Next Article in Journal
Arterioportal Fistulas (APFs) in Pediatric Patients: Single Center Experience with Interventional Radiological versus Conservative Management and Clinical Outcomes
Previous Article in Journal
The Association between Vitamin D and Nonalcoholic Fatty Liver Disease Assessed by Controlled Attenuation Parameter
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Preoperative Routine Laboratory Markers for Predicting Postoperative Recurrence and Death in Patients with Breast Cancer

1
Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
2
Devision of Breast Cancer, Department of General Surgery, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
3
Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, 211 Eonju-ro, Gangnam-gu, Seoul 06273, Korea
4
Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Gangnam Severance Hospital, 211 Eonju-ro, Gangnam-gu, Seoul 06273, Korea
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(12), 2610; https://doi.org/10.3390/jcm10122610
Submission received: 12 March 2021 / Revised: 26 April 2021 / Accepted: 11 June 2021 / Published: 13 June 2021
(This article belongs to the Section Obstetrics & Gynecology)

Abstract

:
Simple, convenient, and reliable preoperative prognostic indicators are needed to estimate the future risk of recurrences and guide the treatment decisions associated with breast cancer. We evaluated preoperative hematological markers related to recurrence and mortality and investigated independent risk factors for recurrence and mortality in patients after breast cancer surgery. We reviewed electronic medical records of patients with invasive breast cancer diagnosed at our tertiary institution between November 2005 and December 2010 and followed them until 2015. We compared two groups of patients classified according to recurrence or death and identified risk factors for postoperative outcomes. Data from 1783 patients were analyzed ultimately. Cancer antigen (CA) 15-3 and red cell distribution width (RDW) had the highest area under the curve values among several preoperative hematological markers for disease-free survival and overall survival (0.590 and 0.637, respectively). Patients with both preoperative CA 15-3 levels over 11.4 and RDW over 13.5 had a 1.7-fold higher risk of recurrence (hazard ratio (HR): 1.655; 95% confidence interval (CI): 1.154–2.374; p = 0.007) and mortality (HR: 1.723; 95% CI: 1.098–2.704; p = 0.019). In conclusion, relatively high preoperative RDW (>13.5) and CA 15-3 levels (>11.4) had the highest predictive power for mortality and recurrence, respectively. When RDW and CA 15-3 exceeded the cut-off value, the risk of recurrence and death also increased approximately 1.7 times.

1. Introduction

Breast cancer accounts for approximately 1 in 3 cancers [1,2] and is now regarded as the most common cancer globally; its incidence increased from 1.7 million in 2005 to 2.4 million cases in 2015 [3]. Above all, breast cancer is currently the main cause of cancer-related mortality in women [3]. Mercifully, overall breast cancer-related death rates decreased by 36% from 1989 to 2012 due to enhancements in early detection and systemic treatments [4,5]. However, nearly 20% of breast cancer patients are diagnosed at advanced stages and experience recurrence or distant metastasis within 5 years [6]. Both ipsilateral breast tumor recurrence and other locoregional recurrence are associated with significantly increased risk of distant disease and death [7,8]. Hence, for patients with breast cancer, outcome assessment is essential because it certainly impacts treatment decisions and prognosis.
The traditional prognostic factors for breast cancer are age, axillary lymph node status, tumor size, histological features (especially histological grade and lympho-vascular invasion), and molecular subtype including hormone receptor status and human epidermal growth factor receptor 2 expression [9,10,11,12]. In addition to demographic and basic clinical information, an increasing number of novel prognostic markers including hematological markers and perioperative anesthetic information have been explored and identified [13,14]. Recently, incorporation of various genetic and molecular biomarkers was attempted to develop new prognostic models for breast cancer [15]. However, most markers are not applied in clinical practice routinely, and their applicability may be limited due to high cost and the need for specialized expertise and equipment. A quick diagnosis of breast cancer is based on identification of inexpensive biomarkers [16]. Recently, accumulating evidence demonstrated that cancer-related systemic inflammation plays a significant role in the development and progression of several neoplastic diseases including breast cancer [17]. Therefore, there is a need to establish low-cost and simple prognostic indicators for breast cancer using routine hematological markers in a complete blood count.
We aimed to investigate preoperative hematological predictors for disease-free survival (DFS) and overall survival (OS) and identify independent risk factors for recurrence and mortality after breast cancer surgery.

2. Materials and Methods

2.1. Study Design

Our study was approved by the Hospital Research Ethics Committee and Institutional Review Board of Severance Hospital, Yonsei University Health System (approval number 3-2020-0134). The informed consent was waived in view of the retrospective nature of this study. We collected clinical data from the electronic medical records of consecutive patients with curative breast cancer surgery between November 2005 and December 2010. The follow-up documents were reviewed until December 2015. Patients who underwent multiple procedures, had incomplete information for anesthesia or surgery, or lacked preoperative hematological data were excluded.

2.2. Data Collection

Data related to the following patients’ demographic information were collected and reviewed: age; body mass index (BMI); presence of comorbidities; values of preoperative routine hematologic markers within 30 days before surgery, i.e., hemoglobin, hematocrit, red blood cell (RBC) count, RBC distribution width (RDW), platelet distribution width (PDW), white blood cell (WBC) count, neutrophil, lymphocyte, neutrophil-to-lymphocyte ratio (NLR), platelet, mean platelet volume (MPV), prothrombin time (PT), alkaline phosphatase (ALP), carcinoembryonic antigen (CEA), and cancer antigen (CA) 15-3; and administered anesthetics and analgesics. Surgical and histopathological information was also collected, including the conducted surgical procedures and the operating time, tumor receptor expression status including estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor (HER) 2, tumor-node-metastasis (TNM) stage, type of tumor, histological type, and any chemotherapy or radiotherapy performed. We used the American Joint Committee on Cancer 7th edition criteria to identify the TNM stage [18]. Postoperative assessments included medical history, physical examination, and laboratory and imaging tests to detect any evidence of recurrence. Local metastasis was deemed to have occurred if there was tumor presence in the ipsilateral breast, regional lymph nodes, and/or chest wall. Any recurrence at a distant site, including the contralateral axillary or supraclavicular lymph nodes, was defined as distant metastasis. Recurrence-free survival (RFS) was calculated from the date of surgery to the date when locoregional or distant metastasis was first detected. OS was defined as the interval between the date of the first curative surgery and the date of the last follow-up or death.

2.3. Statistical Analysis

Patients were categorized into two groups depending on whether they had recurrence (or died) or not. These two groups were compared using the independent two-sample t-test and χ2 test (Fisher’s exact test) for continuous variables and categorical variables, respectively.
The Contal and O’Quigley method was used to designate the point where the Kaplan–Meier survival curves of the two groups differed the most with the log-rank test and survival optimal cut-off values obtained. Sensitivity and specificity were calculated for each point using Youden Index cut-off values in the receiver operating characteristic (ROC) curve and area under curve (AUC) where the sum that reached the maximum was assessed. Additionally, predictive power was calculated using Harrell’s C-index with the bootstrapping method. The closer the C-index value was to 1.0, the greater the predictive power was. All potential confounders (chosen based on their clinical significance as reported in the literature) associated with recurrence and mortality after breast cancer surgery, which were age, BMI, co-morbidity, anesthetic agent, analgesics, transfusion, steroid, surgical data, surgical procedure, TNM stage, three receptor status, radiotherapy, neoadjuvant chemotherapy, adjuvant therapy, and histological grade, were analyzed using univariate and multivariate Cox regression methods. First, we performed univariate analysis to identify potential risk factors for postoperative recurrence and mortality; variables with p-values < 0.05 were further subjected to multivariate analysis, following which hazard ratio (HR) and the associated 95% confidence interval (CI) were calculated. Multivariate Cox regression processes were additively adjusted for several variables that could be confounding factors. For stepwise multivariate Cox regression analysis, variables including surgical procedure, TNM stage, estrogen receptor, and histological grade were adjusted.
We also conducted propensity score matching analysis for sensitivity test to assess the robustness of our findings regarding the relationship between variables and recurrence (or mortality). After factors including age, BMI, and comorbidities were matched; the greedy heuristic algorithm was used to identify the optimally matched groups without dropouts. This aided in excluding cases with differences that exceeded twice the standard deviation (SD) while matching similar propensity scores. As a result, a 1 to 5 matching score was chosen because it carried the strongest statistical power.
All statistical analyses were conducted with SAS, version 9.4 (SAS Institute Inc., Cary, NC, USA), except for Kaplan–Meier curves, which were constructed using the R statistics (R studio, Boston, MA, USA), version 3.6.2 (www.r-project.org, accessed on 13 June 2020).

3. Results

3.1. Subjects

We collected the data of 2645 patients who underwent surgery after breast cancer during the study period. Thirty patients who underwent multiple surgeries, 27 with unclear anesthetic methods, and 805 who lacked laboratory profiles within the 30 days before the surgery were excluded. Finally, 1783 patients were analyzed. The mean (SD) follow-up period for our study population was 68.7 (17.2) months. During the study period, the postoperative recurrence rate was 7.5% and the all-cause mortality rate was 5.7% (Figure 1).

3.2. Comparison of Patients’ Characteristics

A comparison of the characteristics of patients who had recurrence (n = 134) with those who did not have recurrence (n = 1649) is shown in Table 1. There were no significant differences regarding the demographic data and anesthetic information. Values of hemoglobin and CA 15-3 and surgical information including conducted procedure, TNM staging, estrogen and progesterone receptor status, histological grade, and status of neo/adjuvant chemotherapy were significantly different between the two groups even after 1 to 5 propensity score matching.
A comparison of the characteristics of patients who died (n = 101) and those who survived (n = 1682) is shown in Table 2. The groups were comparable in terms of demographic data and anesthetic information. Hemoglobin, hematocrit, RBC count, RDW, MPV, ALP, and CA 15-3 were significantly different between the two groups. Moreover, surgical factors including the procedure performed, TNM stage, estrogen and progesterone receptor status, histological grade, and status of neo/adjuvant chemotherapy were also significantly different between the two groups even after 1 to 5 propensity score matching.

3.3. Predictors for Postoperative Recurrence and Mortality after Breast Cancer Surgery

Figure 2 shows the Kaplan–Meier curve with a survival optimal cut-off value and ROC curve of CA 15-3, which was the biomarker with the highest predictive power for postoperative recurrence. When the survival optimal cut-off value of CA15-3 was 11.4 (p value < 0.001), the Harrell’s C-index (AUC) was 0.590. RDW showed the highest predictive power regarding postoperative mortality, and the survival optimal cut-off value was 13.5 (p value < 0.001). The Harrell’s C-index of RDW was 0.637 (Figure 3).

3.4. Independent Risk Factors for Postoperative Recurrence and Mortality in Breast Cancer Surgery

Table 3 lists the factors that affected postoperative breast cancer recurrence as revealed by competing risk analysis. Multivariate Cox regression with 1 to 5 propensity score matching showed that perioperative CA 15-3 level > 11.4 (HR, 1.665; 95% CI, 1.154–2.374; p = 0.007), mastectomy (HR, 2.169; 95% CI, 1.419–3.314; p < 0.001) compared with breast-conserving surgery, TNM stage 3 (HR, 3.481; 95% CI, 2.136–5.672; p < 0.001) compared with TNM stage 1, neoadjuvant chemotherapy (HR, 1.957; 95% CI, 1.127–3.397; p = 0.045), and moderate (HR, 2.071; 95% CI, 1.139–3.766; p = 0.017) or poor histological grade (HR, 3.878; 95% CI, 2.044–7.357; p < 0.001) compared with good histological grade increased the risks for cancer recurrence.
The factors associated with postoperative mortality after breast cancer surgery are presented in Table 4. Multivariate Cox regression analysis with 1 to 5 propensity score matching showed that RDW > 13.5 (HR, 1.723; 95% CI, 1.098–2.704; p = 0.019), NLR > 2.82 (HR, 1.771; 95% CI, 1.108–2.832; p = 0.017), ALP > 76 (HR, 2.257; 95% CI, 1.412–3.607; p < 0.001), CEA > 1.57 (HR, 1.553; 95% CI, 1.007–2.395; p = 0.047), mastectomy (HR, 2.993; 95% CI, 1.750–5.118; p < 0.001) compared with breast-conserving surgery, TNM stage 2 (HR, 2.310; 95% CI, 1.239–4.306; p = 0.009) and 3 (HR, 6.563; 95% CI, 3.482–12.370; p < 0.001) compared with TNM stage 1, neoadjuvant chemotherapy (HR, 1.442; 95% CI, 1.048–3.409; p = 0.035), and moderate (HR, 3.253; 95% CI, 1.412–7.491; p = 0.006) and poor histological grade (HR, 6.455; 95% CI, 2.757–15.113; p < 0.001) compared with good histological grade remained significant factors for an increased risk of postoperative mortality.

4. Discussion

In our study, factors including preoperative routine hematologic markers affecting the prognosis after breast cancer surgery were investigated through follow-up for over 5 years. Compared with previous studies related to the outcomes of oncologic patients using blood tests, our study calculated the survival optimal cut-off value and HR of hematological markers as well as the predictive power of long-term recurrence and mortality after breast surgery using a large sample size. Preoperative relatively high RDW (>13.5) had the highest predictive power for postoperative mortality, and preoperative relatively high CA 15-3 levels (>11.4) had the highest predictive power for recurrence. When these two factors exceeded the cut-off value the risk of recurrence and death also increased approximately 1.7 times.
Even though many novel prognostic markers have been explored and identified, the main problem in most of these studies is that the biomarkers depend on complex molecular or genetic tests [12,19,20,21,22]. In our study, considering that CA 15-3 and RDW are easily available and inexpensive markers and parameters that are routinely assessed in blood examination, they could be used as potential novel accurate and reproducible indices to identify breast cancer patients with poorer outcomes.
Complete blood count is essentially used to assess anemia-related diseases, which usually have increased risk for surgical mortality and can often be treated before surgery [23,24]. We have used RDW, which describes the size variability of circulating red blood cells, for the workup of anemia and it has recently been suggested as an inflammatory biomarker [25]. Another mechanism for the association between chronic inflammation and elevated RDW could be increased red blood cell fragility in inflammatory states secondary to the rise of lipid oxidation [26]. Additionally, oxidation of hemoglobin from exposure to free radicals [27] signals erythrocyte removal and increases RDW. Other explanations for the relationship include the increased release and binding of free histones to erythrocytes, which increases their fragility [28]. Similar to inflammation, the status of oxidative stress may reduce RBC survival and lead to elevated RDW [29].
Inflammation in the tumor microenvironment promotes tumor growth, invasion, angiogenesis, and, ultimately, metastasis [30,31,32,33]. It is now extensively recognized that smoldering inflammation plays an important role in the initiation and progression of cancer [31,33]. In the same manner, Seretis et al. [23] reported RDW to be a useful biomarker in distinguishing benign and malignant breast tumors. RDW has also recently been reported as a potential prognostic factor in breast cancer [34,35]. Furthermore, RDW is considered a biomarker that influences the increased mortality rate in non-cardiac surgery because anemia, malnutrition, and oxidative stress may be indicated by high RDW [36]. However, our RDW cut-off value (13.5) was relatively low compared to those reported in other studies (13.75, 13.82, and 13.7). Accordingly, we should pay more attention and monitor patients that have higher preoperational RDW and try to optimize RDW before surgery providing interventions such as nutritional support or anti-inflammatory medications [36]. RDW, which is a potential biomarker of inflammation, might be utilized to risk-stratify cancer surgical patients and monitor the efficacy of risk-modifying strategies.
Preoperative levels of CA15-3 have an independent association with prognosis in patients with early-stage breast cancer [37]. It has been shown to be extremely sensitive for distant metastases, especially those in the bone and liver [38,39,40]. However, there is currently no clear clinical cut-off for considering CA15-3 abnormal; cut-offs reported in other studies have varied from 22 to 60 U/mL [41,42]. The choice of cut-off to classify CA15-3 could indeed affect the conclusions; a lower cut-off would likely decrease the predictive value, while a higher cut-off may increase the predictive value [43]. In our study, CA 15-3 was associated with the risk of recurrence and had predictive power. The cut-off value was 11.4, and the predicted power was relatively low, with an AUC of 0.59 (negative predictive value 94.8%, positive predictive value 11.4%). Although it was a cut-off value in our population, a slight increase in CA 15-3 before surgery may indicate an increased risk of recurrence. Persistently elevated tumor markers including CA 15-3 are associated with recurrence or treatment resistance, which may be useful in monitoring treatment effects [44].
CA 15-3 can be easily and inexpensively tested, but its sensitivity is relatively low [45,46]. The role of CA 15-3 in breast cancer surveillance remains controversial. Moreover, there is no definite guideline on how often to test; this test alone can increase the patient’s anxiety enough. Because there were insufficient data including prospective randomized clinical trials to provide evidence on whether detection and treatment of occult or asymptomatic metastases using CA 15-3 impact on the most significant prognosis, ASCO guidelines for follow-up and management of patients with breast cancer [47] have not recommended the use of CA 15.3 for detecting recurrence routinely. On the contrary, European Group on Tumor Markers has recommended the use of CA15-3 levels for assessing prognosis, early detection of disease progression, and treatment monitoring in breast cancer [44]. It was also reported that the preoperative elevation of CA15-3 was associated with aggressive characteristics and worse overall survival in breast cancer patients without distant metastasis [48,49,50]. Consequently, routine follow-ups of CA 15-3 to check for relapse after treatment of breast cancer have not yet been not recommended by major international guidelines [40,47,51] because of its high frequency of false negative and false positive prediction.
The Nottingham Prognostic Index (NPI), known for prognostic index, is also widely used for predicting survival in operable primary patients with breast cancer [52,53]. It is based on tumor size, histologic grade, and lymph node status [54] and is simple and easy for clinical use. It shows suboptimal performance in predicting tumor recurrence. The AUCs were 0.66 and 0.63 in the training and test cohorts, respectively [55,56,57,58]. In our findings, RDW had a similar predictive power as that of NPI indicating that information can be easily obtained through blood tests that can approximately predict a patient’s prognosis. Biomarkers alone, however, are insufficient to predict the postoperative prognosis in patients with breast cancer, but correction of RDW before surgery may lower the risk of postoperative death. Moreover, CA 15-3 is highly related to recurrence, and it can act as a useful marker for cancer tracing.
Our study has some limitations, including its retrospective cohort design, which was associated with the selecting bias inclusion of an entire population. Although the propensity matching score carefully balanced age, sex, and a large host of preoperative comorbidities that might affect the relationship between RDW and mortality, there still might be unbalanced preoperative comorbidities or medications explaining the relationship. These could include differences in the cofactors of hemoglobin production or ischemic diseases. Therefore, it needs future investigation in a less heterogeneous cohort. Another limitation is that we did not investigate precise mechanisms including molecular or genetic explanations regarding how CA 15-3 and RDW act on the oncological outcomes, especially on breast cancer. In addition, the association scores of multiple variables should be more clearly identified because CA 15-3 was one of the several significant variables included in the multivariate Cox regression analysis including propensity matching. Further prospective studies are needed to determine the role of hematological markers for predictors.
The calculated RDW cut-off value related to the risk of death was 13.5, which was lower than that reported previously. This shows that even a slight increase in RDW in breast cancer patients may have a correlation with postoperative mortality. There may be several reasons for the increase in RDW; therefore, efforts to normalize RDW before surgery are necessary at least in patients with breast cancer. CA15-3 was associated solely with the risk of breast cancer recurrence in our study. Although the AUC was relatively low for predicting recurrence, monitoring of CA 15-3 levels would be somewhat helpful in the follow-ups for breast cancer recurrence or metastasis.

Author Contributions

Conceptualization, Y.-C.Y. and M.-H.K.; Data curation, S.P., H.-J.K., H.-E.J., J.-Y.K., and M.-H.K.; Formal analysis, Y.-C.Y. and M.-H.K.; Investigation, Y.-C.Y. and M.-H.K.; Methodology, Y.-C.Y., S.P. and M.-H.K.; Supervision, M.-H.K.; Writing—original draft: Y.-C.Y. and M.-H.K.; Writing—review and editing, Y.-C.Y. and M.-H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Our study was approved by the Hospital Research Ethics Committee and Institutional Review Board of Severance Hospital, Yonsei University Health System (approval number 3-2020-0134).

Informed Consent Statement

The informed consent was waived in view of the retrospective nature of this study.

Data Availability Statement

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

Acknowledgments

We greatly thank the Biostatistics Collaboration Unit and the Electronic Medical Record Team at Yonsei University College of Medicine for their contributions to this study.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

RBCred blood cell
WBCwhite blood cell
RDWred blood cell distribution width
PDWplatelet distribution width
NLRneutrophil-to-lymphocyte ratio
MPVmean platelet volume
PTprothrombin time
ALPalkaline phosphatase
CEAcarcinoembryonic antigen
CAcancer antigen
TIVAtotal intravenous anesthesia
NSAIDsnon-steroidal anti-inflammatory drugs
BCSbreast-conserving surgery
TNMtumor-node-metastasis
HER2human epidermal growth factor receptor 2
CIconfidence interval
HRhazard ratio
ROCreceiver operating characteristic

