Skip to main content
Top
Published in: Breast Cancer Research 1/2018

Open Access 01-12-2018 | Research article

Cleavage of the extracellular domain of junctional adhesion molecule-A is associated with resistance to anti-HER2 therapies in breast cancer settings

Authors: Astrid O. Leech, Sri HariKrishna Vellanki, Emily J. Rutherford, Aoife Keogh, Hanne Jahns, Lance Hudson, Norma O’Donovan, Siham Sabri, Bassam Abdulkarim, Katherine M. Sheehan, Elaine W. Kay, Leonie S. Young, Arnold D. K. Hill, Yvonne E. Smith, Ann M. Hopkins

Published in: Breast Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Junctional adhesion molecule-A (JAM-A) is an adhesion molecule whose overexpression on breast tumor tissue has been associated with aggressive cancer phenotypes, including human epidermal growth factor receptor-2 (HER2)-positive disease. Since JAM-A has been described to regulate HER2 expression in breast cancer cells, we hypothesized that JAM-dependent stabilization of HER2 could participate in resistance to HER2-targeted therapies.

Methods

Using breast cancer cell line models resistant to anti-HER2 drugs, we investigated JAM-A expression and the effect of JAM-A silencing on biochemical/functional parameters. We also tested whether altered JAM-A expression/processing underpinned differences between drug-sensitive and -resistant cells and acted as a biomarker of patients who developed resistance to HER2-targeted therapies.

Results

Silencing JAM-A enhanced the anti-proliferative effects of anti-HER2 treatments in trastuzumab- and lapatinib-resistant breast cancer cells and further reduced HER2 protein expression and Akt phosphorylation in drug-treated cells. Increased epidermal growth factor receptor expression observed in drug-resistant models was normalized upon JAM-A silencing. JAM-A was highly expressed in all of a small cohort of HER2-positive patients whose disease recurred following anti-HER2 therapy. High JAM-A expression also correlated with metastatic disease at the time of diagnosis in another patient cohort resistant to trastuzumab therapy. Importantly, cleavage of JAM-A was increased in drug-resistant cell lines in conjunction with increased expression of ADAM-10 and -17 metalloproteases. Pharmacological inhibition or genetic silencing studies suggested a particular role for ADAM-10 in reducing JAM-A cleavage and partially re-sensitizing drug-resistant cells to the anti-proliferative effects of HER2-targeted drugs. Functionally, recombinant cleaved JAM-A enhanced breast cancer cell invasion in vitro and both invasion and proliferation in a semi-in vivo model. Finally, cleaved JAM-A was detectable in the serum of a small cohort of HER2-positive patients and correlated significantly with resistance to HER2-targeted therapy.

Conclusions

Collectively, our data suggest a novel model whereby increased expression and cleavage of JAM-A drive tumorigenic behavior and act as a biomarker and potential therapeutic target for resistance to HER2-targeted therapies.
Appendix
Available only for authorised users
Literature
1.
go back to reference Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human-breast cancer - correlation of relapse and survival with amplification of the her-2 neu oncogene. Science. 1987;235:177–82.CrossRef Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human-breast cancer - correlation of relapse and survival with amplification of the her-2 neu oncogene. Science. 1987;235:177–82.CrossRef
2.
go back to reference Middleton LP, Price KM, Puig P, Heydon LJ, Tarco E, Sneige N, et al. Implementation of American Society of Clinical Oncology/College of American Pathologists HER2 Guideline Recommendations in a tertiary care facility increases HER2 immunohistochemistry and fluorescence in situ hybridization concordance and decreases the number of inconclusive cases. Arch Pathol Lab Med. 2009;133:775–80.PubMed Middleton LP, Price KM, Puig P, Heydon LJ, Tarco E, Sneige N, et al. Implementation of American Society of Clinical Oncology/College of American Pathologists HER2 Guideline Recommendations in a tertiary care facility increases HER2 immunohistochemistry and fluorescence in situ hybridization concordance and decreases the number of inconclusive cases. Arch Pathol Lab Med. 2009;133:775–80.PubMed
3.
go back to reference Rakha EA, Pinder SE, Bartlett JM, Ibrahim M, Starczynski J, Carder PJ, et al. Updated UK Recommendations for HER2 assessment in breast cancer. J Clin Pathol. 2015, 68:93–99.CrossRef Rakha EA, Pinder SE, Bartlett JM, Ibrahim M, Starczynski J, Carder PJ, et al. Updated UK Recommendations for HER2 assessment in breast cancer. J Clin Pathol. 2015, 68:93–99.CrossRef
4.
go back to reference Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN. The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist. 2009;14:320–68.CrossRef Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN. The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist. 2009;14:320–68.CrossRef
5.
go back to reference Yardley DA, Kaufman PA, Brufsky A, Yood MU, Rugo H, Mayer M, et al. Treatment patterns and clinical outcomes for patients with de novo versus recurrent HER2-positive metastatic breast cancer. Breast Cancer Res Treat. 2014;145:725–34.CrossRef Yardley DA, Kaufman PA, Brufsky A, Yood MU, Rugo H, Mayer M, et al. Treatment patterns and clinical outcomes for patients with de novo versus recurrent HER2-positive metastatic breast cancer. Breast Cancer Res Treat. 2014;145:725–34.CrossRef
6.
go back to reference Ahmed S, Sami A, Xiang J. HER2-directed therapy: current treatment options for HER2-positive breast cancer. Breast Cancer. 2015;22:101–16.CrossRef Ahmed S, Sami A, Xiang J. HER2-directed therapy: current treatment options for HER2-positive breast cancer. Breast Cancer. 2015;22:101–16.CrossRef
7.
go back to reference Araki K, Fukada I, Horii R, Takahashi S, Akiyama F, Iwase T, et al. Trastuzumab Rechallenge After Lapatinib- and Trastuzumab-Resistant Disease Progression in HER2-Positive Breast Cancer. Clin Breast Cancer. 2015;15:432–9.CrossRef Araki K, Fukada I, Horii R, Takahashi S, Akiyama F, Iwase T, et al. Trastuzumab Rechallenge After Lapatinib- and Trastuzumab-Resistant Disease Progression in HER2-Positive Breast Cancer. Clin Breast Cancer. 2015;15:432–9.CrossRef
8.
go back to reference O'Brien NA, Browne BC, Chow L, Wang Y, Ginther C, Arboleda J, et al. Activated phosphoinositide 3-kinase/AKT signaling confers resistance to trastuzumab but not lapatinib. Mol Cancer Ther. 2010;9:1489–502.CrossRef O'Brien NA, Browne BC, Chow L, Wang Y, Ginther C, Arboleda J, et al. Activated phosphoinositide 3-kinase/AKT signaling confers resistance to trastuzumab but not lapatinib. Mol Cancer Ther. 2010;9:1489–502.CrossRef
9.
go back to reference Guarneri V, Dieci MV, Frassoldati A, Maiorana A, Ficarra G, Bettelli S, et al. Prospective Biomarker Analysis of the Randomized CHER-LOB Study Evaluating the Dual Anti-HER2 Treatment With Trastuzumab and Lapatinib Plus Chemotherapy as Neoadjuvant Therapy for HER2-Positive Breast Cancer. Oncologist 2015, 20:1001–1010.CrossRef Guarneri V, Dieci MV, Frassoldati A, Maiorana A, Ficarra G, Bettelli S, et al. Prospective Biomarker Analysis of the Randomized CHER-LOB Study Evaluating the Dual Anti-HER2 Treatment With Trastuzumab and Lapatinib Plus Chemotherapy as Neoadjuvant Therapy for HER2-Positive Breast Cancer. Oncologist 2015, 20:1001–1010.CrossRef
10.
go back to reference de Melo Gagliato D, Jardim DL, Marchesi MS, Hortobagyi GN. Mechanisms of resistance and sensitivity to anti-HER2 therapies in HER2+ breast cancer. Oncotarget. 2016;7:64431–46.PubMedPubMedCentral de Melo Gagliato D, Jardim DL, Marchesi MS, Hortobagyi GN. Mechanisms of resistance and sensitivity to anti-HER2 therapies in HER2+ breast cancer. Oncotarget. 2016;7:64431–46.PubMedPubMedCentral
11.
