Cargando…

Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent

High levels of expression of glycoprotein non-metastatic B (gpNMB) in triple negative breast cancer (TNBC) and its association with metastasis and recurrence make it an attractive target for therapy with the antibody drug conjugate, glembatumumab vedotin (CDX-011). This report describes the developm...

Descripción completa

Detalles Bibliográficos
Autores principales: Marquez-Nostra, Bernadette V., Lee, Supum, Laforest, Richard, Vitale, Laura, Nie, Xingyu, Hyrc, Krzysztof, Keler, Tibor, Hawthorne, Thomas, Hoog, Jeremy, Li, Shunqiang, Dehdashti, Farrokh, Ma, Cynthia X., Lapi, Suzanne E.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5732808/
https://www.ncbi.nlm.nih.gov/pubmed/29262642
http://dx.doi.org/10.18632/oncotarget.22228
_version_ 1783286785493696512
author Marquez-Nostra, Bernadette V.
Lee, Supum
Laforest, Richard
Vitale, Laura
Nie, Xingyu
Hyrc, Krzysztof
Keler, Tibor
Hawthorne, Thomas
Hoog, Jeremy
Li, Shunqiang
Dehdashti, Farrokh
Ma, Cynthia X.
Lapi, Suzanne E.
author_facet Marquez-Nostra, Bernadette V.
Lee, Supum
Laforest, Richard
Vitale, Laura
Nie, Xingyu
Hyrc, Krzysztof
Keler, Tibor
Hawthorne, Thomas
Hoog, Jeremy
Li, Shunqiang
Dehdashti, Farrokh
Ma, Cynthia X.
Lapi, Suzanne E.
author_sort Marquez-Nostra, Bernadette V.
collection PubMed
description High levels of expression of glycoprotein non-metastatic B (gpNMB) in triple negative breast cancer (TNBC) and its association with metastasis and recurrence make it an attractive target for therapy with the antibody drug conjugate, glembatumumab vedotin (CDX-011). This report describes the development of a companion PET-based diagnostic imaging agent using (89)Zr-labeled glembatumumab ([(89)Zr]DFO-CR011) to potentially aid in the selection of patients most likely to respond to targeted treatment with CDX-011. [(89)Zr]DFO-CR011 was characterized for its pharmacologic properties in TNBC cell lines. Preclinical studies determined that [(89)Zr]DFO-CR011 binds specifically to gpNMB with high affinity (Kd = 25 ± 5 nM), immunoreactivity of 2.2-fold less than the native CR011, and its cellular uptake correlates with gpNMB expression (r = 0.95). In PET studies at the optimal imaging timepoint of 7 days p.i., the [(89)Zr]DFO-CR011 tumor uptake in gpNMB-expressing MDA-MB-468 xenografts had a mean SUV of 2.9, while significantly lower in gpNMB-negative MDA-MB-231 tumors with a mean SUV of 1.9. [(89)Zr]DFO-CR011 was also evaluated in patient-derived xenograft models of TNBC, where tumor uptake in vivo had a positive correlation with total gpNMB protein expression via ELISA (r = 0.79), despite the heterogeneity of gpNMB expression within the same group of PDX mice. Lastly, the radiation dosimetry calculated from biodistribution studies in MDA-MB-468 xenografts determined the effective dose for human use would be 0.54 mSv/MBq. Overall, these studies demonstrate that [(89)Zr]DFO-CR011 is a potential companion diagnostic imaging agent for CDX-011 which targets gpNMB, an emerging biomarker for TNBC.
format Online
Article
Text
id pubmed-5732808
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-57328082017-12-19 Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent Marquez-Nostra, Bernadette V. Lee, Supum Laforest, Richard Vitale, Laura Nie, Xingyu Hyrc, Krzysztof Keler, Tibor Hawthorne, Thomas Hoog, Jeremy Li, Shunqiang Dehdashti, Farrokh Ma, Cynthia X. Lapi, Suzanne E. Oncotarget Research Paper High levels of expression of glycoprotein non-metastatic B (gpNMB) in triple negative breast cancer (TNBC) and its association with metastasis and recurrence make it an attractive target for therapy with the antibody drug conjugate, glembatumumab vedotin (CDX-011). This report describes the development of a companion PET-based diagnostic imaging agent using (89)Zr-labeled glembatumumab ([(89)Zr]DFO-CR011) to potentially aid in the selection of patients most likely to respond to targeted treatment with CDX-011. [(89)Zr]DFO-CR011 was characterized for its pharmacologic properties in TNBC cell lines. Preclinical studies determined that [(89)Zr]DFO-CR011 binds specifically to gpNMB with high affinity (Kd = 25 ± 5 nM), immunoreactivity of 2.2-fold less than the native CR011, and its cellular uptake correlates with gpNMB expression (r = 0.95). In PET studies at the optimal imaging timepoint of 7 days p.i., the [(89)Zr]DFO-CR011 tumor uptake in gpNMB-expressing MDA-MB-468 xenografts had a mean SUV of 2.9, while significantly lower in gpNMB-negative MDA-MB-231 tumors with a mean SUV of 1.9. [(89)Zr]DFO-CR011 was also evaluated in patient-derived xenograft models of TNBC, where tumor uptake in vivo had a positive correlation with total gpNMB protein expression via ELISA (r = 0.79), despite the heterogeneity of gpNMB expression within the same group of PDX mice. Lastly, the radiation dosimetry calculated from biodistribution studies in MDA-MB-468 xenografts determined the effective dose for human use would be 0.54 mSv/MBq. Overall, these studies demonstrate that [(89)Zr]DFO-CR011 is a potential companion diagnostic imaging agent for CDX-011 which targets gpNMB, an emerging biomarker for TNBC. Impact Journals LLC 2017-11-01 /pmc/articles/PMC5732808/ /pubmed/29262642 http://dx.doi.org/10.18632/oncotarget.22228 Text en Copyright: © 2017 Marquez-Nostra et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0/) 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Marquez-Nostra, Bernadette V.
Lee, Supum
Laforest, Richard
Vitale, Laura
Nie, Xingyu
Hyrc, Krzysztof
Keler, Tibor
Hawthorne, Thomas
Hoog, Jeremy
Li, Shunqiang
Dehdashti, Farrokh
Ma, Cynthia X.
Lapi, Suzanne E.
Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent
title Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent
title_full Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent
title_fullStr Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent
title_full_unstemmed Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent
title_short Preclinical PET imaging of glycoprotein non-metastatic melanoma B in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent
title_sort preclinical pet imaging of glycoprotein non-metastatic melanoma b in triple negative breast cancer: feasibility of an antibody-based companion diagnostic agent
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5732808/
https://www.ncbi.nlm.nih.gov/pubmed/29262642
http://dx.doi.org/10.18632/oncotarget.22228
work_keys_str_mv AT marqueznostrabernadettev preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT leesupum preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT laforestrichard preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT vitalelaura preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT niexingyu preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT hyrckrzysztof preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT kelertibor preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT hawthornethomas preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT hoogjeremy preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT lishunqiang preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT dehdashtifarrokh preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT macynthiax preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent
AT lapisuzannee preclinicalpetimagingofglycoproteinnonmetastaticmelanomabintriplenegativebreastcancerfeasibilityofanantibodybasedcompaniondiagnosticagent