Open Access

Preoperative carcinoembryonic antigen to body mass index ratio contributes to prognosis prediction in colorectal cancer

  • Authors:
    • Jia Xiang
    • Mengyao Ding
    • Jixing Lin
    • Tianhui Xue
    • Qianwen Ye
    • Bing Yan
  • View Affiliations

  • Published online on: September 29, 2022     https://doi.org/10.3892/ol.2022.13536
  • Article Number: 416
  • Copyright: © Xiang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Both carcinoembryonic antigen (CEA) level and body mass index (BMI) are traditional prognostic markers in colorectal cancer (CRC); however, to the best of our knowledge, the value of the CEA to BMI ratio (CBR) has never been addressed. In the present study, 191 patients with CRC treated using radical resection were retrospectively included, and the significance of the CBR in predicting disease‑free survival (DFS) or overall survival (OS) rates was calculated. The prognostic efficacy of the CBR in predicting OS was compared with individual CEA and BMI values. The survival differences of the subgroups were calculated by Kaplan‑Meier analysis, and corresponding risk factors were then estimated by a Cox proportional hazards model. As a result, 29.84% (57/191) of the patients were assigned to the high CBR group (cut‑off, ≥0.28); the CBR had a sensitivity of 56.50 and 68.90%, and a specificity of 80.60 and 80.10% for DFS and OS, respectively. Patients with a high CBR more commonly underwent laparotomy and exhibited advanced T stages, the presence of tumor deposits and advanced Tumor‑Node‑Metastasis stages (stage II or III). The CBR was more efficient than the CEA or BMI alone in predicting OS. In addition, patients with a high CBR presented with a significantly worse outcome than patients with a low CBR. Finally, the CBR was an independent risk factor for both DFS and OS. In conclusion, the CBR was a more robust prognostic factor in CRC, and patients with a relatively high CBR exhibited poorer survival.

Introduction

Colorectal cancer (CRC) remains a life-threatening disease worldwide (1), with an incidence rate that increases annually by ~2% and a death rate that increases annually by ~1.3% among those <50 years of age (2). In recent years, a group of prognostic markers obtained from patients' routine blood tests, such as the neutrophil to lymphocyte ratio (NLR) (3) and the lymphocyte to monocyte ratio (LMR) (4), have been established. These indicators are non-invasive and easily accessible in practice; however, their prognostic efficacies have not been conventionally compared with traditional markers.

Body mass index (BMI), which is calculated as weight in kilograms divided by height in meters squared (kg/m2), is a traditional prognostic marker for numerous malignancies, including breast (5), ovarian (6), esophageal (7), lung (8) and gastric (9) cancer, as well as CRC (1012). However, there are still unresolved problems regarding the prognostic value of BMI in CRC. On the one hand, the BMI criteria to group patients have been inconsistent in previous studies; for example, Guercio et al (13) divided patients into 5 subgroups [<21 (underweight), 21–24.9 (normal), 25–29.9, 30–34.9 and 35 kg/m2], as did Chiu et al (11); however, in the latter study, underweight (<18.50 kg/m2) and normal weight (18.50-24.99 kg/m2) patients were not in line with those in the former study. Furthermore, Song et al found that 20.2 kg/m2 was the best discrimination point for survival (12). In addition, the final conclusion for the role of BMI in CRC is conflicting, with some studies indicating that patients with an underweight BMI (<18.50 kg/m2) would have poor survival (1012), but other studies failing to reproduce these results (13) or even yielding opposite results (14). Based on this background, it is reasonable to improve the prognostic efficacy of BMI with a combination of other markers. In fact, other studies have tried to combine BMI with other markers, including the NLR (15,16); however, these studies did not routinely report the prognostic efficacy of the new indicators or statistically compare them when it was generated.

The carcinoembryonic antigen (CEA) level is another long-term established tumor marker that is recommended by the American Society of Clinical Oncology for CRC (17) and has been incorporated into the Tumor-Node-Metastasis (TNM) system (the so-called C-stage) to provide additional prognostic information (18). However, apart from the fact that only 21–36% of patients are positive for CEA at diagnosis (19), limitations such as a low sensitivity of a single CEA value for prognosis cannot be ignored (20), and its efficacy could be largely attenuated in conditions such as type II diabetes (21) or smoking (22). Notably, in previous studies, the serum concentration of CEA was closely correlated with BMI; for example, an increase or decrease in BMI in patients with cancer was correlated with a fluctuation in systemic inflammatory factors such as interleukin-6 (IL-6) (23,24), which can promote the secretion of CEA in CRC cells (25,26). Based on these factors, it is reasonable to explore the prognostic value of the CEA to BMI ratio (CBR) in CRC, as the significance of CEA levels could be balanced to some extent by BMI in these patients; however, to the best of our knowledge, associated reports are still absent.

The present study aimed to explore the prognostic value of the CBR, as well as its prognostic efficacy when compared with individual CEA, BMI and other inflammatory prognostic indicators in CRC.

