TBCRC 002: a phase II, randomized, open-label trial of preoperative letrozole with or without bevacizumab in postmenopausal women with newly diagnosed stage 2/3 hormone receptor-positive and HER2-negative breast cancer

Christos Vaklavas, Brian S Roberts, Katherine E Varley, Nancy U Lin, Minetta C Liu, Hope S Rugo, Shannon Puhalla, Rita Nanda, Anna Maria Storniolo, Lisa A Carey, Mansoor N Saleh, Yufeng Li, Jennifer F Delossantos, William E Grizzle, Albert F LoBuglio, Richard M Myers, Andres Forero-Torres, Translational Breast Cancer Research Consortium (TBCRC), Christos Vaklavas, Brian S Roberts, Katherine E Varley, Nancy U Lin, Minetta C Liu, Hope S Rugo, Shannon Puhalla, Rita Nanda, Anna Maria Storniolo, Lisa A Carey, Mansoor N Saleh, Yufeng Li, Jennifer F Delossantos, William E Grizzle, Albert F LoBuglio, Richard M Myers, Andres Forero-Torres, Translational Breast Cancer Research Consortium (TBCRC)

Abstract

Background: In preclinical studies, the expression of vascular endothelial growth factor (VEGF) in hormone receptor-positive breast cancer is associated with estrogen-independent tumor growth and resistance to endocrine therapies. This study investigated whether the addition of bevacizumab, a monoclonal antibody against VEGF, to letrozole enhanced the antitumor activity of the letrozole in the preoperative setting.

Methods: Postmenopausal women with newly diagnosed stage 2 or 3 estrogen and/or progesterone receptor-positive, HER2-negative breast cancer were randomly assigned (2:1) between letrozole 2.5 mg PO daily plus bevacizumab 15 mg/kg IV every 3 weeks (Let/Bev) and letrozole 2.5 mg PO daily (Let) for 24 weeks prior to definitive surgery. Primary objective was within-arm pathologic complete remission (pCR) rate. Secondary objectives were safety, objective response, and downstaging rate.

Results: Seventy-five patients were randomized (Let/Bev n = 50, Let n = 25). Of the 45 patients evaluable for pathological response in the Let/Bev arm, 5 (11%; 95% CI, 3.7-24.1%) achieved pCR and 4 (9%; 95% CI, 2.5-21.2%) had microscopic residual disease; no pCRs or microscopic residual disease was seen in the Let arm (0%; 95% CI, 0-14.2%). The rates of downstaging were 44.4% (95% CI, 29.6-60.0%) and 37.5% (95% CI, 18.8-59.4%) in the Let/Bev and Let arms, respectively. Adverse events typically associated with letrozole (hot flashes, arthralgias, fatigue, myalgias) occurred in similar frequencies in the two arms. Hypertension, headache, and proteinuria were seen exclusively in the Let/Bev arm. The rates of grade 3 and 4 adverse events and discontinuation due to adverse events were 18% vs 8% and 16% vs none in the Let/Bev and Let arms, respectively. A small RNA-based classifier predictive of response to preoperative Let/Bev was developed and confirmed on an independent cohort.

Conclusion: In the preoperative setting, the addition of bevacizumab to letrozole was associated with a pCR rate of 11%; no pCR was seen with letrozole alone. There was additive toxicity with the incorporation of bevacizumab. Responses to Let/Bev can be predicted from the levels of 5 small RNAs in a pretreatment biopsy.

Trial registration: This trial is registered with ClinicalTrials.gov (Identifier: NCT00161291), first posted on September 12, 2005, and is completed.

Keywords: Bevacizumab; Breast cancer; Hormone receptor-positive breast cancer; Letrozole; Luminal breast cancer; Postmenopausal; Preoperative therapy.

