Inhibition of iNOS as a novel effective targeted therapy against triple-negative breast cancer

Sergio Granados-Principal, Yi Liu, Maria L Guevara, Elvin Blanco, Dong Soon Choi, Wei Qian, Tejal Patel, Angel A Rodriguez, Joseph Cusimano, Heidi L Weiss, Hong Zhao, Melissa D Landis, Bhuvanesh Dave, Steven S Gross, Jenny C Chang, Sergio Granados-Principal, Yi Liu, Maria L Guevara, Elvin Blanco, Dong Soon Choi, Wei Qian, Tejal Patel, Angel A Rodriguez, Joseph Cusimano, Heidi L Weiss, Hong Zhao, Melissa D Landis, Bhuvanesh Dave, Steven S Gross, Jenny C Chang

Abstract

Introduction: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer with no effective targeted therapy. Inducible nitric oxide synthase (iNOS) is associated with poor survival in patients with breast cancer by increasing tumor aggressiveness. This work aimed to investigate the potential of iNOS inhibitors as a targeted therapy for TNBC. We hypothesized that inhibition of endogenous iNOS would decrease TNBC aggressiveness by reducing tumor initiation and metastasis through modulation of epithelial-mesenchymal transition (EMT)-inducing factors.

Methods: iNOS protein levels were determined in 83 human TNBC tissues and correlated with clinical outcome. Proliferation, mammosphere-forming efficiency, migration, and EMT transcription factors were assessed in vitro after iNOS inhibition. Endogenous iNOS targeting was evaluated as a potential therapy in TNBC mouse models.

Results: High endogenous iNOS expression was associated with worse prognosis in patients with TNBC by gene expression as well as immunohistochemical analysis. Selective iNOS (1400 W) and pan-NOS (L-NMMA and L-NAME) inhibitors diminished cell proliferation, cancer stem cell self-renewal, and cell migration in vitro, together with inhibition of EMT transcription factors (Snail, Slug, Twist1, and Zeb1). Impairment of hypoxia-inducible factor 1α, endoplasmic reticulum stress (IRE1α/XBP1), and the crosstalk between activating transcription factor 3/activating transcription factor 4 and transforming growth factor β was observed. iNOS inhibition significantly reduced tumor growth, the number of lung metastases, tumor initiation, and self-renewal.

Conclusions: Considering the effectiveness of L-NMMA in decreasing tumor growth and enhancing survival rate in TNBC, we propose a targeted therapeutic clinical trial by re-purposing the pan-NOS inhibitor L-NMMA, which has been extensively investigated for cardiogenic shock as an anti-cancer therapeutic.

Figures

Figure 1
Figure 1
Enhanced nitric oxide synthase 2 (NOS2) expression correlates with poor patient survival in invasive triple-negative breast cancer (TNBC). (A) Higher NOS2 mRNA expression in invasive TNBC versus non-TNBC (P = 3.85 × 10−5). (B) High NOS2 expression correlates with death at 5 years in invasive breast carcinoma (P = 0.037). Kaplan-Meier survival analyses in (C) Van de Vijver (n = 69; P = 0.04) and (D) Curtis (n = 260; P = 0.01) breast databases show that high NOS2 expression correlates with worse overall survival of patients with TNBC. (E) Immunohistochemical analysis of TNBC human samples for inducible nitric oxide synthase (iNOS) protein expression. Weak-to-moderate (3 or 4), moderate-to-strong (5 or 6), and strong (7) were the cutoffs established for further analysis of survival. Several samples showed expression in both tumor (T) and stromal (S) cells (original optical objective: 20×). MDA-MB-231 cells transfected with either NOS2-directed shRNA (shRNA1) or empty vector (EV) were used as negative and positive control of iNOS staining, respectively (original optical objective: 10×). Counterstain: hematoxylin. (F) Increased iNOS expression is associated with less patient survival when compared with low iNOS expression. Kaplan-Meier survival analysis of TNBC human patient samples (n = 83) (P = 0.05). shRNA1, small hairpin RNA 1; TCGA, The Cancer Genome Atlas.
Figure 2
Figure 2
Effects of inducible nitric oxide synthase (iNOS) inhibitors on tumor cell proliferation, migration, and mammosphere formation in triple-negative breast cancer (TNBC) cell lines. Proliferation (A, B) and primary (C) and (D) secondary mammosphere, and migration index of MDA-MB-231 and SUM159 cell lines treated with 1400 W (E) and L-NMMA (F) for 96 hours. Results were normalized to vehicle. Data are presented as mean ± standard error of the mean. *P <0.05, **P <0.01, ***P <0.001, ****P <0.0001. 1400 W, N-[[3-(aminomethyl)phenyl]methyl]-ethanimidamide; L-NMMA, NG-monomethyl-L-arginine; MSFE, mammosphere-forming efficiency.
Figure 3
Figure 3
iNOS knockdown impairs tumorigenicity and EMT by a dual impact on HIF1α and ER stress/TGFβ/AFT4/ATF3 crosstalk. Proliferation (A), migration (B), and mammosphere-forming efficiency (MSFE) (C) in MDA-MB-231 cells transiently transfected with two different NOS2-directed siRNAs (siRNA1 and siRNA2) compared with scrambled control. Western blot analysis of NOS isoforms (iNOS, eNOS, and nNOS) and EMT transcription factors in MDA-MB-231 and SUM159 cell lines treated with (D) 1400 W and (E) siRNA-mediated NOS2 knockdown. (F) Selective iNOS inhibition reduced hypoxia (HIF1α) and ER stress markers (IRE1α and ATF4). (G) Phospho-Smad2/3, Smad2/3, and mature TGFβ protein levels in MDA-MB-231 and SUM159 cells. (H) Recombinant TGFβ1 (10 ng/mL for 7 days) activates the PERK/eIF2α/ATF4/ATF3 axis in MCF10A. (I) Effects on the PERK/eIF2α/ATF4/ATF3 axis by co-treatment of recombinant TGFβ1 (10 ng/mL, 7 days) and 1400 W (4 mM) for 24 hours in MCF10A cells. iNOS, ATF4, ATF3, and mature TGFβ protein levels in siRNA-mediated NOS2 knockdown (siRNA2) MCF10A cells for 96 hours. (J) Selective iNOS inhibition is postulated to impair EMT and tumor cell migration by an impact on HIF1α, ER stress (IRE1α/XBP1), and the crosstalk between ATF4, ATF3, and TGFβ. Results were normalized to scrambled. Data are presented as mean ± standard error of the mean. ****P <0.0001. 