Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 10(12)2022 12.
Article in English | MEDLINE | ID: mdl-36460333

ABSTRACT

BACKGROUND: In situ vaccination (ISV) is a cancer immunotherapy strategy in which immunostimulatory reagents are introduced directly into a tumor to stimulate antitumor immunity both against the treated tumor and systemically against untreated tumors. Recently, we showed that cowpea mosaic virus (CPMV) is a potent multi-toll-like receptor (TLR) agonist with potent efficacy for treating tumors in mice and dogs by ISV. However, ISV with CPMV alone does not uniformly treat all mouse tumor models tested, however this can be overcome through strategic combinations. More insight is needed to delineate potency and mechanism of systemic antitumor immunity and abscopal effect. METHOD: We investigated the systemic efficacy (abscopal effect) of CPMV ISV with a two-tumor mouse model using murine tumor lines B16F10, 4T1, CT26 and MC38. Flow cytometry identified changes in cell populations responsible for systemic efficacy of CPMV. Transgenic knockout mice and depleting antibodies validated the role of relevant candidate cell populations and cytokines. We evaluated these findings and engineered a multicomponent combination therapy to specifically target the candidate cell population and investigated its systemic efficacy, acquired resistance and immunological memory in mouse models. RESULTS: ISV with CPMV induces systemic antitumor T-cell-mediated immunity that inhibits growth of untreated tumors and requires conventional type-1 dendritic cells (cDC1s). Furthermore, using multiple tumor mouse models resistant to anti-programmed death 1 (PD-1) therapy, we tested the hypothesis that CPMV along with local activation of antigen-presenting cells with agonistic anti-CD40 can synergize and strengthen antitumor efficacy. Indeed, this combination ISV strategy induces an influx of CD8+ T cells, triggers regression in both treated local and untreated distant tumors and potentiates tumor responses to anti-PD-1 therapy. Moreover, serial ISV overcomes resistance to anti-PD-1 therapy and establishes tumor-specific immunological memory. CONCLUSIONS: These findings provide new insights into in situ TLR activation and cDC1 recruitment as effective strategies to overcome resistance to immunotherapy in treated and untreated tumors.


Subject(s)
Comovirus , Immune Checkpoint Inhibitors , Mice , Animals , Dogs , CD8-Positive T-Lymphocytes , Immunotherapy , Vaccination , Adjuvants, Immunologic , Dendritic Cells , Disease Models, Animal
2.
mSphere ; 6(5): e0053721, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34550005

ABSTRACT

Defective interfering (DI) genomes restrict viral replication and induce type I interferon. Since DI genomes have been proposed as vaccine adjuvants or therapeutic antiviral agents, it is important to understand their generation, delineate their mechanism of action, develop robust production capacities, assess their safety and in vivo longevity, and determine their long-term effects. To address this, we generated a recombinant canine distemper virus (rCDV) from an entirely synthetic molecular clone designed using the genomic sequence from a clinical isolate obtained from a free-ranging raccoon with distemper. rCDV was serially passaged in vitro to identify DI genomes that naturally arise during rCDV replication. Defective genomes were identified by Sanger and next-generation sequencing techniques, and predominant genomes were synthetically generated and cloned into T7-driven plasmids. Fully encapsidated DI particles (DIPs) were then generated using a rationally attenuated rCDV as a producer virus to drive DI genome replication. We demonstrate that these DIPs interfere with rCDV replication in a dose-dependent manner in vitro. Finally, we show sustained replication of a fluorescent DIP in experimentally infected ferrets over a period of 14 days. Most importantly, DIPs were isolated from the lymphoid tissues, which are a major site of CDV replication. Our established pipeline for detection, generation, and assaying DIPs is transferable to highly pathogenic paramyxoviruses and will allow qualitative and quantitative assessment of the therapeutic effects of DIP administration on disease outcome. IMPORTANCE Defective interfering (DI) genomes have long been considered inconvenient artifacts that suppressed viral replication in vitro. However, advances in sequencing technologies have led to DI genomes being identified in clinical samples, implicating them in disease progression and outcome. It has been suggested that DI genomes might be harnessed therapeutically. Negative-strand RNA virus research has provided a rich pool of natural DI genomes over many years, and they are probably the best understood in vitro. Here, we demonstrate the identification, synthesis, production, and experimental inoculation of novel CDV DI genomes in highly susceptible ferrets. These results provide important evidence that rationally designed and packaged DI genomes can survive the course of a wild-type virus infection.


Subject(s)
Distemper Virus, Canine/genetics , Distemper Virus, Canine/physiology , Animals , Cell Line , Chlorocebus aethiops , Defective Viruses , Dogs , Ferrets , Genome, Viral , Male , Raccoons/virology , Vero Cells , Virus Replication/genetics , Virus Replication/physiology
3.
Cancer Res ; 80(18): 3795-3796, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934025

ABSTRACT

IL6 is targeted as part of treatment in adoptive cell therapy (ACT) because of its protumor effects and its role in the cytokine release syndrome. However, another major role of IL6 is to polarize naïve CD4+ T cells from Tregs to Th17 cells. While Th17 T cells are associated with autoimmunity, they are present around many different solid tumor cancers and their role in tumor microenvironments is unclear. In this issue of Cancer Research, Knochelmann and colleagues show that Th17 cells with less in vitro expansion in IL6-driven Th17 ACT provide greater solid tumor control and robust immune memory, highlighting advancement in the field of ACT application to solid tumor immunotherapy.See related article by Knochelmann et al., p. 3920.


Subject(s)
Interleukin-6 , Neoplasms , Cell- and Tissue-Based Therapy , Humans , Immunotherapy , Neoplasms/therapy , Th17 Cells , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL