Early prediction of pathologic response to neoadjuvant treatment of breast cancer: use of a cell-loss metric based on serum thymidine kinase 1 and tumour volume

Bernhard Tribukait, Bernhard Tribukait

Abstract

Background: After neoadjuvant chemotherapy of breast cancer pathologic complete response (pCR) indicates a favorable prognosis. Among non-selected patients, pCR is, however, achieved in only 10-30%. Early evaluation of tumour response to treatment would facilitate individualized therapy, with ineffective chemotherapy interrupted or changed. The methodology for this purpose is still limited. Tumour imaging and analysis of macromolecules, released from disrupted tumour cells, are principal alternatives.

Objective: To investigate whether a metric of cell-loss, defined as the ratio between serum concentration of thymidine kinase1 (sTK1, ng x ml- 1) and tumour volume, can be used for early prediction of pathologic response.

Methods: One hunred four women with localized breast cancer received neoadjuvant epirubicin/docetaxel in 6 cycles, supplemented with bevacizumab in cycles 3-6. The cell-loss metric was established at baseline (n = 104), 48 h after cycle 2 (n = 104) and prior to cycle 2 (n = 57). The performance of the metric was evaluated by association with pathologic tumour response at surgery 4 months later.

Results: Treatment caused a rise in sTK1, a reduction in tumour volume and a marked increase in the cell-loss metric. Patients were subdivided into quartiles according to the baseline cell-loss metric. For these groups, baseline means were 0.0016, 0.0042, 0.0062, 0.0178 units. After subtraction of baselines, means for the quartiles 48 h after treatment 2 were 0.002, 0.011, 0.030 and 0.357 units. pCR was achieved in 24/104, their distribution in the quartiles (11, 11, 23 and 46%) differed significantly (p = 0.01). In 80 patients with remaining tumour, tumour size was inversely related to the metric (p = 0.002). In 57 patients studied before treatment 2, positive and negative predictive values of the metric were 77.8 and 83.3%, compared to 40.5 and 88.7% 48 h after treatment 2.

Conclusion: A cell-loss metric, based on serum levels of TK1, released from disrupted tumour cells, and tumour volume, reveal tumour response early during neoadjuvant treatment. The metric reflect tumour properties that differ greatly between patients and determine the sensitivity to cytotoxic treatment. The findings point to the significance of cell loss for tumour growth rate. The metric should be considered in personalized oncology and in the evaluation of new therapeutic modalities.

Trial registration: PROMIX (Clinical Trials.govNCT000957125).

Trial registration: ClinicalTrials.gov NCT00957125.

Keywords: Biomarker; Breast cancer; Cell-loss; Circulating thymidine kinase 1; Treatment response.

Conflict of interest statement

BT is a shareholder in AroCell Ab. The manuscript is written completely independent of the company.

Figures

Fig. 1
Fig. 1
Receiver operating characteristic for distinguishing pCR from remaining tumour in 104 women, based on the cell-loss metric 48 h after the 2nd treatment cycle. At a cut-off value of 0.026 for the cell-loss metric, 1-specificity and sensitivity were 0.31 and 0.71, respectively. ROC Area = 0.714, p = 0.02
Fig. 2
Fig. 2
Cell-loss metric 48 h after the 2nd treatment cycle in relation to pathologic tumour volume at surgery after six treatment cycles (p = 0.002)
Fig. 3
Fig. 3
Percentage of pathological complete response in the breast after six cycles of chemotherapy among 104 women, grouped into quartiles according to the cell-loss metric obtained 48 h after the 2nd treatment cycle

