Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.

<h4>Background</h4>Molecularly-guided trials (i.e. PMed) now seek to aid clinical decision-making by matching cancer targets with therapeutic options. Progress has been hampered by the lack of cancer models that account for individual-to-individual heterogeneity within and across cancer...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Melissa Paoloni, Craig Webb, Christina Mazcko, David Cherba, William Hendricks, Susan Lana, E J Ehrhart, Brad Charles, Heather Fehling, Leena Kumar, David Vail, Michael Henson, Michael Childress, Barbara Kitchell, Christopher Kingsley, Seungchan Kim, Mark Neff, Barbara Davis, Chand Khanna, Jeffrey Trent
Formato: article
Lenguaje:EN
Publicado: Public Library of Science (PLoS) 2014
Materias:
R
Q
Acceso en línea:https://doaj.org/article/55faf0c024234b4cb92c74818bed7656
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:55faf0c024234b4cb92c74818bed7656
record_format dspace
spelling oai:doaj.org-article:55faf0c024234b4cb92c74818bed76562021-11-18T08:27:51ZProspective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.1932-620310.1371/journal.pone.0090028https://doaj.org/article/55faf0c024234b4cb92c74818bed76562014-01-01T00:00:00Zhttps://www.ncbi.nlm.nih.gov/pmc/articles/pmid/24637659/?tool=EBIhttps://doaj.org/toc/1932-6203<h4>Background</h4>Molecularly-guided trials (i.e. PMed) now seek to aid clinical decision-making by matching cancer targets with therapeutic options. Progress has been hampered by the lack of cancer models that account for individual-to-individual heterogeneity within and across cancer types. Naturally occurring cancers in pet animals are heterogeneous and thus provide an opportunity to answer questions about these PMed strategies and optimize translation to human patients. In order to realize this opportunity, it is now necessary to demonstrate the feasibility of conducting molecularly-guided analysis of tumors from dogs with naturally occurring cancer in a clinically relevant setting.<h4>Methodology</h4>A proof-of-concept study was conducted by the Comparative Oncology Trials Consortium (COTC) to determine if tumor collection, prospective molecular profiling, and PMed report generation within 1 week was feasible in dogs. Thirty-one dogs with cancers of varying histologies were enrolled. Twenty-four of 31 samples (77%) successfully met all predefined QA/QC criteria and were analyzed via Affymetrix gene expression profiling. A subsequent bioinformatics workflow transformed genomic data into a personalized drug report. Average turnaround from biopsy to report generation was 116 hours (4.8 days). Unsupervised clustering of canine tumor expression data clustered by cancer type, but supervised clustering of tumors based on the personalized drug report clustered by drug class rather than cancer type.<h4>Conclusions</h4>Collection and turnaround of high quality canine tumor samples, centralized pathology, analyte generation, array hybridization, and bioinformatic analyses matching gene expression to therapeutic options is achievable in a practical clinical window (<1 week). Clustering data show robust signatures by cancer type but also showed patient-to-patient heterogeneity in drug predictions. This lends further support to the inclusion of a heterogeneous population of dogs with cancer into the preclinical modeling of personalized medicine. Future comparative oncology studies optimizing the delivery of PMed strategies may aid cancer drug development.Melissa PaoloniCraig WebbChristina MazckoDavid CherbaWilliam HendricksSusan LanaE J EhrhartBrad CharlesHeather FehlingLeena KumarDavid VailMichael HensonMichael ChildressBarbara KitchellChristopher KingsleySeungchan KimMark NeffBarbara DavisChand KhannaJeffrey TrentPublic Library of Science (PLoS)articleMedicineRScienceQENPLoS ONE, Vol 9, Iss 3, p e90028 (2014)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
Melissa Paoloni
Craig Webb
Christina Mazcko
David Cherba
William Hendricks
Susan Lana
E J Ehrhart
Brad Charles
Heather Fehling
Leena Kumar
David Vail
Michael Henson
Michael Childress
Barbara Kitchell
Christopher Kingsley
Seungchan Kim
Mark Neff
Barbara Davis
Chand Khanna
Jeffrey Trent
Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.
description <h4>Background</h4>Molecularly-guided trials (i.e. PMed) now seek to aid clinical decision-making by matching cancer targets with therapeutic options. Progress has been hampered by the lack of cancer models that account for individual-to-individual heterogeneity within and across cancer types. Naturally occurring cancers in pet animals are heterogeneous and thus provide an opportunity to answer questions about these PMed strategies and optimize translation to human patients. In order to realize this opportunity, it is now necessary to demonstrate the feasibility of conducting molecularly-guided analysis of tumors from dogs with naturally occurring cancer in a clinically relevant setting.<h4>Methodology</h4>A proof-of-concept study was conducted by the Comparative Oncology Trials Consortium (COTC) to determine if tumor collection, prospective molecular profiling, and PMed report generation within 1 week was feasible in dogs. Thirty-one dogs with cancers of varying histologies were enrolled. Twenty-four of 31 samples (77%) successfully met all predefined QA/QC criteria and were analyzed via Affymetrix gene expression profiling. A subsequent bioinformatics workflow transformed genomic data into a personalized drug report. Average turnaround from biopsy to report generation was 116 hours (4.8 days). Unsupervised clustering of canine tumor expression data clustered by cancer type, but supervised clustering of tumors based on the personalized drug report clustered by drug class rather than cancer type.<h4>Conclusions</h4>Collection and turnaround of high quality canine tumor samples, centralized pathology, analyte generation, array hybridization, and bioinformatic analyses matching gene expression to therapeutic options is achievable in a practical clinical window (<1 week). Clustering data show robust signatures by cancer type but also showed patient-to-patient heterogeneity in drug predictions. This lends further support to the inclusion of a heterogeneous population of dogs with cancer into the preclinical modeling of personalized medicine. Future comparative oncology studies optimizing the delivery of PMed strategies may aid cancer drug development.
format article
author Melissa Paoloni
Craig Webb
Christina Mazcko
David Cherba
William Hendricks
Susan Lana
E J Ehrhart
Brad Charles
Heather Fehling
Leena Kumar
David Vail
Michael Henson
Michael Childress
Barbara Kitchell
Christopher Kingsley
Seungchan Kim
Mark Neff
Barbara Davis
Chand Khanna
Jeffrey Trent
author_facet Melissa Paoloni
Craig Webb
Christina Mazcko
David Cherba
William Hendricks
Susan Lana
E J Ehrhart
Brad Charles
Heather Fehling
Leena Kumar
David Vail
Michael Henson
Michael Childress
Barbara Kitchell
Christopher Kingsley
Seungchan Kim
Mark Neff
Barbara Davis
Chand Khanna
Jeffrey Trent
author_sort Melissa Paoloni
title Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.
title_short Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.
title_full Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.
title_fullStr Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.
title_full_unstemmed Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.
title_sort prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (pmed) trials.
publisher Public Library of Science (PLoS)
publishDate 2014
url https://doaj.org/article/55faf0c024234b4cb92c74818bed7656
work_keys_str_mv AT melissapaoloni prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT craigwebb prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT christinamazcko prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT davidcherba prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT williamhendricks prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT susanlana prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT ejehrhart prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT bradcharles prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT heatherfehling prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT leenakumar prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT davidvail prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT michaelhenson prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT michaelchildress prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT barbarakitchell prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT christopherkingsley prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT seungchankim prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT markneff prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT barbaradavis prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT chandkhanna prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
AT jeffreytrent prospectivemolecularprofilingofcaninecancersprovidesaclinicallyrelevantcomparativemodelforevaluatingpersonalizedmedicinepmedtrials
_version_ 1718421740460179456