Conflict of interest statement

C.V. consulting or advisory board: Daiichi-Sankyo, Genentech (unpaid); research funding (paid to the University of Alabama at Birmingham): Roche, Pfizer, Incyte, Novartis, Pharmacyclics, Tracon, Innocrin, H3 Biomedicine, Zymeworks. A.F.T received support from Genentech to conduct clinical and laboratory research (paid to the University of Alabama at Birmingham). A.F.T. is currently an employee of Seattle Genetics. N.U.L.—consulting or advisory board: Puma, Daiichi-Sankyo, Seattle Genetics, Genentech; research funding (paid to institution): Genentech, Pfizer, Novartis, Seattle Genetics, Array Biopharma. The remaining authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
CONSORT diagram
Fig. 2
Fig. 2
Waterfall plot of response by treatment arm. a Pathologic responses were assessed by comparing the maximum cumulative diameter of the target lesion(s) at the time of diagnosis as assessed by imaging studies with the size of the tumor in the final surgical pathology. b Radiological responses were assessed according to RECIST by breast ultrasound or breast MRI in selected cases (whichever modality was consistently obtained throughout the protocol therapy and most accurately assessed the status of the tumor)
Fig. 3
Fig. 3
a Quantile-quantile plot of small RNA p values for association with letrozole/bevacizumab response. The observed p values from the association of small RNA expression values with letrozole/bevacizumab response (see the “Methods” section) are plotted against the null (uniform) distribution. Small RNAs selected by LASSO regression for an optimal classifier model (see the “Methods” section) are marked by black dots and labeled. b Classifier model performance on three cohorts. Classifier values calculated from qPCR data for each patient tumor sample (x-axis) across three cohorts are plotted on the y-axis (see the “Methods” section). Bar color indicates responder status. The dotted line indicates the common threshold used to calculate model accuracy. The “discovery cohort” is the cohort of patients who received letrozole/bevacizumab in the present study. The “validation cohort” is the cohort of patients who received the same treatment in a separate independent clinical trial [15]. The “letrozole cohort” consists of patients who received letrozole in the present study. The model was trained on sequencing data from the discovery cohort

References

    1. Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 1996;86(3):353–364. doi: 10.1016/S0092-8674(00)80108-7.
    1. Zhang HT, Craft P, Scott PA, Ziche M, Weich HA, Harris AL, et al. Enhancement of tumor growth and vascular density by transfection of vascular endothelial cell growth factor into MCF-7 human breast carcinoma cells. J Natl Cancer Inst. 1995;87(3):213–219. doi: 10.1093/jnci/87.3.213.
    1. Guo P, Fang Q, Tao HQ, Schafer CA, Fenton BM, Ding I, et al. Overexpression of vascular endothelial growth factor by MCF-7 breast cancer cells promotes estrogen-independent tumor growth in vivo. Cancer Res. 2003;63(15):4684–4691.
    1. Qu Z, Van Ginkel S, Roy AM, Westbrook L, Nasrin M, Maxuitenko Y, et al. Vascular endothelial growth factor reduces tamoxifen efficacy and promotes metastatic colonization and desmoplasia in breast tumors. Cancer Res. 2008;68(15):6232–6240. doi: 10.1158/0008-5472.CAN-07-5654.
    1. Pinto MP, Badtke MM, Dudevoir ML, Harrell JC, Jacobsen BM, Horwitz KB. Vascular endothelial growth factor secreted by activated stroma enhances angiogenesis and hormone-independent growth of estrogen receptor-positive breast cancer. Cancer Res. 2010;70(7):2655–2664. doi: 10.1158/0008-5472.CAN-09-4373.
    1. Losordo DW, Isner JM. Estrogen and angiogenesis: a review. Arterioscler Thromb Vasc Biol. 2001;21(1):6–12. doi: 10.1161/01.ATV.21.1.6.
    1. Hyder SM, Stancel GM. Regulation of angiogenic growth factors in the female reproductive tract by estrogens and progestins. Mol Endocrinol (Baltimore) 1999;13(6):806–811. doi: 10.1210/mend.13.6.0308.
    1. Hyder SM, Nawaz Z, Chiappetta C, Stancel GM. Identification of functional estrogen response elements in the gene coding for the potent angiogenic factor vascular endothelial growth factor. Cancer Res. 2000;60(12):3183–3190.
    1. Buteau-Lozano H, Ancelin M, Lardeux B, Milanini J, Perrot-Applanat M. Transcriptional regulation of vascular endothelial growth factor by estradiol and tamoxifen in breast cancer cells: a complex interplay between estrogen receptors alpha and beta. Cancer Res. 2002;62(17):4977–4984.
    1. Mueller MD, Vigne JL, Minchenko A, Lebovic DI, Leitman DC, Taylor RN. Regulation of vascular endothelial growth factor (VEGF) gene transcription by estrogen receptors alpha and beta. Proc Natl Acad Sci U S A. 2000;97(20):10972–10977. doi: 10.1073/pnas.200377097.
    1. Applanat MP, Buteau-Lozano H, Herve MA, Corpet A. Vascular endothelial growth factor is a target gene for estrogen receptor and contributes to breast cancer progression. Adv Exp Med Biol. 2008;617:437–444. doi: 10.1007/978-0-387-69080-3_42.
    1. Foekens JA, Peters HA, Grebenchtchikov N, Look MP, Meijer-van Gelder ME, Geurts-Moespot A, et al. High tumor levels of vascular endothelial growth factor predict poor response to systemic therapy in advanced breast cancer. Cancer Res. 2001;61(14):5407–14.
    1. Linderholm B, Grankvist K, Wilking N, Johansson M, Tavelin B, Henriksson R. Correlation of vascular endothelial growth factor content with recurrences, survival, and first relapse site in primary node-positive breast carcinoma after adjuvant treatment. J Clin Oncol. 2000;18(7):1423–1431. doi: 10.1200/JCO.2000.18.7.1423.
    1. Linderholm B, Tavelin B, Grankvist K, Henriksson R. Vascular endothelial growth factor is of high prognostic value in node-negative breast carcinoma. J Clin Oncol. 1998;16(9):3121–3128. doi: 10.1200/JCO.1998.16.9.3121.
