Impact of inducible nitric oxide synthase (iNOS) expression on triple negative breast cancer outcome and activation of EGFR and ERK signaling pathways

Pablo Garrido, Aliaa Shalaby, Elaine M Walsh, Nessa Keane, Mark Webber, Maccon M Keane, Francis J Sullivan, Michael J Kerin, Grace Callagy, Aideen E Ryan, Sharon A Glynn, Pablo Garrido, Aliaa Shalaby, Elaine M Walsh, Nessa Keane, Mark Webber, Maccon M Keane, Francis J Sullivan, Michael J Kerin, Grace Callagy, Aideen E Ryan, Sharon A Glynn

Abstract

Inflammation is implicated in triple negative breast cancer (TNBC) progression. TNBC carries a worse prognosis than other breast cancer subtypes, and with the clinical and molecular heterogeneity of TNBC, there is a lack of effective therapeutic targets available. Identification of molecular targets for TNBC subtypes is crucial towards personalized patient stratification. Inducible nitric oxide synthase (iNOS) has been shown to induce p53 mutation accumulation, basal-like gene signature enrichment and transactivation of the epidermal growth factor receptor (EGFR) via s-nitrosylation. Herein we report that iNOS is associated with disease recurrence, distant metastasis and decreased breast cancer specific survival in 209 cases of TNBC. Employing TNBC cell lines representing normal basal breast, and basal-like 1 and basal-like 2 tumors, we demonstrate that nitric oxide (NO) induces EGFR-dependent ERK phosphorylation in basal-like TNBC cell lines. Moreover NO mediated cell migration and cell invasion was found to be dependent on EGFR and ERK activation particularly in basal-like 2 TBNC cells. This occurred in conjunction with NF-κB activation and increased secretion of pro-inflammatory cytokines IL-8, IL-1β and TNF-α. This provides substantial evidence for EGFR as a therapeutic target to be taken into consideration in the treatment of a specific subset of basal-like TNBC overexpressing iNOS.

Keywords: EGFR; inflammation; metastasis; nitric oxide; triple negative breast cancer.

Conflict of interest statement

CONFLICTS OF INTEREST Authors declare no conflicts of interest.

Figures

Figure 1. iNOS expression immunohistochemistry in TNBC
Figure 1. iNOS expression immunohistochemistry in TNBC
Representative images of TNBC tumor with (A) negative, (B) low, (C) moderate, and (D) strong iNOS tumor epithelial staining.
Figure 2. iNOS expression is associated with…
Figure 2. iNOS expression is associated with decreased recurrence free survival, distant metastasis free survival and breast cancer survival in TNBC
(A) Kaplan-Meier cumulative recurrence free survival curves for TNBC patients by iNOS status (n = 199). Patients with weak iNOS expression (n = 79) or moderate iNOS expression (n = 59) were significantly associated with decreased recurrence free survival compared to patients with negative iNOS expression (n = 27) (P=0.036 & P=0.007, log-rank test). Patients with strong iNOS expression (n = 34) showed a trend toward worse survival compared to patients with negative iNOS expression (n = 27), but did not reach significance (P=0.078, log-rank test). (B) Kaplan-Meier cumulative distant metastasis free survival curves for TNBC patients by iNOS status (n = 199). Patients with weak iNOS expression (n = 79) or moderate levels of iNOS expression (n = 59) were significantly associated with increased risk of distant metastasis compared to patients with negative iNOS expression (n = 27) (P=0.017 & P=0.002, log-rank test). Patients with strong iNOS expression (n = 34) showed a trend toward increased risk of distant metastasis compared to patients with negative iNOS expression (n = 27), but did not reach significance (P=0.052, log-rank test). (C) Kaplan-Meier cumulative breast cancer–specific survival curves for TNBC patients by iNOS status (n = 200). Patients with weak iNOS expression (n = 78) or moderate levels of iNOS expression (n = 59) were significantly associated with increased risk of death due to breast cancer compared to patients with negative iNOS expression (n = 28) (P=0.012 & P=0.007, log-rank test). Patients with strong iNOS expression (n = 35) showed a trend toward increased risk of distant metastasis compared to patients with negative iNOS expression (n = 28), but did not reach significance (P=0.093, log-rank test).
Figure 3. NO induces increased EGFR and…
Figure 3. NO induces increased EGFR and ERK phosphorylation in TNBC cell lines
Phosphorylation status of EGFR (A) in the MCF-10A, MDA-MB-468 and HCC1806 cell lines after 24 hours exposure to increasing doses of DETA/NO alone or in combination with 100nM of PD153035 (EGFR inhibitor). (B) Phosphorylation status of the MAP kinases ERK1 and ERK2 after 24 hours exposure to 0.5mM of DETA/NO alone or in combination with 100nM of PD153035 or 200nM PD198306 (MEK inhibitor).
Figure 4. Densitometry analysis of EGFR phosphorylation…
Figure 4. Densitometry analysis of EGFR phosphorylation in response to DETA/NO
Quantification of western blots (Figure 3A) examining EGFR phosphorylation at Y1068, Y1173 and Y1045 and total EGFR expression in the MCF-10A, MDA-MB-468 and HCC1806 cell lines after 24 hours exposure to increasing doses of DETA/NO alone or in combination with 100nM of PD153035 (EGFR inhibitor).
Figure 5. Densitometry analysis of ERK phosphorylation…
Figure 5. Densitometry analysis of ERK phosphorylation in response to DETA/NO
Quantification of western blots (Figure 3B) examining ERK phosphorylation at Y204 and total ERK expression in the MCF-10A, MDA-MB-468 and HCC1806 cell lines after 24 hours exposure to increasing doses of DETA/NO alone or in combination with 100nM of PD153035 (EGFR inhibitor) or 200nM PD198306 (MEK inhibitor).
Figure 6. Induction of COX-2 by NO…
Figure 6. Induction of COX-2 by NO in HCC1806 is suppressed by EGFR or MEK inhibitiors
COX-2 protein levels in MCF-10A, MDA-MB-468 and HCC1806 after 24 hours exposure to 0.5mM of DETA/NO alone or in combination with 100nM of PD153035 or 200nM PD198306.
Figure 7. NO induction of IL-1β and…
Figure 7. NO induction of IL-1β and IL-8 secretion is perturbed by EGFR inhibition in HCC1806
Cytokine profiling of released IL-6 (A) IL-1β (B) and Il-8 (C) in MCF-10A, MDA-MB-468 and HCC1806 after 24 hours exposure to vehicle or 0.5mM of DETA/NO alone or in combination with 100nM of PD153035. * p<0.05 vs vehicle; # p<0.05 vs DETA/NO plus PD153035
Figure 8. NO induces increased TNFα secretion…
Figure 8. NO induces increased TNFα secretion and activation of NF-κB in HCC1806
TNFα released (A) in the three cell lines after 24 hours exposure to vehicle or 0.5mM of DETA/NO alone or in combination with 100nM of PD153035. (B) Luciferase activity of NF-κB reporter after 24 hours exposure to vehicle or 0.5mM of DETA/NO alone or in combination with 50μg/ml of Infliximab (TNFα blocking antibody), 10ng/ml of recombinant TNFα were used as a positive control. * p<0.05 vs vehicle; # p<0.05 vs DETA/NO plus PD153035.
Figure 9. NO induces increased MMP expression…
Figure 9. NO induces increased MMP expression and activity
(A) qPCR showing the mRNA expression levels of MMP1, MMP2 and MMP9 after 24 hours exposure to vehicle or 0.5mM of DETA/NO alone or in combination with 100nM of PD153035. (B) Zymography showing the gelatinase activity of MMP9 and MMP2 after 24 hours exposure to vehicle or 0.5mM of DETA/NO alone or in combination with 100nM of PD153035.
Figure 10. Impact of EGFR inhibition on…
Figure 10. Impact of EGFR inhibition on NO induction of cell migration and invasion
(A) Cell migration, or invasion through (B) gelatin and (C) collagen after 24 hours exposure to vehicle or 0.5mM of DETA/NO alone or in combination with 100nM of PD153035 or 200nM PD198306.* p<0.05 vs vehicle; # p<0.05 vs DETA/NO plus PD153035.
Figure 11. Representative images of NO effects…
Figure 11. Representative images of NO effects on migration and invasion
Representative images of migratory and invasion potential (A) and invasive potential of the cell lines to invade through a 0.2% gelatin (B) or 1mg/ml collagen (C) of the cell lines after 24 hours exposure to vehicle or 0.5mM of DETA/NO alone or in combination with 100nM of PD153035 or 200nM PD198306.

References

    1. Carey LA, Dees EC, Sawyer L, Gatti L, Moore DT, Collichio F, Ollila DW, Sartor CI, Graham ML, Perou CM. The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res. 2007;13:2329–2334.
    1. Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P, Narod SA. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. 2007;13:4429–4434.
    1. Tischkowitz M, Brunet JS, Begin LR, Huntsman DG, Cheang MC, Akslen LA, Nielsen TO, Foulkes WD. Use of immunohistochemical markers can refine prognosis in triple negative breast cancer. BMC Cancer. 2007;7:134.
    1. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121:2750–2767.
    1. Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, Savage MI, Osborne CK, Hilsenbeck SG, Chang JC, Mills GB, Lau CC, Brown PH. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 2015;21:1688–1698.
    1. Lehmann BD, Jovanovic B, Chen X, Estrada MV, Johnson KN, Shyr Y, Moses HL, Sanders ME, Pietenpol JA. Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection. PLoS One. 2016;11:e0157368.
    1. Burke AJ, Sullivan FJ, Giles FJ, Glynn SA. The yin and yang of nitric oxide in cancer progression. Carcinogenesis. 2013;34:503–512.
    1. Ambs S, Glynn SA. Candidate pathways linking inducible nitric oxide synthase to a basal-like transcription pattern and tumor progression in human breast cancer. Cell Cycle. 2011;10:619–624.
    1. Switzer CH, Glynn SA, Cheng RY, Ridnour LA, Green JE, Ambs S, Wink DA. S-nitrosylation of EGFR and Src activates an oncogenic signaling network in human basal-like breast cancer. Mol Cancer Res. 2012;10:1203–1215.
    1. Wink DA, Vodovotz Y, Laval J, Laval F, Dewhirst MW, Mitchell JB. The multifaceted roles of nitric oxide in cancer. Carcinogenesis. 1998;19:711–721.
    1. Pervin S, Singh R, Hernandez E, Wu G, Chaudhuri G. Nitric oxide in physiologic concentrations targets the translational machinery to increase the proliferation of human breast cancer cells: involvement of mammalian target of rapamycin/eIF4E pathway. Cancer Res. 2007;67:289–299.
    1. Shami PJ, Sauls DL, Weinberg JB. Schedule and concentration-dependent induction of apoptosis in leukemia cells by nitric oxide. Leukemia. 1998;12:1461–1466.
    1. Nathan C, Xie QW. Nitric oxide synthases: roles, tolls, and controls. Cell. 1994;78:915–918.
    1. Glynn SA, Boersma BJ, Dorsey TH, Yi M, Yfantis HG, Ridnour LA, Martin DN, Switzer CH, Hudson RS, Wink DA, Lee DH, Stephens RM, Ambs S. Increased NOS2 predicts poor survival in estrogen receptor-negative breast cancer patients. J Clin Invest. 2010;120:3843–3854.
    1. Krishnamurti U, Wetherilt CS, Yang J, Peng L, Li X. Tumor-infiltrating lymphocytes are significantly associated with better overall survival and disease-free survival in triple negative but not estrogen receptor positive breast cancers. Hum Pathol. 2017;64:7–12.
    1. Bellizzi A, Greco MR, Rubino R, Paradiso A, Forciniti S, Zeeberg K, Cardone RA, Reshkin SJ. The scaffolding protein NHERF1 sensitizes EGFR-dependent tumor growth, motility and invadopodia function to gefitinib treatment in breast cancer cells. Int J Oncol. 2015;46:1214–1224.
    1. Park HS, Jang MH, Kim EJ, Kim HJ, Lee HJ, Kim YJ, Kim JH, Kang E, Kim SW, Kim IA, Park SY. High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. Mod Pathol. 2014;27:1212–1222.
    1. Thomas DD, Ridnour LA, Isenberg JS, Flores-Santana W, Switzer CH, Donzelli S, Hussain P, Vecoli C, Paolocci N, Ambs S, Colton CA, Harris CC, Roberts DD, et al. The chemical biology of nitric oxide: implications in cellular signaling. Free Radic Biol Med. 2008;45:18–31.
    1. Wink DA, Hines HB, Cheng RY, Switzer CH, Flores-Santana W, Vitek MP, Ridnour LA, Colton CA. Nitric oxide and redox mechanisms in the immune response. J Leukoc Biol. 2011;89:873–891.
    1. Thippeswamy T, McKay JS, Morris R, Quinn J, Wong LF, Murphy D. Glial-mediated neuroprotection: evidence for the protective role of the NO-cGMP pathway via neuron-glial communication in the peripheral nervous system. Glia. 2005;49:197–210.
    1. Nyati MK, Morgan MA, Feng FY, Lawrence TS. Integration of EGFR inhibitors with radiochemotherapy. Nat Rev Cancer. 2006;6:876–885.
    1. Ripple MO, Kim N, Springett R. Acute mitochondrial inhibition by mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK) 1/2 inhibitors regulates proliferation. J Biol Chem. 2013;288:2933–2940.
    1. Heinecke JL, Ridnour LA, Cheng RY, Switzer CH, Lizardo MM, Khanna C, Glynn SA, Hussain SP, Young HA, Ambs S, Wink DA. Tumor microenvironment-based feed-forward regulation of NOS2 in breast cancer progression. Proc Natl Acad Sci U S A. 2014;111:6323–6328.
    1. Basudhar D, Somasundaram V, de Oliveira GA, Kesarwala A, Heinecke JL, Cheng RY, Glynn SA, Ambs S, Wink DA, Ridnour LA. Nitric Oxide Synthase-2-Derived Nitric Oxide Drives Multiple Pathways of Breast Cancer Progression. Antioxid Redox Signal. 2016;26:1044–1058.
    1. Glynn SA, Prueitt RL, Ridnour LA, Boersma BJ, Dorsey TM, Wink DA, Goodman JE, Yfantis HG, Lee DH, Ambs S. COX-2 activation is associated with Akt phosphorylation and poor survival in ER-negative, HER2-positive breast cancer. BMC Cancer. 2010;10:626.
    1. Attiga FA, Fernandez PM, Weeraratna AT, Manyak MJ, Patierno SR. Inhibitors of prostaglandin synthesis inhibit human prostate tumor cell invasiveness and reduce the release of matrix metalloproteinases. Cancer Res. 2000;60:4629–4637.
    1. Grosch S, Maier TJ, Schiffmann S, Geisslinger G. Cyclooxygenase-2 (COX-2)-independent anticarcinogenic effects of selective COX-2 inhibitors. J Natl Cancer Inst. 2006;98:736–747.
    1. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539–545.
    1. Grellner W, Georg T, Wilske J. Quantitative analysis of proinflammatory cytokines (IL-1beta, IL-6, TNF-alpha) in human skin wounds. Forensic Sci Int. 2000;113:251–264.
    1. Zidi I, Mestiri S, Bartegi A, Amor NB. TNF-alpha and its inhibitors in cancer. Med Oncol. 2010;27:185–198.
    1. Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, Mayo MW. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004;23:2369–2380.
    1. Wu Y, Zhou BP. Inflammation: a driving force speeds cancer metastasis. Cell Cycle. 2009;8:3267–3273.
    1. Coffelt SB, de Visser KE. Cancer: Inflammation lights the way to metastasis. Nature. 2014;507:48–49.
    1. Jovanovic M, Stefanoska I, Radojcic L, Vicovac L. Interleukin-8 (CXCL8) stimulates trophoblast cell migration and invasion by increasing levels of matrix metalloproteinase (MMP)2 and MMP9 and integrins alpha5 and beta1. Reproduction. 2010;139:789–798.
    1. Liu CJ, Kuo FC, Wang CL, Kuo CH, Wang SS, Chen CY, Huang YB, Cheng KH, Yokoyama KK, Chen CL, Lu CY, Wu DC. Suppression of IL-8-Src signalling axis by 17beta-estradiol inhibits human mesenchymal stem cells-mediated gastric cancer invasion. J Cell Mol Med. 2016;20:962–972.
    1. Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2:563–572.
    1. Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3:362–374.
    1. Saha P, Nanda R. Concepts and targets in triple-negative breast cancer: recent results and clinical implications. Ther Adv Med Oncol. 2016;8:351–359.
    1. Granados-Principal S, Liu Y, Guevara ML, Blanco E, Choi DS, Qian W, Patel T, Rodriguez AA, Cusimano J, Weiss HL, Zhao H, Landis MD, Dave B, et al. Inhibition of iNOS as a novel effective targeted therapy against triple-negative breast cancer. Breast Cancer Res. 2015;17:25.
    1. Bos M, Mendelsohn J, Kim YM, Albanell J, Fry DW, Baselga J. PD153035, a tyrosine kinase inhibitor, prevents epidermal growth factor receptor activation and inhibits growth of cancer cells in a receptor number-dependent manner. Clin Cancer Res. 1997;3:2099–2106.
    1. Bartholomeusz C, Gonzalez-Angulo AM, Liu P, Hayashi N, Lluch A, Ferrer-Lozano J, Hortobagyi GN. High ERK protein expression levels correlate with shorter survival in triple-negative breast cancer patients. Oncologist. 2012;17:766–774.
    1. Kim S, Lee J, Jeon M, Lee JE, Nam SJ. MEK-dependent IL-8 induction regulates the invasiveness of triple-negative breast cancer cells. Tumour Biol. 2016;37:4991–4999.
    1. Bartholomeusz C, Xie X, Pitner MK, Kondo K, Dadbin A, Lee J, Saso H, Smith PD, Dalby KN, Ueno NT. MEK Inhibitor Selumetinib (AZD6244; ARRY-142886) Prevents Lung Metastasis in a Triple-Negative Breast Cancer Xenograft Model. Mol Cancer Ther. 2015;14:2773–2781.
    1. Gupta S, Srivastava M, Ahmad N, Bostwick DG, Mukhtar H. Over-expression of cyclooxygenase-2 in human prostate adenocarcinoma. Prostate. 2000;42:73–78.
    1. Rubio J, Ramos D, Lopez-Guerrero JA, Iborra I, Collado A, Solsona E, Almenar S, Llombart-Bosch A. Immunohistochemical expression of Ki-67 antigen, cox-2 and Bax/Bcl-2 in prostate cancer; prognostic value in biopsies and radical prostatectomy specimens. Eur Urol. 2005;48:745–751.
    1. Tsujii M, Kawano S, Tsuji S, Sawaoka H, Hori M, DuBois RN. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell. 1998;93:705–716.
    1. Brown JR, DuBois RN. COX-2: a molecular target for colorectal cancer prevention. J Clin Oncol. 2005;23:2840–2855.
    1. Mantovani A, Bonecchi R, Locati M. Tuning inflammation and immunity by chemokine sequestration: decoys and more. Nat Rev Immunol. 2006;6:907–918.
    1. Kaler P, Augenlicht L, Klampfer L. Macrophage-derived IL-1beta stimulates Wnt signaling and growth of colon cancer cells: a crosstalk interrupted by vitamin D3. Oncogene. 2009;28:3892–3902.
    1. Balkwill F. Tumour necrosis factor and cancer. Nat Rev Cancer. 2009;9:361–371.
