We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleOpen Accesscc iconby iconnc iconnd icon

Effect of preoperative peripheral blood platelet volume index on prognosis in patients with invasive breast cancer

    Kai Huang‡

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Suosu Wei‡

    Department of Scientific Cooperation of Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Zhen Huang

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    ,
    Yujie Xie

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    ,
    Chunyu Wei

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    ,
    Jinan Xu

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    ,
    Lingguang Dong

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    ,
    Quanqing Zou

    *Author for correspondence:

    E-mail Address: zouquanqing@163.com

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    &
    Jianrong Yang

    **Author for correspondence:

    E-mail Address: gandansurgery2014@163.com

    Department of Breast & Thyroid Surgery, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China

    Published Online:https://doi.org/10.2217/fon-2022-0930

    Abstract

    Aim: This study was designed to investigate the prognostic value of the platelet volume index in patients with invasive breast cancer (IBC). Methods: A total of 524 patients with IBC were enrolled in this study, with a median follow-up time of 6.76 years. The relationship between platelet volume indices and breast cancer prognosis was analyzed. Results: There is a strong correlation between a higher platelet distribution width-to-platelet count ratio (PDW/P) and poorer disease-free survival (DFS) in patients with IBC. The DFS rate was significantly lower among individuals with elevated PDW/P ratios compared with those with lower ratios. Conclusion: The PDW/P ratio is an independent risk factor for predicting DFS in patients with IBC.

    Tweetable abstract

    Platelet volume index is an inexpensive and convenient biomarker to measure in clinical practice, with the potential to aid in prognostic risk assessment and guide personalized diagnosis and treatment choices for invasive breast cancer.

    Breast cancer (BC) is a frequent malignant tumor in females occurring in the mammary epithelial tissue. It has the characteristics of incidence and rapid disease progression, which gravely threatens the lives and healthcare of women worldwide. According to the latest estimates from the WHO's International Agency for Research on Cancer [1], worldwide, there were 2.08 million new BC cases and 630,000 women died from BC in 2018. At present, early BC is mainly treated by surgery. Although such patients can obtain a good prognosis, the risk of metastasis and recurrence remains difficult to predict [2].

    Traditional prognostic factors of BC, such as molecular typing and clinical staging, have poor accuracy, while new prognostic tools and genes for BC, such as Oncotype Dx and BC susceptibility gene BRCA, respectively, are expensive, which limits their clinical application. With increasing attention being paid to the interaction between the tumor inflammatory environment and tumor cells, the relationship between activated platelets and tumors has become a hot research field. The mutual interaction between activated platelets and cancer cells results in tumor growth, abnormal angiogenesis and metastasis [3,4]. Activated platelets can escape shear-induced injury by binding to tumor cells, thereby promoting tumor cell colonization [5]. In recent years, it has been reported that platelet count [6] and platelet-to-lymphocyte ratio (PLR) [7] have predictive value for the long-term prognosis of BC. In addition, larger platelets have been reported to store more particles and receptors and to have greater adhesivity compared with smaller platelets. Therefore, the activity of activated platelets is more accurately expressed by their volume than by count [8]. Therefore, platelet volume indices (PVI) are the best way to assess platelet activation. Platelet distribution width (PDW) and mean platelet volume (MPV) are markers of platelet activation and morphology [9] associated with various inflammatory states [10]. Recent studies have shown that in BC, esophageal cancer, hepatocellular carcinoma and non-small-cell lung cancer (NSCLC), the level of MPV-to-platelet count ratio (MPV/P) was related to poor prognosis [11–14]. PDW and PDW-to-platelet count ratio (PDW/P) are related to poor prognostic value in endometrial cancer, melanoma, gastric cancer as well as NSCLC [15–18], Meanwhile, Okuturlar et al. [19] indicated that there were no statistical differences in the univariate analysis of patients using MPV, PDW, neutrophil-to-lymphocyte ratio (NLR), PLR, MPV/P and other inflammatory cells between patients with metastatic BC and those without. However, Huang et al. [20] reported that PDW/P was not statistically meaningful in the long-term prognosis of BC in univariate analysis of overall survival, and MPV/P, PDW/P and MPV had no statistical significance in the multivariate analysis.