References

  1. Wang, H.; Naghavi, M.; Allen, C.; Barber, R.M.; Bhutta, Z.A.; Casey, D.C.; Charlson, F.J.; Chen, A.Z.; Coates, M.M.; Coggeshall, M.; et al. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet 2016, 388, 1459–1544. [Google Scholar] [CrossRef] [Green Version]
  2. DeSantis, C.; Ma, J.; Bryan, L.; Jemal, A. Breast cancer statistics, 2013. CA A Cancer J. Clin. 2013, 64, 52–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fitzmaurice, C.; Allen, C.; Barber, R.M.; Barregard, L.; Bhutta, Z.A.; Brenner, H.; Satpathy, M. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2017, 3, 524–548. [Google Scholar]
  4. Amoroso, V.; Generali, D.; Buchholz, T.; Cristofanilli, M.; Pedersini, R.; Curigliano, G.; Daidone, M.G.; Di Cosimo, S.; Dowsett, M.; Fox, S.; et al. International Expert Consensus on Primary Systemic Therapy in the Management of Early Breast Cancer: Highlights of the Fifth Symposium on Primary Systemic Therapy in the Management of Operable Breast Cancer, Cremona, Italy (2013). J. Natl. Cancer Inst. Monogr. 2015, 2015, 90–96. [Google Scholar] [CrossRef]
  5. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.-J.; Albain, K.S.; Andre, F.; Bergh, J.; et al. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223. [Google Scholar] [CrossRef] [PubMed]
  6. Peto, R.; Davies, C.; Godwin, J.; Gray, R.; Pan, H.C.; Clarke, M.; Pritchard, K. Comparisons between different polychemotherapy regimens for early breast cancer: Meta-analyses of long-term outcome among 100,000 women in 123 randomised trials. Lancet 2012, 379, 432–444. [Google Scholar]
  7. Wapnir, I.L.; Anderson, S.J.; Mamounas, E.P.; Jeong, J.; Tan-Chiu, E.; Fisher, B.; Wolmark, N. Prognosis After Ipsilateral Breast Tumor Recurrence and Locoregional Recurrences in Five National Surgical Adjuvant Breast and Bowel Project Node-Positive Adjuvant Breast Cancer Trials. J. Clin. Oncol. 2006, 24, 2028–2037. [Google Scholar] [CrossRef] [PubMed]
  8. Fisher, B.; Anderson, S.; Bryant, J.; Margolese, R.G.; Deutsch, M.; Fisher, E.R.; Jeong, J.; Wolmark, N. Twenty-Year Follow-up of a Randomized Trial Comparing Total Mastectomy, Lumpectomy, and Lumpectomy plus Irradiation for the Treatment of Invasive Breast Cancer. N. Engl. J. Med. 2002, 347, 1233–1241. [Google Scholar] [CrossRef] [Green Version]
  9. Schnitt, S.J. Classification and prognosis of invasive breast cancer: From morphology to molecular taxonomy. Mod. Pathol. 2010, 23, S60–S64. [Google Scholar] [CrossRef] [Green Version]
  10. Ito, M.; Shien, T.; Omori, M.; Mizoo, T.; Iwamoto, T.; Nogami, T.; Motoki, T.; Taira, N.; Doihara, H.; Miyoshi, S. Evaluation of aldehyde dehydrogenase 1 and transcription factors in both primary breast cancer and axillary lymph node metastases as a prognostic factor. Breast Cancer 2015, 23, 437–444. [Google Scholar] [CrossRef]
  11. Watanabe, G.; Ishida, T.; Furuta, A.; Takahashi, S.; Watanabe, M.; Nakata, H.; Kato, S.; Ishioka, C.; Ohuchi, N. Combined Immunohistochemistry of PLK1, p21, and p53 for Predicting TP53 Status. Am. J. Surg. Pathol. 2015, 39, 1026–1034. [Google Scholar] [CrossRef]
  12. Chen, B.; Tang, H.; Liu, X.; Liu, P.; Yang, L.; Xie, X.; Ye, F.; Song, C.; Xie, X.; Wei, W. miR-22 as a prognostic factor targets glucose transporter protein type 1 in breast cancer. Cancer Lett. 2015, 356, 410–417. [Google Scholar] [CrossRef] [PubMed]
  13. Wall, T.; Sherwin, A.; Ma, D.; Buggy, D. Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: A narrative review. Br. J. Anaesth. 2019, 123, 135–150. [Google Scholar] [CrossRef] [PubMed]
  14. Bajwa, S.J.S.; Anand, S.; Kaur, G. Anesthesia and cancer recurrences: The current knowledge and evidence. J. Cancer Res. Ther. 2015, 11, 528–534. [Google Scholar] [CrossRef] [PubMed]
  15. Rivenbark, A.G.; O’Connor, S.M.; Coleman, W.B. Molecular and cellular heterogeneity in breast cancer: Challenges for personalized medicine. Am. J. Pathol. 2013, 183, 1113–1124. [Google Scholar] [CrossRef] [Green Version]
  16. Stegner, D.; Dütting, S.; Nieswandt, B. Mechanistic explanation for platelet contribution to cancer metastasis. Thromb. Res. 2014, 133, S149–S157. [Google Scholar] [CrossRef]
  17. Koh, C.-H.; Bhoopathy, N.; Ng, K.-L.; Jabir, R.S.; Tan, G.-H.; See, M.H.; Jamaris, S.; Taib, N. Utility of pre-treatment neutrophil–lymphocyte ratio and platelet–lymphocyte ratio as prognostic factors in breast cancer. Br. J. Cancer 2015, 113, 150–158. [Google Scholar] [CrossRef]
  18. Hammond, M.E.H.; Hayes, D.F.; Dowsett, M.; Allred, D.C.; Hagerty, K.L.; Badve, S.; Fitzgibbons, P.L.; Francis, G.; Goldstein, N.S.; Hayes, M.; et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Immunohistochemical Testing of Estrogen and Progesterone Receptors in Breast Cancer. J. Clin. Oncol. 2010, 28, 2784–2795. [Google Scholar] [CrossRef] [Green Version]
  19. Zwart, W.; Flach, K.D.; Rudraraju, B.; Abdel-Fatah, T.M.; Gojis, O.; Canisius, S.; Moore, D.; Nevedomskaya, E.; Opdam, M.; Droog, M.; et al. SRC3 Phosphorylation at Serine 543 Is a Positive Independent Prognostic Factor in ER-Positive Breast Cancer. Clin. Cancer Res. 2016, 22, 479–491. [Google Scholar] [CrossRef] [Green Version]
  20. Rui, X.; Li, Y.; Jin, F.; Li, F. TMPRSS3 is a novel poor prognostic factor for breast cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 5435–5442. [Google Scholar] [PubMed]
  21. Dong, L.-L.; Chen, L.-M.; Wang, W.-M.; Zhang, L.-M. Decreased expression of microRNA-124 is an independent unfavorable prognostic factor for patients with breast cancer. Diagn. Pathol. 2015, 10, 1–5. [Google Scholar] [CrossRef] [Green Version]