go back to reference Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, et al. Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol. 2013, 24:2278–2284.CrossRef Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, et al. Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol. 2013, 24:2278–2284.CrossRef
12.
go back to reference Swain SM, Baselga J, Kim SB, Ro J, Semiglazov V, Campone M, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015, 372:724–734.CrossRef Swain SM, Baselga J, Kim SB, Ro J, Semiglazov V, Campone M, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015, 372:724–734.CrossRef
13.
go back to reference Babinska A, Kedees MH, Athar H, Sobocki T, Sobocka MB, Ahmed T, et al. Two regions of the human platelet F11-receptor (F11R) are critical for platelet aggregation, potentiation and adhesion. Thromb Haemost. 2002;87:712–21.CrossRef Babinska A, Kedees MH, Athar H, Sobocki T, Sobocka MB, Ahmed T, et al. Two regions of the human platelet F11-receptor (F11R) are critical for platelet aggregation, potentiation and adhesion. Thromb Haemost. 2002;87:712–21.CrossRef
14.
go back to reference Götte M, Mohr C, Koo CY, Stock C, Vaske AK, Viola M, et al. miR-145-dependent targeting of junctional adhesion molecule A and modulation of fascin expression are associated with reduced breast cancer cell motility and invasiveness. Oncogene 2010, 29:6569–6580.CrossRef Götte M, Mohr C, Koo CY, Stock C, Vaske AK, Viola M, et al. miR-145-dependent targeting of junctional adhesion molecule A and modulation of fascin expression are associated with reduced breast cancer cell motility and invasiveness. Oncogene 2010, 29:6569–6580.CrossRef
15.
go back to reference McSherry EA, Brennan K, Hudson L, Hill AD, Hopkins AM. Breast cancer cell migration is regulated through junctional adhesion molecule-A-mediated activation of Rap1 GTPase. Breast Cancer Res. 2011;13:R31.CrossRef McSherry EA, Brennan K, Hudson L, Hill AD, Hopkins AM. Breast cancer cell migration is regulated through junctional adhesion molecule-A-mediated activation of Rap1 GTPase. Breast Cancer Res. 2011;13:R31.CrossRef
16.
go back to reference Murakami M, Giampietro C, Giannotta M, Corada M, Torselli I, Orsenigo F, et al. Abrogation of junctional adhesion molecule-A expression induces cell apoptosis and reduces breast cancer progression. PLoS One 2011, 6:e21242.CrossRef Murakami M, Giampietro C, Giannotta M, Corada M, Torselli I, Orsenigo F, et al. Abrogation of junctional adhesion molecule-A expression induces cell apoptosis and reduces breast cancer progression. PLoS One 2011, 6:e21242.CrossRef
17.
go back to reference Goetsch L, Haeuw JF, Beau-Larvor C, Gonzalez A, Zanna L, Malissard M, et al. A novel role for junctional adhesion molecule-A in tumor proliferation: modulation by an anti-JAM-A monoclonal antibody. Int J Cancer 2013, 132:1463–1474.CrossRef Goetsch L, Haeuw JF, Beau-Larvor C, Gonzalez A, Zanna L, Malissard M, et al. A novel role for junctional adhesion molecule-A in tumor proliferation: modulation by an anti-JAM-A monoclonal antibody. Int J Cancer 2013, 132:1463–1474.CrossRef
18.
go back to reference Brennan K, McSherry EA, Hudson L, Kay EW, Hill ADK, Young LS, et al. Junctional adhesion molecule-A is co-expressed with HER2 in breast tumors and acts as a novel regulator of HER2 protein degradation and signaling. Oncogene. 2013;32:2799–804.CrossRef Brennan K, McSherry EA, Hudson L, Kay EW, Hill ADK, Young LS, et al. Junctional adhesion molecule-A is co-expressed with HER2 in breast tumors and acts as a novel regulator of HER2 protein degradation and signaling. Oncogene. 2013;32:2799–804.CrossRef
19.
