Inflammation-Based Scores Increase the Prognostic Value of Circulating Tumor Cells in Primary Breast Cancer

Svetlana Miklikova, Gabriel Minarik, Tatiana Sedlackova, Jana Plava, Marina Cihova, Silvia Jurisova, Katarina Kalavska, Marian Karaba, Juraj Benca, Bozena Smolkova, Michal Mego, Svetlana Miklikova, Gabriel Minarik, Tatiana Sedlackova, Jana Plava, Marina Cihova, Silvia Jurisova, Katarina Kalavska, Marian Karaba, Juraj Benca, Bozena Smolkova, Michal Mego

Abstract

A correlation between circulating tumor cells (CTCs) and monocytes in metastatic breast cancer (BC), where CTCs and monocyte-to-lymphocyte ratio (MLR) were predictors of overall survival (OS), was recently shown. Herein, we aimed to assess the association between CTCs and the complete blood count (CBC)-derived inflammation-based scores in 284 primary BC patients. CTCs were determined in CD45-depleted peripheral blood mononuclear cells by real time-PCR. This method allowed us to detect a subset of CTCs with an epithelial-to-mesenchymal transition phenotype (CTC EMT), previously associated with inferior outcomes in primary BC. In the present study, CTC EMT positivity (hazard ratio (HR) = 2.4; 95% CI 1.20-4.66, p = 0.013) and elevated neutrophil-to-lymphocyte ratio (NLR) (HR = 2.20; 95% CI 1.07-4.55; p = 0.033) were associated with shorter progression-free survival (PFS) in primary BC patients. Multivariate analysis showed that CTC EMT-positive patients with NLR ≥ 3 had 8.6 times increased risk of disease recurrence (95% CI 2.35-31.48, p = 0.001) compared with CTC EMT-negative patients with NLR < 3. Similarly, disease recurrence was 13.14 times more likely in CTC EMT-positive patients with MLR ≥ 0.34 (95% CI 4.35-39.67, p < 0.001). Given its low methodological and financial demands, the CBC-derived inflammation-based score determination could, after broader validation, significantly improve the prognostication of BC patients.

Keywords: breast cancer; circulating tumor cells; monocyte-to-lymphocyte ratio; neutrophil-to-lymphocyte ratio; platelet-to-lymphocyte ratio; systemic immune-inflammation index.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Kaplan–Meier PFS estimates for CTC EMT. CTC EMT-positive patients had significantly shorter progression-free survival (PFS) than CTC EMT-negative patients (p = 0.010 by Log-rank test).
Figure 2
Figure 2
Kaplan–Meier PFS estimates for NLR (a) and MLR (b) status. PFS was significantly reduced in patients with NLR ≥ 3 compared to those with NLR < 3 (p = 0.029 by Log-rank test), while the difference was not significant for MLR (p = 0.109 by Log-rank test).
Figure 3
Figure 3
Kaplan–Meier PFS estimates for combinations of CTC EMT and NLR index. CTC EMT-positive patients with NLR ≥ 3 had shorter PFS compared to patients with any other combination of CTC EMT and NLR (p = 0.003 by Log-rank test).
Figure 4
Figure 4
Kaplan–Meier PFS estimates for combinations of CTC EMT and MLR index. CTC EMT-positive patients with MLR ≥ 0.34 had shorter PFS compared to patients with all other combinations of CTC EMT and MLR (p < 0.001 by Log-rank test).

References

    1. Harbeck N., Penault-Llorca F., Cortes J., Gnant M., Houssami N., Poortmans P., Ruddy K., Tsang J., Cardoso F. Breast cancer. Nat. Rev. Dis. Prim. 2019;5:66. doi: 10.1038/s41572-019-0111-2.
    1. Arciero C.A., Styblo T.M. The Breast. Elsevier; Amsterdam, The Netherlands: 2018. Clinically Established Prognostic Factors in Breast Cancer; pp. 250–257.
    1. Greten F.R., Grivennikov S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity. 2019;51:27–41. doi: 10.1016/j.immuni.2019.06.025.
    1. Saini M., Szczerba B.M., Aceto N. Circulating Tumor Cell-Neutrophil Tango along the Metastatic Process. Cancer Res. 2019;79:6067–6073. doi: 10.1158/0008-5472.CAN-19-1972.
    1. Coffelt S.B., Wellenstein M.D., de Visser K.E. Neutrophils in cancer: Neutral no more. Nat. Rev. Cancer. 2016;16:431. doi: 10.1038/nrc.2016.52.
    1. Finisguerra V., Di Conza G., Di Matteo M., Serneels J., Costa S., Thompson A.A., Wauters E., Walmsley S., Prenen H., Granot Z., et al. MET is required for the recruitment of anti-tumoural neutrophils. Nature. 2015;522:349–353. doi: 10.1038/nature14407.
    1. Galdiero M.R., Bonavita E., Barajon I., Garlanda C., Mantovani A., Jaillon S. Tumor associated macrophages and neutrophils in cancer. Immunobiology. 2013;218:1402–1410. doi: 10.1016/j.imbio.2013.06.003.
    1. Sagiv J.Y., Michaeli J., Assi S., Mishalian I., Kisos H., Levy L., Damti P., Lumbroso D., Polyansky L., Sionov R.V., et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015;10:562–573. doi: 10.1016/j.celrep.2014.12.039.
    1. Ponzetta A., Carriero R., Carnevale S., Barbagallo M., Molgora M., Perucchini C., Magrini E., Gianni F., Kunderfranco P., Polentarutti N., et al. Neutrophils Driving Unconventional T Cells Mediate Resistance against Murine Sarcomas and Selected Human Tumors. Cell. 2019;178:346–360. doi: 10.1016/j.cell.2019.05.047.
    1. Fridlender Z.G., Albelda S.M. Tumor-associated neutrophils: Friend or foe? Carcinogenesis. 2012;33:949–955. doi: 10.1093/carcin/bgs123.
    1. Gregory A.D., Houghton A.M. Tumor-associated neutrophils: New targets for cancer therapy. Cancer Res. 2011;71:2411–2416. doi: 10.1158/0008-5472.CAN-10-2583.
    1. Piccard H., Muschel R.J., Opdenakker G. On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Crit. Rev. Oncol. Hematol. 2012;82:296–309. doi: 10.1016/j.critrevonc.2011.06.004.
    1. Kowanetz M., Wu X., Lee J., Tan M., Hagenbeek T., Qu X., Yu L., Ross J., Korsisaari N., Cao T., et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl. Acad. Sci. USA. 2010;107:21248–21255. doi: 10.1073/pnas.1015855107.
    1. Granot Z., Henke E., Comen E.A., King T.A., Norton L., Benezra R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 2011;20:300–314. doi: 10.1016/j.ccr.2011.08.012.
    1. Huh S.J., Liang S., Sharma A., Dong C., Robertson G.P. Transiently entrapped circulating tumor cells interact with neutrophils to facilitate lung metastasis development. Cancer Res. 2010;70:6071–6082. doi: 10.1158/0008-5472.CAN-09-4442.
    1. Pantel K., Speicher M.R. The biology of circulating tumor cells. Oncogene. 2016;35:1216–1224. doi: 10.1038/onc.2015.192.
    1. Mego M., Cierna Z., Janega P., Karaba M., Minarik G., Benca J., Sedlackova T., Sieberova G., Gronesova P., Manasova D., et al. Relationship between circulating tumor cells and epithelial to mesenchymal transition in early breast cancer. BMC Cancer. 2015;15:533. doi: 10.1186/s12885-015-1548-7.
    1. Mego M., Gao H., Lee B.N., Cohen E.N., Tin S., Giordano A., Wu Q., Liu P., Nieto Y., Champlin R.E., et al. Prognostic Value of EMT-Circulating Tumor Cells in Metastatic Breast Cancer Patients Undergoing High-Dose Chemotherapy with Autologous Hematopoietic Stem Cell Transplantation. J. Cancer. 2012;3:369–380. doi: 10.7150/jca.5111.
    1. Mego M., Karaba M., Minarik G., Benca J., Silvia J., Sedlackova T., Manasova D., Kalavska K., Pindak D., Cristofanilli M., et al. Circulating Tumor Cells with Epithelial-to-mesenchymal Transition Phenotypes Associated With Inferior Outcomes in Primary Breast Cancer. Anticancer. Res. 2019;39:1829–1837. doi: 10.21873/anticanres.13290.
    1. De Giorgi U., Mego M., Scarpi E., Giordano A., Giuliano M., Valero V., Alvarez R.H., Ueno N.T., Cristofanilli M., Reuben J.M. Association between circulating tumor cells and peripheral blood monocytes in metastatic breast cancer. Ther. Adv. Med. Oncol. 2019;11:1758835919866065. doi: 10.1177/1758835919866065.
    1. Olingy C.E., Dinh H.Q., Hedrick C.C. Monocyte heterogeneity and functions in cancer. J. Leukoc. Biol. 2019;106:309–322. doi: 10.1002/JLB.4RI0818-311R.
    1. Wang L., Simons D.L., Lu X., Tu T.Y., Avalos C., Chang A.Y., Dirbas F.M., Yim J.H., Waisman J., Lee P.P. Breast cancer induces systemic immune changes on cytokine signaling in peripheral blood monocytes and lymphocytes. EBioMedicine. 2020;52:102631. doi: 10.1016/j.ebiom.2020.102631.
    1. Zhong J.H., Huang D.H., Chen Z.Y. Prognostic role of systemic immune-inflammation index in solid tumors: A systematic review and meta-analysis. Oncotarget. 2017;8:75381–75388. doi: 10.18632/oncotarget.18856.
    1. Huszno J., Kolosza Z. Prognostic value of the neutrophil-lymphocyte, platelet-lymphocyte and monocyte-lymphocyte ratio in breast cancer patients. Oncol. Lett. 2019;18:6275–6283. doi: 10.3892/ol.2019.10966.
    1. Ethier J.-L., Desautels D., Templeton A., Shah P.S., Amir E. Prognostic role of neutrophil-to-lymphocyte ratio in breast cancer: A systematic review and meta-analysis. Breast Cancer Res. 2017;19:2. doi: 10.1186/s13058-016-0794-1.
    1. Cuello-Lopez J., Fidalgo-Zapata A., Lopez-Agudelo L., Vasquez-Trespalacios E. Platelet-to-lymphocyte ratio as a predictive factor of complete pathologic response to neoadjuvant chemotherapy in breast cancer. PLoS ONE. 2018;13:e0207224. doi: 10.1371/journal.pone.0207224.
    1. Mao Y., Chen D., Duan S., Zhao Y., Wu C., Zhu F., Chen C., Chen Y. Prognostic impact of pretreatment lymphocyte-to-monocyte ratio in advanced epithelial cancers: A meta-analysis. Cancer Cell Int. 2018;18:201. doi: 10.1186/s12935-018-0698-5.
    1. Jia W., Wu J., Jia H., Yang Y., Zhang X., Chen K., Su F. 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:e0143061. doi: 10.1371/journal.pone.0143061.
    1. Pantel K., Alix-Panabières C. Circulating tumour cells in cancer patients: Challenges and perspectives. Trends Mol. Med. 2010;16:398–406. doi: 10.1016/j.molmed.2010.07.001.
    1. Fabisiewicz A., Grzybowska E. CTC clusters in cancer progression and metastasis. Med. Oncol. 2017;34:12. doi: 10.1007/s12032-016-0875-0.
    1. Thery L., Meddis A., Cabel L., Proudhon C., Latouche A., Pierga J.-Y., Bidard F.-C. Circulating tumor cells in early breast cancer. JNCI Cancer Spectr. 2019;3:pkz026. doi: 10.1093/jncics/pkz026.
    1. Cristofanilli M., Pierga J.Y., Reuben J., Rademaker A., Davis A.A., Peeters D.J., Fehm T., Nole F., Gisbert-Criado R., Mavroudis D., et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): International expert consensus paper. Crit. Rev. Oncol. Hematol. 2019;134:39–45. doi: 10.1016/j.critrevonc.2018.12.004.
    1. Janni W.J., Rack B., Terstappen L.W., Pierga J.-Y., Taran F.-A., Fehm T., Hall C., De Groot M.R., Bidard F.-C., Friedl T.W. Pooled analysis of the prognostic relevance of circulating tumor cells in primary breast cancer. Clin. Cancer Res. 2016;22:2583–2593. doi: 10.1158/1078-0432.CCR-15-1603.
    1. Grover P.K., Cummins A.G., Price T.J., Roberts-Thomson I.C., Hardingham J.E. Circulating tumour cells: The evolving concept and the inadequacy of their enrichment by EpCAM-based methodology for basic and clinical cancer research. Ann. Oncol. 2014;25:1506–1516. doi: 10.1093/annonc/mdu018.
    1. Jie X.-X., Zhang X.-Y., Xu C.-J. Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: Mechanisms and clinical applications. Oncotarget. 2017;8:81558–81571. doi: 10.18632/oncotarget.18277.
    1. Papadaki M.A., Stoupis G., Theodoropoulos P.A., Mavroudis D., Georgoulias V., Agelaki S. Circulating Tumor Cells with Stemness and Epithelial-to-Mesenchymal Transition Features Are Chemoresistant and Predictive of Poor Outcome in Metastatic Breast Cancer. Mol. Cancer Ther. 2019;18:437–447. doi: 10.1158/1535-7163.MCT-18-0584.
    1. Zhang S., Wu T., Peng X., Liu J., Liu F., Wu S., Liu S., Dong Y., Xie S., Ma S. Mesenchymal phenotype of circulating tumor cells is associated with distant metastasis in breast cancer patients. Cancer Manag. Res. 2017;9:691–700. doi: 10.2147/CMAR.S149801.
    1. Guan X., Ma F., Li C., Wu S., Hu S., Huang J., Sun X., Wang J., Luo Y., Cai R., et al. The prognostic and therapeutic implications of circulating tumor cell phenotype detection based on epithelial–mesenchymal transition markers in the first-line chemotherapy of HER2-negative metastatic breast cancer. Cancer Commun. 2019;39:1. doi: 10.1186/s40880-018-0346-4.
    1. Bulfoni M., Gerratana L., Del Ben F., Marzinotto S., Sorrentino M., Turetta M., Scoles G., Toffoletto B., Isola M., Beltrami C.A. In patients with metastatic breast cancer the identification of circulating tumor cells in epithelial-to-mesenchymal transition is associated with a poor prognosis. Breast Cancer Res. 2016;18:30. doi: 10.1186/s13058-016-0687-3.
    1. Hayes D.F., Cristofanilli M., Budd G.T., Ellis M.J., Stopeck A., Miller M.C., Matera J., Allard W.J., Doyle G.V., Terstappen L.W. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin. Cancer Res. 2006;12:4218–4224. doi: 10.1158/1078-0432.CCR-05-2821.
    1. Fischer K.R., Durrans A., Lee S., Sheng J., Li F., Wong S.T., Choi H., El Rayes T., Ryu S., Troeger J. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527:472–476. doi: 10.1038/nature15748.
    1. Ye X., Brabletz T., Kang Y., Longmore G.D., Nieto M.A., Stanger B.Z., Yang J., Weinberg R.A. Upholding a role for EMT in breast cancer metastasis. Nature. 2017;547:E1–E3. doi: 10.1038/nature22816.
    1. Yu M., Bardia A., Wittner B.S., Stott S.L., Smas M.E., Ting D.T., Isakoff S.J., Ciciliano J.C., Wells M.N., Shah A.M. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339:580–584. doi: 10.1126/science.1228522.
    1. Heeke S., Mograbi B., Alix-Panabières C., Hofman P. Never Travel Alone: The Crosstalk of Circulating Tumor Cells and the Blood Microenvironment. Cells. 2019;8:714. doi: 10.3390/cells8070714.
    1. Coussens L.M., Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. doi: 10.1038/nature01322.
    1. Walsh S.R., Cook E.J., Goulder F., Justin T.A., Keeling N.J. Neutrophil-lymphocyte ratio as a prognostic factor in colorectal cancer. J. Surg. Oncol. 2005;91:181–184. doi: 10.1002/jso.20329.
    1. Cao J., Zhu X., Zhao X., Li X.F., Xu R. Neutrophil-to-Lymphocyte Ratio Predicts PSA Response and Prognosis in Prostate Cancer: A Systematic Review and Meta-Analysis. PLoS ONE. 2016;11:e0158770. doi: 10.1371/journal.pone.0158770.
    1. Sarraf K.M., Belcher E., Raevsky E., Nicholson A.G., Goldstraw P., Lim E. Neutrophil/lymphocyte ratio and its association with survival after complete resection in non–small cell lung cancer. J. Thorac. Cardiovasc. Surg. 2009;137:425–428. doi: 10.1016/j.jtcvs.2008.05.046.
    1. Wei B., Yao M., Xing C., Wang W., Yao J., Hong Y., Liu Y., Fu P. The neutrophil lymphocyte ratio is associated with breast cancer prognosis: An updated systematic review and meta-analysis. Onco Targets Ther. 2016;9:5567–5575. doi: 10.2147/OTT.S108419.
    1. Suppan C., Bjelic-Radisic V., La Garde M., Groselj-Strele A., Eberhard K., Samonigg H., Loibner H., Dandachi N., Balic M. Neutrophil/Lymphocyte ratio has no predictive or prognostic value in breast cancer patients undergoing preoperative systemic therapy. BMC Cancer. 2015;15:1027. doi: 10.1186/s12885-015-2005-3.
    1. Zhang M., Huang X.-Z., Song Y.-X., Gao P., Sun J.-X., Wang Z.-N. High platelet-to-lymphocyte ratio predicts poor prognosis and clinicopathological characteristics in patients with breast cancer: A meta-analysis. BioMed Res. Int. 2017;2017:1–11. doi: 10.1155/2017/9503025.
    1. Hernandez C.M., Madrona A.P., Vazquez P.G., Fernández P.G., Merino G.R., Romero J.A., Paricio P.P. 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;20:476–483. doi: 10.1007/s12094-017-1732-0.
    1. Guo W., Lu X., Liu Q., Zhang T., Li P., Qiao W., Deng M. Prognostic value of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio for breast cancer patients: An updated meta-analysis of 17079 individuals. Cancer Med. 2019;8:4135–4148. doi: 10.1002/cam4.2281.
    1. Yao M., Liu Y., Jin H., Liu X., Lv K., Wei H., Du C., Wang S., Wei B., Fu P. Prognostic value of preoperative inflammatory markers in Chinese patients with breast cancer. Onco Targets Ther. 2014;7:1743.
    1. Krenn-Pilko S., Langsenlehner U., Thurner E.M., Stojakovic T., Pichler M., Gerger A., Kapp K.S., Langsenlehner T. The elevated preoperative platelet-to-lymphocyte ratio predicts poor prognosis in breast cancer patients. Br. J. Cancer. 2014;110:2524–2530. doi: 10.1038/bjc.2014.163.
    1. Geng S.K., Fu S.M., Fu Y.P., Zhang H.W. Neutrophil to lymphocyte ratio is a prognostic factor for disease free survival in patients with breast cancer underwent curative resection. Medicine (Baltimore) 2018;97:e11898. doi: 10.1097/MD.0000000000011898.
    1. Ivars Rubio A., Yufera J.C., de la Morena P., Fernández Sánchez A., Navarro Manzano E., García Garre E., García Martinez E., Marín Zafra G., Sánchez Cánovas M., García Torralba E., et al. Neutrophil-lymphocyte ratio in metastatic breast cancer is not an independent predictor of survival, but depends on other variables. Sci. Rep. 2019;9:16979. doi: 10.1038/s41598-019-53606-3.
    1. Wen J., Ye F., Huang X., Li S., Yang L., Xiao X., Xie X. Prognostic Significance of Preoperative Circulating Monocyte Count in Patients with Breast Cancer: Based on a Large Cohort Study. Medicine (Baltimore) 2015;94:e2266. doi: 10.1097/MD.0000000000002266.
    1. Szczerba B.M., Castro-Giner F., Vetter M., Krol I., Gkountela S., Landin J., Scheidmann M.C., Donato C., Scherrer R., Singer J., et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature. 2019;566:553–557. doi: 10.1038/s41586-019-0915-y.
    1. Wculek S.K., Malanchi I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature. 2015;528:413–417. doi: 10.1038/nature16140.
    1. Hamilton G., Rath B., Klameth L., Hochmair M.J. Small cell lung cancer: Recruitment of macrophages by circulating tumor cells. Oncoimmunology. 2016;5:e1093277. doi: 10.1080/2162402X.2015.1093277.
    1. Wyckoff J.B., Wang Y., Lin E.Y., Li J.F., Goswami S., Stanley E.R., Segall J.E., Pollard J.W., Condeelis J. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007;67:2649–2656. doi: 10.1158/0008-5472.CAN-06-1823.
    1. Williams C.B., Yeh E.S., Soloff A.C. Tumor-associated macrophages: Unwitting accomplices in breast cancer malignancy. NPJ Breast Cancer. 2016;2:15025. doi: 10.1038/npjbcancer.2015.25.
    1. Mu Z., Wang C., Ye Z., Rossi G., Sun C., Li L., Zhu Z., Yang H., Cristofanilli M. Prognostic values of cancer associated macrophage-like cells (CAML) enumeration in metastatic breast cancer. Breast Cancer Res. Treat. 2017;165:733–741. doi: 10.1007/s10549-017-4372-8.
    1. Reduzzi C., Vismara M., Gerratana L., Silvestri M., De Braud F., Raspagliesi F., Verzoni E., Di Cosimo S., Locati L.D., Cristofanilli M., et al. The curious phenomenon of dual-positive circulating cells: Longtime overlooked tumor cells. Semin. Cancer Biol. 2020;60:344–350. doi: 10.1016/j.semcancer.2019.10.008.
    1. Gast C.E., Silk A.D., Zarour L., Riegler L., Burkhart J.G., Gustafson K.T., Parappilly M.S., Roh-Johnson M., Goodman J.R., Olson B. Cell fusion potentiates tumor heterogeneity and reveals circulating hybrid cells that correlate with stage and survival. Sci. Adv. 2018;4:eaat7828. doi: 10.1126/sciadv.aat7828.
    1. Cierna Z., Mego M., Janega P., Karaba M., Minarik G., Benca J., Sedlácková T., Cingelova S., Gronesova P., Manasova D., et al. Matrix metalloproteinase 1 and circulating tumor cells in early breast cancer. BMC Cancer. 2014;14:472. doi: 10.1186/1471-2407-14-472.
    1. Mego M., Mani S.A., Lee B.N., Li C., Evans K.W., Cohen E.N., Gao H., Jackson S.A., Giordano A., Hortobagyi G.N., et al. Expression of epithelial-mesenchymal transition-inducing transcription factors in primary breast cancer: The effect of neoadjuvant therapy. Int. J. Cancer. 2012;130:808–816. doi: 10.1002/ijc.26037.

Source: PubMed

3
订阅