    1. Ben-Baruch A. The Tumor-Promoting Flow of Cells Into, Within and Out of the Tumor Site: Regulation by the Inflammatory Axis of TNFalpha and Chemokines. Cancer Microenviron. 2012;5:151–164.
    1. Karin M, Lin A. NF-kappaB at the crossroads of life and death. Nat Immunol. 2002;3:221–227.
    1. Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2:301–310.
    1. Fernando RI, Castillo MD, Litzinger M, Hamilton DH, Palena C. IL-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells. Cancer Res. 2011;71:5296–5306.
    1. Shemesh CS, Hardy CW, Yu DS, Fernandez B, Zhang H. Indocyanine green loaded liposome nanocarriers for photodynamic therapy using human triple negative breast cancer cells. Photodiagnosis Photodyn Ther. 2014;11:193–203.
    1. Huber MC, Mall R, Braselmann H, Feuchtinger A, Molatore S, Lindner K, Walch A, Gross E, Schmitt M, Falkenberg N, Aubele M. uPAR enhances malignant potential of triple-negative breast cancer by directly interacting with uPA and IGF1R. BMC Cancer. 2016;16:615.
    1. Anantaraju HS, Battu MB, Viswanadha S, Sriram D, Yogeeswari P. Cathepsin D inhibitors as potential therapeutics for breast cancer treatment: Molecular docking and bioevaluation against triple-negative and triple-positive breast cancers. Mol Divers. 2016;20:521–535.
    1. Ingthorsson S, Andersen K, Hilmarsdottir B, Maelandsmo GM, Magnusson MK, Gudjonsson T. HER2 induced EMT and tumorigenicity in breast epithelial progenitor cells is inhibited by coexpression of EGFR. Oncogene. 2016;35:4244–4255.
    1. Ferraro DA, Gaborit N, Maron R, Cohen-Dvashi H, Porat Z, Pareja F, Lavi S, Lindzen M, Ben-Chetrit N, Sela M, Yarden Y. Inhibition of triple-negative breast cancer models by combinations of antibodies to EGFR. Proc Natl Acad Sci U S A. 2013;110:1815–1820.
    1. Nabholtz JM, Chalabi N, Radosevic-Robin N, Dauplat MM, Mouret-Reynier MA, Van Praagh I, Servent V, Jacquin JP, Benmammar KE, Kullab S, Bahadoor MR, Kwiatkowski F, Cayre A, et al. Multicentric neoadjuvant pilot Phase II study of cetuximab combined with docetaxel in operable triple negative breast cancer. Int J Cancer. 2016;138:2274–2280.
    1. Baselga J, Gomez P, Greil R, Braga S, Climent MA, Wardley AM, Kaufman B, Stemmer SM, Pego A, Chan A, Goeminne JC, Graas MP, Kennedy MJ, et al. Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J Clin Oncol. 2013;31:2586–2592.
    1. Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX, Liu MC, Storniolo AM, Rimawi MF, Forero-Torres A, Wolff AC, Hobday TJ, Ivanova A, et al. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol. 2012;30:2615–2623.
    1. Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13:674–690.
    1. Prueitt RL, Boersma BJ, Howe TM, Goodman JE, Thomas DD, Ying L, Pfiester CM, Yfantis HG, Cottrell JR, Lee DH, Remaley AT, Hofseth LJ, Wink DA, et al. Inflammation and IGF-I activate the Akt pathway in breast cancer. Int J Cancer. 2007;120:796–805.
    1. Wallace TA, Downey RF, Seufert CJ, Schetter A, Dorsey TH, Johnson CA, Goldman R, Loffredo CA, Yan P, Sullivan FJ, Giles FJ, Wang-Johanning F, Ambs S, et al. Elevated HERV-K mRNA expression in PBMC is associated with a prostate cancer diagnosis particularly in older men and smokers. Carcinogenesis. 2014;35:2074–2083.

Source: PubMed

3
Prenumerera