Published in

American Association for Cancer Research, Cancer Research, 13_Supplement(78), p. 789-789, 2018

DOI: 10.1158/1538-7445.am2018-789

Links

Tools

Export citation

Search in Google Scholar

Abstract 789: Nuclear IGFBP-3 is a potential biomarker for response to EGFR-sphingosine kinase targeted therapy in basal-like triple-negative breast cancer (TNBC)

Journal article published in 2018 by Sohel M. Julovi ORCID, Janet L. Martin, Robert C. Baxter
This paper is made freely available by the publisher.
This paper is made freely available by the publisher.

Full text: Download

Green circle
Preprint: archiving allowed
Orange circle
Postprint: archiving restricted
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

Abstract Background: TNBC comprises about 15% of breast cancer cases and, with no approved targeted therapy, is commonly treated with adjuvant chemotherapy. We have described a novel targeted therapy for basal-like TNBC combining EGFR and sphingosine kinase (SphK) inhibitors, which induce a strongly synergistic cytostatic effect in six TNBC cell lines (Martin et al, Breast Cancer Res 19:90, 2017). IGFBP-3, which is highly expressed in basal-like TNBC and prognostic for poor recurrence-free survival, initiates an oncogenic pathway involving SphK1 and EGFR activation. Since IGFBP-3 downregulation in vitro prevents the synergistic cytostasis caused by EGFR and SphK inhibitors, we aimed in this study to evaluate nuclear IGFBP-3 as a response biomarker in TNBC tumors after treatment with the inhibitor combination, with or without doxorubicin (dox), a common component of TNBC chemotherapy. Methods: Human basal-like TNBC cell lines HCC1806 and MDA-MB-468 were grown as orthotopic xenograft tumors in female BALB/c nude mice, and treated when tumors reached 100 mm3 with the SphK inhibitor and S1P receptor modulator, fingolimod, plus the EGFR kinase inhibitor, gefitinib (F+G) ± dox at the maximum tolerated dose of 2 mg/Kg weekly. Tumors were analyzed by immunohistochemistry (IHC). Cell proliferation was studied in vitro by IncuCyte live-cell imaging. Results: In both cell lines in vitro, F+G at doses that caused minimal cytostasis, acted synergistically with a sub-cytostatic dox dose (10 nM), to cause almost complete inhibition of proliferation. In contrast, in xenograft mouse models, in which F+G administered 3x weekly significantly inhibited tumor growth and enhanced mouse survival, dox administration for 6 weeks showed minimal growth inhibition alone, and no significant incremental effect with F+G. Examined by IHC, TNBC tumor IGFBP-3 staining showed predominantly nuclear localization, was positively correlated with Ki67, and was significantly downregulated by F+G treatment, with no added dox effect. By Kaplan-Meier analysis, high (> median) nuclear IGFBP-3 IHC scores were strongly associated with worse mouse survival, similar to high Ki67 scores, while high apoptosis (cleaved caspase-3) scores were associated with better survival. Conclusion: The synergism between dox and combination F+G, observed in vitro, was not seen in vivo, possibly because of dose-limiting dox toxicity. This might be improved by alternative dox delivery methods. However, the F+G combination alone is highly inhibitory to basal-like TNBC tumor growth in both xenograft models. IGFBP-3 is known to interact with nuclear hormone receptors and DNA damage response intermediates in the nucleus. Nuclear IGFBP-3 staining may have utility as a biomarker of treatment response in TNBC, alone or together with Ki67 and cleaved caspase-3. Supported by Cancer Council NSW. Citation Format: Sohel M. Julovi, Janet L. Martin, Robert C. Baxter. Nuclear IGFBP-3 is a potential biomarker for response to EGFR-sphingosine kinase targeted therapy in basal-like triple-negative breast cancer (TNBC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 789.