author_facet Croessmann, Sarah
Formisano, Luigi
Kinch, Lisa N.
Gonzalez-Ericsson, Paula I.
Sudhan, Dhivya R.
Nagy, Rebecca J.
Mathew, Aju
Bernicker, Eric H.
Cristofanilli, Massimo
He, Jie
Cutler, Richard E.
Lalani, Alshad S.
Miller, Vincent A.
Lanman, Richard B.
Grishin, Nick V.
Arteaga, Carlos L.
Croessmann, Sarah
Formisano, Luigi
Kinch, Lisa N.
Gonzalez-Ericsson, Paula I.
Sudhan, Dhivya R.
Nagy, Rebecca J.
Mathew, Aju
Bernicker, Eric H.
Cristofanilli, Massimo
He, Jie
Cutler, Richard E.
Lalani, Alshad S.
Miller, Vincent A.
Lanman, Richard B.
Grishin, Nick V.
Arteaga, Carlos L.
author Croessmann, Sarah
Formisano, Luigi
Kinch, Lisa N.
Gonzalez-Ericsson, Paula I.
Sudhan, Dhivya R.
Nagy, Rebecca J.
Mathew, Aju
Bernicker, Eric H.
Cristofanilli, Massimo
He, Jie
Cutler, Richard E.
Lalani, Alshad S.
Miller, Vincent A.
Lanman, Richard B.
Grishin, Nick V.
Arteaga, Carlos L.
spellingShingle Croessmann, Sarah
Formisano, Luigi
Kinch, Lisa N.
Gonzalez-Ericsson, Paula I.
Sudhan, Dhivya R.
Nagy, Rebecca J.
Mathew, Aju
Bernicker, Eric H.
Cristofanilli, Massimo
He, Jie
Cutler, Richard E.
Lalani, Alshad S.
Miller, Vincent A.
Lanman, Richard B.
Grishin, Nick V.
Arteaga, Carlos L.
Clinical Cancer Research
Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
Cancer Research
Oncology
author_sort croessmann, sarah
spelling Croessmann, Sarah Formisano, Luigi Kinch, Lisa N. Gonzalez-Ericsson, Paula I. Sudhan, Dhivya R. Nagy, Rebecca J. Mathew, Aju Bernicker, Eric H. Cristofanilli, Massimo He, Jie Cutler, Richard E. Lalani, Alshad S. Miller, Vincent A. Lanman, Richard B. Grishin, Nick V. Arteaga, Carlos L. 1078-0432 1557-3265 American Association for Cancer Research (AACR) Cancer Research Oncology http://dx.doi.org/10.1158/1078-0432.ccr-18-1544 <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>We examined the role of ERBB2-activating mutations in endocrine therapy resistance in estrogen receptor positive (ER+) breast cancer.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>ERBB2 mutation frequency was determined from large genomic databases. Isogenic knock-in ERBB2 mutations in ER+ MCF7 cells and xenografts were used to investigate estrogen-independent growth. Structural analysis was used to determine the molecular interaction of HERL755S with HER3. Small molecules and siRNAs were used to inhibit PI3Kα, TORC1, and HER3.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>Genomic data revealed a higher rate of ERBB2 mutations in metastatic versus primary ER+ tumors. MCF7 cells with isogenically incorporated ERBB2 kinase domain mutations exhibited resistance to estrogen deprivation and to fulvestrant both in vitro and in vivo, despite maintaining inhibition of ERα transcriptional activity. Addition of the irreversible HER2 tyrosine kinase inhibitor neratinib restored sensitivity to fulvestrant. HER2-mutant MCF7 cells expressed higher levels of p-HER3, p-AKT, and p-S6 than cells with wild-type HER2. Structural analysis of the HER2L755S variant implicated a more flexible active state, potentially allowing for enhanced dimerization with HER3. Treatment with a PI3Kα inhibitor, a TORC1 inhibitor or HER3 siRNA, but not a MEK inhibitor, restored sensitivity to fulvestrant and to estrogen deprivation. Inhibition of mutant HER2 or TORC1, when combined with fulvestrant, equipotently inhibited growth of MCF7/ERBB2V777L xenografts, suggesting a role for TORC1 in antiestrogen resistance induced by ERBB2 mutations.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>ERBB2 mutations hyperactivate the HER3/PI3K/AKT/mTOR axis, leading to antiestrogen resistance in ER+ breast cancer. Dual blockade of the HER2 and ER pathways is required for the treatment of ER+/HER2 mutant breast cancers.</jats:p> </jats:sec> Combined Blockade of Activating <i>ERBB2</i> Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer Clinical Cancer Research
doi_str_mv 10.1158/1078-0432.ccr-18-1544
facet_avail Online
Free
finc_class_facet Medizin
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xMDc4LTA0MzIuY2NyLTE4LTE1NDQ
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xMDc4LTA0MzIuY2NyLTE4LTE1NDQ
institution DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
DE-L229
imprint American Association for Cancer Research (AACR), 2019
