Cargando…

Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy

Rationale: Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) is a co-inhibitory checkpoint receptor that is expressed by naïve T-cells in lymph nodes (LNs) to inhibit activation against “self” antigens (Ags). In cancer, anti-CTLA-4 blocks inhibitory action, enabling robust activation of T-cells a...

Descripción completa

Detalles Bibliográficos
Autores principales: Kwon, Sunkuk, Velasquez, Fred Christian, Rasmussen, John C., Greives, Matthew R., Turner, Kelly D., Morrow, John R., Hwu, Wen-Jen, Ross, Russell F., Zhang, Songlin, Sevick-Muraca, Eva M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6857054/
https://www.ncbi.nlm.nih.gov/pubmed/31754400
http://dx.doi.org/10.7150/thno.35280
_version_ 1783470688706756608
author Kwon, Sunkuk
Velasquez, Fred Christian
Rasmussen, John C.
Greives, Matthew R.
Turner, Kelly D.
Morrow, John R.
Hwu, Wen-Jen
Ross, Russell F.
Zhang, Songlin
Sevick-Muraca, Eva M.
author_facet Kwon, Sunkuk
Velasquez, Fred Christian
Rasmussen, John C.
Greives, Matthew R.
Turner, Kelly D.
Morrow, John R.
Hwu, Wen-Jen
Ross, Russell F.
Zhang, Songlin
Sevick-Muraca, Eva M.
author_sort Kwon, Sunkuk
collection PubMed
description Rationale: Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) is a co-inhibitory checkpoint receptor that is expressed by naïve T-cells in lymph nodes (LNs) to inhibit activation against “self” antigens (Ags). In cancer, anti-CTLA-4 blocks inhibitory action, enabling robust activation of T-cells against tumor Ags presented in tumor draining LNs (TDLNs). However, anti-CTLA-4 is administered intravenously with limited exposure within TDLNs and immune related adverse events (irAEs) are associated with over-stimulation of the immune system. Methods: Herein, we first deliver anti-CTLA-4 in an orthotopic mammary carcinoma murine model using a nanotopographical microneedle-array device to compare its anti-tumor response to that from systemic administration. Additionally, to demonstrate the feasibility of lymphatic delivery in humans using the device, we use near-infrared fluorescence imaging to image delivery of ICG to LNs. Results: Our data show that lymphatic infusion results in more effective tumor growth inhibition, arrest of metastases, increased tumor infiltrating lymphocytes and complete responses when compared to conventional systemic administration. In clinical studies, we demonstrate for the first time that nanotopographic infusion can deliver ICG through the lymphatics directly to the axilla and inguinal LNs of healthy human volunteers. Conclusion: Taken together, these results suggest that regional delivery using a nanotopography-based microneedle array could revolutionize checkpoint blockade immunotherapy by reducing systemic drug exposure and maximizing drug delivery to TDLNs where tumor Ags present. Future work is needed to determine whether lymphatic delivery of anti-CTLA-4 can alleviate irAEs that occur with systemic dosing.
format Online
Article
Text
id pubmed-6857054
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-68570542019-11-21 Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy Kwon, Sunkuk Velasquez, Fred Christian Rasmussen, John C. Greives, Matthew R. Turner, Kelly D. Morrow, John R. Hwu, Wen-Jen Ross, Russell F. Zhang, Songlin Sevick-Muraca, Eva M. Theranostics Research Paper Rationale: Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) is a co-inhibitory checkpoint receptor that is expressed by naïve T-cells in lymph nodes (LNs) to inhibit activation against “self” antigens (Ags). In cancer, anti-CTLA-4 blocks inhibitory action, enabling robust activation of T-cells against tumor Ags presented in tumor draining LNs (TDLNs). However, anti-CTLA-4 is administered intravenously with limited exposure within TDLNs and immune related adverse events (irAEs) are associated with over-stimulation of the immune system. Methods: Herein, we first deliver anti-CTLA-4 in an orthotopic mammary carcinoma murine model using a nanotopographical microneedle-array device to compare its anti-tumor response to that from systemic administration. Additionally, to demonstrate the feasibility of lymphatic delivery in humans using the device, we use near-infrared fluorescence imaging to image delivery of ICG to LNs. Results: Our data show that lymphatic infusion results in more effective tumor growth inhibition, arrest of metastases, increased tumor infiltrating lymphocytes and complete responses when compared to conventional systemic administration. In clinical studies, we demonstrate for the first time that nanotopographic infusion can deliver ICG through the lymphatics directly to the axilla and inguinal LNs of healthy human volunteers. Conclusion: Taken together, these results suggest that regional delivery using a nanotopography-based microneedle array could revolutionize checkpoint blockade immunotherapy by reducing systemic drug exposure and maximizing drug delivery to TDLNs where tumor Ags present. Future work is needed to determine whether lymphatic delivery of anti-CTLA-4 can alleviate irAEs that occur with systemic dosing. Ivyspring International Publisher 2019-10-22 /pmc/articles/PMC6857054/ /pubmed/31754400 http://dx.doi.org/10.7150/thno.35280 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
Kwon, Sunkuk
Velasquez, Fred Christian
Rasmussen, John C.
Greives, Matthew R.
Turner, Kelly D.
Morrow, John R.
Hwu, Wen-Jen
Ross, Russell F.
Zhang, Songlin
Sevick-Muraca, Eva M.
Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy
title Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy
title_full Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy
title_fullStr Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy
title_full_unstemmed Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy
title_short Nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy
title_sort nanotopography-based lymphatic delivery for improved anti-tumor responses to checkpoint blockade immunotherapy
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6857054/
https://www.ncbi.nlm.nih.gov/pubmed/31754400
http://dx.doi.org/10.7150/thno.35280
work_keys_str_mv AT kwonsunkuk nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT velasquezfredchristian nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT rasmussenjohnc nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT greivesmatthewr nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT turnerkellyd nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT morrowjohnr nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT hwuwenjen nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT rossrussellf nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT zhangsonglin nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy
AT sevickmuracaevam nanotopographybasedlymphaticdeliveryforimprovedantitumorresponsestocheckpointblockadeimmunotherapy