  22. Seong, M.-K.; Lee, J.-Y.; Byeon, J.; Sohn, Y.-J.; Seol, H.; Lee, J.-K.; Kim, E.-K.; Kim, H.-A.; Noh, W.C. Bcl-2 is a highly significant prognostic marker of hormone-receptor-positive, human epidermal growth factor receptor-2-negative breast cancer. Breast Cancer Res. Treat. 2015, 150, 141–148. [Google Scholar] [CrossRef]
  23. Feely, M.; Collins, C.S.; Daniels, P.R.; Kebede, E.B.; Jatoi, A.; Mauck, K.F. Preoperative testing before noncardiac surgery: Guidelines and recommendations. Am. Fam. Physician 2013, 87, 414–418. [Google Scholar] [PubMed]
  24. Burns, C.D.; Brown, J.P.; Corwin, H.L.; Gross, I.; Ozawa, S.J.; Shander, A. Special Report From the Society for the Advancement of Blood Management: The Choosing Wisely Campaign. Anesth. Analg. 2019, 129, 1381–1386. [Google Scholar] [CrossRef]
  25. Lippi, G.; Targher, G.; Montagnana, M.; Salvagno, G.L.; Zoppini, G.; Guidi, G.C. Relation between red blood cell distribution width and inflammatory biomarkers in a large cohort of unselected outpatients. Arch. Pathol. Lab. Med. 2009, 133, 628–632. [Google Scholar] [CrossRef] [PubMed]
  26. Maurya, P.K.; Kumar, P.; Chandra, P. Biomarkers of oxidative stress in erythrocytes as a function of human age. World J. Methodol. 2015, 5, 216–222. [Google Scholar] [CrossRef] [Green Version]
  27. Mohanty, J.G.; Nagababu, E.; Rifkind, J.M. Red blood cell oxidative stress impairs oxygen delivery and induces red blood cell aging. Front. Physiol. 2014, 5, 84. [Google Scholar] [CrossRef] [Green Version]
  28. Kordbacheh, F.; O’Meara, C.H.; Coupland, L.A.; Lelliott, P.M.; Parish, C. Extracellular histones induce erythrocyte fragility and anemia. Blood 2017, 130, 2884–2888. [Google Scholar] [CrossRef] [PubMed]
  29. Abrahan, I.L.L.; Ramos, J.D.A.; Cunanan, E.L.; Tiongson, M.D.A.; Punzalan, F.E.R. Red Cell Distribution Width and Mortality in Patients With Acute Coronary Syndrome: A Meta-Analysis on Prognosis. Cardiol. Res. 2018, 9, 144–152. [Google Scholar] [CrossRef]
  30. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F.R. Cancer-related inflammation. Nat. Cell Biol. 2008, 454, 436–444. [Google Scholar] [CrossRef]
  31. Balkwill, F.R.; Mantovani, A. Cancer-related inflammation: Common themes and therapeutic opportunities. Semin. Cancer Biol. 2012, 22, 33–40. [Google Scholar] [CrossRef]
  32. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Allen, M.D.; Jones, L.J. The role of inflammation in progression of breast cancer: Friend or foe? Int. J. Oncol. 2015, 47, 797–805. [Google Scholar] [CrossRef] [PubMed]
  34. Huang, D.-P.; Ma, R.-M.; Xiang, Y.-Q. Utility of Red Cell Distribution Width as a Prognostic Factor in Young Breast Cancer Patients. Medicine 2016, 95, e3430. [Google Scholar] [CrossRef] [PubMed]
  35. Yao, D.; Wang, Z.; Cai, H.; Li, Y.; Li, B. Relationship between red cell distribution width and prognosis in patients with breast cancer after operation: A retrospective cohort study. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Olafsson, H.B.; Sigurdarson, G.A.; Christopher, K.B.; Karason, S.; Sigurdsson, G.H.; Sigurdsson, M.I. A retrospective cohort study on the association between elevated preoperative red cell distribution width and all-cause mortality after noncardiac surgery. Br. J. Anaesth. 2020, 124, 718–725. [Google Scholar] [CrossRef] [PubMed]
  37. Buamah, P.K.; Bent, D.J.; Bodger, W.A.H.; Skillen, A.W. A profile of serum CA 15-3, carcinoembryonic antigen, alkaline phosphatase, and γ-glutamyl transferase levels in patients with breast cancer. J. Surg. Oncol. 1993, 53, 84–87. [Google Scholar] [CrossRef] [PubMed]
  38. Basuyau, J.P.; Brunelle, P.; Charrot, P.; Chevallier, B.; Delapierre, F.; Graic, Y.; Julien, J.P.; Veyret, C. CA 15.3 and early diagnosis of recurrence in breast cancer. Bull Cancer 1993, 80, 213–218. [Google Scholar] [PubMed]
  39. Lauro, S.; Trasatti, L.; Bordin, F.; Lanzetta, G.; Bria, E.; Gelibter, A.; Reale, M.G.; Vecchione, A. Comparison of CEA, MCA, CA 15-3 and CA 27-29 in follow-up and monitoring therapeutic response in breast cancer patients. Anticancer Res. 1999, 19, 3511–3515. [Google Scholar]
  40. NCCN Clinical Practice Guidelines in Oncology; Breast Cancer Version 5; NCCN Guidelines®: Plymouth Meeting, PA, USA, 2020.
  41. Hayes, D.F.; Zurawski, V.R.; Kufe, D.W. Comparison of circulating CA15-3 and carcinoembryonic antigen levels in patients with breast cancer. J. Clin. Oncol. 1986, 4, 1542–1550. [Google Scholar] [CrossRef]
  42. Molina, R.; Jo, J.; Filella, X.; Zanón, G.; Farrus, B.; Muñoz, M.; Latre, M.L.; Pahisa, J.; Velasco, M.; Fernandez, P.; et al. C-erbB-2, CEA and CA 15.3 serum levels in the early diagnosis of recurrence of breast cancer patients. Anticancer Res. 1999, 19, 2551–2555. [Google Scholar]
  43. Wu, S.-G.; He, Z.-Y.; Zhou, J.; Sun, J.-Y.; Li, F.-Y.; Lin, Q.; Guo, L.; Lin, H.-X. Serum levels of CEA and CA15-3 in different molecular subtypes and prognostic value in Chinese breast cancer. Breast 2014, 23, 88–93. [Google Scholar] [CrossRef]
  44. Uehara, M.; Kinoshita, T.; Hojo, T.; Akashi-Tanaka, S.; Iwamoto, E.; Fukutomi, T. Long-term prognostic study of carcinoembryonic antigen (CEA) and carbohydrate antigen 15-3 (CA 15-3) in breast cancer. Int. J. Clin. Oncol. 2008, 13, 447–451. [Google Scholar] [CrossRef] [PubMed]