go back to reference McSherry EA, McGee SF, Jirstrom K, Doyle EM, Brennan DJ, Landberg G, et al. JAM-A expression positively correlates with poor prognosis in breast cancer patients. Int J Cancer. 2009;125:1343–51.CrossRef McSherry EA, McGee SF, Jirstrom K, Doyle EM, Brennan DJ, Landberg G, et al. JAM-A expression positively correlates with poor prognosis in breast cancer patients. Int J Cancer. 2009;125:1343–51.CrossRef
20.
go back to reference Leech AO, Cruz RG, Hill AD, Hopkins AM. Paradigms lost-an emerging role for over-expression of tight junction adhesion proteins in cancer pathogenesis. Annals Transl Med. 2015;3:184. Leech AO, Cruz RG, Hill AD, Hopkins AM. Paradigms lost-an emerging role for over-expression of tight junction adhesion proteins in cancer pathogenesis. Annals Transl Med. 2015;3:184.
21.
go back to reference McDermott MSJ, Browne BC, Conlon NT, O'Brien NA, Slamon DJ, Henry M, et al. PP2A inhibition overcomes acquired resistance to HER2 targeted therapy. Mol Cancer. 2014;13:157.CrossRef McDermott MSJ, Browne BC, Conlon NT, O'Brien NA, Slamon DJ, Henry M, et al. PP2A inhibition overcomes acquired resistance to HER2 targeted therapy. Mol Cancer. 2014;13:157.CrossRef
22.
go back to reference Browne BC, Crown J, Venkatesan N, Duffy MJ, Clynes M, Slamon D, et al. Inhibition of IGF1R activity enhances response to trastuzumab in HER-2-positive breast cancer cells. Ann Oncol. 2011;22:68–73.CrossRef Browne BC, Crown J, Venkatesan N, Duffy MJ, Clynes M, Slamon D, et al. Inhibition of IGF1R activity enhances response to trastuzumab in HER-2-positive breast cancer cells. Ann Oncol. 2011;22:68–73.CrossRef
23.
go back to reference Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang GR, Rahmeh M, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–9.CrossRef Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang GR, Rahmeh M, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:1630–9.CrossRef
24.
go back to reference Lesniak D, Xu Y, Deschenes J, Lai R, Thoms J, Murray D, et al. Beta1-integrin circumvents the antiproliferative effects of trastuzumab in human epidermal growth factor receptor-2-positive breast cancer. Cancer Res. 2009;69:8620–8.CrossRef Lesniak D, Xu Y, Deschenes J, Lai R, Thoms J, Murray D, et al. Beta1-integrin circumvents the antiproliferative effects of trastuzumab in human epidermal growth factor receptor-2-positive breast cancer. Cancer Res. 2009;69:8620–8.CrossRef
25.
go back to reference Krop IE, Flores L, Najita JS, Mayer IA, Hobday TJ, Falkson CI, et al. The role of EGFR amplification in trastuzumab resistance: A correlative analysis of TBCRC003. J Clin Oncol. 2011;29(no. 15_suppl):528.CrossRef Krop IE, Flores L, Najita JS, Mayer IA, Hobday TJ, Falkson CI, et al. The role of EGFR amplification in trastuzumab resistance: A correlative analysis of TBCRC003. J Clin Oncol. 2011;29(no. 15_suppl):528.CrossRef
26.
go back to reference Iida M, Brand TM, Starr MM, Huppert EJ, Luthar N, Bahrar H, et al. Overcoming acquired resistance to cetuximab by dual targeting HER family receptors with antibody-based therapy. Mol Cancer 2014, 13:242.CrossRef Iida M, Brand TM, Starr MM, Huppert EJ, Luthar N, Bahrar H, et al. Overcoming acquired resistance to cetuximab by dual targeting HER family receptors with antibody-based therapy. Mol Cancer 2014, 13:242.CrossRef
27.
