Your browser doesn't support javascript.
loading
Systemic delivery of a Gli inhibitor via polymeric nanocarriers inhibits tumor-induced bone disease.
Vanderburgh, Joseph P; Kwakwa, Kristin A; Werfel, Thomas A; Merkel, Alyssa R; Gupta, Mukesh K; Johnson, Rachelle W; Guelcher, Scott A; Duvall, Craig L; Rhoades, Julie A.
Afiliação
  • Vanderburgh JP; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
  • Kwakwa KA; Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
  • Werfel TA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
  • Merkel AR; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232,
  • Gupta MK; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
  • Johnson RW; Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
  • Guelcher SA; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA; Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineer
  • Duvall CL; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
  • Rhoades JA; Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA; Department of Biomedical Engineering, Van
J Control Release ; 311-312: 257-272, 2019 10.
Article em En | MEDLINE | ID: mdl-31494183
ABSTRACT
Solid tumors frequently metastasize to bone and induce bone destruction leading to severe pain, fractures, and other skeletal-related events (SREs). Osteoclast inhibitors such as bisphosphonates delay SREs but do not prevent skeletal complications or improve overall survival. Because bisphosphonates can cause adverse side effects and are contraindicated for some patients, we sought an alternative therapy to reduce tumor-associated bone destruction. Our previous studies identified the transcription factor Gli2 as a key regulator of parathyroid hormone-related protein (PTHrP), which is produced by bone metastatic tumor cells to promote osteoclast-mediated bone destruction. In this study, we tested the treatment effect of a Gli antagonist GANT58, which inhibits Gli2 nuclear translocation and PTHrP expression in tumor cells. In initial testing, GANT58 did not have efficacy in vivo due to its low water solubility and poor bioavailability. We therefore developed a micellar nanoparticle (NP) to encapsulate and colloidally stabilize GANT58, providing a fully aqueous, intravenously injectable formulation based on the polymer poly(propylene sulfide)135-b-poly[(oligoethylene glycol)9 methyl ether acrylate]17 (PPS135-b-POEGA17). POEGA forms the hydrophilic NP surface while PPS forms the hydrophobic NP core that sequesters GANT58. In response to reactive oxygen species (ROS), PPS becomes hydrophilic and degrades to enable drug release. In an intratibial model of breast cancer bone metastasis, treatment with GANT58-NPs decreased bone lesion area by 49% (p<.01) and lesion number by 38% (p<.05) and resulted in a 2.5-fold increase in trabecular bone volume (p<.001). Similar results were observed in intracardiac and intratibial models of breast and lung cancer bone metastasis, respectively. Importantly, GANT58-NPs reduced tumor cell proliferation but did not alter mesenchymal stem cell proliferation or osteoblast mineralization in vitro, nor was there evidence of cytotoxicity after repeated in vivo treatment. Thus, inhibition of Gli2 using GANT58-NPs is a potential therapy to reduce bone destruction that should be considered for further testing and development toward clinical translation.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 6_ODS3_enfermedades_notrasmisibles Base de dados: MEDLINE Assunto principal: Piridinas / Tiofenos / Neoplasias Ósseas / Portadores de Fármacos / Neoplasias Mamárias Animais / Nanopartículas / Antineoplásicos Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Revista: J Control Release Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 6_ODS3_enfermedades_notrasmisibles Base de dados: MEDLINE Assunto principal: Piridinas / Tiofenos / Neoplasias Ósseas / Portadores de Fármacos / Neoplasias Mamárias Animais / Nanopartículas / Antineoplásicos Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Revista: J Control Release Ano de publicação: 2019 Tipo de documento: Article