Cargando…

Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer

Tumor-associated macrophages (TAMs) enhance tumor growth in mice and are correlated with a worse prognosis for breast cancer patients. While early therapies sought to deplete all macrophages, current therapeutics aim to reprogram pro-tumor macrophages (M2) and preserve those necessary for anti-tumor...

Descripción completa

Detalles Bibliográficos
Autores principales: Esser, Alison K., Ross, Michael H., Fontana, Francesca, Su, Xinming, Gabay, Ariel, Fox, Gregory C., Xu, Yalin, Xiang, Jingyu, Schmieder, Anne H., Yang, Xiaoxia, Cui, Grace, Scott, Michael, Achilefu, Samuel, Chauhan, Jay, Fletcher, Steven, Lanza, Gregory M., Weilbaecher, Katherine N.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7359087/
https://www.ncbi.nlm.nih.gov/pubmed/32685002
http://dx.doi.org/10.7150/thno.44523
_version_ 1783558974040178688
author Esser, Alison K.
Ross, Michael H.
Fontana, Francesca
Su, Xinming
Gabay, Ariel
Fox, Gregory C.
Xu, Yalin
Xiang, Jingyu
Schmieder, Anne H.
Yang, Xiaoxia
Cui, Grace
Scott, Michael
Achilefu, Samuel
Chauhan, Jay
Fletcher, Steven
Lanza, Gregory M.
Weilbaecher, Katherine N.
author_facet Esser, Alison K.
Ross, Michael H.
Fontana, Francesca
Su, Xinming
Gabay, Ariel
Fox, Gregory C.
Xu, Yalin
Xiang, Jingyu
Schmieder, Anne H.
Yang, Xiaoxia
Cui, Grace
Scott, Michael
Achilefu, Samuel
Chauhan, Jay
Fletcher, Steven
Lanza, Gregory M.
Weilbaecher, Katherine N.
author_sort Esser, Alison K.
collection PubMed
description Tumor-associated macrophages (TAMs) enhance tumor growth in mice and are correlated with a worse prognosis for breast cancer patients. While early therapies sought to deplete all macrophages, current therapeutics aim to reprogram pro-tumor macrophages (M2) and preserve those necessary for anti-tumor immune responses (M1). Recent studies have shown that c-MYC (MYC) is induced in M2 macrophages in vitro and in vivo where it regulates the expression of tumor-promoting genes. In a myeloid lineage MYC KO mouse model, MYC had important roles in macrophage maturation and function leading to reduced tumor growth. We therefore hypothesized that targeted delivery of a MYC inhibitor to established M2 TAMs could reduce polarization toward an M2 phenotype in breast cancer models. Methods: In this study, we developed a MYC inhibitor prodrug (MI3-PD) for encapsulation within perfluorocarbon nanoparticles, which can deliver drugs directly to the cytosol of the target cell through a phagocytosis independent mechanism. We have previously shown that M2-like TAMs express significant levels of the vitronectin receptor, integrin β3, and in vivo targeting and therapeutic potential was evaluated using αvβ3 integrin targeted rhodamine-labeled nanoparticles (NP) or integrin αvβ3-MI3-PD nanoparticles. Results: We observed that rhodamine, delivered by αvβ3-rhodamine NP, was incorporated into M2 tumor promoting macrophages through both phagocytosis-independent and dependent mechanisms, while NP uptake in tumor suppressing M1 macrophages was almost exclusively through phagocytosis. In a mouse model of breast cancer (4T1-GFP-FL), M2-like TAMs were significantly reduced with αvβ3-MI3-PD NP treatment. To validate this effect was independent of drug delivery to tumor cells and was specific to the MYC inhibitor, mice with integrin β3 knock out tumors (PyMT-Bo1 β3KO) were treated with αvβ3-NP or αvβ3-MI3-PD NP. M2 macrophages were significantly reduced with αvβ3-MI3-PD nanoparticle therapy but not αvβ3-NP treatment. Conclusion: These data suggest αvβ3-NP-mediated drug delivery of a c-MYC inhibitor can reduce protumor M2-like macrophages while preserving antitumor M1-like macrophages in breast cancer.
format Online
Article
Text
id pubmed-7359087
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-73590872020-07-17 Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer Esser, Alison K. Ross, Michael H. Fontana, Francesca Su, Xinming Gabay, Ariel Fox, Gregory C. Xu, Yalin Xiang, Jingyu Schmieder, Anne H. Yang, Xiaoxia Cui, Grace Scott, Michael Achilefu, Samuel Chauhan, Jay Fletcher, Steven Lanza, Gregory M. Weilbaecher, Katherine N. Theranostics Research Paper Tumor-associated macrophages (TAMs) enhance tumor growth in mice and are correlated with a worse prognosis for breast cancer patients. While early therapies sought to deplete all macrophages, current therapeutics aim to reprogram pro-tumor macrophages (M2) and preserve those necessary for anti-tumor immune responses (M1). Recent studies have shown that c-MYC (MYC) is induced in M2 macrophages in vitro and in vivo where it regulates the expression of tumor-promoting genes. In a myeloid lineage MYC KO mouse model, MYC had important roles in macrophage maturation and function leading to reduced tumor growth. We therefore hypothesized that targeted delivery of a MYC inhibitor to established M2 TAMs could reduce polarization toward an M2 phenotype in breast cancer models. Methods: In this study, we developed a MYC inhibitor prodrug (MI3-PD) for encapsulation within perfluorocarbon nanoparticles, which can deliver drugs directly to the cytosol of the target cell through a phagocytosis independent mechanism. We have previously shown that M2-like TAMs express significant levels of the vitronectin receptor, integrin β3, and in vivo targeting and therapeutic potential was evaluated using αvβ3 integrin targeted rhodamine-labeled nanoparticles (NP) or integrin αvβ3-MI3-PD nanoparticles. Results: We observed that rhodamine, delivered by αvβ3-rhodamine NP, was incorporated into M2 tumor promoting macrophages through both phagocytosis-independent and dependent mechanisms, while NP uptake in tumor suppressing M1 macrophages was almost exclusively through phagocytosis. In a mouse model of breast cancer (4T1-GFP-FL), M2-like TAMs were significantly reduced with αvβ3-MI3-PD NP treatment. To validate this effect was independent of drug delivery to tumor cells and was specific to the MYC inhibitor, mice with integrin β3 knock out tumors (PyMT-Bo1 β3KO) were treated with αvβ3-NP or αvβ3-MI3-PD NP. M2 macrophages were significantly reduced with αvβ3-MI3-PD nanoparticle therapy but not αvβ3-NP treatment. Conclusion: These data suggest αvβ3-NP-mediated drug delivery of a c-MYC inhibitor can reduce protumor M2-like macrophages while preserving antitumor M1-like macrophages in breast cancer. Ivyspring International Publisher 2020-06-12 /pmc/articles/PMC7359087/ /pubmed/32685002 http://dx.doi.org/10.7150/thno.44523 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Esser, Alison K.
Ross, Michael H.
Fontana, Francesca
Su, Xinming
Gabay, Ariel
Fox, Gregory C.
Xu, Yalin
Xiang, Jingyu
Schmieder, Anne H.
Yang, Xiaoxia
Cui, Grace
Scott, Michael
Achilefu, Samuel
Chauhan, Jay
Fletcher, Steven
Lanza, Gregory M.
Weilbaecher, Katherine N.
Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer
title Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer
title_full Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer
title_fullStr Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer
title_full_unstemmed Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer
title_short Nanotherapy delivery of c-myc inhibitor targets Protumor Macrophages and preserves Antitumor Macrophages in Breast Cancer
title_sort nanotherapy delivery of c-myc inhibitor targets protumor macrophages and preserves antitumor macrophages in breast cancer
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7359087/
https://www.ncbi.nlm.nih.gov/pubmed/32685002
http://dx.doi.org/10.7150/thno.44523
work_keys_str_mv AT esseralisonk nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT rossmichaelh nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT fontanafrancesca nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT suxinming nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT gabayariel nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT foxgregoryc nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT xuyalin nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT xiangjingyu nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT schmiederanneh nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT yangxiaoxia nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT cuigrace nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT scottmichael nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT achilefusamuel nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT chauhanjay nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT fletchersteven nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT lanzagregorym nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer
AT weilbaecherkatherinen nanotherapydeliveryofcmycinhibitortargetsprotumormacrophagesandpreservesantitumormacrophagesinbreastcancer