References

    1. Kaufmann M, von Minckwitz G, Mamounas EP, Cameron D, Carey L, Christofanilli M, et al. Recommendations from an international consensus conference on the current status and future of neoadjuvant systemic therapy in primary breast cancer. Ann Surg Oncol. 2012;19(5):1508–1516. doi: 10.1245/s10434-011-2108-2.
    1. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384:164–172. doi: 10.1016/S0140-6736(13)62422-8.
    1. Broglio KR, Quintana M, Foster M, Olinger M, McGlothlin A, Berry SM, et al. Association of pathologic complete response to neoadjuvant therapy in HER2-positive breast cancer with long-term outcomes. JAMA Oncol. 2016;2(6):751–760. doi: 10.1001/jamaoncol.2015.6113.
    1. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. 2018;19:27–39. doi: 10.1016/S1470-2045(17)30777-5.
    1. Fisher B, Bryant J, Wolmark N, Mamounas E, Brown A, Fisher ER, et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol. 1998;16(8):2672–2685. doi: 10.1200/JCO.1998.16.8.2672.
    1. van der Hage JA, van de Velde CJH, Julien J-P, Tubiana-Hulin M, Vandervelden C, Duchateau L. and Cooperating Investigators. Preoperative chemotherapy in primary operable breast cancer: results from the European organization for research and treatment of cancer trial 10902. J Clin Oncol. 2001;19(22):4224–4237. doi: 10.1200/JCO.2001.19.22.4224.
    1. US Food and drug administration. Guidelines for industry. Pathologic complete response in neoadjuvant treatment of high-risk early-stage breast cancer: use as an endpoint to support accelerating approval. .
    1. Chavez-MacGregor M, Litton J, Chen H, Giordano SH, Hudis CA, Wolff AC, et al. Pathologic complete response in breast cancer patients receiving anthracycline- and taxane-based neoadjuvant chemotherapy. Cancer. 2010;116:4168–4177. doi: 10.1002/cncr.25296.
    1. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121(7):2750–2767. doi: 10.1172/JCI45014.
    1. Kalimutho M, Parsons K, Mittal D, López JA, Srihari S, Khanna KK. Targeted Therapies for Triple-Negative Breast Cancer: Combating a Stubborn Disease. Trends in Pharmacological Sciences. 2015;36(12):822–846. doi: 10.1016/j.tips.2015.08.009.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45(2):228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Avril N, Sassen S, Roylance R. Response to therapy in breast cancer. J Nucl Med. 2009;50:55S–63S. doi: 10.2967/jnumed.108.057240.
    1. Alix-Panabieres C, Pantel K. Clinical applications of circulating tumour cells and circulating tumour DNA as liquid biopsy. Cancer Discov. 2016;10:479–491. doi: 10.1158/-15-1483.
    1. Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–238. doi: 10.1038/nrc.2017.7.
    1. Munch-Petersen B. Enzymatic regulation of cytosolic thymidine kinase 1 and mitochondrial thymidine kinase 2: a mini review. Nucleosides, Nucleotides and Nucleic Acids. 2010;29:363–369. doi: 10.1080/15257771003729591.
    1. Ке P-Y, Chang Z-F. Mitotic degradation of human thymidine kinase 1 is dependent on the anaphase-promoting complex/cyclosome-Cdh1-mediated pathway. Mol Cell Biol. 2004;24(2):514–526. doi: 10.1128/MCB.24.2.514-526.2004.
    1. Topolcan O, Holubec L., Jr The role of thymidine kinase in cancer diseases. Expert Opin Med Diagn. 2008;2(2):129–141. doi: 10.1517/17530059.2.2.129.
    1. Kumar J, Aronsson A, Pilko G. Zupan M, Kumer K, Fabjan T, et al. A clinical evaluation of the TK 210 ELISA in sera from breast cancer patients demonstrates high sensitivity and specificity in all stages of disease.Tumor Biology. 2016;37(9):11937-11945.
    1. Tribukait B, Jagarlamudi KK, Bergh J, Hatschek T. Quantification of cell-loss in breast cancer during neoadjuvant treatment (NACT) assessed by serum thymidine kinase protein concentration (sTK1). Ann Oncol. 2017;28 suppl.10:81.
    1. Kimbung S, Markholm I, Bjöhle J, Lekberg T, von Wachenfeldt A, Azavedo E, et al. For the PROMIX Trialists Group. Assessment of early response biomarkers in relation to long-term survival in patients with HER2-negative breast cancer receiving neoadjuvant chemotherapy plus bevacizumab: Results from the phase II PROMIX trial. Int J Cancer. 2018;142:618–628. doi: 10.1002/ijc.31070.
    1. Tubiana M. Tumor cell proliferation kinetics and tumour growth rate. Acta Oncol. 1989;28(1):113–121. doi: 10.3109/02841868909111193.
    1. Weber WA. Assessing tumour response to therapy. J Nucl Med. 2009;50:1s-10s.
    1. Alison JD, Tannock IF. Repopulation of tumour cells between cycles of chemotherapy: a neglected factor. Lancet Oncol. 2000;1:86–93. doi: 10.1016/S1470-2045(00)00019-X.
    1. Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R, Frabetti F, et al. An estimation of the number of cells in the human body. Ann Hum Biol. 2013;40(6):463–471. doi: 10.3109/03014460.2013.807878.
    1. Smaaland R, Laerum OD, Lote K, Sletvold O, Sothern RB, Bjerknes R. DNA synthesis in human bone marrow is circadian stage dependent. Blood. 1991;77(12):2603–2611. doi: 10.1182/blood.V77.12.2603.2603.
    1. Nagata S. Apoptosis and clearance of apoptotic cells. Annu Rev Immunol. 2018;36:487–517. doi: 10.1146/annurev-immunol-042617-053010.
    1. Choi J-J, Reich CD, III, Pisetsky DS. The role of macrophages in the in vitro generation of extracellular DNA from apoptotic and necrotic cells. Immunology. 2005;115:55–62. doi: 10.1111/j.1365-2567.2005.02130.x.
    1. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996;271(5256):1734–1736. doi: 10.1126/science.271.5256.1734.
    1. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–3895. doi: 10.1002/j.1460-2075.1992.tb05481.x.
    1. Duffy MJ, Crown J. Precision treatment for cancer: Role of prognostic and predictive markers. Crit Rev Clin Lab Sci. 2014;51(1):30–45. doi: 10.3109/10408363.2013.865700.
    1. Smith IC, Heys SD, Hutcheon AW, Miller ID, Payne S, Gilbert FJ, et al. Neoadjuvant chemotherapy in breast cancer: significantly enhanced response with docetaxel. J Clin Oncol. 2002;20(6):1456–1466. doi: 10.1200/JCO.2002.20.6.1456.
    1. Von Minckwitz G, Blohmer JU, Costa SD, Denkert C, Eidtmann H, Eiermann W, et al. Response-guided neoadjuvant chemotherapy for breast cancer. J Clin Oncol. 2013;31(29):3623–3630. doi: 10.1200/JCO.2012.45.0940.
    1. Nisman B, Nechushtan H, Biran H, Gantz-Sorotsky H, Peled N, Gronowitz S, et al. Serum thymidine kinase 1 activity in the prognosis and monitoring of chemotherapy in lung cancer patients: A brief report. J Thorac Oncol. 2014;9:1568–1572. doi: 10.1097/JTO.0000000000000276.
    1. Topolcan O, Holubec L, Jr, Finek J, Stieber P, Holdenrieder S, Lamerz R, et al. Changes of thymidine kinase (TK) during adjuvant and palliative chemotherapy. Anticancer Res. 2005;25(3A):1831–1833.
    1. Tie J, Kinde I, Wang Y, Wong HL, Roebert J, Christie M, et al. Circulating tumour DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann Oncol. 2015;26(8):1715–1722. doi: 10.1093/annonc/mdv177.
    1. Marchetti A, Palma JF, Felicioni L, De Pas TM, Chiari R, Grammastro MD, et al. Early pediction of response to tyrosine kinase inhibitors by quantification of EGFR mutations in plasma of NSCLC patients. J Thorac Oncol. 2015;10:1437–1443. doi: 10.1097/JTO.0000000000000643.
    1. Holdenrieder S, Stieber P, von Pawel J, Raith H, Nagel D, Feldmann K, et al. Circulating nucleosomes predict the response of chemotherapy in patients with advanced non-small cell lung cancer. Clin Cancer Res. 2004;10:5981–5987. doi: 10.1158/1078-0432.CCR-04-0625.
    1. Stoetzeг OJ, Fersching DMI, Salat C, Steinkohl O, Gabka CJ, Hamann U, et al. Prediction of response to neoadjuvant chemotherapy in breast cancer patients by circulating apoptoic biomarkers nucleosomes, DNAse, cytokeratin-18 fragments and surviving. Cancer Letters. 2013;336:140–148. doi: 10.1016/j.canlet.2013.04.013.
    1. Hägg Olofsson M, Ueno T, Pan Y, Xu R, Cai F, Van Der Kuip H, et al. Cytokeratin-18 is a useful serum biomarker for early determination of response of breast carcinomas to chemotherapy. Clin Cancer Res. 2007;13(11):3198–3206. doi: 10.1158/1078-0432.CCR-07-0009.
    1. Riva T, Bidard F-C, Houy A, Saliou A, Madic J, Rampanou A, et al. Patient-specific circulating tumor DNA detection during neoadjuvant chemotherapy in triple-negative breast cancer. Clin.Chem. 2017;63:691–699. doi: 10.1373/clinchem.2016.262337.
    1. Smerage JB, Barlow WE, Hortobagyi GN, Winer EP, Leyland-Jones B, Srkalovic G, et al. Circulating tumour cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol. 2014;32(31):3483–3489. doi: 10.1200/JCO.2014.56.2561.
    1. Peintinger F, Kuerer HM, Anderson K, Boughey JC, Meric-Bernstam F, Singletary SE, et al. Accuracy of the combination of mammography and sonography in predicting tumour response in breast cancer patients after neoadjuvant chemotherapy. Ann Surg Oncol. 2006;13(11):1443–1449. doi: 10.1245/s10434-006-9086-9.

Source: PubMed

3
Sottoscrivi