Pre-treatment systemic immune-inflammation index is a useful prognostic indicator in patients with breast cancer undergoing neoadjuvant chemotherapy

Li Chen, Xiangyi Kong, Zhongzhao Wang, Xiangyu Wang, Yi Fang, Jing Wang, Li Chen, Xiangyi Kong, Zhongzhao Wang, Xiangyu Wang, Yi Fang, Jing Wang

Abstract

The systemic immune-inflammation index (SII = N × P/L) based on neutrophil (N), platelet (P) and lymphocyte (L) counts is used to predict the survival of patients with malignant tumours and can fully reflect the balance between host inflammatory and immune status. This study is conducted to explore the potential prognostic significance of SII in patients with breast cancer undergoing neoadjuvant chemotherapy (NACT). A total of 262 patients with breast cancer received NACT were enrolled in this study. According to the receiver operating characteristic curve, the optimal cut-off value of SII was divided into two groups: low SII group (<602 × 109 /L) and high SII group (≥602 × 109 /L). The associations between breast cancer and clinicopathological variables by SII were determined by chi-squared test or Fisher's exact test. The Kaplan-Meier plots and log-rank test were used to determine clinical outcomes of disease-free survival (DFS) and overall survival (OS). The prognostic value of SII was analysed by univariate and multivariate Cox proportional hazards regression models. The toxicity of NACT was accessed by National Cancer Institute Common Toxicity Criteria (NCICTC). According to univariate and multivariate Cox regression survival analyses, the results showed that the value of SII had prognostic significance for DFS and OS. The patients with low SII value had longer DFS and OS than those with high SII value (31.11 vs 40.76 months, HR: 1.075, 95% CI: 0.718-1.610, P = .006; 44.47 vs 53.68 months, HR: 1.051, 95% CI: 0.707-1.564, P = .005, respectively). The incidence of DFS and OS in breast cancer patients with low SII value was higher than that in those patients with high SII value in 3-, 5- and 10-year rates. The common toxicities after NACT were haematological and gastrointestinal reaction, and there were no differences by SII for the assessment of side effects of neoadjuvant chemotherapy. Meanwhile, the results also proved that breast cancer patients with low SII value and high Miller and Payne grade (MPG) survived longer than those breast cancer with high SII value and low MPG grade. In patients without lymph vessel invasion, these breast cancer patients with low SII value had better prognosis and lower recurrence rates than those with high SII value. Pre-treatment SII with the advantage of reproducible, convenient and non-invasive was a useful prognostic indicator for breast cancer patients undergoing neoadjuvant chemotherapy and is a promising biomarker for breast cancer on treatment strategy decisions.

Keywords: breast cancer; neoadjuvant chemotherapy; prognosis; survival; systemic immune-inflammation index (SII).

Conflict of interest statement

The authors declare no competing financial interests.

© 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
DFS and OS of patients with breast cancer. A, Kaplan‐Meier analysis of DFS of all patients with breast cancer. B, Kaplan‐Meier analysis of OS of all patients with breast cancer. C, Kaplan‐Meier analysis of DFS for the SII of all patients with breast cancer. D, Kaplan‐Meier analysis of OS for the SII of all patients with breast cancer. SII is a novel systemic immune‐inflammation index (SII = N×P/L), which is based on neutrophil (N), platelet (P) and lymphocyte (L) counts
Figure 2
Figure 2
The 3‐, 5‐ and 10‐year rates of DFS and OS in patients with breast cancer. A, The 3‐, 5‐ and 10‐year rates of DFS in all patients with breast cancer. B, The 3‐, 5‐ and 10‐year rates of OS in all patients with breast cancer. C, The 3‐, 5‐ and 10‐year rates of DFS in all patients by SII with breast cancer. D, The 3‐, 5‐ and 10‐year rates of OS in all patients by SII with breast cancer
Figure 3
Figure 3
DFS and OS of patients for the SII by molecular subtypes with breast cancer. A, Kaplan‐Meier analysis of DFS of patients by Luminal A subtype with breast cancer. B, Kaplan‐Meier analysis of OS of patients by Luminal A subtype with breast cancer. C, Kaplan‐Meier analysis of DFS of patients by Luminal B HER2‐positive subtype with breast cancer. D, Kaplan‐Meier analysis of OS of patients by Luminal B HER2‐positive subtype with breast cancer. E, Kaplan‐Meier analysis of DFS of patients by Luminal B HER2‐negative subtype with breast cancer. F, Kaplan‐Meier analysis of OS of patients by Luminal B HER2‐negative subtype with breast cancer. G, Kaplan‐Meier analysis of DFS of patients by HER2‐enriched subtype with breast cancer. H, Kaplan‐Meier analysis of OS of patients by HER2‐enriched subtype with breast cancer. I, Kaplan‐Meier analysis of DFS of patients by Triple negative subtype with breast cancer. J, Kaplan‐Meier analysis of OS of patients by Triple negative subtype with breast cancer
Figure 4
Figure 4
DFS and OS of patients for the SII by Miller and Payne grade (MPG) with breast cancer. A, Kaplan‐Meier analysis of DFS of patients by MPG‐A group with breast cancer. B, Kaplan‐Meier analysis of OS of patients by MPG‐A group with breast cancer. C, Kaplan‐Meier analysis of DFS of patients by MPG‐B group with breast cancer. D, Kaplan‐Meier analysis of OS of patients by MPG‐B group with breast cancer. E, Kaplan‐Meier analysis of DFS of patients by MPG‐C group with breast cancer. F, Kaplan‐Meier analysis of OS of patients by MPG‐C group with breast cancer
Figure 5
Figure 5
DFS and OS of patients for the SII by ER status with breast cancer. A, Kaplan‐Meier analysis of DFS of patients by ER negative with breast cancer in core needle biopsy. B, Kaplan‐Meier analysis of OS of patients by ER negative with breast cancer in core needle biopsy. C, Kaplan‐Meier analysis of DFS of patients by ER positive with breast cancer in core needle biopsy. D, Kaplan‐Meier analysis of OS of patients by ER positive with breast cancer in core needle biopsy. E, Kaplan‐Meier analysis of DFS of patients by ER negative with breast cancer in post‐operative pathology IHC. F, Kaplan‐Meier analysis of OS of patients by ER negative with breast cancer in post‐operative pathology IHC. G, Kaplan‐Meier analysis of DFS of patients by ER positive with breast cancer in post‐operative pathology IHC. H, Kaplan‐Meier analysis of OS of patients by ER positive with breast cancer in post‐operative pathology IHC
Figure 6
Figure 6
DFS and OS of patients for the SII by Ki‐67 status with breast cancer. A, Kaplan‐Meier analysis of DFS of patients by Ki‐67 negative with breast cancer. B, Kaplan‐Meier analysis of OS of patients by Ki‐67 negative with breast cancer. C, Kaplan‐Meier analysis of DFS of patients by Ki‐67 positive with breast cancer. D, Kaplan‐Meier analysis of OS of patients by Ki‐67 positive with breast cancer
Figure 7
Figure 7
DFS and OS of patients for the SII by lymph vessel invasion status with breast cancer. A, Kaplan‐Meier analysis of DFS of patients by lymph vessel invasion negative with breast cancer. B, Kaplan‐Meier analysis of OS of patients by lymph vessel invasion negative with breast cancer. C, Kaplan‐Meier analysis of DFS of patients by lymph vessel invasion positive with breast cancer. D, Kaplan‐Meier analysis of OS of patients by lymph vessel invasion positive with breast cancer

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7‐34.
    1. Iqbal J, Amir E, Rochon PA, Giannakeas V, Sun P, Narod SA. Association of the timing of pregnancy with survival in women with breast cancer. JAMA Oncol. 2017;3(5):659‐665.
    1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359‐E386.
    1. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115‐132.
    1. Chen W, Zheng R, Zhang S, et al. Cancer incidence and mortality in China in 2013: an analysis based on urbanization level. Chin J Cancer Res. 2017;29(1):1‐10.
    1. Patel DA, Xi J, Luo J, et al. Neutrophil‐to‐lymphocyte ratio as a predictor of survival in patients with triple‐negative breast cancer. Breast Cancer Res Treat. 2019;174(2):443‐452.
    1. Wang M, Hou L, Chen M, et al. Neoadjuvant chemotherapy creates surgery opportunities for inoperable locally advanced breast cancer. Sci Rep. 2017;7:44673.
    1. Sinacki M, Badzio A, Wełnicka‐Jaśkiewicz M, et al. Pattern of care in locally advanced breast cancer: focus on local therapy. Breast. 2011;20(2):145‐150.
    1. Matuschek C, Bölke E, Haussmann J, et al. The benefit of adjuvant radiotherapy after breast conserving surgery in older patients with low risk breast cancer‐ a meta‐analysis of randomized trials. Radiat Oncol. 2017;12(1):60.
    1. He Z, Song A, Zhang Z, et al. Comparative efficacy of non‐pharmacological adjuvant therapies for quality of life in the patients with breast cancer receiving chemo‐ or radio‐therapy: a protocol for systematic review and Bayesian network meta‐analysis. Medicine (Baltimore). 2018;97(35):e12096.
    1. Brackstone M, Palma D, Tuck AB, et al. Concurrent neoadjuvant chemotherapy and radiation therapy in locally advanced breast cancer. Int J Radiat Oncol Biol Phys. 2017;99(4):769‐776.
    1. Derks MGM, van de Velde CJH. Neoadjuvant chemotherapy in breast cancer: more than just downsizing. Lancet Oncol. 2018;19(1):2‐3.
    1. DeMichele A, Yee D, Esserman L. Mechanisms of resistance to neoadjuvant chemotherapy in breast cancer. N Engl J Med. 2017;377(23):2287‐2289.
    1. Xie H, Liu J, Yu S, et al. Patterns of use of docetaxel‐containing adjuvant chemotherapy among Chinese patients with operable breast cancer: a multicenter observational study. Adv Ther. 2019;36(1):131‐146.
    1. Franceschini G, Di Leone A, Natale M, et al. Conservative surgery after neoadjuvant chemotherapy in patients with operable breast cancer. Ann Ital Chir. 2018;89:290.
    1. Pathak M, Dwivedi SN, Deo SVS, Thakur B, Sreenivas V, Rath GK. Neoadjuvant chemotherapy regimens in treatment of breast cancer: a systematic review and network meta‐analysis protocol. Syst Rev. 2018;7(1):89.
    1. Wu D, Chen T, Jiang H, et al. Comparative efficacy and tolerability of neoadjuvant immunotherapy regimens for patients with HER2‐positive breast cancer: a network meta‐analysis. J Oncol. 2019;2019:3406972.
    1. Medeiros B, Allan AL. Molecular mechanisms of breast cancer metastasis to the lung: clinical and experimental perspectives. Int J Mol Sci. 2019;20(9):pii: E2272.
    1. Karn T, Jiang T, Hatzis C, et al. Association between genomic metrics and immune infiltration in triple‐negative breast cancer. JAMA Oncol. 2017;3(12):1707‐1711.
    1. van Vulpen JK, Schmidt ME, Velthuis MJ, et al. Effects of physical exercise on markers of inflammation in breast cancer patients during adjuvant chemotherapy. Breast Cancer Res Treat. 2018;168(2):421‐431.
    1. Elyasinia F, Keramati MR, Ahmadi F, et al. Neutrophil‐lymphocyte ratio in different stages of breast cancer. Acta Med Iran. 2017;55(4):228‐232.
    1. Ma JY, Hu G, Liu Q. Prognostic significance of the lymphocyte‐to‐monocyte ratio in bladder cancer undergoing radical cystectomy: a meta‐analysis of 5638 individuals. Dis Markers. 2019;2019:7593560.
    1. Chen L, Hao Y, Cong X, et al. Peripheral venous blood platelet‐to‐lymphocyte ratio (PLR) for predicting the survival of patients with gastric cancer treated with SOX or XELOX regimen neoadjuvant chemotherapy. Technol Cancer Res Treat. 2019;18:1533033819829485.
    1. Li S, Xu H, Wang W, et al. The systemic inflammation response index predicts survival and recurrence in patients with resectable pancreatic ductal adenocarcinoma. Cancer Manag Res. 2019;11:3327‐3337.
    1. Chen JH, Zhai ET, Yuan YJ, et al. Systemic immune‐inflammation index for predicting prognosis of colorectal cancer. World J Gastroenterol. 2017;23(34):6261‐6272.
    1. Wang K, Diao F, Ye Z, et al. Prognostic value of systemic immune‐inflammation index in patients with gastric cancer. Chin J Cancer. 2017;36(1):75.
    1. Abdel‐Rahman O. Validation of the 8th AJCC prognostic staging system for breast cancer in a population‐based setting. Breast Cancer Res Treat. 2018;168(1):269‐275.
    1. Fouad TM, Barrera AMG, Reuben JM, et al. Inflammatory breast cancer: a proposed conceptual shift in the UICC‐AJCC TNM staging system. Lancet Oncol. 2017;18(4):e228‐e232.
    1. Sandberg LJ, Clemens MW, Symmans WF, et al. Molecular profiling using breast cancer subtype to plan for breast reconstruction. Plast Reconstr Surg. 2017;139(3):586e‐596e.
    1. Soldevilla‐Gallardo I, Medina‐Ornelas SS, Villarreal‐Garza C, et al. Usefulness of positron emission mammography in the evaluation of response to neoadjuvant chemotherapy in patients with breast cancer. Am J Nucl Med Mol Imaging. 2018;8(5):341‐350.
    1. Phukan JP, Sinha A, Deka JP. Cytological grading of breast carcinoma on fine needle aspirates and its relation with histological grading. South Asian J Cancer. 2015;4(1):32‐34.
    1. Singh K, He X, Kalife ET, et al. Relationship of histologic grade and histologic subtype with oncotype Dx recurrence score; retrospective review of 863 breast cancer oncotype Dx results. Breast Cancer Res Treat. 2018;168(1):29‐34.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228‐247.
    1. Magbanua MJM, Rugo HS, Wolf DM, et al. Expanded genomic profiling of circulating tumor cells in metastatic breast cancer patients to assess biomarker status and biology over time (CALGB 40502 and CALGB 40503, alliance). Clin Cancer Res. 2018;24(6):1486‐1499.
    1. Hodorowicz‐Zaniewska D, Herman K. Principles of follow‐up assessments in patients with breast cancer after radical treatment. Pol Przegl Chir. 2017;89(3):36‐39.
    1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394‐424.
    1. Vaidya JS, Massarut S, Vaidya HJ, et al. Rethinking neoadjuvant chemotherapy for breast cancer. BMJ. 2018;360:j5913.
    1. Harbeck N, Gluz O. Neoadjuvant therapy for triple negative and HER2‐positive early breast cancer. Breast. 2017;34(Suppl 1):S99‐S103.
    1. Shinko D, Diakos CI, Clarke SJ, et al. Cancer‐related systemic inflammation: the challenges and therapeutic opportunities for personalized medicine. Clin Pharmacol Ther. 2017;102(4):599‐610.
    1. Romero‐Cordoba S, Meneghini E, Sant M, et al. Decoding immune heterogeneity of triple negative breast cancer and its association with systemic inflammation. Cancers (Basel). 2019;11(7) pii: E911
    1. Zhao LY, Yang DD, Ma XK, et al. The prognostic value of aspartate aminotransferase to lymphocyte ratio and systemic immune‐inflammation index for overall survival of hepatocellular carcinoma patients treated with palliative treatments. J Cancer. 2019;10(10):2299‐2311.
    1. Zhou T, Zhao Y, Zhao S, et al. Comparison of the prognostic value of systemic inflammation response markers in small cell lung cancer patients. J Cancer. 2019;10(7):1685‐1692.
    1. Lerman I, Hammes SR. Neutrophil elastase in the tumor microenvironment. Steroids. 2018;133:96‐101.
    1. Walz W, Cayabyab FS. Neutrophil infiltration and matrix metalloproteinase‐9 in lacunar Infarction. Neurochem Res. 2017;42(9):2560‐2565.
    1. Fusella F, Seclì L, Busso E, et al. The IKK/NF‐κB signaling pathway requires Morgana to drive breast cancer metastasis. Nat Commun. 2017;8(1):1636.
    1. Tan KW, Chong SZ, Wong FH, et al. Neutrophils contribute to inflammatory lymphangiogenesis by increasing VEGF‐A bioavailability and secreting VEGF‐D. Blood. 2013;122(22):3666‐3677.
    1. Xiao P, Long X, Zhang L, et al. Neurotensin/IL‐8 pathway orchestrates local inflammatory response and tumor invasion by inducing M2 polarization of tumor‐associated macrophages and epithelial‐mesenchymal transition of hepatocellular carcinoma cells. Oncoimmunology. 2018;7(7):e1440166.
    1. Ronca R, Benkheil M, Mitola S, et al. Tumor angiogenesis revisited: regulators and clinical implications. Med Res Rev. 2017;37(6):1231‐1274.
    1. Huang C, Li Z, Li N, et al. Interleukin 35 expression correlates with microvessel density in pancreatic ductal adenocarcinoma, recruits monocytes, and promotes growth and angiogenesis of xenograft tumors in mice. Gastroenterology. 2018;154(3):675‐688.
    1. Laviron M, Combadière C, Boissonnas A. Tracking monocytes and macrophages in tumors with live imaging. Front Immunol. 2019;10:1201.
    1. Jiang L, Luan Y, Miao X, et al. Platelet releasate promotes breast cancer growth and angiogenesis via VEGF‐integrin cooperative signaling. Br J Cancer. 2017;117(5):695‐703.
    1. Kanikarla‐Marie P, Lam M, Menter DG, Kopetz S. circulating tumor cells, and the circulome. Cancer Metastasis Rev. 2017;36(2):235‐248.
    1. Menter DG, Kopetz S, Hawk E, et al. Platelet "first responders" in wound response, cancer, and metastasis. Cancer Metastasis Rev. 2017;36(2):199‐213.
    1. Shankaran V, Ikeda H, Bruce AT, et al. IFN gamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature. 2001;410(6832):1107‐1111.
    1. Mohme M, Riethdorf S, Pantel K. Circulating and disseminated tumour cells‐mechanisms of immune surveillance and escape. Nat Rev Clin Oncol. 2017;14(3):155‐167.
    1. Velaei K, Samadi N, Barazvan B, et al. Tumor microenvironment‐mediated chemoresistance in breast cancer. Breast. 2016;30:92‐100.
    1. Droin N, Hendra JB, Ducoroy P, et al. Human defensins as cancer biomarkers and antitumour molecules. J Proteomics. 2009;72(6):918‐927.
    1. Templeton AJ, Rodríguez‐Lescure Á, Ruíz A, et al. Prognostic role for the derived neutrophil‐to‐lymphocyte ratio in early breast cancer: a GEICAM/9906 substudy. Clin Transl Oncol. 2018;20(12):1548‐1556.
    1. Chen J, Pan Y, He B, et al. Meta‐analysis of prognostic value of inflammation parameter in breast cancer. J Cancer Res Ther. 2018;14(Supplement):S85‐S89.
    1. Marín Hernández C, Piñero Madrona A, Gil Vázquez PJ, et al. Usefulness of lymphocyte‐to‐monocyte, neutrophil‐to‐monocyte and neutrophil‐to‐lymphocyte ratios as prognostic markers in breast cancer patients treated with neoadjuvant chemotherapy. Clin Transl Oncol. 2018;20(4):476‐483.
    1. Goto W, Kashiwagi S, Asano Y, et al. Predictive value of lymphocyte‐to‐monocyte ratio in the preoperative setting for progression of patients with breast cancer. BMC Cancer. 2018;18(1):1137.
    1. Jia W, Wu J, Jia H, et al. The peripheral blood neutrophil‐to‐lymphocyte ratio is superior to the lymphocyte‐to‐monocyte ratio for predicting the long‐term survival of triple‐negative breast cancer patients. PLoS ONE. 2015;10(11):e0143061.
    1. Hu RJ, Liu Q, Ma JY, et al. Preoperative lymphocyte‐to‐monocyte ratio predicts breast cancer outcome: a meta‐analysis. Clin Chim Acta. 2018;484:1‐6.
    1. Asano Y, Kashiwagi S, Onoda N, et al. Platelet‐lymphocyte ratio as a useful predictor of the therapeutic effect of neoadjuvant chemotherapy in breast cancer. PLoS ONE. 2016;11(7):e0153459.
    1. Rafee S, McHugh D, Greally M, et al. Neutrophil‐to‐lymphocyte ratio (NLR) and platelet‐to‐lymphocyte ratio (PLR) as predictive biomarkers of pathologic complete response (pCR) in neoadjuvant breast cancer: an Irish Clinical Oncology Group study (ICORG 16–20). Ann Oncol. 2016;27(suppl 6):1538P.
    1. Cuello‐López J, Fidalgo‐Zapata A, López‐Agudelo L, et al. Platelet‐to‐lymphocyte ratio as a predictive factor of complete pathologic response to neoadjuvant chemotherapy in breast cancer. PLoS ONE. 2018;13(11):e0207224.
    1. Chen L, Yan Y, Zhu L, et al. Systemic immune‐inflammation index as a useful prognostic indicator predicts survival in patients with advanced gastric cancer treated with neoadjuvant chemotherapy. Cancer Manag Res. 2017;9:849‐867.
    1. Fan L, Wang R, Chi C, et al. Systemic immune‐inflammation index predicts the combined clinical outcome after sequential therapy with abiraterone and docetaxel for metastatic castration‐resistant prostate cancer patients. Prostate. 2018;78(4):250‐256.
    1. Tong YS, Tan J, Zhou XL, et al. Systemic immune‐inflammation index predicting chemoradiation resistance and poor outcome in patients with stage III non‐small cell lung cancer. J Transl Med. 2017;15(1):221.
    1. Aziz MH, Sideras K, Aziz NA, et al. The systemic‐immune‐inflammation index independently predicts survival and recurrence in resectable pancreatic cancer and its prognostic value depends on bilirubin levels: a retrospective multicenter cohort study. Ann Surg. 2019;270(1):139‐146.
    1. Robertson S, Stålhammar G, Darai‐Ramqvist E, et al. Prognostic value of Ki67 analysed by cytology or histology in primary breast cancer. J Clin Pathol. 2018;71(9):787‐794.
    1. Ellis MJ, Suman VJ, Hoog J, et al. Ki67 proliferation index as a tool for chemotherapy decisions during and after neoadjuvant aromatase inhibitor treatment of breast cancer: results from the American college of surgeons oncology group Z1031 trial (Alliance). J Clin Oncol. 2017;35(10):1061‐1069.
    1. Karaman S, Detmar M. Mechanisms of lymphatic metastasis. J Clin Invest. 2014;124(3):922‐928.
    1. Kato T, Kameoka S, Kimura T, et al. The combination of angiogenesis and blood vessel invasion as a prognostic indicator in primary breast cancer. Br J Cancer. 2003;88(12):1900‐1908.

Source: PubMed

3
Abonner