1400 W, N-[[3-(aminomethyl)phenyl]methyl]-ethanimidamide; ATF3, activating transcription factor 3; ATF4, activating transcription factor 4; EMT, epithelial-mesenchymal transition; ER, endoplasmic reticulum; HIF1α, hypoxia-inducible factor 1α; iNOS, inducible nitric oxide synthase; siRNA, small interfering RNA; TGFβ, transforming growth factor β.
Figure 4
Figure 4
Decrease in tumor initiation and lung metastases in MDA-MB-231 xenografts. (A) Tumor volume of MDA-MB-231 breast xenografts (n = 5 per group) after daily injection of L-NAME (80 mg/kg, intraperitoneal). (B) Primary and secondary mammosphere of cancer cells isolated from tumor tissue. (C) Luminescence of MDA-MB-231 L/G tumor cells in lungs of vehicle- and L-NAME-treated mice. (D) Tumor-initiating capacity of tumor cells (limiting dilution assay). Results were normalized to vehicle. Data are presented as mean ± standard error of the mean. *P <0.05, **P <0.01, ***P <0.001. L-NAME, N5-[imino(nitroamino)methyl]-L-ornithine methyl ester; MSFE, mammosphere-forming efficiency.
Figure 5
Figure 5
In vivoeffects of L-NMMA in MDA-MB-231 xenografts. (A) Tumor volume of MDA-MB-231 breast xenografts (n = 10 per group) treated with vehicle, L-NMMA, docetaxel, and combination. (B) Illustrative images of Ki67 staining in the vehicle, L-NMMA, docetaxel, and combination groups. Original optical objective: 10×. Counterstain: hematoxylin. (C) Cell proliferation of tumor xenografts with Ki67 immunostaining. (D) Nuclear cleaved caspase-3 staining in the chemo and combo groups. (E) Primary and secondary mammosphere of breast cancer cells isolated from tumor tissue. (F) Tumor-initiating capacity of tumor cells. Results were normalized to vehicle. For proliferation and apoptosis, 1,000 cells were counted from 10 different fields, and the percentage was determined. Data are presented as mean ± standard error of the mean. *P <0.05, **P <0.01, ***P <0.001, ****P <0.0001. L-NMMA, NG-monomethyl-L-arginine; MSFE, mammosphere-forming efficiency.
Figure 6
Figure 6
Clinically relevant dose regimen of L-NMMA in orthotopic mouse models of triple-negative breast cancer. (A) Mean systolic pressure of mice (n = 5) giving one cycle of the dose rate proposed in this study. (B) Mean systolic pressure of mice 30 minutes and 24 hours after the last injection of one cycle treatment (n = 5). (C) Tumor volume of MDA-MB-231 xenografts (n = 10 per group) treated with vehicle, docetaxel, and combination (docetaxel + amlodipine + L-NMMA). (D) Kaplan-Meier survival curve of vehicle-, chemotherapy-, and combo-treated MDA-MB-231 xenograft-bearing mice. (E) Tumor volume of SUM159 xenografts (n = 10 per group) treated with vehicle, docetaxel, and combination. (F) Effects of amlodipine on tumor volume in MDA-MB-231 xenografts (n = 5 per group). Data are presented as mean ± standard error of the mean. *P <0.05, **P <0.01, ***P <0.001, ****P <0.0001. L-NMMA, NG-monomethyl-L-arginine; ns, not significant.

References

    1. Chang JC, Wooten EC, Tsimelzon A, Hilsenbeck SG, Gutierrez MC, Tham YL, et al. Patterns of resistance and incomplete response to docetaxel by gene expression profiling in breast cancer patients. J Clin Oncol. 2005;23:1169–77. doi: 10.1200/JCO.2005.03.156.
    1. Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci U S A. 2009;106:13820–5. doi: 10.1073/pnas.0905718106.
    1. Dave B, Mittal V, Tan NM, Chang JC. Epithelial-mesenchymal transition, cancer stem cells and treatment resistance. Breast Cancer Res. 2012;14:202. doi: 10.1186/bcr2938.
    1. Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, Krop I, et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19:1512–24. doi: 10.1158/1078-0432.CCR-11-3326.
    1. Burke AJ, Sullivan FJ, Giles FJ, Glynn SA. The yin and yang of nitric oxide in cancer progression. Carcinogenesis. 2013;34:503–12. doi: 10.1093/carcin/bgt034.
    1. Bulut AS, Erden E, Sak SD, Doruk H, Kursun N, Dincol D. Significance of inducible nitric oxide synthase expression in benign and malignant breast epithelium: an immunohistochemical study of 151 cases. Virchows Arch. 2005;447:24–30. doi: 10.1007/s00428-005-1250-2.
    1. Glynn SA, Boersma BJ, Dorsey TH, Yi M, Yfantis HG, Ridnour LA, et al. Increased NOS2 predicts poor survival in estrogen receptor-negative breast cancer patients. J Clin Invest. 2010;120:3843–54. doi: 10.1172/JCI42059.
    1. Loibl S, Buck A, Strank C, von Minckwitz G, Roller M, Sinn HP, et al. The role of early expression of inducible nitric oxide synthase in human breast cancer. Eur J Cancer. 2005;41:265–71. doi: 10.1016/j.ejca.2004.07.010.
    1. Thomsen LL, Miles DW, Happerfield L, Bobrow LG, Knowles RG, Moncada S. Nitric oxide synthase activity in human breast cancer. Br J Cancer. 1995;72:41–4. doi: 10.1038/bjc.1995.274.
    1. Okayama H, Saito M, Oue N, Weiss JM, Stauffer J, Takenoshita S, et al. NOS2 enhances KRAS-induced lung carcinogenesis, inflammation and microRNA-21 expression. Int J Cancer. 2013;132:9–18. doi: 10.1002/ijc.27644.
    1. Ambs S, Merriam WG, Bennett WP, Felley-Bosco E, Ogunfusika MO, Oser SM, et al. Frequent nitric oxide synthase-2 expression in human colon adenomas: implication for tumor angiogenesis and colon cancer progression. Cancer Res. 1998;58:334–41.
    1. Massi D, Franchi A, Sardi I, Magnelli L, Paglierani M, Borgognoni L, et al. Inducible nitric oxide synthase expression in benign and malignant cutaneous melanocytic lesions. J Pathol. 2001;194:194–200. doi: 10.1002/1096-9896(200106)194:2<194::AID-PATH851>;2-S.
    1. Eyler CE, Wu Q, Yan K, MacSwords JM, Chandler-Militello D, Misuraca KL, et al. Glioma stem cell proliferation and tumor growth are promoted by nitric oxide synthase-2. Cell. 2011;146:53–66. doi: 10.1016/j.cell.2011.06.006.
    1. Switzer CH, Cheng RY, Ridnour LA, Glynn SA, Ambs S, Wink DA. Ets-1 is a transcriptional mediator of oncogenic nitric oxide signaling in estrogen receptor-negative breast cancer. Breast Cancer Res. 2012;14:R125. doi: 10.1186/bcr3319.
    1. Alexander JH, Reynolds HR, Stebbins AL, Dzavik V, Harrington RA, Van de Werf F, et al. Effect of tilarginine acetate in patients with acute myocardial infarction and cardiogenic shock: the TRIUMPH randomized controlled trial. JAMA. 2007;297:1657–66. doi: 10.1001/jama.297.15.joc70035.
    1. Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, et al. Oncomine 3.0: genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 2007;9:166–80. doi: 10.1593/neo.07112.
    1. van de Vijver MJ, He YD, van’t Veer LJ, Dai H, Hart AA, Voskuil DW, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999–2009. doi: 10.1056/NEJMoa021967.
    1. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–52.