    1. Forero-Torres A, Saleh MN, Galleshaw JA, Jones CF, Shah JJ, Percent IJ, et al. Pilot trial of preoperative (neoadjuvant) letrozole in combination with bevacizumab in postmenopausal women with newly diagnosed estrogen receptor- or progesterone receptor-positive breast cancer. Clin Breast Cancer. 2010;10(4):275–280. doi: 10.3816/CBC.2010.n.035.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92(3):205–216. doi: 10.1093/jnci/92.3.205.
    1. FDA. Draft guidance for industry: pathologic complete response in neoadjuvant treatment of high-risk early-stage breast cancer: use as an endpoint to support accelerated approval 2012. Available from: . Accessed 23 July 2012.
    1. Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351(8):781–791. doi: 10.1056/NEJMoa040766.
    1. Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10(20):6897–6904. doi: 10.1158/1078-0432.CCR-04-0378.
    1. Garcia JA, Rosenberg JE, Weinberg V, Scott J, Frohlich M, Park JW, et al. Evaluation and significance of circulating epithelial cells in patients with hormone-refractory prostate cancer. BJU Int. 2007;99(3):519–524. doi: 10.1111/j.1464-410X.2007.06659.x.
    1. Rugo HS, Chien AJ, Franco SX, Stopeck AT, Glencer A, Lahiri S, et al. A phase II study of lapatinib and bevacizumab as treatment for HER2-overexpressing metastatic breast cancer. Breast Cancer Res Treat. 2011;134(1):13–20. doi: 10.1007/s10549-011-1918-z.
    1. Roberts BS, Hardigan AA, Kirby MK, Fitz-Gerald MB, Wilcox CM, Kimberly RP, et al. Blocking of targeted microRNAs from next-generation sequencing libraries. Nucleic Acids Res. 2015;43(21):e145.
    1. Baselga J, Semiglazov V, van Dam P, Manikhas A, Bellet M, Mayordomo J, et al. Phase II randomized study of neoadjuvant everolimus plus letrozole compared with placebo plus letrozole in patients with estrogen receptor-positive breast cancer. J Clin Oncol. 2009;27(16):2630–2637. doi: 10.1200/JCO.2008.18.8391.
    1. Ellis MJ, Tao Y, Luo J, A'Hern R, Evans DB, Bhatnagar AS, et al. Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst. 2008;100(19):1380–1388. doi: 10.1093/jnci/djn309.
    1. Saura C, Hlauschek D, Oliveira M, Zardavas D, Jallitsch-Halper A, de la Pena L, et al. Neoadjuvant letrozole plus taselisib versus letrozole plus placebo in postmenopausal women with oestrogen receptor-positive, HER2-negative, early-stage breast cancer (LORELEI): a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2019.
    1. Roberts BS, Hardigan AA, Moore DE, Ramaker RC, Jones AL, Fitz-Gerald MB, et al. Discovery and validation of circulating biomarkers of colorectal adenoma by high-depth small RNA sequencing. Clin Cancer Res. 2018;24(9):2092–2099. doi: 10.1158/1078-0432.CCR-17-1960.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. doi: 10.1186/s13059-014-0550-8.
    1. Friedman J, Hastie T, Tibshirani R. Regularization paths for generalized linear models via coordinate descent. J Stat Softw. 2010;33(1):1–22. doi: 10.18637/jss.v033.i01.
    1. Johnson K, Sarma D, Hwang ES. Lobular breast cancer series: imaging. Breast Cancer Res. 2015;17:94. doi: 10.1186/s13058-015-0605-0.
    1. Mulrane L, Madden SF, Brennan DJ, Gremel G, McGee SF, McNally S, et al. miR-187 is an independent prognostic factor in breast cancer and confers increased invasive potential in vitro. Clin Cancer Res. 2012;18(24):6702–6713. doi: 10.1158/1078-0432.CCR-12-1420.
    1. von Minckwitz G, Eidtmann H, Rezai M, Fasching PA, Tesch H, Eggemann H, et al. Neoadjuvant chemotherapy and bevacizumab for HER2-negative breast cancer. N Engl J Med. 2012;366(4):299–309. doi: 10.1056/NEJMoa1111065.
    1. Spring LM, Gupta A, Reynolds KL, Gadd MA, Ellisen LW, Isakoff SJ, et al. Neoadjuvant endocrine therapy for estrogen receptor-positive breast cancer: a systematic review and meta-analysis. JAMA Oncol. 2016;2(11):1477–1486. doi: 10.1001/jamaoncol.2016.1897.
    1. Ma CX, Gao F, Luo J, Northfelt DW, Goetz M, Forero A, et al. NeoPalAna: neoadjuvant palbociclib, a cyclin-dependent kinase 4/6 inhibitor, and anastrozole for clinical stage 2 or 3 estrogen receptor-positive breast cancer. Clin Cancer Res. 2017;23(15):4055–4065. doi: 10.1158/1078-0432.CCR-16-3206.
    1. Cottu P, D'Hondt V, Dureau S, Lerebours F, Desmoulins I, Heudel PE, et al. Letrozole and palbociclib versus chemotherapy as neoadjuvant therapy of high-risk luminal breast cancer. Ann Oncol. 2018;29(12):2334–2340. doi: 10.1093/annonc/mdy448.
    1. Davis DW, Inoue K, Dinney CP, Hicklin DJ, Abbruzzese JL, McConkey DJ. Regional effects of an antivascular endothelial growth factor receptor monoclonal antibody on receptor phosphorylation and apoptosis in human 253J B-V bladder cancer xenografts. Cancer Res. 2004;64(13):4601–4610. doi: 10.1158/0008-5472.CAN-2879-2.
    1. Dickler MN, Barry WT, Cirrincione CT, Ellis MJ, Moynahan ME, Innocenti F, et al. Phase III trial evaluating letrozole as first-line endocrine therapy with or without bevacizumab for the treatment of postmenopausal women with hormone receptor-positive advanced-stage breast cancer: CALGB 40503 (Alliance) J Clin Oncol. 2016;34(22):2602–2609. doi: 10.1200/JCO.2015.66.1595.
    1. Martin M, Loibl S, Hyslop T, De la Haba-Rodriguez J, Aktas B, Cirrincione CT, et al. Evaluating the addition of bevacizumab to endocrine therapy as first-line treatment for hormone receptor-positive metastatic breast cancer: a pooled analysis from the LEA (GEICAM/2006-11_GBG51) and CALGB 40503 (Alliance) trials. Eur J Cancer. 2019;117:91–98. doi: 10.1016/j.ejca.2019.06.002.
    1. Martin M, Loibl S, von Minckwitz G, Morales S, Martinez N, Guerrero A, et al. Phase III trial evaluating the addition of bevacizumab to endocrine therapy as first-line treatment for advanced breast cancer: the letrozole/fulvestrant and avastin (LEA) study. J Clin Oncol. 2015;33(9):1045–1052. doi: 10.1200/JCO.2014.57.2388.
    1. Earl HM, Hiller L, Dunn JA, Blenkinsop C, Grybowicz L, Vallier AL, et al. Efficacy of neoadjuvant bevacizumab added to docetaxel followed by fluorouracil, epirubicin, and cyclophosphamide, for women with HER2-negative early breast cancer (ARTemis): an open-label, randomised, phase 3 trial. Lancet Oncol. 2015;16(6):656–666. doi: 10.1016/S1470-2045(15)70137-3.
    1. Bear HD, Tang G, Rastogi P, Geyer CE, Jr, Robidoux A, Atkins JN, et al. Bevacizumab added to neoadjuvant chemotherapy for breast cancer. N Engl J Med. 2012;366(4):310–320. doi: 10.1056/NEJMoa1111097.
    1. Nahleh ZA, Barlow WE, Hayes DF, Schott AF, Gralow JR, Sikov WM, et al. SWOG S0800 (NCI CDR0000636131): addition of bevacizumab to neoadjuvant nab-paclitaxel with dose-dense doxorubicin and cyclophosphamide improves pathologic complete response (pCR) rates in inflammatory or locally advanced breast cancer. Breast Cancer Res Treat. 2016;158(3):485–495. doi: 10.1007/s10549-016-3889-6.
    1. Nahleh Z, Botrus G, Dwivedi A, Jennings M, Nagy S, Tfayli A. Bevacizumab in the neoadjuvant treatment of human epidermal growth factor receptor 2-negative breast cancer: a meta-analysis of randomized controlled trials. Mol Clin Oncol. 2019;10(3):357–365.
    1. Earl HM, Hiller L, Dunn JA, Blenkinsop C, Grybowicz L, Vallier AL, et al. Disease-free and overall survival at 3.5 years for neoadjuvant bevacizumab added to docetaxel followed by fluorouracil, epirubicin and cyclophosphamide, for women with HER2 negative early breast cancer: ARTemis trial. Ann Oncol. 2017;28(8):1817–1824. doi: 10.1093/annonc/mdx173.
    1. Bear HD, Tang G, Rastogi P, Geyer CE, Jr, Liu Q, Robidoux A, et al. Neoadjuvant plus adjuvant bevacizumab in early breast cancer (NSABP B-40 [NRG oncology]): secondary outcomes of a phase 3, randomised controlled trial. Lancet Oncol. 2015;16(9):1037–1048. doi: 10.1016/S1470-2045(15)00041-8.
    1. von Minckwitz G, Loibl S, Untch M, Eidtmann H, Rezai M, Fasching PA, et al. Survival after neoadjuvant chemotherapy with or without bevacizumab or everolimus for HER2-negative primary breast cancer (GBG 44-GeparQuinto)dagger. Ann Oncol. 2014;25(12):2363–2372. doi: 10.1093/annonc/mdu455.
    1. Miles D, Cameron D, Bondarenko I, Manzyuk L, Alcedo JC, Lopez RI, et al. Bevacizumab plus paclitaxel versus placebo plus paclitaxel as first-line therapy for HER2-negative metastatic breast cancer (MERiDiAN): a double-blind placebo-controlled randomised phase III trial with prospective biomarker evaluation. Eur J Cancer. 2017;70:146–155. doi: 10.1016/j.ejca.2016.09.024.
    1. Schneider BP, Gray RJ, Radovich M, Shen F, Vance G, Li L, et al. Prognostic and predictive value of tumor vascular endothelial growth factor gene amplification in metastatic breast cancer treated with paclitaxel with and without bevacizumab; results from ECOG 2100 trial. Clin Cancer Res. 2013;19(5):1281–1289. doi: 10.1158/1078-0432.CCR-12-3029.
    1. Jain RK, Duda DG, Willett CG, Sahani DV, Zhu AX, Loeffler JS, et al. Biomarkers of response and resistance to antiangiogenic therapy. Nat Rev Clin Oncol. 2009;6(6):327–338. doi: 10.1038/nrclinonc.2009.63.
    1. Lucci A, Hall CS, Lodhi AK, Bhattacharyya A, Anderson AE, Xiao L, et al. Circulating tumour cells in non-metastatic breast cancer: a prospective study. Lancet Oncol. 2012;13(7):688–695. doi: 10.1016/S1470-2045(12)70209-7.
    1. Choi SK, Kim HS, Jin T, Hwang EH, Jung M, Moon WK. Overexpression of the miR-141/200c cluster promotes the migratory and invasive ability of triple-negative breast cancer cells through the activation of the FAK and PI3K/AKT signaling pathways by secreting VEGF-A. BMC Cancer. 2016;16:570. doi: 10.1186/s12885-016-2620-7.
    1. Cheung AK, Ko JM, Lung HL, Chan KW, Stanbridge EJ, Zabarovsky E, et al. Cysteine-rich intestinal protein 2 (CRIP2) acts as a repressor of NF-kappaB-mediated proangiogenic cytokine transcription to suppress tumorigenesis and angiogenesis. Proc Natl Acad Sci U S A. 2011;108(20):8390–8395. doi: 10.1073/pnas.1101747108.
    1. Shi W, Bruce J, Lee M, Yue S, Rowe M, Pintilie M, et al. MiR-449a promotes breast cancer progression by targeting CRIP2. Oncotarget. 2016;7(14):18906–18918. doi: 10.18632/oncotarget.7753.
    1. Williams GT, Farzaneh F. Are snoRNAs and snoRNA host genes new players in cancer? Nat Rev Cancer. 2012;12(2):84–88. doi: 10.1038/nrc3195.
    1. Bastide A, Karaa Z, Bornes S, Hieblot C, Lacazette E, Prats H, et al. An upstream open reading frame within an IRES controls expression of a specific VEGF-A isoform. Nucleic Acids Res. 2008;36(7):2434–2445. doi: 10.1093/nar/gkn093.
    1. Marcel V, Ghayad SE, Belin S, Therizols G, Morel AP, Solano-Gonzalez E, et al. p53 acts as a safeguard of translational control by regulating fibrillarin and rRNA methylation in cancer. Cancer Cell. 2013;24(3):318–330. doi: 10.1016/j.ccr.2013.08.013.
    1. Ji X, Lu H, Zhou Q, Luo K. LARP7 suppresses P-TEFb activity to inhibit breast cancer progression and metastasis. Elife. 2014;3:e02907. doi: 10.7554/eLife.02907.

Source: PubMed

Подписаться