go back to reference Dua R, Zhang J, Nhonthachit P, Penuel E, Petropoulos C, Parry G. EGFR over-expression and activation in high HER2, ER negative breast cancer cell line induces trastuzumab resistance. Breast Cancer Res Treat. 2010;122:685–97.CrossRef Dua R, Zhang J, Nhonthachit P, Penuel E, Petropoulos C, Parry G. EGFR over-expression and activation in high HER2, ER negative breast cancer cell line induces trastuzumab resistance. Breast Cancer Res Treat. 2010;122:685–97.CrossRef
28.
go back to reference Koenen RR, Pruessmeyer J, Soehnlein O, Fraemohs L, Zernecke A, Schwarz N, et al. Regulated release and functional modulation of junctional adhesion molecule A by disintegrin metalloproteinases. Blood. 2009;113:4799–809.CrossRef Koenen RR, Pruessmeyer J, Soehnlein O, Fraemohs L, Zernecke A, Schwarz N, et al. Regulated release and functional modulation of junctional adhesion molecule A by disintegrin metalloproteinases. Blood. 2009;113:4799–809.CrossRef
29.
go back to reference Lokman NA, Elder AS, Ricciardelli C, Oehler MK. Chick chorioallantoic membrane (CAM) assay as an in vivo model to study the effect of newly identified molecules on ovarian cancer invasion and metastasis. Int J Mol Sci. 2012;13:9959–70.CrossRef Lokman NA, Elder AS, Ricciardelli C, Oehler MK. Chick chorioallantoic membrane (CAM) assay as an in vivo model to study the effect of newly identified molecules on ovarian cancer invasion and metastasis. Int J Mol Sci. 2012;13:9959–70.CrossRef
30.
go back to reference Vogler M, Walczak H, Stadel D, Haas TL, Genze F, Jovanovic M, et al. Targeting XIAP bypasses Bcl-2-mediated resistance to TRAIL and cooperates with TRAIL to suppress pancreatic cancer growth in vitro and in vivo. Cancer Res. 2008;68:7956–65.CrossRef Vogler M, Walczak H, Stadel D, Haas TL, Genze F, Jovanovic M, et al. Targeting XIAP bypasses Bcl-2-mediated resistance to TRAIL and cooperates with TRAIL to suppress pancreatic cancer growth in vitro and in vivo. Cancer Res. 2008;68:7956–65.CrossRef
31.
go back to reference Kuefer R, Hofer MD, Altug V, Zorn C, Genze F, Kunzi-Rapp K, et al. Sodium butyrate and tributyrin induce in vivo growth inhibition and apoptosis in human prostate cancer. Br J Cancer. 2004;90:535–41.CrossRef Kuefer R, Hofer MD, Altug V, Zorn C, Genze F, Kunzi-Rapp K, et al. Sodium butyrate and tributyrin induce in vivo growth inhibition and apoptosis in human prostate cancer. Br J Cancer. 2004;90:535–41.CrossRef
32.
go back to reference Vogler M, Giagkousiklidis S, Genze F, Gschwend JE, Debatin KM, Fulda S. Inhibition of clonogenic tumor growth: a novel function of Smac contributing to its antitumor activity. Oncogene. 2005;24:7190–202.CrossRef Vogler M, Giagkousiklidis S, Genze F, Gschwend JE, Debatin KM, Fulda S. Inhibition of clonogenic tumor growth: a novel function of Smac contributing to its antitumor activity. Oncogene. 2005;24:7190–202.CrossRef
33.
go back to reference Patanaphan V, Salazar OM, Risco R. BREAST-CANCER - METASTATIC PATTERNS AND THEIR PROGNOSIS. South Med J. 1988;81:1109–12.CrossRef Patanaphan V, Salazar OM, Risco R. BREAST-CANCER - METASTATIC PATTERNS AND THEIR PROGNOSIS. South Med J. 1988;81:1109–12.CrossRef
34.
go back to reference Guth U, Magaton I, Huang DJ, Fisher R, Schotzau A, Vetter M. Primary and secondary distant metastatic breast cancer: Two sides of the same coin. Breast. 2014;23:26–32.CrossRef Guth U, Magaton I, Huang DJ, Fisher R, Schotzau A, Vetter M. Primary and secondary distant metastatic breast cancer: Two sides of the same coin. Breast. 2014;23:26–32.CrossRef
35.
go back to reference Menyhart O, Santarpia L, Gyorffy B. A Comprehensive Outline of Trastuzumab Resistance Biomarkers in HER2 Overexpressing Breast Cancer. Curr Cancer Drug Targets. 2015;15:665–83.CrossRef Menyhart O, Santarpia L, Gyorffy B. A Comprehensive Outline of Trastuzumab Resistance Biomarkers in HER2 Overexpressing Breast Cancer. Curr Cancer Drug Targets. 2015;15:665–83.CrossRef
36.
go back to reference Nunes J, Zhang H, Angelopoulos N, Chhetri J, Osipo C, Grothey A, et al. ATG9A loss confers resistance to trastuzumab via c-Cbl mediated Her2 degradation. Oncotarget. 2016;7:27599–612.CrossRef Nunes J, Zhang H, Angelopoulos N, Chhetri J, Osipo C, Grothey A, et al. ATG9A loss confers resistance to trastuzumab via c-Cbl mediated Her2 degradation. Oncotarget. 2016;7:27599–612.CrossRef
37.
go back to reference Liu XD, Fridman JS, Wang Q, Caulder E, Yang GJ, Covington M, et al. Selective inhibition of ADAM metalloproteases blocks HER-2 extracellular domain (ECD) cleavage and potentiates the anti-tumor effects of trastuzumab. Cancer Biol Ther. 2006;5:648–56.CrossRef Liu XD, Fridman JS, Wang Q, Caulder E, Yang GJ, Covington M, et al. Selective inhibition of ADAM metalloproteases blocks HER-2 extracellular domain (ECD) cleavage and potentiates the anti-tumor effects of trastuzumab. Cancer Biol Ther. 2006;5:648–56.CrossRef
38.
go back to reference Kang SH, Kang KW, Kim KH, Kwon B, Kim SK, Lee HY, et al. Upregulated HSP27 in human breast cancer cells reduces Herceptin susceptibility by increasing Her2 protein stability. BMC Cancer. 2008;8:286.CrossRef Kang SH, Kang KW, Kim KH, Kwon B, Kim SK, Lee HY, et al. Upregulated HSP27 in human breast cancer cells reduces Herceptin susceptibility by increasing Her2 protein stability. BMC Cancer. 2008;8:286.CrossRef
39.
go back to reference Chandarlapaty S, Sakr RA, Giri D, Patil S, Heguy A, Morrow M, et al. Frequent mutational activation of the PI3K-AKT pathway in trastuzumab-resistant breast cancer. Clin Cancer Res. 2012, 18:6784–6791.CrossRef Chandarlapaty S, Sakr RA, Giri D, Patil S, Heguy A, Morrow M, et al. Frequent mutational activation of the PI3K-AKT pathway in trastuzumab-resistant breast cancer. Clin Cancer Res. 2012, 18:6784–6791.CrossRef
40.
go back to reference Nagata Y, Lan KH, Zhou XY, Tan M, Esteva FJ, Sahin AA, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell. 2004;6:117–27.CrossRef Nagata Y, Lan KH, Zhou XY, Tan M, Esteva FJ, Sahin AA, et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell. 2004;6:117–27.CrossRef
41.
go back to reference Berns K, Horlings HM, Hennessy BT, Madiredjo M, Hijmans EM, Beelen K, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell. 2007;12:395–402.CrossRef Berns K, Horlings HM, Hennessy BT, Madiredjo M, Hijmans EM, Beelen K, et al. A functional genetic approach identifies the PI3K pathway as a major determinant of trastuzumab resistance in breast cancer. Cancer Cell. 2007;12:395–402.CrossRef
42.