    1. Chen Q, Park HC, Goligorsky MS, Chander P, Fischer SM, Gross SS. Untargeted plasma metabolite profiling reveals the broad systemic consequences of xanthine oxidoreductase inactivation in mice. PLoS One. 2012;7:e37149. doi: 10.1371/journal.pone.0037149.
    1. Babykutty S, Suboj P, Srinivas P, Nair AS, Chandramohan K, Gopala S. Insidious role of nitric oxide in migration/invasion of colon cancer cells by upregulating MMP-2/9 via activation of cGMP-PKG-ERK signaling pathways. Clin Exp Metastasis. 2012;29:471–92. doi: 10.1007/s10585-012-9464-6.
    1. Pang Y, Gara SK, Achyut BR, Li Z, Yan HH, Day CP, et al. TGF-β signaling in myeloid cells is required for tumor metastasis. Cancer Discov. 2013;3:936–51. doi: 10.1158/-12-0527.
    1. Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488:522–6. doi: 10.1038/nature11287.
    1. Driessens G, Beck B, Caauwe A, Simons BD, Blanpain C. Defining the mode of tumour growth by clonal analysis. Nature. 2012;488:527–30. doi: 10.1038/nature11344.
    1. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90. doi: 10.1016/j.cell.2009.11.007.
    1. Matrone C, Pignataro G, Molinaro P, Irace C, Scorziello A, Di Renzo GF, et al. HIF-1alpha reveals a binding activity to the promoter of iNOS gene after permanent middle cerebral artery occlusion. J Neurochem. 2004;90:368–78. doi: 10.1111/j.1471-4159.2004.02483.x.
    1. Yang MH, Wu KJ. TWIST activation by hypoxia inducible factor-1 (HIF-1): implications in metastasis and development. Cell Cycle. 2008;7:2090–6. doi: 10.4161/cc.7.14.6324.
    1. Zhong Q, Zhou B, Ann DK, Minoo P, Liu Y, Banfalvi A, et al. Role of endoplasmic reticulum stress in epithelial-mesenchymal transition of alveolar epithelial cells: effects of misfolded surfactant protein. Am J Respir Cell Mol Biol. 2011;45:498–509. doi: 10.1165/rcmb.2010-0347OC.
    1. Yin X, Wolford CC, Chang YS, McConoughey SJ, Ramsey SA, Aderem A, et al. ATF3, an adaptive-response gene, enhances TGF{beta} signaling and cancer-initiating cell features in breast cancer cells. J Cell Sci. 2010;123:3558–65. doi: 10.1242/jcs.064915.
    1. Lian N, Lin T, Liu W, Wang W, Li L, Sun S, et al. Transforming growth factor β suppresses osteoblast differentiation via the vimentin activating transcription factor 4 (ATF4) axis. J Biol Chem. 2012;287:35975–84. doi: 10.1074/jbc.M112.372458.
    1. Kilbourn RG, Szabó C, Traber DL. Beneficial versus detrimental effects of nitric oxide synthase inhibitors in circulatory shock: lessons learned from experimental and clinical studies. Shock. 1997;7:235–46. doi: 10.1097/00024382-199704000-00001.
    1. Kilbourn RG, Fonseca GA, Trissel LA, Griffith OW. Strategies to reduce side effects of interleukin-2: evaluation of the antihypotensive agent NG-monomethyl-L-arginine. Cancer J Sci Am. 2000;6:21–30.
    1. Dave B, Granados-Principal S, Zhu R, Benz S, Rabizadeh S, Soon-Shiong P, et al. Targeting RPL39 and MLF2 reduces tumor initiation and metastasis in breast cancer by inhibiting nitric oxide synthase signaling. Proc Natl Acad Sci U S A. 2014;111:8838–43. doi: 10.1073/pnas.1320769111.
    1. Vakkala M, Kahlos K, Lakari E, Pääkkö P, Kinnula V, Soini Y. Inducible nitric oxide synthase expression, apoptosis, and angiogenesis in in situ and invasive breast carcinomas. Clin Cancer Res. 2000;6:2408–16.
    1. Takaoka K, Hidaka S, Hashitani S, Segawa E, Yamamura M, Tanaka N, et al. Effect of a nitric oxide synthase inhibitor and a CXC chemokine receptor-4 antagonist on tumor growth and metastasis in a xenotransplanted mouse model of adenoid cystic carcinoma of the oral floor. Int J Oncol. 2013;43:737–45.
    1. Chinje EC, Williams KJ, Telfer BA, Wood PJ, van der Kogel AJ, Stratford IJ. 17beta-Oestradiol treatment modulates nitric oxide synthase activity in MDA231 tumour with implications on growth and radiation response. Br J Cancer. 2002;86:136–42. doi: 10.1038/sj.bjc.6600032.
    1. Radisavljevic Z. Inactivated tumor suppressor Rb by nitric oxide promotes mitosis in human breast cancer cells. J Cell Biochem. 2004;92:1–5. doi: 10.1002/jcb.20063.
    1. Sen S, Kawahara B, Chaudhuri G. Mitochondrial-associated nitric oxide synthase activity inhibits cytochrome c oxidase: implications for breast cancer. Free Radic Biol Med. 2013;57:210–20. doi: 10.1016/j.freeradbiomed.2012.10.545.
    1. Kim RK, Suh Y, Cui YH, Hwang E, Lim EJ, Yoo KC, et al. Fractionated radiation-induced nitric oxide promotes expansion of glioma stem-like cells. Cancer Sci. 2013;104:1172–7. doi: 10.1111/cas.12207.
    1. Edwards P, Cendan JC, Topping DB, Moldawer LL, MacKay S, Copeland EMIII, et al. Tumor cell nitric oxide inhibits cell growth in vitro, but stimulates tumorigenesis and experimental lung metastasis in vivo. J Surg Res. 1996;63:49–52. doi: 10.1006/jsre.1996.0221.
    1. Jadeski LC, Hum KO, Chakraborty C, Lala PK. Nitric oxide promotes murine mammary tumour growth and metastasis by stimulating tumour cell migration, invasiveness and angiogenesis. Int J Cancer. 2000;86:30–9. doi: 10.1002/(SICI)1097-0215(20000401)86:1<30::AID-IJC5>;2-I.
    1. Siegert A, Rosenberg C, Schmitt WD, Denkert C, Hauptmann S. Nitric oxide of human colorectal adenocarcinoma cell lines promotes tumour cell invasion. Br J Cancer. 2002;86:1310–5. doi: 10.1038/sj.bjc.6600224.
    1. Yasuoka H, Tsujimoto M, Yoshidome K, Nakahara M, Kodama R, Sanke T, et al. Cytoplasmic CXCR4 expression in breast cancer: induction by nitric oxide and correlation with lymph node metastasis and poor prognosis. BMC Cancer. 2008;8:340. doi: 10.1186/1471-2407-8-340.
    1. Voutsadakis IA. The ubiquitin-proteasome system and signal transduction pathways regulating Epithelial Mesenchymal transition of cancer. J Biomed Sci. 2012;19:67. doi: 10.1186/1423-0127-19-67.
    1. Carlisle RE, Heffernan A, Brimble E, Liu L, Jerome D, Collins CA, et al. TDAG51 mediates epithelial-to-mesenchymal transition in human proximal tubular epithelium. Am J Physiol Renal Physiol. 2012;303:F467–81. doi: 10.1152/ajprenal.00481.2011.
    1. Tanjore H, Cheng DS, Degryse AL, Zoz DF, Abdolrasulnia R, Lawson WE, et al. Alveolar epithelial cells undergo epithelial-to-mesenchymal transition in response to endoplasmic reticulum stress. J Biol Chem. 2011;286:30972–80. doi: 10.1074/jbc.M110.181164.
    1. Ulianich L, Garbi C, Treglia AS, Punzi D, Miele C, Raciti GA, et al. ER stress is associated with dedifferentiation and an epithelial-to-mesenchymal transition-like phenotype in PC Cl3 thyroid cells. J Cell Sci. 2008;121:477–86. doi: 10.1242/jcs.017202.
    1. Chowdhury R, Godoy LC, Thiantanawat A, Trudel LJ, Deen WM, Wogan GN. Nitric oxide produced endogenously is responsible for hypoxia-induced HIF-1α stabilization in colon carcinoma cells. Chem Res Toxicol. 2012;25:2194–202. doi: 10.1021/tx300274a.
    1. Nagelkerke A, Bussink J, Mujcic H, Wouters BG, Lehmann S, Sweep FC, et al. Hypoxia stimulates migration of breast cancer cells via the PERK/ATF4/LAMP3-arm of the unfolded protein response. Breast Cancer Res. 2013;15:R2. doi: 10.1186/bcr3373.
    1. Pan YX, Chen H, Thiaville MM, Kilberg MS. Activation of the ATF3 gene through a co-ordinated amino acid-sensing response programme that controls transcriptional regulation of responsive genes following amino acid limitation. Biochem J. 2007;401:299–307. doi: 10.1042/BJ20061261.
    1. López A, Lorente JA, Steingrub J, Bakker J, McLuckie A, Willatts S, et al. Multiple-center, randomized, placebo-controlled, double-blind study of the nitric oxide synthase inhibitor 546C88: effect on survival in patients with septic shock. Crit Care Med. 2004;32:21–30. doi: 10.1097/01.CCM.0000105581.01815.C6.

Source: PubMed

3
구독하다