author_facet Eigenmann, Miro J.
Frances, Nicolas
Hoffmann, Gerhard
Lavé, Thierry
Walz, Antje-Christine
Eigenmann, Miro J.
Frances, Nicolas
Hoffmann, Gerhard
Lavé, Thierry
Walz, Antje-Christine
author Eigenmann, Miro J.
Frances, Nicolas
Hoffmann, Gerhard
Lavé, Thierry
Walz, Antje-Christine
spellingShingle Eigenmann, Miro J.
Frances, Nicolas
Hoffmann, Gerhard
Lavé, Thierry
Walz, Antje-Christine
Molecular Cancer Therapeutics
Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
Cancer Research
Oncology
author_sort eigenmann, miro j.
spelling Eigenmann, Miro J. Frances, Nicolas Hoffmann, Gerhard Lavé, Thierry Walz, Antje-Christine 1535-7163 1538-8514 American Association for Cancer Research (AACR) Cancer Research Oncology http://dx.doi.org/10.1158/1535-7163.mct-16-0076 <jats:title>Abstract</jats:title> <jats:p>We quantitatively compare the efficacy of two approved EGFR tyrosine kinase inhibitors, erlotinib and gefitinib, based on in vivo and in vitro data and show how a modeling approach can be used to scale from animal to humans. Gefitinib shows a higher tumor uptake in cancer patients, and we explored the potential impact on pharmacologic and antitumor activity in in vitro and in xenograft mice. Tumor growth inhibition was monitored, and the pharmacokinetics (PK) in plasma and tumor, as well as temporal changes of phospho-Erk (pErk) signals were examined in patient-derived tumor xenograft mice. These data were integrated in a translational PKPD model, allowing us to project an efficacious human dose, which we retrospectively compared with prescribed doses for cancer patients. In vitro experiments showed that cell-cycle arrest was similar for erlotinib and gefitinib. Similar pERK biomarker responses were obtained despite a 6.6-fold higher total tumor exposure for gefitinib. The PKPD model revealed a 3.7-fold higher in vivo potency for gefitinib, which did not translate into a lower anticipated efficacious dose in humans. The model-based dose prediction matched the recommended clinical doses well. These results suggest that despite having lower total tumor-to-plasma ratios, active drug exposure at target site is higher for erlotinib. Considering the PK properties, this translates in a 50% lower recommended daily dose of erlotinib in cancer patients. In summary, total exposure at target site is not suitable to rank compounds, and an integrated modeling and experimental approach can assess efficacy more accurately. Mol Cancer Ther; 15(12); 3110–9. ©2016 AACR.</jats:p> Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib Molecular Cancer Therapeutics
doi_str_mv 10.1158/1535-7163.mct-16-0076
facet_avail Online
Free
finc_class_facet Medizin
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xNTM1LTcxNjMubWN0LTE2LTAwNzY
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xNTM1LTcxNjMubWN0LTE2LTAwNzY
institution DE-Brt1
DE-Zwi2
DE-D161
DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
DE-L229
DE-D275
DE-Bn3
imprint American Association for Cancer Research (AACR), 2016
imprint_str_mv American Association for Cancer Research (AACR), 2016
issn 1535-7163
1538-8514
issn_str_mv 1535-7163
1538-8514
language English
mega_collection American Association for Cancer Research (AACR) (CrossRef)
match_str eigenmann2016combiningnonclinicalexperimentswithtranslationalpkpdmodelingtodifferentiateerlotinibandgefitinib
publishDateSort 2016
publisher American Association for Cancer Research (AACR)
recordtype ai
record_format ai
series Molecular Cancer Therapeutics
source_id 49
title Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_unstemmed Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_full Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_fullStr Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_full_unstemmed Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_short Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_sort combining nonclinical experiments with translational pkpd modeling to differentiate erlotinib and gefitinib
topic Cancer Research
Oncology
url http://dx.doi.org/10.1158/1535-7163.mct-16-0076
publishDate 2016
physical 3110-3119
description <jats:title>Abstract</jats:title> <jats:p>We quantitatively compare the efficacy of two approved EGFR tyrosine kinase inhibitors, erlotinib and gefitinib, based on in vivo and in vitro data and show how a modeling approach can be used to scale from animal to humans. Gefitinib shows a higher tumor uptake in cancer patients, and we explored the potential impact on pharmacologic and antitumor activity in in vitro and in xenograft mice. Tumor growth inhibition was monitored, and the pharmacokinetics (PK) in plasma and tumor, as well as temporal changes of phospho-Erk (pErk) signals were examined in patient-derived tumor xenograft mice. These data were integrated in a translational PKPD model, allowing us to project an efficacious human dose, which we retrospectively compared with prescribed doses for cancer patients. In vitro experiments showed that cell-cycle arrest was similar for erlotinib and gefitinib. Similar pERK biomarker responses were obtained despite a 6.6-fold higher total tumor exposure for gefitinib. The PKPD model revealed a 3.7-fold higher in vivo potency for gefitinib, which did not translate into a lower anticipated efficacious dose in humans. The model-based dose prediction matched the recommended clinical doses well. These results suggest that despite having lower total tumor-to-plasma ratios, active drug exposure at target site is higher for erlotinib. Considering the PK properties, this translates in a 50% lower recommended daily dose of erlotinib in cancer patients. In summary, total exposure at target site is not suitable to rank compounds, and an integrated modeling and experimental approach can assess efficacy more accurately. Mol Cancer Ther; 15(12); 3110–9. ©2016 AACR.</jats:p>
container_issue 12
container_start_page 3110
container_title Molecular Cancer Therapeutics
container_volume 15
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_ 1792348045675855872
geogr_code not assigned
last_indexed 2024-03-01T18:04:57.01Z
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=Combining+Nonclinical+Experiments+with+Translational+PKPD+Modeling+to+Differentiate+Erlotinib+and+Gefitinib&rft.date=2016-12-01&genre=article&issn=1538-8514&volume=15&issue=12&spage=3110&epage=3119&pages=3110-3119&jtitle=Molecular+Cancer+Therapeutics&atitle=Combining+Nonclinical+Experiments+with+Translational+PKPD+Modeling+to+Differentiate+Erlotinib+and+Gefitinib&aulast=Walz&aufirst=Antje-Christine&rft_id=info%3Adoi%2F10.1158%2F1535-7163.mct-16-0076&rft.language%5B0%5D=eng
SOLR
_version_ 1792348045675855872
author Eigenmann, Miro J., Frances, Nicolas, Hoffmann, Gerhard, Lavé, Thierry, Walz, Antje-Christine
author_facet Eigenmann, Miro J., Frances, Nicolas, Hoffmann, Gerhard, Lavé, Thierry, Walz, Antje-Christine, Eigenmann, Miro J., Frances, Nicolas, Hoffmann, Gerhard, Lavé, Thierry, Walz, Antje-Christine
author_sort eigenmann, miro j.
container_issue 12
container_start_page 3110
container_title Molecular Cancer Therapeutics
container_volume 15
description <jats:title>Abstract</jats:title> <jats:p>We quantitatively compare the efficacy of two approved EGFR tyrosine kinase inhibitors, erlotinib and gefitinib, based on in vivo and in vitro data and show how a modeling approach can be used to scale from animal to humans. Gefitinib shows a higher tumor uptake in cancer patients, and we explored the potential impact on pharmacologic and antitumor activity in in vitro and in xenograft mice. Tumor growth inhibition was monitored, and the pharmacokinetics (PK) in plasma and tumor, as well as temporal changes of phospho-Erk (pErk) signals were examined in patient-derived tumor xenograft mice. These data were integrated in a translational PKPD model, allowing us to project an efficacious human dose, which we retrospectively compared with prescribed doses for cancer patients. In vitro experiments showed that cell-cycle arrest was similar for erlotinib and gefitinib. Similar pERK biomarker responses were obtained despite a 6.6-fold higher total tumor exposure for gefitinib. The PKPD model revealed a 3.7-fold higher in vivo potency for gefitinib, which did not translate into a lower anticipated efficacious dose in humans. The model-based dose prediction matched the recommended clinical doses well. These results suggest that despite having lower total tumor-to-plasma ratios, active drug exposure at target site is higher for erlotinib. Considering the PK properties, this translates in a 50% lower recommended daily dose of erlotinib in cancer patients. In summary, total exposure at target site is not suitable to rank compounds, and an integrated modeling and experimental approach can assess efficacy more accurately. Mol Cancer Ther; 15(12); 3110–9. ©2016 AACR.</jats:p>
doi_str_mv 10.1158/1535-7163.mct-16-0076
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-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE1OC8xNTM1LTcxNjMubWN0LTE2LTAwNzY
imprint American Association for Cancer Research (AACR), 2016
imprint_str_mv American Association for Cancer Research (AACR), 2016
institution DE-Brt1, DE-Zwi2, DE-D161, DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1, DE-L229, DE-D275, DE-Bn3
issn 1535-7163, 1538-8514
issn_str_mv 1535-7163, 1538-8514
language English
last_indexed 2024-03-01T18:04:57.01Z
match_str eigenmann2016combiningnonclinicalexperimentswithtranslationalpkpdmodelingtodifferentiateerlotinibandgefitinib
mega_collection American Association for Cancer Research (AACR) (CrossRef)
physical 3110-3119
publishDate 2016
publishDateSort 2016
publisher American Association for Cancer Research (AACR)
record_format ai
recordtype ai
series Molecular Cancer Therapeutics
source_id 49
spelling Eigenmann, Miro J. Frances, Nicolas Hoffmann, Gerhard Lavé, Thierry Walz, Antje-Christine 1535-7163 1538-8514 American Association for Cancer Research (AACR) Cancer Research Oncology http://dx.doi.org/10.1158/1535-7163.mct-16-0076 <jats:title>Abstract</jats:title> <jats:p>We quantitatively compare the efficacy of two approved EGFR tyrosine kinase inhibitors, erlotinib and gefitinib, based on in vivo and in vitro data and show how a modeling approach can be used to scale from animal to humans. Gefitinib shows a higher tumor uptake in cancer patients, and we explored the potential impact on pharmacologic and antitumor activity in in vitro and in xenograft mice. Tumor growth inhibition was monitored, and the pharmacokinetics (PK) in plasma and tumor, as well as temporal changes of phospho-Erk (pErk) signals were examined in patient-derived tumor xenograft mice. These data were integrated in a translational PKPD model, allowing us to project an efficacious human dose, which we retrospectively compared with prescribed doses for cancer patients. In vitro experiments showed that cell-cycle arrest was similar for erlotinib and gefitinib. Similar pERK biomarker responses were obtained despite a 6.6-fold higher total tumor exposure for gefitinib. The PKPD model revealed a 3.7-fold higher in vivo potency for gefitinib, which did not translate into a lower anticipated efficacious dose in humans. The model-based dose prediction matched the recommended clinical doses well. These results suggest that despite having lower total tumor-to-plasma ratios, active drug exposure at target site is higher for erlotinib. Considering the PK properties, this translates in a 50% lower recommended daily dose of erlotinib in cancer patients. In summary, total exposure at target site is not suitable to rank compounds, and an integrated modeling and experimental approach can assess efficacy more accurately. Mol Cancer Ther; 15(12); 3110–9. ©2016 AACR.</jats:p> Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib Molecular Cancer Therapeutics
spellingShingle Eigenmann, Miro J., Frances, Nicolas, Hoffmann, Gerhard, Lavé, Thierry, Walz, Antje-Christine, Molecular Cancer Therapeutics, Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib, Cancer Research, Oncology
title Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_full Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_fullStr Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_full_unstemmed Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_short Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
title_sort combining nonclinical experiments with translational pkpd modeling to differentiate erlotinib and gefitinib
title_unstemmed Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib
topic Cancer Research, Oncology
url http://dx.doi.org/10.1158/1535-7163.mct-16-0076