    It can be concluded that the clinical significance of peripheral blood PVIs in BC remains obscure. To the best of our knowledge, PVIs in the Chinese population in the processing of information on how BC prognosis has changed remain restricted. Therefore, the intention of this study was to assess the preoperative influence of PVIs on disease-free survival (DFS) in invasive BC with the largest possible sample and longer follow-up time to ascertain their proportional meaning.

    Materials & methods

    Study population

    The current review cohort study included all hospitalized BC patients first diagnosed at the People's Hospital of Guangxi Zhuang Autonomous Region (PHGZAR), China, between March 2013 and December 2016 (registration number: ChiCTR2200058542; http://www.chictr.org.cn/index.aspx).

    After approval by the Ethics Committee of the PHGZAR, a historical cohort was constructed from March 2013 to December 2016, using the International Classification of Diseases code C50 and limiting all adult inpatients to individuals of Asian ethnic background. This study used the resource database of the PHGZAR, which is an advanced medical data management system in China. It connects and indexes the hospital's diagnostic and treatment records. The electronic data include demographic characteristics, medical diagnosis codes and services, surgical codes, medication prescriptions and death information. A series of 674 female participants were recruited for this study. A total of 22 patients with preoperative recurrence and metastasis, 58 patients with preoperative neoadjuvant therapy and incomplete pathological data, 66 patients with noninvasive cancer and no operative therapy in the authors' hospital, two male BC patients and two patients with no follow-up visits were excluded (Figure 1). A series of 524 invasive BC patients were enrolled in this research and were followed up until 30 June 2022. The median follow-up period was 6.76 years. Lymph node metastases, primary tumor size, distant metastases, histological grade, estrogen receptor (ER) status, progesterone receptor (PR) status and HER2 status were examined and recorded for each patient. The primary end point of this research was DFS, which was defined as the interval from the initial treatment date to initial observed disease recurrence.

    Figure 1. Patient selection flowchart.

    Immunohistochemistry & HER2 analysis

    The evaluation of ER and PR status was performed using immunohistochemistry (IHC). Samples were considered ER and PR positive if at least 1% immunostained tumor nuclei of the nucleus of an immune-stained tumor was detected (Pathology Department, PHGZAR, China). Detection of HER2 status positive by IHC or FISH and IHC score 3+ or HER2 score 2+ in tumor cells was considered positive when HER2 amplification was confirmed by FISH (Pathology Department, PHGZAR, Guangxi Province, China).

    Clinical hematological examination

    Fasting blood samples were collected via peripheral vein puncture before any treatment commenced. Platelets, MPV and PDW were measured with an automatic blood and fluid analyzer (Sysmex XN-9000, Kobe, Japan). PDW/P and MPV/P values were calculated as the PDW and MPV, respectively, divided by the platelet count. Receiver operating characteristic (ROC) curves were used to determine thresholds.

    Statistical analysis

    Median and interquartile range or frequency (percentage) were used for continuous variables and categorical variables are expressed in terms of frequency (percentage). Statistical comparisons of categorical variables by χ2 test and Wilcoxon rank sum test were employed to detect differences between two groups of normally distributed continuous variables. The optimal threshold values for PDW, MPV, MPV/P and PDW/P were determined from ROC curves. The estimated hazard ratio (HR) using Cox analysis is the relative risk reported with a corresponding 95% CI. Kaplan-Meier curves were utilized to compare survival differences between the cohorts. All statistical analyses were completed and validated by SPSS 18.0 (Windows version. IBM Corp. NY, USA); p < 0.05 was assumed to be statistically meaningful.

    Data collection & ethics compliance

    Since this study was a retrospective study, it was authorized by the Ethics Review Committee of the PHGZAR. The medical data for all trial subjects were anonymized. The PHGZAR and Guangxi Academy of Medical Sciences announced the research plan and the option to freely reject participation. Patients were deemed to have agreed to the study unless they explicitly refused to participate.

    Results

    Subjects characteristics

    Table 1 summarizes patient characteristics. The median age was 51.87 ± 11.59 years (range: 22–88 years). There were 290 patients (55.34%) >50 years of age and 234 patients (44.66%) <50 years of age. There were 215 patients (41.03%) with axillary lymph node metastasis, 288 patients (54.96%) without axillary lymph node metastasis and 21 patients (4.01%) without confirmed axillary lymph node metastasis. A total of 105 patients (20.04%) had luminal A and 172 patients (32.82%) had luminal B (HER2-), which accounted for the majority of molecular typing. There were 55 HER2+ (HR+) patients (10.50%); 76 HER2+ (HR-) patients (14.50%); 72 triple-negative patients (13.74%) and 44 clinically unconfirmed (8.40%).

    Table 1. Basic characteristics of enrolled patients.
    CharacteristicsNo. (%)
    Age
      <50234 (44.66%)
      ≥50290 (55.34%)
    Size
     ≤20 mm209 (39.89%)
      >20 mm290 (55.34%)
      Unknown25 (4.77%)
    Histological
      Low305 (58.21%)
      High124 (23.66%)
      Unknown95 (18.13%)
    ALNM
      No288 (54.96%)
      Yes215 (41.03%)
      Unknown21 (4.01%)
    KI67
      <14%132 (25.19%)
      14%≤KI67≤30%183 (34.92%)
      >30%169 (32.25%)
      Unknown40 (7.63%)
    ER
      Negative163 (31.11%)
      Positive352 (67.18%)
      Unknown9 (1.72%)
    PR
      Negative211 (40.27%)
      Positive303 (57.82%)
      Unknown10 (1.91%)
    HER2
      Negative357 (68.13%)
      Positive131 (25.00%)
      Unknown36 (6.87%)
    PDW/P
      <0.6046465 (88.74%)
      ≥0.604659 (11.26%)
    PDW
      <10.0500113 (21.56%)
      ≥10.0500411 (78.44%)
    MPV/P
      <0.5330458 (87.40%)
      ≥0.533066 (12.60%)
    MPV
      <10.0500271 (51.72%)
      ≥10.0500253 (48.28%)
    Molecular
      Luminal A105 (20.04%)
      Luminal B (HER2-)172 (32.82%)
      HER2+ (HR+)55 (10.50%)
      HER2+ (HR-)76 (14.50%)
      TNBC72 (13.74%)
      Unknown44 (8.40%)
    Platelet count254.00 (219.00–291.25)

    ALNM: Axillary lymph node metastasis; ER: Estrogen receptor; MPV: Mean platelet volume; MPV/P: Mean platelet volume-to-platelet count ratio; PR: Progesterone receptor; PDW: Platelet distribution width; PDW/P: Platelet distribution width-to-platelet count ratio; TNBC: Triple-negative breast cancer.

    According to the ROC curve analysis (Table 2), the optimal cutoff values for PDW, MPV, PDW/P and MPV/P were 10.05, 10.05, 0.06 and 0.53, respectively. Table 3 shows the associations between PVIs and clinicopathological variables. Increases in PDW/P and MPV/P were dramatically related to age (p < 0.05).

    Table 2. Receiver operating characteristic analyses of platelet volume indices in breast cancer patients.
    VariablesCut off valueSpecificitySensitivity
    PDW10.050.230.88
    MPV10.050.530.53
    PDW/P0.600.910.22
    MPV/P0.530.890.22

    MPV: Mean platelet volume; MPV/P: Mean platelet volume-to-platelet count ratio; PDW: Platelet distribution width; PDW/P: Platelet distribution width-to-platelet count ratio.

    Table 3. Association between platelet volume indices and clinicopathological factors in patients with breast cancer.
    VariablesPDW/PMPV/P
    AverageSDp-valueAverageSDp-value
    Age (years)
      <500.420.120.0010.390.1<0.001
      ≥500.470.160.420.13  
    Size
      ≤20 mm0.440.130.4940.40.110.493
      >20 mm0.450.16 0.410.13 
    Histological
      Low0.460.150.0810.420.120.065
      High0.440.14 0.40.11 
    ALNM
      No0.450.160.870.410.130.807
      Yes0.440.13 0.40.11 
    KI67
      <14%0.440.140.1110.40.120.118
      14%≤KI67≤30%0.450.16 0.410.13 
      >30%0.430.13 0.40.1 
    ER
      Negative0.450.150.790.410.120.725
      Positive0.440.15 0.40.12 
    PR
      Negative0.450.140.960.410.120.844
      Positive0.450.15 0.40.12 
    HER2
      Negative0.450.140.5850.410.120.622
      Positive0.440.15 0.40.12 
    Molecular  0.642  0.567
      Luminal A0.440.13 0.40.11 
      Luminal B (HER2-)0.450.15 0.410.12 
      HER2+ (HR+)0.430.13 0.40.12 
      HER2+ (HR-)0.430.16 0.40.13 
      TNBC0.470.14 0.420.12 

    ALNM: Axillary lymph node metastasis; ER: Estrogen receptor; MPV: Mean platelet volume; MPV/P: Mean platelet volume-to-platelet count ratio; PDW: Platelet distribution width; PDW/P: Platelet distribution width-to-platelet count ratio; PR: Progesterone receptor; SD: Standard deviation; TNBC: Triple-negative breast cancer.

