Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Mol Cancer ; 20(1): 69, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33858437

ABSTRACT

Synthetic mRNA provides a template for the synthesis of any given protein, protein fragment or peptide and lends itself to a broad range of pharmaceutical applications, including different modalities of cancer immunotherapy. With the ease of rapid, large scale Good Manufacturing Practice-grade mRNA production, mRNA is ideally poised not only for off-the shelf cancer vaccines but also for personalized neoantigen vaccination. The ability to stimulate pattern recognition receptors and thus an anti-viral type of innate immune response equips mRNA-based vaccines with inherent adjuvanticity. Nucleoside modification and elimination of double-stranded RNA can reduce the immunomodulatory activity of mRNA and increase and prolong protein production. In combination with nanoparticle-based formulations that increase transfection efficiency and facilitate lymphatic system targeting, nucleoside-modified mRNA enables efficient delivery of cytokines, costimulatory receptors, or therapeutic antibodies. Steady but transient production of the encoded bioactive molecule from the mRNA template can improve the pharmacokinetic, pharmacodynamic and safety properties as compared to the respective recombinant proteins. This may be harnessed for applications that benefit from a higher level of expression control, such as chimeric antigen receptor (CAR)-modified adoptive T-cell therapies. This review highlights the advancements in the field of mRNA-based cancer therapeutics, providing insights into key preclinical developments and the evolving clinical landscape.


Subject(s)
Genetic Therapy , Immunotherapy , Neoplasms/etiology , Neoplasms/therapy , RNA, Messenger/administration & dosage , Animals , Antibodies/genetics , Antibodies/immunology , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biomarkers, Tumor , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Therapy/methods , Humans , Immunologic Factors/genetics , Immunotherapy/methods , Neoplasms/pathology , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Cell Rep Methods ; 2(3): 100182, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35475224

ABSTRACT

Single-cell ATAC sequencing (scATAC-seq) is a powerful and increasingly popular technique to explore the regulatory landscape of heterogeneous cellular populations. However, the high noise levels, degree of sparsity, and scale of the generated data make its analysis challenging. Here, we present PeakVI, a probabilistic framework that leverages deep neural networks to analyze scATAC-seq data. PeakVI fits an informative latent space that preserves biological heterogeneity while correcting batch effects and accounting for technical effects, such as library size and region-specific biases. In addition, PeakVI provides a technique for identifying differential accessibility at a single-region resolution, which can be used for cell-type annotation as well as identification of key cis-regulatory elements. We use public datasets to demonstrate that PeakVI is scalable, stable, robust to low-quality data, and outperforms current analysis methods on a range of critical analysis tasks. PeakVI is publicly available and implemented in the scvi-tools framework.


Subject(s)
Chromatin , Regulatory Sequences, Nucleic Acid , Chromatin/genetics , Gene Library
3.
Hum Vaccin Immunother ; 18(1): 2024416, 2022 12 31.
Article in English | MEDLINE | ID: mdl-35130105

ABSTRACT

After one year of absence, the 18th Annual Meeting of the Association for Cancer Immunotherapy (CIMT), Europe's cancer immunotherapy meeting, took place virtually from 10 to 12 May 2021. Over 850 academic and clinical professionals from 30 countries met to discuss the recent advancements in cancer immunotherapy and the current progress in COVID19-related research. This meeting report summarizes the highlights of CIMT2021.


Subject(s)
COVID-19 , Neoplasms , Humans , Immunotherapy , Neoplasms/therapy , Tumor Microenvironment
4.
Hum Vaccin Immunother ; 14(12): 2864-2873, 2018.
Article in English | MEDLINE | ID: mdl-30111232

ABSTRACT

The 16th Annual Meeting of the Association for Cancer Immunotherapy (CIMT), Europe's largest meeting series of its kind, took place in Mainz, Germany from 15-17 May, 2018. Cutting-edge advancements in cancer immunotherapy were discussed among more than 700 scientists under the motto "Pushing Frontiers in Cancer Immunotherapy". This meeting report is a summary of some of the CIMT 2018 highlights.


Subject(s)
Immunotherapy , Neoplasms/therapy , Animals , Congresses as Topic , Disease Models, Animal , Drug Therapy, Combination , Humans , Tumor Microenvironment , Vaccination , Xenograft Model Antitumor Assays
5.
Oncotarget ; 7(37): 59589-59603, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27449098

ABSTRACT

Prostate cancer is a heterogeneous disease. MiR-375 is a marker for prostate cancer progression, but its cellular function is not characterized. Here, we provide the first comprehensive investigation of miR-375 in prostate cancer. We show that miR-375 is enriched in prostate cancer compared to normal cells. Furthermore, miR-375 enhanced proliferation, migration and invasion in vitro and induced tumor growth and reduced survival in vivo showing that miR-375 has oncogenic properties in prostate cancer. On the molecular level, we provide the targetome and genome-wide transcriptional changes of miR-375 expression by applying a generalized linear model for Ago-RIP-Seq and RNA-Seq, and show that miR-375 is involved in tumorigenic networks and Polycomb regulation. Integration of tissue and gene ontology data prioritized miR-375 targets and identified the tumor suppressor gene CBX7, a member of Polycomb repressive complex 1, as a major miR-375 target. MiR-375-mediated repression of CBX7 was accompanied by increased expression of its homolog CBX8 and activated transcriptional programs linked to malignant progression in prostate cancer cells. Tissue analysis showed association of CBX7 loss with advanced prostate cancer. Our study indicates that miR-375 exerts its tumor-promoting role in prostate cancer by influencing the epigenetic regulation of transcriptional programs through its ability to directly target the Polycomb complex member CBX7.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Polycomb Repressive Complex 1/genetics , Prostatic Neoplasms/genetics , Animals , Cell Line, Tumor , Disease Progression , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Male , Mice, Nude , Polycomb Repressive Complex 1/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL