.8) | 9.46 (6.69–17.58) | ||||||||
Neural invasion | Nerve (–) | 10.4 (8.8–12.7) | 0.039 | 40.3 (35.2–43.5) | 0.014 | 66.6 (60.6–72.0) | 0.892 | 9.05 (6.58–13.77) | 0.036 | |||
Nerve (+) | 11.2 (8.9–15.9) | 38.1 (34.9–42.3) | 67.1 (59.9–71.5) | 9.80 (6.79–18.77) | ||||||||
T stage | T1–2 | 10.2 (8.2–12.0) | <0.001 | 40.2 (35.2–43.3) | 0.536 | 66.2 (59.8–70.9) | 0.331 | 9.18 (6.58–13.97) | 0.290 | |||
T3–4 | 11.4 (9.1–15.3) | 39.2 (34.9–43.0) | 67.1 (60.8–71.9) | 9.26 (6.81–16.14) | ||||||||
Stage | Stage I–II | 10.3 (8.9–12.4) | 0.004 | 40.5 (35.6–43.6) | 0.005 | 66.2 (60.0–71.9) | 0.452 | 8.81 (6.55–13.65) | 0.005 | |||
Stage III–IV | 12.0 (8.8–16.1) | 38.0 (34.8–42.1) | 67.4 (60.8–71.8) | 10.0 (6.82–19.97) |
FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125; PM, peritoneal metastasis; lQR, interquartile range.
Table 4 Clinical Data of Patients Included in the Training and Validation Groups
Covariates | Type | Train set (n=198) | Validation set (n=198) | p-value |
---|---|---|---|---|
Age | ≤62 yr | 100 (50.51) | 102 (51.52) | 0.920 |
>62 yr | 98 (49.49) | 96 (48.48) | ||
Sex | Male | 124 (62.63) | 126 (63.64) | 0.917 |
Female | 74 (37.37) | 72 (36.36) | ||
Vessel invasion | No | 85 (42.93) | 91 (45.96) | 0.613 |
Yes | 113 (57.07) | 107 (54.04) | ||
Nerve invasion | No | 111 (56.06) | 110 (55.56) | 0.998 |
Yes | 87 (43.94) | 88 (44.44) | ||
T stage | T1 | 28 (14.14) | 30 (15.15) | 0.854 |
T2 | 39 (19.70) | 42 (21.21) | ||
T3 | 85 (42.93) | 87 (43.94) | ||
T4 | 46 (23.23) | 39 (19.70) | ||
N stage | N0 | 96 (48.48) | 116 (58.58) | 0.094 |
N1 | 30 (15.15) | 20 (10.10) | ||
N2 | 47 (23.74) | 47 (23.74) | ||
N3 | 25 (12.63) | 15 (7.58) | ||
M stage | M0 | 178 (89.90) | 184 (92.93) | 0.369 |
M1 | 20 (10.10) | 14 (7.07) | ||
Stage | I | 47 (23.74) | 62 (31.31) | 0.293 |
II | 59 (29.80) | 59 (29.80) | ||
III | 72 (36.36) | 63 (31.82) | ||
IV | 20 (10.10) | 14 (7.07) | ||
Tumor | Gastric cancer | 66 (33.33) | 57 (28.79) | 0.385 |
Intestinal cancer | 132 (66.67) | 141 (71.21) | ||
Peritoneal metastasis | No | 178 (89.90) | 185 (93.43) | 0.275 |
Yes | 20 (10.10) | 13 (6.57) |
Data are presented as number (%).
Table 5 Basic Parameters of the Predictive Model for Determining Peritoneal Metastasis in Patients with Gastrointestinal Malignancies
SE | Z-value | OR | 95% CI | p-value | |
---|---|---|---|---|---|
Intercept | 1.895 | –2.443 | 0.01 | 0.00–0.40 | 0.014 |
FR+-CTC | 0.026 | 4.593 | 1.13 | 1.07–1.18 | <0.001 |
Albumin | 0.057 | –3.036 | 0.84 | 0.75–0.94 | 0.002 |
Total protein | 0.036 | 2.780 | 1.10 | 1.03–1.18 | 0.005 |
CA-125 | 0.005 | 3.468 | 1.02 | 1.01–1.03 | 0.001 |
SE, standard error; OR, odds ratio; CI, confidence interval; FR+-CTC, folate receptor-positive circulating tumor cells; CA-125, cancer antigen 125.