    Survival

    The median follow-up was 6.76 years, during which 81 patients (15.46%) experienced a recurrence. Clinicopathological factors and the values of PVIs for the long-term prognosis of invasive BC were further explored. For tumor size, patients with a size >20 mm had a worse DFS HR of 2.23 (95% CI: 1.36–3.66; p < 0.05). In multivariate analysis, tumor size was not statistically significant (Table 4).

    Table 4. Survival analyses of clinicopathological factors and platelet volume indices.
    VariablesUnivariate analysisMultivariate analysis
     Hazard ratio (95% CI)p-valueHazard ratio (95% CI)p-value
    Age (years)
      <501 1 
      ≥501.39 (0.89–2.17)0.14851.01 (0.99–1.03)0.2772
    Size
      ≤20 mm1   
      >20 mm2.23 (1.36–3.66)0.00161.13 (0.83–1.52)0.4326
    Histological
      Low1   
      High1.17 (0.70–1.95)0.5601  
    ALNM
      No1   
      Yes2.01 (1.29–3.15)0.00221.27 (0.97–1.65)0.0794
    KI67
      <14%1   
      14%≤KI67≤30%0.99 (0.55–1.77)0.9701  
      >30%1.13 (0.64–2.02)0.6732  
    ER
      Negative1   
      Positive0.79 (0.50–1.25)0.3079  
    PR
      Negative1   
      Positive0.70 (0.45–1.09)0.1139  
    HER2
      Negative1   
      Positive1.04 (0.62–1.73)0.8944  
    PDW/P
      <0.60461 1 
      ≥0.60462.42 (1.43–4.09)0.00092.61 (1.14–5.99)0.023
    PDW
      <10.05001 1 
      ≥10.05002.03 (1.05–3.93)0.03620.98 (0.82–1.18)0.8323
    MPV/P
      <0.53301 1 
      ≥0.53302.10 (1.24–3.54)0.00570.68 (0.05–9.13)0.773
    MPV
      <10.05001   
      ≥10.05001.21 (0.78–1.87)0.3947  
    Molecular
      Luminal A1   
      Luminal B (HER2-)1.06 (0.56–2.01)0.8584  
      HER2+ (HR+)0.94 (0.38–2.30)0.8871  
      HER2+ (HR-)1.35 (0.64–2.83)0.4318  
      TNBC1.40 (0.67–2.94)0.374  

    ALNM: Axillary lymph node metastasis; ER: Estrogen receptor; MPV: Mean platelet volume; MPV/P: Mean platelet volume-to-platelet count ratio; PDW: Platelet distribution width; PDW/P: Platelet distribution width-to-platelet count ratio; PR: Progesterone receptor; TNBC: Triple-negative breast cancer.

    The presence of axillary lymph node metastases was related to a worse long-term prognosis than the absence of axillary lymph node metastases, with an HR of 2.01 (95% CI: 1.29–3.15; p < 0.05). Axillary lymph node metastasis was not statistically significant in the multivariate analysis. However, clinicopathological factors such as age, histological grade and tumor immunological indicators were not statistically significant in relation to DFS in these patients (Table 4). For PVIs, using the optimum threshold as a cutoff point, higher PDW/P, PDW and MPV/P were significantly associated with poor DFS prognosis, with HRs of 2.42 (95% CI: 1.43–4.09; p < 0.05), 2.03 (95% CI: 1.05–3.93; p < 0.05) and 2.10 (95% CI: 1.24–3.54; p < 0.05). In multifactorial analysis, only PDW/P was statistically significant at an HR of 2.61 (95% CI: 1.14–5.99; p < 0.05; Table 4). To further demonstrate the prognostic long-term efficiency of PDW/P for invasive BC, survival analysis was performed using Kaplan-Meier curves showing that subjects with higher PDW/P had a worse long-term prognosis compared with subjects with low PDW/P (5-year survival rate: 74.17 vs 88.39%; p < 0.05; Figure 2).