  45. Li, X.; Dai, D.; Chen, B.; Tang, H.; Xie, X.; Wei, W. Clinicopathological and Prognostic Significance of Cancer Antigen 15-3 and Carcinoembryonic Antigen in Breast Cancer: A Meta-Analysis including 12,993 Patients. Dis. Markers 2018, 2018, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Molina, R.; Barak, V.; Van Dalen, A.; Duffy, M.J.; Einarsson, R.; Gion, M.; Goike, H.; Lamerz, R.; Nap, M.; Sölétormos, G.; et al. Tumor Markers in Breast Cancer—European Group on Tumor Markers Recommendations. Tumor Biol. 2005, 26, 281–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Harris, L.; Fritsche, H.; Mennel, R.; Norton, L.; Ravdin, P.; Taube, S.; Somerfield, M.R.; Hayes, D.F.; Bast, R.C., Jr. American society of clinical oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J. Clin. Oncol. 2007, 25, 5287–5312. [Google Scholar] [CrossRef] [Green Version]
  48. Park, B.-W.; Oh, J.-W.; Kim, J.-H.; Park, S.H.; Kim, K.-S.; Lee, K.S. Preoperative CA 15-3 and CEA serum levels as predictor for breast cancer outcomes. Ann. Oncol. 2007, 19, 675–681. [Google Scholar] [CrossRef] [PubMed]
  49. Lee, J.S.; Park, S.; Park, J.M.; Cho, J.H.; Kim, S.I.; Park, B.-W. Elevated levels of preoperative CA 15-3 and CEA serum levels have independently poor prognostic significance in breast cancer. Ann. Oncol. 2012, 24, 1225–1231. [Google Scholar] [CrossRef]
  50. Nam, S.E.; Lim, W.; Jeong, J.; Lee, S.; Choi, J.; Park, H.; Jung, Y.; Jung, S.P.; Bae, S.Y. The prognostic significance of preoperative tumor marker (CEA, CA15-3) elevation in breast cancer patients: Data from the Korean Breast Cancer Society Registry. Breast Cancer Res. Treat. 2019, 177, 669–678. [Google Scholar] [CrossRef]
  51. Senkus, E.; Kyriakides, S.; Ohno, S.; Penault-Llorca, F.; Poortmans, P.; Rutgers, E.; Zackrisson, S.; Cardoso, F. Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2015, 26, v8–v30. [Google Scholar] [CrossRef]
  52. Malmström, P.; Bendahl, P.-O.; Boiesen, P.; Brünner, N.; Idvall, I.; Fernö, M. S-Phase Fraction and Urokinase Plasminogen Activator Are Better Markers for Distant Recurrences Than Nottingham Prognostic Index and Histologic Grade in a Prospective Study of Premenopausal Lymph Node–Negative Breast Cancer. J. Clin. Oncol. 2001, 19, 2010–2019. [Google Scholar] [CrossRef] [PubMed]
  53. Blamey, R.; Ellis, I.; Pinder, S.; Lee, A.; Macmillan, R.; Morgan, D.; Robertson, J.; Mitchell, M.; Ball, G.; Haybittle, J.; et al. Survival of invasive breast cancer according to the Nottingham Prognostic Index in cases diagnosed in 1990–1999. Eur. J. Cancer 2007, 43, 1548–1555. [Google Scholar] [CrossRef]
  54. Galea, M.H.; Blamey, R.W.; Elston, C.E.; Ellis, I.O. The Nottingham prognostic index in primary breast cancer. Breast Cancer Res. Treat. 1992, 22, 207–219. [Google Scholar] [CrossRef] [PubMed]
  55. Tan, W.; Luo, W.; Jia, W.; Liang, G.; Xie, X.; Zheng, W.; Song, E.; Su, F.; Gong, C. A combination of Nottingham prognostic index and IHC4 score predicts pathological complete response of neoadjuvant chemotherapy in estrogen receptor positive breast cancer. Oncotarget 2016, 7, 87312–87322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Wu, X.; Ye, Y.; Barcenas, C.H.; Chow, W.-H.; Meng, Q.H.; Chavez-MacGregor, M.; Hildebrandt, M.A.T.; Zhao, H.; Gu, X.; Deng, Y.; et al. Personalized Prognostic Prediction Models for Breast Cancer Recurrence and Survival Incorporating Multidimensional Data. J. Natl. Cancer Inst. 2017, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Drukker, C.A.; Nijenhuis, M.V.; Bueno-De-Mesquita, J.M.; Retèl, V.; Van Harten, W.H.; Van Tinteren, H.; Wesseling, J.; Schmidt, M.K.; Veer, L.J.V.; Sonke, G.S.; et al. Optimized outcome prediction in breast cancer by combining the 70-gene signature with clinical risk prediction algorithms. Breast Cancer Res. Treat. 2014, 145, 697–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Kim, W.; Kim, K.S.; Lee, J.E.; Noh, D.-Y.; Kim, S.-W.; Jung, Y.S.; Park, M.Y.; Park, R.W. Development of Novel Breast Cancer Recurrence Prediction Model Using Support Vector Machine. J. Breast Cancer 2012, 15, 230–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. CONSORT Flow Diagram.
Figure 1. CONSORT Flow Diagram.
Jcm 10 02610 g001
Figure 2. (A) Kaplan–Meier survival curve and (B) ROC curve for postoperative recurrence in association with CA 15-3 in patients with breast cancer surgery.
Figure 2. (A) Kaplan–Meier survival curve and (B) ROC curve for postoperative recurrence in association with CA 15-3 in patients with breast cancer surgery.
Jcm 10 02610 g002
Figure 3. (A) Kaplan–Meier survival curve and (B) ROC curve for postoperative mortality in association with RDW in patients with breast cancer surgery.
Figure 3. (A) Kaplan–Meier survival curve and (B) ROC curve for postoperative mortality in association with RDW in patients with breast cancer surgery.
Jcm 10 02610 g003
Table 1. Patient characteristics based on postoperative recurrence.
Table 1. Patient characteristics based on postoperative recurrence.
Before 1 to 5 Propensity Score MatchingAfter 1 to 5 Propensity Score Matching
ParametersNon-Recurrence
(n = 1649)
Recurrence
(n = 134)
p-ValueNon-Recurrence
(n = 645)
Recurrence
(n = 129)
p-Value
Demographic data
Age (yr)50.1 ± 9.949.7 ± 11.20.68348.7 ± 9.4949.69 ± 10.880.333
Body mass index (kg/m2)23.3 ± 3.223.0 ± 3.00.24423.0 ± 3.123.1 ± 3.00.851
Comorbidities
Hypertension321 (19.5)30 (22.4)0.413114 (17.7)27 (20.9)0.382
Diabetes mellitus110 (6.7)13 (9.7)0.18337 (5.7)12 (9.3)0.129
Cardiac43 (2.6)4 (3.0)0.77718 (2.8)4 (3.1)0.775
Pulmonary27 (1.6)4 (3.0)0.28710 (1.5)0 (0.0)0.384
Endocrine 79 (4.8)7 (5.2)0.82226 (4.0)7 (5.4)0.474