imprint_str_mv American Association for Cancer Research (AACR), 2019
issn 1078-0432
1557-3265
issn_str_mv 1078-0432
1557-3265
language English
mega_collection American Association for Cancer Research (AACR) (CrossRef)
match_str croessmann2019combinedblockadeofactivatingerbb2mutationsanderresultsinsyntheticlethalityoferher2mutantbreastcancer
publishDateSort 2019
publisher American Association for Cancer Research (AACR)
recordtype ai
record_format ai
series Clinical Cancer Research
source_id 49
title Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_unstemmed Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_full Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_fullStr Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_full_unstemmed Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_short Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_sort combined blockade of activating <i>erbb2</i> mutations and er results in synthetic lethality of er+/her2 mutant breast cancer
topic Cancer Research
Oncology
url http://dx.doi.org/10.1158/1078-0432.ccr-18-1544
publishDate 2019
physical 277-289
description <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>We examined the role of ERBB2-activating mutations in endocrine therapy resistance in estrogen receptor positive (ER+) breast cancer.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>ERBB2 mutation frequency was determined from large genomic databases. Isogenic knock-in ERBB2 mutations in ER+ MCF7 cells and xenografts were used to investigate estrogen-independent growth. Structural analysis was used to determine the molecular interaction of HERL755S with HER3. Small molecules and siRNAs were used to inhibit PI3Kα, TORC1, and HER3.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>Genomic data revealed a higher rate of ERBB2 mutations in metastatic versus primary ER+ tumors. MCF7 cells with isogenically incorporated ERBB2 kinase domain mutations exhibited resistance to estrogen deprivation and to fulvestrant both in vitro and in vivo, despite maintaining inhibition of ERα transcriptional activity. Addition of the irreversible HER2 tyrosine kinase inhibitor neratinib restored sensitivity to fulvestrant. HER2-mutant MCF7 cells expressed higher levels of p-HER3, p-AKT, and p-S6 than cells with wild-type HER2. Structural analysis of the HER2L755S variant implicated a more flexible active state, potentially allowing for enhanced dimerization with HER3. Treatment with a PI3Kα inhibitor, a TORC1 inhibitor or HER3 siRNA, but not a MEK inhibitor, restored sensitivity to fulvestrant and to estrogen deprivation. Inhibition of mutant HER2 or TORC1, when combined with fulvestrant, equipotently inhibited growth of MCF7/ERBB2V777L xenografts, suggesting a role for TORC1 in antiestrogen resistance induced by ERBB2 mutations.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>ERBB2 mutations hyperactivate the HER3/PI3K/AKT/mTOR axis, leading to antiestrogen resistance in ER+ breast cancer. Dual blockade of the HER2 and ER pathways is required for the treatment of ER+/HER2 mutant breast cancers.</jats:p> </jats:sec>
container_issue 1
container_start_page 277
container_title Clinical Cancer Research
container_volume 25
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792347838554832897
geogr_code not assigned
last_indexed 2024-03-01T18:01:39.367Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=Combined+Blockade+of+Activating+ERBB2+Mutations+and+ER+Results+in+Synthetic+Lethality+of+ER%2B%2FHER2+Mutant+Breast+Cancer&rft.date=2019-01-01&genre=article&issn=1557-3265&volume=25&issue=1&spage=277&epage=289&pages=277-289&jtitle=Clinical+Cancer+Research&atitle=Combined+Blockade+of+Activating+%3Ci%3EERBB2%3C%2Fi%3E+Mutations+and+ER+Results+in+Synthetic+Lethality+of+ER%2B%2FHER2+Mutant+Breast+Cancer&aulast=Arteaga&aufirst=Carlos+L.&rft_id=info%3Adoi%2F10.1158%2F1078-0432.ccr-18-1544&rft.language%5B0%5D=eng
SOLR
_version_ 1792347838554832897
author Croessmann, Sarah, Formisano, Luigi, Kinch, Lisa N., Gonzalez-Ericsson, Paula I., Sudhan, Dhivya R., Nagy, Rebecca J., Mathew, Aju, Bernicker, Eric H., Cristofanilli, Massimo, He, Jie, Cutler, Richard E., Lalani, Alshad S., Miller, Vincent A., Lanman, Richard B., Grishin, Nick V., Arteaga, Carlos L.