Materials and methods

Patients

Between January 2012 and October 2021, patients with CRC who underwent radical resection of primary lesions at Hainan Hospital of Chinese People's Liberation Army (PLA) General Hospital (Sanya, China) were retrospectively included in the present study. Patients meeting any one of the following criteria were excluded: i) Patients with an absence of preoperative laboratory test results or abnormal aminotransferase or serum creatinine levels, since such abnormalities could cause an altered metabolism or excretion of CEA (27,28); ii) patients with distant lesions; iii) patients who were missing any TNM information in their postoperative pathological reports; iv) patients with multiple or recurrent malignancies or in situ lesions; v) and patients with a follow-up time of <36 months. In addition, patients who received neoadjuvant chemotherapy were also excluded, since such therapy could cause problems in being able to accurately confirm the pT/pN stages in post-operative pathological findings, in particular for those who reached a tumor regression grading of 2 or 3 (29). A binary system was applied for patients with cancer with or without mucinous elements, tumor deposits (TDs) and risk factors (with perineural or lymphovascular invasion). Other clinicopathological parameters were recorded as described in previous studies (30,31). The study was performed in line with the principles stated in the Declaration of Helsinki and was approved by the Ethics Committee of Hainan Hospital of Chinese PLA General Hospital (approval no. 301HLFYLS15). All patients or their authorized relatives provided written informed consent.

Determination of the CBR, NLR, LMR, platelet to lymphocyte ratio (PLR) and prognostic nutritional index (PNI), and their correlations

Laboratory tests were performed as described in our previous studies (30,31). The CBR was calculated as the concentration of serum CEA (reference, 0–5 µg/ml) divided by the BMI. In addition, other inflammatory prognostic indicators, including NLR, LMR, PLR and PNI, were also determined according to previous studies (3,4,32,33). The correlation of CBR with NLR, LMR, PLR and PNI was also determined.

Definitions of disease-free survival (DFS) and overall survival (OS)

The follow-up was conducted as previously described (31). Briefly, patients were interviewed every 3–6 months for the first 2 years and then every 6–12 months after for those who survived for >2 years. DFS time was defined as the time from the date of surgery until the date of the first recurrence or metastasis at any location, or death from any reason. OS time was defined from the same initial point to the date of death from any cause. The latest follow-up point was December 2021.

Statistical analysis

Statistical analyses were conducted using SPSS 20.0 (IBM Corp.) and MedCalc v19.0.7 (MedCalc Software bvba). The optimal discriminator point of the CBR was calculated by receiver operating characteristic (ROC) curve analysis, and then the area under the curve (AUC) was compared using the methods described by DeLong et al (34). The differences in the clinicopathological parameters between the CBR subgroups were estimated by χ2 test. The association of CBR with other inflammatory prognostic indicators was determined by Pearson's correlation analysis. Survival differences for the low or high CBR subgroups were determined by Kaplan-Meier analysis followed by log-rank tests. A Cox proportional hazards model was applied to select the risk factors for survival (forward likelihood ratio model). Based on the results of the multivariable analysis, nomograms were established using R (version 4.1.1; http://www.r-project.org) with the survival and RMS package, and the C-index was used to determine the prediction efficacy. P<0.05 (two-sided) was considered to indicate a statistically significant difference.

Results

Demographic features and the prognostic efficacy of the CBR

As shown in Fig. 1, a total of 191 patients (68 females and 123 males) were finally included in the study. The mean age of the patients was 59.3 years (range, 24–85 years), with a mean follow-up period of 51.1 months (range, 1–111 months). By ROC analysis, taking 0.28 as the optimal cutoff point (according to the Youden index), 29.84% (57/191) of the patients were assigned to the high CBR group (≥0.28), and 70.16% (134/191) were assigned to the low CBR group (<0.28). The CBR presented a sensitivity of 56.50 and 68.90%, and a specificity of 80.60 and 80.10% for DFS and OS, respectively (both P<0.01) (Fig. 2). Next, a further comparison of the prognostic efficacy of the CBR in predicting OS with individual CEA (Z=2.35, P=0.02), BMI (Z=2.01, P=0.04), NLR (Z=2.90, P<0.01), LMR (Z=2.42, P=0.02) and PLR (Z=2.49, P=0.01) values indicated significant differences, with the exception of PNI (Z=1.16, P=0.25).

Correlation of CBR with NLR, LMR, PLR and PNI

According to Pearson's correlation analysis, the CBR displayed a significant positive association with NLR (r=0.20, P=0.01) and PLR (r=0.21, P<0.01), whereas a negative association was exhibited with LMR (r=−0.17, P=0.02) and PNI (r=−0.20, P=0.01) (Fig. 3).

Clinicopathological parameter differences between the high- and low-CBR subgroups

According to the χ2 test, patients who underwent a laparotomy (P=0.03), or those with advanced T stages (T3+T4) (P<0.01), the presence of TD(s) (P<0.01) and advanced TNM stages (stage II or III) (P<0.01) were more commonly found in the high CBR group. No significant differences were found for the other clinicopathological parameters between the high and low CBR subgroups (Table I).

Table I.

Differences in clinicopathological parameters among CBR subgroups.

Table I.

Differences in clinicopathological parameters among CBR subgroups.

CBR

CharacteristicTotal patients, nLow, nHigh, nP-value
Age, years 0.35
  <60916724
  ≥601006733
Sex 0.74
  Female684919
  Male1238538
Type of resection 0.03a
  Laparotomy291514
  Laparoscopy16211943
Tumor location 0.25
  Right422616
  Left14910841
Histological grade 0.38
  Well + moderate16211646
  Poor291811
Mucinous element 0.16
  Present372215
  Absent15411242
T stages <0.01a
  T1 + T251447
  T3 + T41409050
N stages 0.11
  N01158629
  N1 + N2764828
Tumor deposits <0.01a
  Present17112645
  Absent20812
TNM stages <0.01a
  I40382
  II754827
  III764828
Risk factors 0.09
  Present241311
  Absent16712146

a P<0.05. TNM, Tumor-Node-Metastasis; CBR, carcinoembryonic antigen to body mass index ratio.

Survival differences between the high and low CBR subgroups in terms of DFS and OS

After the 3-year follow-up period, significant differences in DFS (43.86 vs. 83.58%; P<0.01) and OS (56.14 vs. 89.55%; P<0.01) rates were found between the high and low CBR subgroups, respectively (Fig. 4A and B). Subsequently, the survival differences of the low and high CBR subgroups were further compared in different stages, and it was indicated that patients with a low CBR would have a superior DFS and OS in stage II (with a tendency but non-significant result for DFS) and stage III patients but not in stage I cases (Fig. 4).

Univariate and multivariate analyses of the prognostic factors for DFS and OS

Univariate tests indicated that type of resection, T, N and TNM stages, TDs, risk factors, pre-operative NLR, LMR, PLR, PNI and CBR were all factors that could affect DFS (Table II), and that these, in addition to histological grade and mucinous elements, were factors that could affect OS (Table III). When these significant factors (only those P<0.05) were included in the multivariate tests for DFS and OS, the results indicated that the TDs, PNI and CBR were independent prognostic factors for both DFS (CBR: HR, 3.48; 95% CI, 2.04-5.91; P<0.01) and OS (CBR: HR, 3.71; 95% CI, 1.95-7.08; P<0.01). Additionally, N stage was an independent prognostic factor for DFS, and histological grade, mucinous element and risk factors were independent prognostic factor for OS (Tables II and III).

Table II.

Univariate and multivariate analyses of risk factors for disease-free survival.

Table II.

Univariate and multivariate analyses of risk factors for disease-free survival.

UnivariateMultivariate


CharacteristicP-valueHR95% CIP-valueHR95% CI
Age, years
  <601.00
  ≥600.861.050.63-1.72
Sex
  Female1.00
  Male0.271.360.79-2.35
Type of resection
  Laparotomy1.00
  Laparoscopy <0.01a0.390.22-0.68
Tumor location
  Right1.00
  Left0.840.940.52-1.71
Histological grade
  Well + moderate1.00
  Poor0.081.740.94-3.22
Mucinous element
  Present1.00
  Absent0.060.580.33-1.02
T stages
  T1 + T21.00
  T3 + T4 <0.01a3.961.71-9.20
N stages
  N01.00 1.00
  N1 + N2 <0.01a2.851.71-4.74 <0.01a2.301.30-4.08
Tumor deposits
  Present1.00 1.00
  Absent <0.01a0.130.07-0.24 <0.01a0.340.17-0.67
TNM stages
  I1.00
  II0.05a2.981.02-8.69
  III <0.01a6.452.30-18.14
Risk factors
  Present1.00
  Absent0.01a0.430.23-0.79
Preoperative measures
  NLR <0.01a1.081.03-1.14
  LMR0.02a0.820.69-0.97
  PLR0.021.001.00-1.01
  PNI <0.01a0.930.90-0.97 <0.01a0.930.90-0.97
CBR
  Low1.00 1.00
  High <0.01a3.862.33-6.37 <0.01a3.482.04-5.91

a P<0.05. TNM, Tumor-Node-Metastasis; CBR, carcinoembryonic antigen to body mass index ratio; NLR, neutrophil to lymphocyte ratio; LMR, lymphocyte to monocyte ratio; PLR, platelet to lymphocyte ratio; PNI, prognostic nutritional index.

Table III.

Univariate and multivariate tests of risk factors for overall survival.

Table III.

Univariate and multivariate tests of risk factors for overall survival.

UnivariateMultivariate


CharacteristicP-valueHR95% CIP-valueHR95% CI
Age, years
  <601.00
  ≥600.131.600.88-2.93
Sex
  Female1.00
  Male0.201.540.80-2.99
Type of resection
  Laparotomy1.00
  Laparoscopy0.03a0.470.24-0.93
Tumor location
  Right1.00
  Left0.960.980.47-1.98
Histological grade
  Well + moderate1.00 1.00
  Poor0.01a2.321.20-4.500.01a2.541.26-5.11
Mucinous element
  Present1.00 1.00
  Absent0.04a0.500.26-0.950.05a0.490.24-0.98
T stages
  T1 + T21.00
  T3 + T40.01a4.121.48-11.51
N stages
  N01.00
  N1 + N2 <0.01a2.381.32-4.30
Tumor deposits
  Present1.00 1.00
  Absent <0.01a0.120.06-0.23 <0.01a0.240.12-0.47
TNM stages
  I1.00
  II0.03a4.871.12-21.07
  III <0.01a8.231.95-34.70
Risk factors
  Present1.00 1.00
  Absent <0.01a0.340.17-0.670.01a0.370.18-0.78
Preoperative measures
  NLR0.01a1.091.03-1.16
  LMR0.04a0.820.67-0.99
  PLR0.04a1.001.00-1.01
  PNI <0.01a0.910.87-0.95 <0.01a0.890.85-0.94
CBR
  Low1.00 1.00
  High <0.01a5.502.98-10.15 <0.01a3.711.95-7.08

a P<0.05. TNM, Tumor-Node-Metastasis; CBR, carcinoembryonic antigen to body mass index ratio; NLR, neutrophil to lymphocyte ratio; LMR, lymphocyte to monocyte ratio; PLR, platelet to lymphocyte ratio; PNI, prognostic nutritional index.

Development of nomograms for predicting CRC prognosis

To predict DFS and OS rates for the patients, nomograms were constructed based on the multivariate analyses in Tables II and III. Each factor received a corresponding total point according to the nomograms. As shown in Fig. 5, the C-indexes for the prediction of 3-year DFS and OS were 0.81 (95% CI, 0.72-0.91) and 0.84 (95% CI, 0.71-0.96), respectively (Fig. 5A and B), which indicated that the model provided good discrimination. In addition, the calibration curves displayed a good consistency between the predicted results and the actual results (Fig. 5C and D).

Discussion

The present study found that the CBR was significant in the prognosis of CRC, and that the prognostic efficacy of the CBR was superior to individual CEA, BMI, NLR, PLR and LMR values, with a relatively high sensitivity and specificity. Patients with a high CBR had a worse outcome than patients with a low CBR. Taking into consideration that the CBR was less likely to be influenced by common acute complications such as infection, bleeding and obstruction in CRC, it should be considered a robust prognostic indicator in practice. To the best of our knowledge, the present study is the first report concerning the value of the CBR in CRC.

Both CEA levels and BMI are traditional prognostic markers in CRC, but have individual limitations. In previous studies on CEA levels, the positive rate was relatively low (19), and although patients with a normal range of CEA also had prognostic significance based on reduced cutoff points (35,36), the sensitivity was relatively low (46.00%) (37) with a limited AUC in predicting survival that ranged from 0.636 (cutoff, 11 µg/ml) (38) and 0.645 (cutoff, 5 µg/ml) (39) to 0.740 (cutoff, 12.5 µg/ml) (40). A recent study indicated that the CEA to maximum tumor diameter ratio has prognostic value; however, the AUC of the new marker in predicting 3-year OS was reported to be only 0.704 (41). For BMI, its application in prognosis was largely blocked by inconsistent criteria and conflicting results, as aforementioned (1014). Similarly, some reports indicated that combining BMI with other markers, such as lymphocyte counts, could improve the prognostic efficacy for patients with head and neck cancer who underwent radiation therapy (16). Recently, Xie et al (42) conducted a study that included 2,471 patients with CRC and found that the neutrophil-BMI ratio was a useful prognostic marker. In the present study, CBR presented with a relatively high sensitivity and specificity for OS. Furthermore, the prognostic efficacy was markedly stronger than that of individual CEA and BMI values, and even greater than NLR, LMR and PLR values.

Taking into consideration the individual role of CEA levels and BMI in CRC prognosis, these results could be explained from the following perspectives. First, for patients falling in a certain BMI range, a relatively high CEA (equal to a high CBR) would correlate with poor survival, which has been well established by previous clinical studies (35,43). Molecularly, it is well known that up to 90% of CRC cells can release CEA (44,45), and CEA can trigger CRC progression by inducing epithelial-mesenchymal transition, increasing cancer cell invasiveness and inhibiting apoptotic signaling (46). In addition, CEA can also cause radioresistance in CRC cells in the presence of M2 macrophages (47). Based on these facts, a high CBR weighted by a high CEA was likely to be associated with poor outcomes in the patients. Second, for those with a similar concentration of CEA, a lower BMI (also indicating a high CBR) would lead to worse outcomes, which was also in line with previous observations (1012). However, it is also notable that certain studies indicated that a high BMI, particularly for class I (BMI, 30–35 kg/m2) and II (BMI, ≥35 kg/m2) obesity, was correlated with poor survival in CRC (48,49). In fact, previous studies found an increasing incidence of newly diagnosed liver disorder in obese patients (50), and a relationship between BMI and serum liver enzyme activity (51); subsequently, the metabolism or excretion of CEA was altered and had a tendency to be higher in patients with a high BMI (27). However, studies concerning the CEA level in individuals with a high BMI, particularly in those with class I or class II obesity, are rare, and it is still largely unknown whether the CBR is also applicable in such a scenario (only 5 patients had a BMI of >30 in the present study).

Cancer-related inflammation has a profound effect in regulating cancer development and is regarded as a hallmark of it (52). High CBR associated with poor survival could be also understood from the perspective of inflammation. Previous studies indicated that CEA in patients with CRC could induce the secretion of IL-6, and that the levels of CEA and IL-6 were positively correlated (16,53). It was noted that IL-6 could induce fat loss in cancer cachexia via different approaches, such as promoting white adipose tissue lipolysis (54) and decreasing muscle mass (55), which could result in a decreased BMI in these patients (56). Based on these facts, it was plausible that a high CBR indicated a high level of inflammation and a high tendency for reduced BMI, which could present as cachexia and poor survival. The present study also found that a high CBR was associated with advanced T stage (T3+T4), and the presence of TDs. CBD was positively associated with NLR or PLR and negatively associated with LMR or PNI on correlation analysis; in fact, these parameters were also previously reported to be useful prognostic indicators in CRC. For example, Lino-Silva et al (57) included 392 patients of all stages and found that stage I–III patients who presented with TDs displayed similar mortality rates to stage IV patients. Furthermore, Moon et al (58) published a systematic review that included 90,455 stage III patients and found that those who presented with TDs had a worse 5-year DFS rate. In addition to TDs, the association of a low LMR with poor survival was also recorded; for example, Naszai et al (59) conducted a meta-analysis that included 32,788 patients and found that a pre-treatment high NLR was a significant predictor of poor OS. In line with this, Tan et al (4) also conducted a meta-analysis that concluded that a low LMR was a significant predictor of poor OS. All this evidence supports the fact that patients with CRC and a high CBR have poor survival rates.