    Figure 2. Kaplan-Meier analysis of disease-free survival stratified by platelet distribution width-to-platelet count ratio in breast cancer patients.

    PDW/P: Platelet distribution width-to-platelet count ratio.

    Discussion

    Platelets are small pieces of cytoplasm that are shed from the cytoplasm of mature bone marrow megakaryocytes by lysis. Their main functions are coagulation and hemostasis. Recent reports have confirmed that in addition to their regulatory roles in hemostasis, inflammatory responses and host immunity, activated platelets secrete a variety of growth factors. Among them are VEGF, PDGF and PAF, which enhance tumor cell invasiveness, epithelial–mesenchymal transition, and extravasation and promote angiogenesis and vascular remodeling [21–25]. The interaction between tumor cells in the blood and activated platelets can make cancer patients more prone to thrombotic events and further increase the degree of malignancy of tumor cells [26,27]. Platelets also have a vital function in modulating the metastatic efficiency and survival of various tumor cells in the blood [28,29]. They can adhere to circulating tumor cells, support their extravasation, induce their proliferation [30,31] and even activate and recruit inflammatory cells such as neutrophils or macrophages to induce tumor cells to escape natural killer cell lysis [32]. Kang et al. revealed that PDGF facilitates the invasiveness of BC cells through the NFkB signaling pathway [33]. The platelet-specific receptor glycoprotein VI of collagen and fibrin interacts with BC cells and uses ITAM signaling components in platelets to regulate platelet adhesion, aggregation, procoagulant activity and other functions, which is conducive to extravasation of BC cells [34].

    PDW and MPV are morphological indicators of platelet volume size distribution and variability. Generally speaking, the two are inversely proportional [35] and are often used to distinguish between low-yield and high-destructive thrombocytopenia [36]. The quantity of research on the biological relationship between MPV and the long-term prognostic progression of various types of cancer is quite limited. However, recent clinical research by Riedl et al. [37] using data from 1544 participants with various types of cancer revealed that high MPV values were not only connected to a decrease in the risk of venous thromboembolism but also improved survival rates. Moreover, in patients with advanced NSCLC, low levels of MPV predict poor prognosis [14]. On the contrary, other research has shown that increased MPV is statistically significantly related to the poor prognosis of esophageal cancer, BC and hepatocellular cancer [11–13]. Thus, the value of MVP as a prognostic indicator for BC is presently unclear. These conclusions should be verified in larger prospective studies with larger samples and longer follow-ups. However, it is reasonable to estimate the prognosis of patients with malignant tumors by comparing MPV with platelet count as an indicator of platelet activation, since it seems to gather the advantages of two measures of platelet activation. In fact, inflammatory cell ratio indices such as PLR, monocyte-to-lymphocyte ratio or NLR are widely used, in part because the cutoff values for these indirect indices are not unique. Therefore, our cutoff values for MPV, PDW, PDW/P and MPV/P based on the ROC curves are reasonable, although they are not the dividing line between normal and abnormal. PDW is an index reflecting the average change in platelet volume. Because it does not accumulate in response to platelet bulking [38], it is often considered a more concrete marker of an activated platelet object. Compared with MPV, it is possible that PDW is a more accurate and reliable prognostic biomarker. In this study, MPV was not correlated with prognosis in BC. PDW, PDW/P and MPV/P were statistically significant for the prognosis of BC in univariate analysis. In multivariate analysis, only PDW/P had statistical significance for BC prognosis, which is similar to the recent report of Takeuchi et al. [39]. The current study also confirmed that PDW, PDW/P and MPV/P are relevant to the survival of Asian BC patients. Multicenter and large-sample clinical trials in other ethnic groups are needed to determine whether these findings are genetically race-specific.