author_facet Croessmann, Sarah, Formisano, Luigi, Kinch, Lisa N., Gonzalez-Ericsson, Paula I., Sudhan, Dhivya R., Nagy, Rebecca J., Mathew, Aju, Bernicker, Eric H., Cristofanilli, Massimo, He, Jie, Cutler, Richard E., Lalani, Alshad S., Miller, Vincent A., Lanman, Richard B., Grishin, Nick V., Arteaga, Carlos L., Croessmann, Sarah, Formisano, Luigi, Kinch, Lisa N., Gonzalez-Ericsson, Paula I., Sudhan, Dhivya R., Nagy, Rebecca J., Mathew, Aju, Bernicker, Eric H., Cristofanilli, Massimo, He, Jie, Cutler, Richard E., Lalani, Alshad S., Miller, Vincent A., Lanman, Richard B., Grishin, Nick V., Arteaga, Carlos L.
author_sort croessmann, sarah
container_issue 1
container_start_page 277
container_title Clinical Cancer Research
container_volume 25
description <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>We examined the role of ERBB2-activating mutations in endocrine therapy resistance in estrogen receptor positive (ER+) breast cancer.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>ERBB2 mutation frequency was determined from large genomic databases. Isogenic knock-in ERBB2 mutations in ER+ MCF7 cells and xenografts were used to investigate estrogen-independent growth. Structural analysis was used to determine the molecular interaction of HERL755S with HER3. Small molecules and siRNAs were used to inhibit PI3Kα, TORC1, and HER3.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>Genomic data revealed a higher rate of ERBB2 mutations in metastatic versus primary ER+ tumors. MCF7 cells with isogenically incorporated ERBB2 kinase domain mutations exhibited resistance to estrogen deprivation and to fulvestrant both in vitro and in vivo, despite maintaining inhibition of ERα transcriptional activity. Addition of the irreversible HER2 tyrosine kinase inhibitor neratinib restored sensitivity to fulvestrant. HER2-mutant MCF7 cells expressed higher levels of p-HER3, p-AKT, and p-S6 than cells with wild-type HER2. Structural analysis of the HER2L755S variant implicated a more flexible active state, potentially allowing for enhanced dimerization with HER3. Treatment with a PI3Kα inhibitor, a TORC1 inhibitor or HER3 siRNA, but not a MEK inhibitor, restored sensitivity to fulvestrant and to estrogen deprivation. Inhibition of mutant HER2 or TORC1, when combined with fulvestrant, equipotently inhibited growth of MCF7/ERBB2V777L xenografts, suggesting a role for TORC1 in antiestrogen resistance induced by ERBB2 mutations.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>ERBB2 mutations hyperactivate the HER3/PI3K/AKT/mTOR axis, leading to antiestrogen resistance in ER+ breast cancer. Dual blockade of the HER2 and ER pathways is required for the treatment of ER+/HER2 mutant breast cancers.</jats:p> </jats:sec>
doi_str_mv 10.1158/1078-0432.ccr-18-1544
facet_avail Online, Free
finc_class_facet Medizin
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xMDc4LTA0MzIuY2NyLTE4LTE1NDQ
imprint American Association for Cancer Research (AACR), 2019
imprint_str_mv American Association for Cancer Research (AACR), 2019
institution DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161, DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1, DE-L229
issn 1078-0432, 1557-3265
issn_str_mv 1078-0432, 1557-3265
language English
last_indexed 2024-03-01T18:01:39.367Z
match_str croessmann2019combinedblockadeofactivatingerbb2mutationsanderresultsinsyntheticlethalityoferher2mutantbreastcancer
mega_collection American Association for Cancer Research (AACR) (CrossRef)
physical 277-289
publishDate 2019
publishDateSort 2019
publisher American Association for Cancer Research (AACR)
record_format ai
recordtype ai
series Clinical Cancer Research
source_id 49
spelling Croessmann, Sarah Formisano, Luigi Kinch, Lisa N. Gonzalez-Ericsson, Paula I. Sudhan, Dhivya R. Nagy, Rebecca J. Mathew, Aju Bernicker, Eric H. Cristofanilli, Massimo He, Jie Cutler, Richard E. Lalani, Alshad S. Miller, Vincent A. Lanman, Richard B. Grishin, Nick V. Arteaga, Carlos L. 1078-0432 1557-3265 American Association for Cancer Research (AACR) Cancer Research Oncology http://dx.doi.org/10.1158/1078-0432.ccr-18-1544 <jats:title>Abstract</jats:title> <jats:sec> <jats:title>Purpose:</jats:title> <jats:p>We examined the role of ERBB2-activating mutations in endocrine therapy resistance in estrogen receptor positive (ER+) breast cancer.</jats:p> </jats:sec> <jats:sec> <jats:title>Experimental Design:</jats:title> <jats:p>ERBB2 mutation frequency was determined from large genomic databases. Isogenic knock-in ERBB2 mutations in ER+ MCF7 cells and xenografts were used to investigate estrogen-independent growth. Structural analysis was used to determine the molecular interaction of HERL755S with HER3. Small molecules and siRNAs were used to inhibit PI3Kα, TORC1, and HER3.</jats:p> </jats:sec> <jats:sec> <jats:title>Results:</jats:title> <jats:p>Genomic data revealed a higher rate of ERBB2 mutations in metastatic versus primary ER+ tumors. MCF7 cells with isogenically incorporated ERBB2 kinase domain mutations exhibited resistance to estrogen deprivation and to fulvestrant both in vitro and in vivo, despite maintaining inhibition of ERα transcriptional activity. Addition of the irreversible HER2 tyrosine kinase inhibitor neratinib restored sensitivity to fulvestrant. HER2-mutant MCF7 cells expressed higher levels of p-HER3, p-AKT, and p-S6 than cells with wild-type HER2. Structural analysis of the HER2L755S variant implicated a more flexible active state, potentially allowing for enhanced dimerization with HER3. Treatment with a PI3Kα inhibitor, a TORC1 inhibitor or HER3 siRNA, but not a MEK inhibitor, restored sensitivity to fulvestrant and to estrogen deprivation. Inhibition of mutant HER2 or TORC1, when combined with fulvestrant, equipotently inhibited growth of MCF7/ERBB2V777L xenografts, suggesting a role for TORC1 in antiestrogen resistance induced by ERBB2 mutations.</jats:p> </jats:sec> <jats:sec> <jats:title>Conclusions:</jats:title> <jats:p>ERBB2 mutations hyperactivate the HER3/PI3K/AKT/mTOR axis, leading to antiestrogen resistance in ER+ breast cancer. Dual blockade of the HER2 and ER pathways is required for the treatment of ER+/HER2 mutant breast cancers.</jats:p> </jats:sec> Combined Blockade of Activating <i>ERBB2</i> Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer Clinical Cancer Research
spellingShingle Croessmann, Sarah, Formisano, Luigi, Kinch, Lisa N., Gonzalez-Ericsson, Paula I., Sudhan, Dhivya R., Nagy, Rebecca J., Mathew, Aju, Bernicker, Eric H., Cristofanilli, Massimo, He, Jie, Cutler, Richard E., Lalani, Alshad S., Miller, Vincent A., Lanman, Richard B., Grishin, Nick V., Arteaga, Carlos L., Clinical Cancer Research, Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer, Cancer Research, Oncology
title Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_full Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_fullStr Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_full_unstemmed Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_short Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
title_sort combined blockade of activating <i>erbb2</i> mutations and er results in synthetic lethality of er+/her2 mutant breast cancer
title_unstemmed Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer
topic Cancer Research, Oncology
url http://dx.doi.org/10.1158/1078-0432.ccr-18-1544