go back to reference Ritter CA, Perez-Torres M, Rinehart C, Guix M, Dugger T, Engelman JA, et al. Human breast cancer cells selected for resistance to trastuzumab in vivo overexpress epidermal growth factor receptor and ErbB Ligands and remain dependent on the ErbB receptor network. Clin Cancer Res. 2007;13:4909–19.CrossRef Ritter CA, Perez-Torres M, Rinehart C, Guix M, Dugger T, Engelman JA, et al. Human breast cancer cells selected for resistance to trastuzumab in vivo overexpress epidermal growth factor receptor and ErbB Ligands and remain dependent on the ErbB receptor network. Clin Cancer Res. 2007;13:4909–19.CrossRef
43.
go back to reference Miller KD. The role of ErbB inhibitors in trastuzumab resistance. Oncologist. 2004;9:16–9.CrossRef Miller KD. The role of ErbB inhibitors in trastuzumab resistance. Oncologist. 2004;9:16–9.CrossRef
44.
go back to reference Tian Y, Tian Y, Zhang W, Wei F, Yang J, Luo X, et al. Junctional adhesion molecule-A, an epithelial-mesenchymal transition inducer, correlates with metastasis and poor prognosis in human nasopharyngeal cancer. Carcinogenesis. 2015;36:41–8.CrossRef Tian Y, Tian Y, Zhang W, Wei F, Yang J, Luo X, et al. Junctional adhesion molecule-A, an epithelial-mesenchymal transition inducer, correlates with metastasis and poor prognosis in human nasopharyngeal cancer. Carcinogenesis. 2015;36:41–8.CrossRef
45.
go back to reference Ikeo K, Oshima T, Shan J, Matsui H, Tomita T, Fukui H, et al. Junctional Adhesion Molecule-A Promotes Proliferation and Inhibits Apoptosis of Gastric Cancer. Hepato-Gastroenterology. 2015;62:540–5.PubMed Ikeo K, Oshima T, Shan J, Matsui H, Tomita T, Fukui H, et al. Junctional Adhesion Molecule-A Promotes Proliferation and Inhibits Apoptosis of Gastric Cancer. Hepato-Gastroenterology. 2015;62:540–5.PubMed
46.
go back to reference Duffy MJ, Crown J, Mullooly M. ADAM10 and ADAM17: New Players in Trastuzumab Resistance. Oncotarget. 2014;5:10963.CrossRef Duffy MJ, Crown J, Mullooly M. ADAM10 and ADAM17: New Players in Trastuzumab Resistance. Oncotarget. 2014;5:10963.CrossRef
47.
go back to reference Feldinger K, Generali D, Kramer-Marek G, Gijsen M, Ng TB, Wong JH, et al. ADAM10 mediates trastuzumab resistance and is correlated with survival in HER2 positive breast cancer. Oncotarget. 2014;5:6633–46.CrossRef Feldinger K, Generali D, Kramer-Marek G, Gijsen M, Ng TB, Wong JH, et al. ADAM10 mediates trastuzumab resistance and is correlated with survival in HER2 positive breast cancer. Oncotarget. 2014;5:6633–46.CrossRef
48.
go back to reference Moss ML. The Knockout Punch for HER2 Positive Breast Cancer: Addition of an ADAM10 Inhibitor to the combination therapy of Trastuzumab and Lapatinib. In: Mol Enzyme Drug Targets, vol. 2; 2016. p. 6. Moss ML. The Knockout Punch for HER2 Positive Breast Cancer: Addition of an ADAM10 Inhibitor to the combination therapy of Trastuzumab and Lapatinib. In: Mol Enzyme Drug Targets, vol. 2; 2016. p. 6.
49.
go back to reference Witters L, Scherle P, Friedman S, Fridman J, Caulder E, Newton R, et al. Synergistic inhibition with a dual epidermal growth factor receptor/HER-2/neu tyrosine kinase inhibitor and a disintegrin and metalloprotease inhibitor. Cancer Res. 2008;68:7083–9.CrossRef Witters L, Scherle P, Friedman S, Fridman J, Caulder E, Newton R, et al. Synergistic inhibition with a dual epidermal growth factor receptor/HER-2/neu tyrosine kinase inhibitor and a disintegrin and metalloprotease inhibitor. Cancer Res. 2008;68:7083–9.CrossRef
50.