Renal10 (0.6)0 (0.0)>0.9990 (0.0)0 (0.0)>0.999
Liver8 (0.5)1 (0.8)0.5063 (0.5)1 (0.8)0.519
Neurological24 (1.5)2 (1.5)>0.9999 (1.4)1 (0.8)>0.999
Others10 (0.6)1 (0.8)0.5783 (0.5)1 (0.8)0.519
Hematologic markers
Hemoglobin (g/dl)12.9 ± 1.212.6 ± 1.40.03212.8 ± 1.212.6 ± 1.50.098
Hematocrit (%)39.0 ± 3.338.2 ± 4.20.02338.9 ± 3.238.1 ± 4.20.070
RBC count (106/µL)4.33 ± 0.384.23 ± 0.490.0184.33 ± 0.374.23 ± 0.500.027
RDW (%)13.16 ± 1.3513.55 ± 1.800.01413.20 ± 1.3713.53 ± 1.800.051
PDW (fL)11.12 ± 1.4510.97 ± 1.500.26511.09 ± 1.4410.97 ± 1.490.358
WBC count (103/µL)6.01 ± 1.746.00 ± 1.830.9836.12 ± 1.886.01 ± 1.850.563
Neutrophil (%)3.67 ± 1.483.75 ± 1.500.5503.75 ± 1.573.76 ± 1.520.982
Lymphocyte (%)1.86 ± 0.601.76 ± 0.650.0671.88 ± 0.611.78 ± 0.660.082
NLR2.16 ± 1.202.41 ± 1.490.0652.18 ± 1.172.40 ± 1.510.109
Platelet (103/µL)258.17 ± 60.17252.90 ± 72.140.413262.55 ± 63.35252.12 ± 72.700.131
MPV (fL)9.77 ± 0.869.57 ± 1.060.0399.75 ± 0.869.57 ± 1.060.079
PT (sec)11.00 ± 0.7611.05 ± 0.740.54011.04 ± 0.8311.05 ± 0.740.853
ALP (IU/L)59.97 ± 20.0263.52 ± 20.290.04959.14 ± 19.2663.22 ± 19.890.030
CEA (ng/mL)1.74 ± 3.041.91 ± 2.880.5391.62 ± 1.771.92 ± 2.930.253
CA 15-3 (U/mL)11.97 ± 6.5513.78 ± 7.660.00911.95 ± 7.0713.75 ± 7.680.010
Anesthetic information
Anesthetic agent 0.554 0.480
Sevoflurane1017 (61.7)76 (56.7) 399 (61.9)75 (58.1)
TIVA20 (1.2)3 (2.2) 7 (1.1)2 (1.6)
Desflurane442 (26.8)38 (28.4) 190 (29.5)36 (27.9)
Isoflurane151 (9.2)14 (10.5) 40 (6.20)13 (10.08)
Enflurane19 (1.2)3 (2.2) 9 (1.40)3 (2.33)
Analgesic agents
NSAIDs1486 (90.1)125(93.3)0.230581 (90.1)120 (93.0)0.296
Opioids1023 (62.0)87(64.9)0.508387 (60.0)86 (66.7)0.157
Tramadol625 (37.9)43(32.1)0.182255 (39.5)43 (33.3)0.187
Dexamethasone138 (8.4)14(10.5)0.40751 (7.9)13 (10.1)0.414
Surgical information
Procedure <0.001 <0.001
BCS851 (51.6)33 (24.6) 329 (51.0)32 (24.8)
Mastectomy798 (48.4)101 (75.4) 316 (49.0)97 (75.2)
Surgical time 207.8 ± 134.5221.3 ± 121.90.263204.6 ± 125.3223.3 ± 123.40.120
TNM stage <0.001 <0.001
1849 (51.5)31 (23.1) 310 (48.1)29 (22.5)
2617 (37.4)49 (36.6) 258 (40.0)47 (36.4)
3183 (11.1)54 (40.3) 77 (11.9)53 (41.1)
Receptor status
Estrogen1185 (71.86)78 (58.2)<0.001469 (72.7)75 (58.1)<0.001
Progesterone1081 (65.55)72 (53.7)0.006429 (66.5)71 (55.0)0.013
HER2455 (27.59)31 (23.1)0.265156 (24.2)31 (24.0)0.970
Histological grade <0.001 <0.001
Well369 (22.4)14 (10.5) 152 (23.6)14 (10.9)
Moderate740 (44.9)59 (44.0) 289 (44.8)55 (42.6)
Poorly369 (22.4)57 (42.5) 142 (22.0)56 (43.4)
Other171 (10.4)4 (3.0) 62 (9.6)4 (3.1)
Neoadjuvant chemotherapy64 (3.9)19 (14.2)<0.00125 (3.9)18 (14.0)<0.001
Adjuvant chemotherapy937 (56.8)98 (73.1)<0.001392 (60.8)96 (74.4)0.004
Radiotherapy1062 (64.4)83 (61.9)0.568429 (66.5)80 (62.0)0.326
Data are presented as mean (standard deviation) or number (percentage). Propensity score analysis was matched by factors including age, body mass index, and comorbidities.
Table 2. Patient characteristics based on postoperative mortality.
Table 2. Patient characteristics based on postoperative mortality.
Before 1 to 5 Propensity Score MatchingAfter 1 to 5 Propensity Score Matching
ParametersNon-Death
(n = 1682)
Death
(n = 101)
p-ValueNon-Death
(n = 445)
Death
(n = 89)
p-Value
Demographic data
Age (yr)49.9 ± 9.852.4 ± 12.70.05450.8 ± 9.850.9 ± 11.80.333
Body mass index (kg/m2)23.3 ± 3.122.8 ± 3.10.10222.9 ± 2.922.9 ± 3.10.851
Comorbidities
Hypertension323 (19.2)28 (27.7)0.037110 (24.7)21 (23.6)0.382
Diabetes mellitus110 (6.5)13 (12.9)0.01544 (9.9)8 (9.0)0.129
Cardiac41 (2.4)6 (5.9)0.0464 (0.9)3 (3.4)0.775
Pulmonary25 (1.5)6 (5.9)0.0071 (0.2)0 (0.0)0.384
Endocrine81 (4.8)5 (5.0)0.81417 (3.8)4 (4.5)0.765
Renal9 (0.54)1 (1.0)0.4433 (0.7)1 (1.1)0.519
Liver7 (0.4)2 (2.0)0.0880 (0.0)0 (0.0)>0.999
Neurological24 (1.4)2 (2.0)0.6568 (1.8)2 (2.3)0.676
Others11 (0.7)0 (0.0)>0.9990 (0.0)0 (0.0)>0.999
Hematologic markers
Hemoglobin (g/dl)12.9 ± 1.212.6 ± 1.50.06912.9 ± 1.212.5 ± 1.60.033
Hematocrit (%)39.0 ± 3.338.1 ± 4.50.04339.0 ± 3.237.8 ± 4.60.018
RBC count (106/µL)4.33 ± 0.384.19 ± 0.520.0074.34 ± 0.364.16 ± 0.530.004
RDW (%)13.15 ± 1.3313.84 ± 2.050.00213.11 ± 1.2213.87 ± 2.170.002
PDW (fL)11.12 ± 1.4510.91 ± 1.530.16111.04 ± 1.4410.85 ± 1.480.283
WBC count (103/µL)5.98 ± 1.736.38 ± 2.010.0536.01 ± 1.576.38 ± 2.090.122
Neutrophil (%)3.66 ± 1.473.95 ± 1.670.0603.67 ± 1.353.98 ± 1.720.116
Lymphocyte (%)1.85 ± 0.591.90 ± 0.750.5591.88 ± 0.571.88 ± 0.770.916
NLR2.17 ± 1.192.44 ± 1.660.1042.14 ± 1.172.50 ± 1.730.057
Platelet (103/µL)257.92 ± 59.95255.32 ± 78.720.745264.28 ± 59.61257.71 ± 80.410.467
MPV (fL)9.77 ± 0.879.42 ± 1.030.0019.73 ± 0.899.41 ± 1.030.003
PT (sec)11.01 ± 0.7511.04 ± 0.780.62710.94 ± 0.7811.01 ± 0.740.407
ALP (IU/L)59.80 ± 19.8167.53 ± 22.740.00260.48 ± 18.5565.62 ± 21.670.039
CEA (ng/mL)1.74 ± 3.091.93 ± 1.580.2841.83 ± 4.091.78 ± 1.410.811
CA 15-3 (U/mL)11.98 ± 6.5314.25 ± 8.120.00712.21 ± 6.1013.98 ± 7.560.040
Anesthetic information
Anesthetic agent 0.107 0.174
Sevoflurane1037 (61.7)56 (55.5) 268 (60.2)50 (56.2)
TIVA21 (1.3)2 (2.0) 4 (0.9)1 (1.1)
Desflurane451 (26.8)29 (28.7) 128 (28.8)26 (29.2)
Isoflurane155 (9.2)10 (9.9) 41 (9.2)8 (9.0)
Enflurane18 (1.1)4 (4.0) 4 (0.9)4 (4.5)
Analgesic agents
NSAIDs1520 (90.4)91 (90.1)0.929404 (90.8)83 (93.3)0.453
Opioids1044 (62.1)66 (65.4)0.510265 (59.6)59 (66.3)0.235
Tramadol643 (38.2)25 (24.8)0.007179 (40.2)25 (28.1)0.032
Dexamethasone144 (8.6)8 (7.9)0.82334 (7.6)6 (6.7)0.769
Surgical information
Procedure <0.001 <0.001
BCS865 (51.4)19 (18.8) 218 (49.0)18 (20.2)
Mastectomy817 (48.6)82 (81.2) 227 (51.0)71 (79.8)
Surgical time209.5 ± 135.7197.7 ± 91.50.224204.6 ± 125.3223.3 ± 123.40.120
TNM stage <0.001 <0.001
1862 (51.3)18 (17.8) 226 (50.8)14 (15.7)
2625 (37.2)41 (40.6) 166 (37.3)37 (41.6)
3195 (11.6)42 (41.6) 53 (11.9)38 (42.7)
Receptor status
Estrogen1215 (72.2)48 (47.5)<0.001336 (75.5)40 (44.9)<0.001
Progesterone1110 (66.0)43 (42.6)<0.001292 (65.6)38 (42.7)<0.001
HER2458 (27.2)28 (27.7)0.914123 (27.6)25 (28.1)0.932
Histological grade <0.001 <0.001
Well373 (22.2)10 (9.9) 116 (26.1)7 (7.8)
Moderate760 (45.2)39 (38.6) 190 (42.7)33 (37.1)
Poorly380 (22.6)46 (45.5) 92 (20.7)43 (48.3)
Other169 (10.1)6 (5.9) 47 (10.6)6 (6.7)
Neoadjuvantchemotherapy63 (3.8)20 (19.8)<0.00116 (3.6)19 (21.4)<0.001
Adjuvantchemotherapy965 (57.4)70 (69.3)0.019268 (60.2)65 (73.0)0.023
Radiotherapy1085 (64.5)60 (59.4)0.299273 (61.4)55 (61.8)0.937
Data are presented as mean (standard deviation) or number (percentage). Propensity score analysis was matched by factors including age, body mass index, and comorbidities.
Table 3. Independent risk factors for patients with breast cancer associated with postoperative recurrence after Cox regression analysis with 1 to 5 propensity score matching.
Table 3. Independent risk factors for patients with breast cancer associated with postoperative recurrence after Cox regression analysis with 1 to 5 propensity score matching.
ParameterUnivariate AnalysisMultivariate Analysis
HR (95% CI)p-ValueHR (95% CI)p-Value
Hematologic markers
RDW (%)>13.41.713 (1.196–2.453)0.0041.238 (0.849–1.806)0.267
ALP (IU/L)>631.475 (1.040–2.093)0.030
CEA (ng/mL)>2.731.653 (1.060–2.577)0.0271.543 (0.988–2.411)0.056
CA 15-3 (U/mL)>11.42.145 (1.513–3.040)<0.0011.655 (1.154–2.374)0.007
Surgical procedureBCS1 (ref) 1 (ref)
Mastectomy2.741 (1.837–4.088)<0.0012.169 (1.419–3.314)<0.001
TNM stage11 (ref) 1 (ref)
21.781 (1.121–2.831)0.0151.173 (0.728–1.891)0.512
35.863 (3.725–9.226)<0.0013.481 (2.136–5.672)<0.001
Estrogen receptorNegative1 (ref) 1 (ref)
Positive0.512 (0.360–0.727)<0.0010.671 (0.455–0.990)0.045
Progesterone receptorNegative1 (ref) 1 (ref)
Positive0.598 (0.422–0.846)0.004
Neoadjuvant chemotherapy 3.090 (1.876–5.089)<0.0011.957 (1.127–3.397)0.018
Adjuvant chemotherapy 1.659 (1.116–2.465)0.013
Histological gradeGood1 (ref) 1 (ref)
Moderate2.152 (1.194–3.876)0.0112.071 (1.139–3.766)0.017
Poor4.165 (2.312–7.503)<0.0013.878 (2.044–7.357)<0.001
Other0.734 (0.242–2.231)0.5860.598 (0.194–1.840)0.370
Propensity score analysis was matched by factors including age, body mass index, and comorbidities.
Table 4. Independent risk factors for patients with breast cancer associated with postoperative mortality after Cox regression analysis with 1 to 5 propensity score matching.
Table 4. Independent risk factors for patients with breast cancer associated with postoperative mortality after Cox regression analysis with 1 to 5 propensity score matching.
ParameterUnivariate AnalysisMultivariate Analysis
HR (95% CI)p-ValueHR (95% CI)p-Value
Age<401 (ref) 1 (ref)
40–500.528 (0.284–0.980)0.043
50–600.622 (0.330–1.170)0.141
60–700.701 (0.354–1.389)0.309
≥701.343 (0.491–3.669)0.566
Hematologic markers
Hemoglobin (g/dl) 0.809 (0.697–0.940)0.006
RDW (%)>13.52.368 (1.546–3.625)<0.0011.723 (1.098–2.704)0.019
WBC count (103/µL) 1.137 (1.009–1.281)0.035
NLR>2.821.936 (1.238–3.029)0.0041.771 (1.108–2.832)0.017
MPV (fL)>8.60.358 (0.213–0.601)<0.0010.469 (0.271–0.811)0.007
ALP (IU/L)>761.729 (1.104–2.708)0.0172.257 (1.412–3.607)<0.001
CEA (ng/mL)>1.571.590 (1.046–2.417)0.0311.553 (1.007–2.395)0.047
CA 15-3 (U/mL)>11.52.126 (1.396–3.236)<0.0011.423 (0.919–2.206)0.115
Surgical procedureBCS1 (ref) 1 (ref)
Mastectomy3.287 (1.959–5.516)<0.0012.993 (1.750–5.118)<0.001
TNM stage11 (ref) 1 (ref)
23.172 (1.714–5.869)<0.0012.310 (1.239–4.306)0.009
38.676 (4.699–16.019)<0.0016.563 (3.482–12.370)<0.001
Estrogen receptorNegative1 (ref) 1 (ref)
Positive0.279 (0.184–0.425)<0.0010.350 (0.219–0.561)<0.001
Progesterone receptorNegative1 (ref) 1 (ref)
Positive0.387 (0.254–0.589)<0.001
Radiotherapy 1.027 (0.669–1.576)0.904
Neoadjuvant chemotherapy 4.403 (2.649–7.319)<0.0011.890 (1.048–3.409)0.035
Adjuvant chemotherapy 1.533(0.959–2.450)0.074
Histological gradeGood1 (ref) 1 (ref)
Moderate2.991 (1.320–6.776)0.0093.253 (1.412–7.491)0.006
Poor7.661 (3.430–17.111)<0.0016.455 (2.757–15.113)<0.001
Other2.012 (0.676–5.991)0.2091.452 (0.480–4.393)0.509
Propensity score analysis was matched by factors including age, body mass index, and comorbidities.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yoo, Y.-C.; Park, S.; Kim, H.-J.; Jung, H.-E.; Kim, J.-Y.; Kim, M.-H. Preoperative Routine Laboratory Markers for Predicting Postoperative Recurrence and Death in Patients with Breast Cancer. J. Clin. Med. 2021, 10, 2610. https://doi.org/10.3390/jcm10122610

AMA Style

Yoo Y-C, Park S, Kim H-J, Jung H-E, Kim J-Y, Kim M-H. Preoperative Routine Laboratory Markers for Predicting Postoperative Recurrence and Death in Patients with Breast Cancer. Journal of Clinical Medicine. 2021; 10(12):2610. https://doi.org/10.3390/jcm10122610

Chicago/Turabian Style

Yoo, Young-Chul, Seho Park, Hyun-Joo Kim, Hyun-Eom Jung, Ji-Young Kim, and Myoung-Hwa Kim. 2021. "Preoperative Routine Laboratory Markers for Predicting Postoperative Recurrence and Death in Patients with Breast Cancer" Journal of Clinical Medicine 10, no. 12: 2610. https://doi.org/10.3390/jcm10122610

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop