Personalized Tumor RNA Loaded Lipid-Nanoparticles Prime the Systemic and Intratumoral Milieu for Response to Cancer Immunotherapy

Nano Lett. 2018 Oct 10;18(10):6195-6206. doi: 10.1021/acs.nanolett.8b02179. Epub 2018 Sep 27.

Abstract

Translation of nanoparticles (NPs) into human clinical trials for patients with refractory cancers has lagged due to unknown biologic reactivities of novel NP designs. To overcome these limitations, simple well-characterized mRNA lipid-NPs have been developed as cancer immunotherapeutic vaccines. While the preponderance of RNA lipid-NPs encoding for tumor-associated antigens or neoepitopes have been designed to target lymphoid organs, they remain encumbered by the profound intratumoral and systemic immunosuppression that may stymie an activated T cell response. Herein, we show that systemic localization of untargeted tumor RNA (derived from whole transcriptome) encapsulated in lipid-NPs, with excess positive charge, primes the peripheral and intratumoral milieu for response to immunotherapy. In immunologically resistant tumor models, these RNA-NPs activate the preponderance of systemic and intratumoral myeloid cells (characterized by coexpression of PD-L1 and CD86). Addition of immune checkpoint inhibitors (ICIs) (to animals primed with RNA-NPs) augments peripheral/intratumoral PD-1+CD8+ cells and mediates synergistic antitumor efficacy in settings where ICIs alone do not confer therapeutic benefit. These synergistic effects are mediated by type I interferon released from plasmacytoid dendritic cells (pDCs). In translational studies, personalized mRNA-NPs were safe and active in a client-owned canine with a spontaneous malignant glioma. In summary, we demonstrate widespread immune activation from tumor loaded RNA-NPs concomitant with inducible PD-L1 expression that can be therapeutically exploited. While immunotherapy remains effective for only a subset of cancer patients, combination therapy with systemic immunomodulating RNA-NPs may broaden its therapeutic potency.

Keywords: RNA nanoparticles; cancer immunotherapy; cancer vaccines; immune checkpoint inhibitors; liposomes; personalized therapy.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • B7-2 Antigen / antagonists & inhibitors
  • B7-2 Antigen / genetics
  • B7-2 Antigen / immunology
  • B7-H1 Antigen / antagonists & inhibitors
  • B7-H1 Antigen / genetics
  • B7-H1 Antigen / immunology
  • CD8-Positive T-Lymphocytes / drug effects
  • CD8-Positive T-Lymphocytes / immunology
  • Cancer Vaccines / immunology
  • Cancer Vaccines / therapeutic use
  • Dendritic Cells / immunology
  • Disease Models, Animal
  • Dogs
  • Glioma / drug therapy*
  • Glioma / immunology
  • Glioma / pathology
  • Glioma / veterinary
  • Humans
  • Immunotherapy*
  • Lipids / administration & dosage*
  • Lipids / chemistry
  • Lipids / immunology
  • Lymphocyte Activation / immunology
  • Nanoparticles / administration & dosage*
  • Nanoparticles / chemistry
  • Precision Medicine*
  • RNA, Neoplasm / chemistry
  • RNA, Neoplasm / genetics
  • RNA, Neoplasm / immunology
  • Transcriptome / genetics

Substances

  • B7-2 Antigen
  • B7-H1 Antigen
  • CD274 protein, human
  • Cancer Vaccines
  • Lipids
  • RNA, Neoplasm