    Currently, the main mechanism linking PVIs to survival is unclear. Changes in PVIs may be accompanied by dysfunction of megakaryocytes and bone marrow cells. More recent research has revealed that cytokines secreted by tumor cells, for example, include granulocyte colony-stimulating factor and macrophage colony-stimulating factor, modulate megakaryocyte cell maturation, thrombocyte release and production as well as the size of thrombocytes [40]. In addition, local inflammatory infiltration plays a crucial function in the tumor microenvironment. Several inflammatory cytokines like IL-1, IL-8, IL-6, IL-18, IL-12, TNF and IFN-gamma are upregulated during the progression of tumor cell infiltration [41]. These cytokines promote abnormal maturation of bone marrow cells and megakaryocytes, which results in the increased generation and emission of premature platelets with different properties and sizes in the cardiovascular system. Subsequent interactions between activated platelets and cancer cells lead to angiogenesis, tumor development, tumor invasion and metastasis, further increasing the malignant degree of tumor cells. The convenience and low cost of measuring PVIs in clinical practice make it a potential biomarker for prognostic risk assessment and individualized treatment options in BC. However, before its clinical application, more quality basic and multicenter prospective studies are needed to better explain why higher PDW, PDW/P and MPV/P lead to adverse prognoses in invasive BC.

    The strengths of this research include the large sample size, long follow-up time, good matching and multivariate analysis. However, there are some drawbacks that cannot be ignored. First, this study is single-center and cannot represent the entire Asian population. Second, the retrospective character of the studies could also lead to data selection and analysis bias. Therefore, high-quality basic studies and well-designed polycentric prospective studies are warranted to further explore the connection between the composite index of peripheral blood platelet volume and the prognosis of invasive BC, and to clarify the specific mechanism.

    Conclusion

    Based on the current findings, tumor size, PDW, MPV/P, PDW/P and axillary lymph node involvement are significant clinical factors that impact the prognosis of BC. Moreover, this analysis suggests that the PDW/P ratio has the potential as an independent predictor of prognostic risk in invasive BC. However, further research, including clinical and molecular/genomic validation, is necessary to determine if this data can explain and improve daily clinical treatment regimens.

    Summary points
    • Increases in platelet distribution width-to-platelet count ratio (PDW/P) and mean platelet volume-to-platelet count ratio (MPV/P) were dramatically associated with age (p < 0.05).

    • In univariate analysis, patients with tumor sizes >20 mm had a worse disease-free survival (DFS) risk of 2.23 (95% CI: 1.36–3.66; p < 0.05). In multivariate analysis, tumor size was not statistically significant.

    • In univariate analysis, the long-term prognosis was worse with axillary lymph node metastasis than without, with a risk ratio of 2.01 (95% CI: 1.29–3.15; p < 0.05). In multivariate analysis, axillary lymph node metastasis was not statistically significant.

    • Clinicopathological factors such as age, histological grade and tumor immunological indicators were not statistically significant in relation to DFS in these patients.

    • For platelet volume indices, using the optimum threshold as a cutoff point, higher PDW/P, PDW and MPV/P were significantly associated with poor DFS, with hazard ratios of 2.42 (95% CI: 1.43 4.09; p < 0.05), 2.03 (95% CI: 1.05–3.93; p < 0.05) and 2.10 (95% CI: 1.24–3.54; p < 0.05).

    • In multifactorial analysis, only PDW/P was statistically significant at a hazard ratio of 2.61 (95% CI: 1.14–5.99; p < 0.05).

    • DFS among elevated PDW/P subjects was significantly lower than among lower PDW/P subjects (5-year survival rates: 74.17 vs 88.39%; p < 0.05).

    Author contributions

    K Huang and S Wei conceived and designed the study and reviewed the manuscript. All authors participated in data collection and verification. H Kai wrote the first draft of the manuscript and S Su-Wei performed the statistical analysis of the data. All authors read and approved the final manuscript.

    Acknowledgments

    The authors thank the staff of the Guangxi Zhuang Autonomous Region People's Hospital Information Network Center and the Medical Record Information Quality Control Department for assisting with the retrieval of medical records.

    Financial & competing interests disclosure

    This work was supported by the Major Project of Science and Technology of Guangxi Zhuang Autonomous Region (grant number: Guike-AA22096018) and the Specific Research Project of Guangxi for Research Bases and Talents (grant number: Guike-AD21220042). The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations. In addition, for investigations involving human subjects, informed consent has been obtained from the participants involved.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68(6), 394–424 (2018).
    • 2. Sung H, Ferlay J, Siegel RL et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71(3), 209–249 (2021).
    • 3. Bambace NM, Holmes CE. The platelet contribution to cancer progression. J. Thromb. Haemost. 9(2), 237–249 (2011).
    • 4. Goubran HA, Stakiw J, Radosevic M, Burnouf T. Platelet-cancer interactions. Semin. Thromb. Hemost. 40(3), 296–305 (2014).
    • 5. Egan K, Cooke N, Kenny D. Living in shear: platelets protect cancer cells from shear induced damage. Clin. Exp. Metastasis 31(6), 697–704 (2014).
    • 6. Li L, Wang J, Meng S et al. Peripheral blood leukocytes and platelets serve as prognostic factors in breast cancer. Cancer Biother. Radiopharm. 36(2), 167–173 (2021).
    • 7. Zenan H, Zixiong L, Zhicheng Y et al. Clinical prognostic evaluation of immunocytes in different molecular subtypes of breast cancer. J. Cell. Physiol. 234(11), 20584–20602 (2019). •• Patients with the intraluminal subtype had better clinical and pathological features than patients with HER2 overexpression and triple-negative breast cancer. White blood cell count and neutrophil counts as well as lymphocyte-to-monocyte ratio, neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio all had a significant impact on prognosis.
    • 8. Martin JF, Bath PM, Burr ML. Influence of platelet size on outcome after myocardial infarction. Lancet 338(8780), 1409–1411 (1991).
    • 9. Bath PM, Missouris CG, Buckenham T, Macgregor GA. Increased platelet volume and platelet mass in patients with atherosclerotic renal artery stenosis. Clin. Sci. (Lond.) 87(2), 253–257 (1994).
    • 10. Kaito K, Otsubo H, Usui N et al. Platelet size deviation width, platelet large cell ratio, and mean platelet volume have sufficient sensitivity and specificity in the diagnosis of immune thrombocytopenia. Br. J. Haematol. 128(5), 698–702 (2005).
    • 11. Zhang F, Chen Z, Wang P, Hu X, Gao Y, He J. Combination of platelet count and mean platelet volume (COP-MPV) predicts postoperative prognosis in both resectable early and advanced stage esophageal squamous cell cancer patients. Tumour Biol. 37(7), 9323–9331 (2016).
    • 12. Gu M, Zhai Z, Huang L et al. Pre-treatment mean platelet volume associates with worse clinicopathologic features and prognosis of patients with invasive breast cancer. Breast Cancer 23(5), 752–760 (2016).
    • 13. Cho SY, Yang JJ, You E et al. Mean platelet volume/platelet count ratio in hepatocellular carcinoma. Platelets 24(5), 375–377 (2013).
    • 14. Inagaki N, Kibata K, Tamaki T, Shimizu T, Nomura S. Prognostic impact of the mean platelet volume/platelet count ratio in terms of survival in advanced non-small cell lung cancer. Lung Cancer 83(1), 97–101 (2014).
    • 15. Kurtoglu E, Kokcu A, Celik H, Sari S, Tosun M. Platelet indices may be useful in discrimination of benign and malign endometrial lesions, and early and advanced stage endometrial cancer. Asian Pac. J. Cancer Prev. 16(13), 5397–5400 (2015).
    • 16. Li N, Diao Z, Huang X et al. Increased platelet distribution width predicts poor prognosis in melanoma patients. Sci. Rep. 7(1), 2970 (2017).
    • 17. Zhang X, Cui MM, Fu S et al. Platelet distribution width correlates with prognosis of gastric cancer. Oncotarget 8(12), 20213–20219 (2017).
    • 18. Cui MM, Li N, Liu X et al. Platelet distribution width correlates with prognosis of non-small cell lung cancer. Sci. Rep. 7(1), 3456 (2017).
    • 19. Okuturlar Y, Gunaldi M, Tiken EE et al. Utility of peripheral blood parameters in predicting breast cancer risk. Asian Pac. J. Cancer Prev. 16(6), 2409–2412 (2015).
    • 20. Huang Y, Cui MM, Huang YX et al. Preoperative platelet distribution width predicts breast cancer survival. Cancer Biomark. 23(2), 205–211 (2018).
    • 21. Strilic B, Offermanns S. Intravascular survival and extravasation of tumor cells. Cancer Cell 32(3), 282–293 (2017).
    • 22. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 20(5), 576–590 (2011).
    • 23. Schumacher D, Strilic B, Sivaraj KK, Wettschureck N, Offermanns S. Platelet-derived nucleotides promote tumor-cell transendothelial migration and metastasis via P2Y2 receptor. Cancer Cell 24(1), 130–137 (2013). • This study identified the endothelial P2Y2 receptor, activated by ATP, as the primary mediator of this action. Mice lacking P2Y2 or lacking platelet ATP secretion showed strongly reduced tumor cell metastasis. These findings demonstrate a mechanism by which platelets promote cancer cell metastasis.
    • 24. Wojtukiewicz MZ, Sierko E, Hempel D, Tucker SC, Honn KV. Platelets and cancer angiogenesis nexus. Cancer Metastasis Rev. 36(2), 249–262 (2017).
    • 25. Stegner D, Dutting S, Nieswandt B. Mechanistic explanation for platelet contribution to cancer metastasis. Thromb. Res. 133(Suppl. 2), S149–S157 (2014).
    • 26. Mammadova-Bach E, Mangin P, Lanza F, Gachet C. Platelets in cancer. From basic research to therapeutic implications. Hamostaseologie 35(4), 325–336 (2015).
    • 27. Jurasz P, Alonso-Escolano D, Radomski MW. Platelet–cancer interactions: mechanisms and pharmacology of tumour cell-induced platelet aggregation. Br. J. Pharmacol. 143(7), 819–826 (2004).
    • 28. Leblanc R, Peyruchaud O. Metastasis: new functional implications of platelets and megakaryocytes. Blood 128(1), 24–31 (2016).
    • 29. Heeke S, Mograbi B, Alix-Panabieres C, Hofman P. Never travel alone: the crosstalk of circulating tumor cells and the blood microenvironment. Cells 8(7), 714 (2019).
    • 30. Coupland LA, Chong BH, Parish CR. Platelets and P-selectin control tumor cell metastasis in an organ-specific manner and independently of NK cells. Cancer Res. 72(18), 4662–4671 (2012).
    • 31. Bendas G, Borsig L. Cancer cell adhesion and metastasis: selectins, integrins, and the inhibitory potential of heparins. Int. J. Cell Biol. 2012, 676731 (2012).
    • 32. Lou XL, Sun J, Gong SQ, Yu XF, Gong R, Deng H. Interaction between circulating cancer cells and platelets: clinical implication. Chin. J. Cancer Res. 27(5), 450–460 (2015).
    • 33. Kang DW, Min Do S. Platelet derived growth factor increases phospholipase D1 but not phospholipase D2 expression via NFkappaB signaling pathway and enhances invasion of breast cancer cells. Cancer Lett. 294(1), 125–133 (2010).
    • 34. Mammadova-Bach E, Gil-Pulido J, Sarukhanyan E et al. Platelet glycoprotein VI promotes metastasis through interaction with cancer cell-derived galectin-3. Blood 135(14), 1146–1160 (2020).
    • 35. Shen J, Ran ZH, Zhang Y et al. Biomarkers of altered coagulation and fibrinolysis as measures of disease activity in active inflammatory bowel disease: a gender-stratified, cohort analysis. Thromb. Res. 123(4), 604–611 (2009).
    • 36. De Luca G, Venegoni L, Iorio S et al. Platelet distribution width and the extent of coronary artery disease: results from a large prospective study. Platelets 21(7), 508–514 (2010).
    • 37. Riedl J, Kaider A, Reitter EM et al. Association of mean platelet volume with risk of venous thromboembolism and mortality in patients with cancer. Results from the Vienna Cancer and Thrombosis Study (CATS). Thromb. Haemost. 111(4), 670–678 (2014).
    • 38. Vagdatli E, Gounari E, Lazaridou E, Katsibourlia E, Tsikopoulou F, Labrianou I. Platelet distribution width: a simple, practical and specific marker of activation of coagulation. Hippokratia 14(1), 28–32 (2010). •• Mean platelet volume and platelet distribution width are simple platelet indices, which increase during platelet activation. Platelet distribution width is a more specific marker of platelet activation since it does not increase during simple platelet swelling.
    • 39. Takeuchi H, Abe M, Takumi Y et al. The prognostic impact of the platelet distribution width-to-platelet count ratio in patients with breast cancer. PLOS ONE 12(12), e0189166 (2017).
    • 40. Kowanetz M, Wu X, Lee J et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl Acad. Sci. USA 107(50), 21248–21255 (2010).
    • 41. Kayacan O, Karnak D, Beder S et al. Impact of TNF-alpha and IL-6 levels on development of cachexia in newly diagnosed NSCLC patients. Am. J. Clin. Oncol. 29(4), 328–335 (2006).