In recent years, a small subgroup of patients with CRC featuring mismatch repair deficiency (dMMR) were demonstrated to exhibit better outcomes than those with mismatch repair-proficient tumors (pMMR) in certain stage cases, such as stage II (60,61), and they also had a significantly good response when receiving immunotherapies in metastatic cases when compared to pMMR cases (62). Notably, a lower BMI was found to be more common in patients with dMMR than in those with pMMR (63); additionally, CEA levels were significantly higher in the pMMR subtype (but only in stage III patients) (64). These results suggested that for patients in certain stages, the CBR would be higher than for those in other stages. However, in the present study, no such differences were found either for BMI (n=112, P=0.64; data not shown) or for CEA levels (n=47, P=0.92; data not shown). Taking into consideration the complex prognostic role of MMR status in CRC, additional studies with larger samples are still needed.

The present study also had some limitations. First, it was conducted with a relatively small sample size; in particular, the patients with a follow-up time of <36 months were excluded since these cases may lack definite OS information and cause problems in calculating the specific survival rate, which may also result in a biased finding. Second, both CEA levels and BMI could present extreme values, and the prognostic value of the CBR was not validated in these cases. Third, definite information for those stage II patients with high risks or the stage III patients on adjuvant therapy was not sufficient, and it was well established that adjuvant chemotherapy could improve the DFS in patients with radical surgery (65,66). However, it was notable that a number of other factors could influence the efficacy of these therapies in the real world, including the delay, early discontinuation and dose reduction of the treatment. This is a complex problem that requires additional elegant studies. Further studies are also needed to further validate the present results.

Overall, the present results indicated that, compared with individual CEA, BMI, NLR and LMR values, the CBR was a more robust prognostic factor in CRC, and patients with a relatively high CBR presented with inferior survival rates.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

BY was responsible for the conception of the work. JX, MD and JL obtained the data and contributed to the data analysis. TX, QY and BY checked and analyzed the data. BY wrote the manuscript. JX and BY confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was performed in line with the principles stated in the Declaration of Helsinki and was approved by the Ethics Committee of Hainan Hospital of Chinese People's Liberation Army General Hospital (approval no. 301HLFYLS15). The patients or their authorized relatives provided written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD, Sauer AG, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2020. CA Cancer J Clin. 70:145–164. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Song Y, Yang Y, Gao P, Chen X, Yu D, Xu Y, Zhao J and Wang Z: The preoperative neutrophil to lymphocyte ratio is a superior indicator of prognosis compared with other inflammatory biomarkers in resectable colorectal cancer. BMC Cancer. 17:7442017. View Article : Google Scholar : PubMed/NCBI

4 

Tan D, Fu Y, Tong W and Li F: Prognostic significance of lymphocyte to monocyte ratio in colorectal cancer: A meta-analysis. Int J Surg. 55:128–138. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Takada K, Kashiwagi S, Asano Y, Goto W, Ishihara S, Morisaki T, Shibutani M, Tanaka H, Hirakawa K and Ohira M: Clinical verification of body mass index and tumor immune response in patients with breast cancer receiving preoperative chemotherapy. BMC Cancer. 21:11292021. View Article : Google Scholar : PubMed/NCBI

6 

Zamorano AS, Hagemann AR, Morrison L, Lee JA, Liao LM, Brinton LA, Park Y and Toriola AT: Pre-diagnosis body mass index, physical activity and ovarian cancer mortality. Gynecol Oncol. 155:105–111. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Wang P, Li Y, Sun H, Liu S, Zhang R, Liu X and Zhu Z: Predictive value of body mass index for short-term outcomes of patients with esophageal cancer after esophagectomy: A meta-analysis. Ann Surg Oncol. 26:2090–2103. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Shukla S, Babcock Z, Pizzi L and Brunetti L: Impact of body mass index on survival and serious adverse events in advanced non-small cell lung cancer treated with bevacizumab: A meta-analysis of randomized clinical trials. Curr Med Res Opin. 37:811–817. 2021. View Article : Google Scholar : PubMed/NCBI

9 

Ye Z, Wei S, Zeng Y, Wang Y, Lin Z, Chen S, Xie Y, Zheng Q and Chen L: Prognostic value of preoperative body mass index for diabetic patients with non-metastasis gastric cancer: A single center experience. BMC Surg. 21:3202021. View Article : Google Scholar : PubMed/NCBI

10 

Lee S, Lee DH, Lee JH, Shin SJ, Lee HS, Park EJ, Baik SH, Lee KY and Kang J: Association of body mass index with survival in Asian patients with colorectal cancer. Cancer Res Treat. 54:860–872. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Chiu CC, Ho CH, Hung CM, Chao CM, Lai CC, Chen CM, Liao KM, Wang JJ, Wu YC, Shi HY, et al: Correlation of body mass index with oncologic outcomes in colorectal cancer patients: A large population-based study. Cancers. 13:35922021. View Article : Google Scholar : PubMed/NCBI

12 

Song N, Huang D, Jang D, Kim MJ, Jeong SY, Shin A and Park JW: Optimal body mass index cut-off point for predicting colorectal cancer survival in an asian population: A national health information database analysis. Cancers (Basel). 12:8302020. View Article : Google Scholar : PubMed/NCBI

13 

Guercio BJ, Zhang S, Venook AP, Ou FS, Niedzwiecki D, Lenz HJ, Innocenti F, Mullen BC, O'Neil BH, Shaw JE, et al: Body mass index and weight loss in metastatic colorectal cancer in CALGB (Alliance)/SWOG 80405. JNCI Cancer Spectr. 4:pkaa0242020. View Article : Google Scholar : PubMed/NCBI

14 

Kwak HD, Ju JK, Kang DW, Baek SJ, Kwak JM, Kim J and Kim SH: Outcomes according to body mass index following laparoscopic surgery in patients with colorectal cancer. J Minim Access Surg. 14:134–139. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Dragomir R, Dragomir AS, Negru A, Săftescu S, Popovici D, Schenker M, Lupușoru R and Negru Ș: Role of combining neutrophil-to-lymphocyte ratio and pretreatment body mass index in predicting progression-free survival in patients with non-small cell lung cancer treated with nivolumab. Exp Ther Med. 21:5262021. View Article : Google Scholar : PubMed/NCBI

16 

Wu YY, Chang KP, Lin CY, Pai PC, Wang HM, Hsu CL, Liao CT, Yen TC, Fang TJ, Huang SF, et al: Prognostic significance of combined pretreatment lymphocyte counts and body mass index in patients with head and neck cancer treated with radiation therapy. Cancer Med. 7:2808–2815. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, Somerfield MR, Hayes DF and Bast RC Jr: ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Onol. 24:5313–5327. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Thirunavukarasu P, Sukumar S, Sathaiah M, Mahan M, Pragatheeshwar KD, Pingpank JF, Zeh H III, Bartels CJ, Lee KKW and Bartlett DL: C-stage in colon cancer: implications of carcinoembryonic antigen biomarker in staging, prognosis, and management. J Natl Cancer Inst. 103:689–697. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Carriquiry LA and Piñeyro A: Should carcinoembryonic antigen be used in the management of patients with colorectal cancer? Dis Colon Rectum. 42:921–929. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Ailawadhi S, Sunga A, Rajput A, Yang GY, Smith J and Fakih M: Chemotherapy-induced carcinoembryonic antigen surge in patients with metastatic colorectal cancer. Oncology. 70:49–53. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Huang CS, Chen CY, Huang LK, Wang WS and Yang SH: Postoperative serum carcinoembryonic antigen levels cannot predict survival in colorectal cancer patients with type II diabetes. J Chin Med Assoc. 83:911–917. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Huang CS, Chen CY, Huang LK, Wang WS and Yang SH: Prognostic value of postoperative serum carcinoembryonic antigen levels in colorectal cancer patients who smoke. PLoS One. 15:e02336872020. View Article : Google Scholar : PubMed/NCBI

23 

Kacel EL, Kirsch JL, Sannes TS, Patidar S, Postupack R, Jensen S, Wong S, Garey S, Dodd S, Ulfig CM, et al: Interleukin-6 and body mass index, tobacco use, and sleep in gynecologic cancers. Health Psychol. 38:866–877. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Khaodhiar L, Ling PR, Blackburn GL and Bistrian BR: Serum levels of interleukin-6 and C-reactive protein correlate with body mass index across the broad range of obesity. JPEN J Parenter Enteral Nutr. 28:410–415. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Ullmann CD, Schlom J and Greiner JW: Interleukin-6 increases carcinoembryonic antigen and histocompatibility leukocyte antigen expression on the surface of human colorectal carcinoma cells. J Immunother (1991). 12:231–241. 1992. View Article : Google Scholar : PubMed/NCBI

26 

Belluco C, Nitti D, Frantz M, Toppan P, Basso D, Plebani M, Lise M and Jessup JM: Interleukin-6 blood level is associated with circulating carcinoembryonic antigen and prognosis in patients with colorectal cancer. Ann Surg Oncol. 7:133–138. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Bullen AW, Losowsky MS, Carter S, Patel S and Neville AM: Diagnostic usefulness of plasma carcinoembryonic antigen levels in acute and chronic liver disease. Gastroenterology. 73:673–678. 1977. View Article : Google Scholar : PubMed/NCBI

28 

Huang CS, Huang LK, Chen CY, Wang WS and Yang SH: Prognostic value of postoperative serum carcinoembryonic antigen levels in colorectal cancer patients with chronic kidney disease. Am J Surg. 221:162–167. 2021. View Article : Google Scholar : PubMed/NCBI

29 

MacGregor TP, Maughan TS and Sharma RA: Pathological grading of regression following neoadjuvant chemoradiation therapy: The clinical need is now. J Clin Pathol. 65:867–871. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Xu R, You YJ, Li F and Yan B: Postoperative fasting blood glucose predicts prognosis in stage I–III colorectal cancer patients undergoing resection. Gastroenterol Res Prac. 2020:24824092020.PubMed/NCBI

31 

Zhang YC, Liu Y, Qiu XM and Yan B: Concurrent comparison of the prognostic values of tumor budding, tumor stroma ratio, tumor infiltrating pattern and lymphocyte-to-monocyte ratio in colorectal cancer patients. Technol Cancer Res Treat. 20:153303382110458262021. View Article : Google Scholar : PubMed/NCBI

32 

Li Z, Zhao R, Cui Y, Zhou Y and Wu X: The dynamic change of neutrophil to lymphocyte ratio can predict clinical outcome in stage I–III colon cancer. Sci Rep. 8:94532018. View Article : Google Scholar : PubMed/NCBI

33 

Xie H, Huang S, Yuan G, Tang S and Gan J: Prognostic significance of preoperative fibrinogen-to-prealbumin ratio in patients with stage I–III colorectal cancer undergoing surgical resection: A retrospective cohort study. Biomed Res Int. 2021:39053532021.PubMed/NCBI

34 

DeLong ER, DeLong DM and Clarke-Pearson DL: Comparing the areas under two or more correlated receiver operating characteristic curves: A nonparametric approach. Biometrics. 44:837–845. 1988. View Article : Google Scholar : PubMed/NCBI

35 

Beom SH, Shin SJ, Kim CG, Kim JH, Hur H, Min BS, Lee KY, Kim NK and Ahn JB: Clinical significance of preoperative serum carcinoembryonic antigen within the normal range in colorectal cancer patients undergoing curative resection. Ann Surg Oncol. 27:2774–2783. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Cai Z, Xiao J, He X, Ke J, Zou Y, Chen Y, Wu X, Li X, Wang L, Wang J, et al: Accessing new prognostic significance of preoperative carcinoembryonic antigen in colorectal cancer receiving tumor resection: More than positive and negative. Cancer Biomark. 19:161–168. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Liu Z, Zhang Y, Niu Y, Li K, Liu X, Chen H and Gao C: A systematic review and meta-analysis of diagnostic and prognostic serum biomarkers of colorectal cancer. PLoS One. 9:e1039102014. View Article : Google Scholar : PubMed/NCBI

38 

Tumay V and Guner OS: The utility and prognostic value of CA 19-9 and CEA serum markers in the long-term follow up of patients with colorectal cancer. A single-center experience over 13 years. Ann Ital Chir. 91:494–503. 2020.PubMed/NCBI

39 

Björkman K, Mustonen H, Kaprio T, Kekki H, Pettersson K, Haglund C and Böckelman C: CA125: A superior prognostic biomarker for colorectal cancer compared to CEA, CA19-9 or CA242. Tumour Biol. 43:57–70. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Iacuzzo C, Germani P, Troian M, Mis TC, Giudici F, Osenda E, Bortul M and de Manzini N: Serum carcinoembryonic antigen pre-operative level in colorectal cancer: Revisiting risk stratification. ANZ J Surg. 91:E367–E374. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Li X, Xiong Z, Xie M, Huang Q, Jin L, Yin S, Chen S, Lan P and Lian L: Prognostic value of the ratio of carcinoembryonic antigen concentration to maximum tumor diameter in patients with stage II colorectal cancer. J Gastrointest Oncol. 12:1470–1481. 2021. View Article : Google Scholar : PubMed/NCBI

42 

Xie W, Huang X, Wei C, Mo X, Ru H, Zhang L, Ge L, Tang W and Liu J: Preoperative neutrophil-BMI ratio as a promising new marker for predicting tumor outcomes in colorectal cancer. Technol Cancer Res Treat. Feb 28–2022.(Epub ahead of print). View Article : Google Scholar

43 

Baqar AR, Wilkins S, Staples M, Lee CH, Oliva K and McMurrick P: The role of preoperative CEA in the management of colorectal cancer: A cohort study from two cancer centres. Int J Surg. 64:10–15. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Gold P and Freedman SO: Demonstration of tumor-specific antigens in huamn colonic carcinoma by immunological tolerance and absorption techniques. J Exp Med. 121:439–462. 1965. View Article : Google Scholar : PubMed/NCBI

45 

Goldstein MJ and Mitchell EP: Carcinoembryonic antigen in the staging and follow-up of patients with colorectal cancer. Cancer Invest. 23:338–351. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Bajenova O, Gorbunova A, Evsyukov I, Rayko M, Gapon S, Bozhokina E, Shishkin A and O'Brien SJ: The genome-wide analysis of carcinoembryonic antigen signaling by colorectal cancer cells using RNA sequencing. PLoS One. 11:e01612562016. View Article : Google Scholar : PubMed/NCBI

47 

Huang EY, Chang JC, Chen HH, Hsu CY, Hsu HC and Wu KL: Carcinoembryonic antigen as a marker of radioresistance in colorectal cancer: A potential role of macrophages. BMC Cancer. 18:3212018. View Article : Google Scholar : PubMed/NCBI

48 

Lee J, Meyerhardt JA, Giovannucci E and Jeon JY: Association between body mass index and prognosis of colorectal cancer: A meta-analysis of prospective cohort studies. PLoS One. 10:e01207062015. View Article : Google Scholar : PubMed/NCBI

49 

Daniel CR, Shu X, Ye Y, Gu J, Raju GS, Kopetz S and Wu X: Severe obesity prior to diagnosis limits survival in colorectal cancer patients evaluated at a large cancer centre. Br J Cancer. 114:103–109. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Meier CR, Krähenbühl S, Schlienger RG and Jick H: Association between body mass index and liver disorders: An epidemiological study. J Hepatol. 37:741–747. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Salvaggio A, Periti M, Miano L, Tavanelli M and Marzorati D: Body mass index and liver enzyme activity in serum. Clin Chem. 37:720–723. 1991. View Article : Google Scholar : PubMed/NCBI

52 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Jessup JM, Laguinge L, Lin S, Samara R, Aufman K, Battle P, Frantz M, Edmiston KH and Thomas P: Carcinoembryonic antigen induction of IL-10 and IL-6 inhibits hepatic ischemic/reperfusion injury to colorectal carcinoma cells. Int J Cancer. 111:332–337. 2004. View Article : Google Scholar : PubMed/NCBI

54 

Han J, Meng Q, Shen L and Wu G: Interleukin-6 induces fat loss in cancer cachexia by promoting white adipose tissue lipolysis and browning. Lipids Health Dis. 17:142018. View Article : Google Scholar : PubMed/NCBI

55 

Carson JA and Baltgalvis KA: Interleukin 6 as a key regulator of muscle mass during cachexia. Exerc Sport Sci Rev. 38:168–176. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Webster JM, Kempen LJAP, Hardy RS and Langen RCJ: Inflammation and skeletal muscle wasting during cachexia. Front Physiol. 11:5976752020. View Article : Google Scholar : PubMed/NCBI

57 

Lino-Silva LS, Anchondo-Nunez P, Chit-Huerta A, Aguilar-Romero E, Morales-Soto J, Salazar-Garcia JA, Guzman-Lopez CJ, Maldonado-Martinez HA, Meneses-Garcia A and Salcedo-Hernández RA: Stage I–III colon cancer patients with tumor deposits behave similarly to stage IV patients. Cross-section analysis of 392 patients. J Surg Oncol. 120:300–307. 2019.PubMed/NCBI

58 

Moon JY, Lee MR and Ha GW: Prognostic value of tumor deposits for long-term oncologic outcomes in patients with stage III colorectal cancer: A systematic review and meta-analysis. Int J Colorectal Dis. 37:141–151. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Naszai M, Kurjan A and Maughan TS: The prognostic utility of pre-treatment neutrophil-to-lymphocyte-ratio (NLR) in colorectal cancer: A systematic review and meta-analysis. Cancer Med. 10:5983–5997. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Gkekas I, Novotny J, Fabian P, Nemecek R, Palmqvist R, Strigard K, Pecen L, Svoboda T, Gurlich R and Gunnarsson U: Deficient mismatch repair as a prognostic marker in stage II colon cancer patients. Eur J Surg Oncol. 45:1854–1861. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Wang B, Li F, Zhou X, Ma Y and Fu W: Is microsatellite instability-high really a favorable prognostic factor for advanced colorectal cancer? A meta-analysis. World J Surg Oncol. 17:1692019. View Article : Google Scholar : PubMed/NCBI

62 

Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al: PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 372:2509–2520. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Flaherty DC, Jalas JR, Sim MS, Stojadinovic A, Protic M, Lee DJ and Bilchik AJ: The negative impact of body mass index on the tumor microenvironment in colon cancer: Results of a prospective trial. Ann Surg Oncol. 25:1374–1380. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Søreide K, Søreide JA and Kørner H: Prognostic role of carcinoembryonic antigen is influenced by microsatellite instability genotype and stage in locally advanced colorectal cancers. World J Surg. 35:888–894. 2011. View Article : Google Scholar : PubMed/NCBI

65 

André T, Boni C, Mounedji-Boudiaf L, Navarro M, Tabernero J, Hickish T, Topham C, Zaninelli M, Clingan P, Bridgewater J, et al: Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med. 350:2343–2345. 2004. View Article : Google Scholar : PubMed/NCBI

66 

André T, Meyerhardt J, Iveson T, Sobrero A, Yoshino T, Souglakos I, Grothey A, Niedzwiecki D, Saunders M, Labianca R, et al: Effect of duration of adjuvant chemotherapy for patients with stage III colon cancer (IDEA collaboration): final results from a prospective, pooled analysis of six randomised, phase 3 trials. Lancet Oncol. 21:1620–1629. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 24 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xiang J, Ding M, Lin J, Xue T, Ye Q and Yan B: Preoperative carcinoembryonic antigen to body mass index ratio contributes to prognosis prediction in colorectal cancer. Oncol Lett 24: 416, 2022
APA
Xiang, J., Ding, M., Lin, J., Xue, T., Ye, Q., & Yan, B. (2022). Preoperative carcinoembryonic antigen to body mass index ratio contributes to prognosis prediction in colorectal cancer. Oncology Letters, 24, 416. https://doi.org/10.3892/ol.2022.13536
MLA
Xiang, J., Ding, M., Lin, J., Xue, T., Ye, Q., Yan, B."Preoperative carcinoembryonic antigen to body mass index ratio contributes to prognosis prediction in colorectal cancer". Oncology Letters 24.5 (2022): 416.
Chicago
Xiang, J., Ding, M., Lin, J., Xue, T., Ye, Q., Yan, B."Preoperative carcinoembryonic antigen to body mass index ratio contributes to prognosis prediction in colorectal cancer". Oncology Letters 24, no. 5 (2022): 416. https://doi.org/10.3892/ol.2022.13536