go back to reference Sperinde J, Jin XG, Banerjee J, Penuel E, Saha A, Diedrich G, et al. Quantitation of p95HER2 in Paraffin Sections by Using a p95-Specific Antibody and Correlation with Outcome in a Cohort of Trastuzumab-Treated Breast Cancer Patients. Clin Cancer Res. 2010;16:4226–35.CrossRef Sperinde J, Jin XG, Banerjee J, Penuel E, Saha A, Diedrich G, et al. Quantitation of p95HER2 in Paraffin Sections by Using a p95-Specific Antibody and Correlation with Outcome in a Cohort of Trastuzumab-Treated Breast Cancer Patients. Clin Cancer Res. 2010;16:4226–35.CrossRef
51.
go back to reference Kocar M, Bozkurtlar E, Telli F, Yumuk F, Kaya H, Kocar H, et al. p95-HER2 and trastuzumab resistance in metastatic breast cancer; is immunohistochemistry appropriate? J Buon. 2014;19:245–9.PubMed Kocar M, Bozkurtlar E, Telli F, Yumuk F, Kaya H, Kocar H, et al. p95-HER2 and trastuzumab resistance in metastatic breast cancer; is immunohistochemistry appropriate? J Buon. 2014;19:245–9.PubMed
52.
go back to reference Severson EA, Parkos CA. Mechanisms of Outside-in Signaling at the Tight Junction by Junctional Adhesion Molecule A. Ann N Y Acad Sci. 2009;1165:10–8.CrossRef Severson EA, Parkos CA. Mechanisms of Outside-in Signaling at the Tight Junction by Junctional Adhesion Molecule A. Ann N Y Acad Sci. 2009;1165:10–8.CrossRef
53.
go back to reference Gijsen M, King P, Perera T, Parker PJ, Harris AL, Larijani B, et al. HER2 phosphorylation is maintained by a PKB negative feedback loop in response to anti-HER2 herceptin in breast cancer. PLoS Biol. 2010;8:e1000563.CrossRef Gijsen M, King P, Perera T, Parker PJ, Harris AL, Larijani B, et al. HER2 phosphorylation is maintained by a PKB negative feedback loop in response to anti-HER2 herceptin in breast cancer. PLoS Biol. 2010;8:e1000563.CrossRef
54.
go back to reference Konecny GE, Agarwal R, Keeney GA, Winterhoff B, Jones MB, Mariani A, et al. Claudin-3 and claudin-4 expression in serous papillary, clear-cell, and endometrioid endometrial cancer. Gynecol Oncol. 2008;109:263–9.CrossRef Konecny GE, Agarwal R, Keeney GA, Winterhoff B, Jones MB, Mariani A, et al. Claudin-3 and claudin-4 expression in serous papillary, clear-cell, and endometrioid endometrial cancer. Gynecol Oncol. 2008;109:263–9.CrossRef
55.
go back to reference McDermott MS, Browne BC, O’Brien NA, Slamon DJ, Henry M, Meleady P, et al. PP2A-a Novel Target for the Treatment of Lapatinib-resistant Breast Cancer. Eur J Cancer. 2012;48:45.CrossRef McDermott MS, Browne BC, O’Brien NA, Slamon DJ, Henry M, Meleady P, et al. PP2A-a Novel Target for the Treatment of Lapatinib-resistant Breast Cancer. Eur J Cancer. 2012;48:45.CrossRef
Metadata
Title
Cleavage of the extracellular domain of junctional adhesion molecule-A is associated with resistance to anti-HER2 therapies in breast cancer settings
Authors
Astrid O. Leech
Sri HariKrishna Vellanki
Emily J. Rutherford
Aoife Keogh
Hanne Jahns
Lance Hudson
Norma O’Donovan
Siham Sabri
Bassam Abdulkarim
Katherine M. Sheehan
Elaine W. Kay
Leonie S. Young
Arnold D. K. Hill
Yvonne E. Smith
Ann M. Hopkins
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2018
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-018-1064-1

Other articles of this Issue 1/2018

Breast Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine