Please use this identifier to cite or link to this item: https://ahro.austin.org.au/austinjspui/handle/1/20535
Full metadata record
DC FieldValueLanguage
dc.contributor.authorYap, May Lin-
dc.contributor.authorMcFadyen, James D-
dc.contributor.authorWang, Xiaowei-
dc.contributor.authorZiegler, Melanie-
dc.contributor.authorChen, Yung-Chih-
dc.contributor.authorWillcox, Abbey-
dc.contributor.authorNowell, Cameron J-
dc.contributor.authorScott, Andrew M-
dc.contributor.authorSloan, Erica K-
dc.contributor.authorHogarth, P Mark-
dc.contributor.authorPietersz, Geoffrey A-
dc.contributor.authorPeter, Karlheinz-
dc.date2019-02-07-
dc.date.accessioned2019-04-02T01:08:22Z-
dc.date.available2019-04-02T01:08:22Z-
dc.date.issued2019-02-07-
dc.identifier.citationTheranostics 2019; 9(4): 1154-1169-
dc.identifier.urihttps://ahro.austin.org.au/austinjspui/handle/1/20535-
dc.description.abstractRationale: Platelets are increasingly recognized as mediators of tumor growth and metastasis. Hypothesizing that activated platelets in the tumor microenvironment provide a targeting epitope for tumor-directed chemotherapy, we developed an antibody-drug conjugate (ADC), comprised of a single-chain antibody (scFv) against the platelet integrin GPIIb/IIIa (scFvGPIIb/IIIa) linked to the potent chemotherapeutic microtubule inhibitor, monomethyl auristatin E (MMAE). Methods: We developed an ADC comprised of three components: 1) A scFv which specifically binds to the high affinity, activated integrin GPIIb/IIIa on activated platelets. 2) A highly potent microtubule inhibitor, monomethyl auristatin E. 3) A drug activation/release mechanism using a linker cleavable by cathepsin B, which we demonstrate to be abundant in the tumor microenvironment. The scFvGPIIb/IIIa-MMAE was first conjugated with Cyanine7 for in vivo imaging. The therapeutic efficacy of the scFvGPIIb/IIIa-MMAE was then tested in a mouse metastasis model of triple negative breast cancer. Results: In vitro studies confirmed that this ADC specifically binds to activated GPIIb/IIIa, and cathepsin B-mediated drug release/activation resulted in tumor cytotoxicity. In vivo fluorescence imaging demonstrated that the newly generated ADC localized to primary tumors and metastases in a mouse xenograft model of triple negative breast cancer, a difficult to treat tumor for which a selective tumor-targeting therapy remains to be clinically established. Importantly, we demonstrated that the scFvGPIIb/IIIa-MMAE displays marked efficacy as an anti-cancer agent, reducing tumor growth and preventing metastatic disease, without any discernible toxic effects. Conclusion: Here, we demonstrate the utility of a novel ADC that targets a potent cytotoxic drug to activated platelets and specifically releases the cytotoxic agent within the confines of the tumor. This unique targeting mechanism, specific to the tumor microenvironment, holds promise as a novel therapeutic approach for the treatment of a broad range of primary tumors and metastatic disease, particularly for tumors that lack specific molecular epitopes for drug targeting.-
dc.language.isoeng-
dc.subjectActivated platelets-
dc.subjectAntibody-drug conjugate-
dc.subjectCancer-
dc.subjectGPIIb/IIIa-
dc.titleActivated platelets in the tumor microenvironment for targeting of antibody-drug conjugates to tumors and metastases.-
dc.typeJournal Article-
dc.identifier.journaltitleTheranostics-
dc.identifier.affiliationDepartment of Hematology, The Alfred Hospital, Melbourne, 3004, Australiaen
dc.identifier.affiliationSchool of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australiaen
dc.identifier.affiliationUniversity of Melbourne, Melbourne, Victoria, Australiaen
dc.identifier.affiliationBaker Heart and Diabetes Institute, Melbourne, 3004, Australiaen
dc.identifier.affiliationDepartment of Medicine, Monash University, Melbourne, 3800, Australiaen
dc.identifier.affiliationDepartment of Immunology, Monash University, Melbourne, 3800, Australiaen
dc.identifier.affiliationDepartment of Clinical Pathology, The University of Melbourne, Melbourne, 3010, Australiaen
dc.identifier.affiliationBurnet Institute, Melbourne, 3004, Australiaen
dc.identifier.affiliationCollege of Health and Biomedicine, Victoria University, Melbourne, 3021, Australiaen
dc.identifier.affiliationDrug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australiaen
dc.identifier.affiliationOlivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia-
dc.identifier.affiliationDepartment of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia-
dc.identifier.doi10.7150/thno.29146-
dc.identifier.orcid0000-0002-6656-295X-
dc.identifier.pubmedid30867822-
dc.type.austinJournal Article-
local.name.researcherScott, Andrew M
item.openairecristypehttp://purl.org/coar/resource_type/c_18cf-
item.openairetypeJournal Article-
item.grantfulltextnone-
item.cerifentitytypePublications-
item.fulltextNo Fulltext-
item.languageiso639-1en-
crisitem.author.deptClinical Haematology-
crisitem.author.deptMolecular Imaging and Therapy-
crisitem.author.deptOlivia Newton-John Cancer Research Institute-
Appears in Collections:Journal articles
Show simple item record

Page view(s)

38
checked on Oct 1, 2024

Google ScholarTM

Check


Items in AHRO are protected by copyright, with all rights reserved, unless otherwise indicated.