Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
ACG Case Rep J ; 8(9): e00661, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34621907
2.
J Proteome Res ; 16(9): 3391-3406, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28768414

ABSTRACT

Myeloid cells play a central role in the context of viral eradication, yet precisely how these cells differentiate throughout the course of acute infections is poorly understood. In this study, we have developed a novel quantitative temporal in vivo proteomics (QTiPs) platform to capture proteomic signatures of temporally transitioning virus-driven myeloid cells directly in situ, thus taking into consideration host-virus interactions throughout the course of an infection. QTiPs, in combination with phenotypic, functional, and metabolic analyses, elucidated a pivotal role for inflammatory CD11b+, Ly6G-, Ly6Chigh-low cells in antiviral immune response and viral clearance. Most importantly, the time-resolved QTiPs data set showed the transition of CD11b+, Ly6G-, Ly6Chigh-low cells into M2-like macrophages, which displayed increased antigen-presentation capacities and bioenergetic demands late in infection. We elucidated the pivotal role of myeloid cells in virus clearance and show how these cells phenotypically, functionally, and metabolically undergo a timely transition from inflammatory to M2-like macrophages in vivo. With respect to the growing appreciation for in vivo examination of viral-host interactions and for the role of myeloid cells, this study elucidates the use of quantitative proteomics to reveal the role and response of distinct immune cell populations throughout the course of virus infection.


Subject(s)
Host-Pathogen Interactions , Macrophages/metabolism , Myeloid Cells/metabolism , Proteomics/methods , Reoviridae Infections/genetics , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Biomarkers/metabolism , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Differentiation , Cell Proliferation , Gene Deletion , Gene Expression Regulation , Gene Ontology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Molecular Sequence Annotation , Myeloid Cells/immunology , Myeloid Cells/virology , Orthoreovirus, Mammalian/growth & development , Orthoreovirus, Mammalian/pathogenicity , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Reoviridae Infections/immunology , Reoviridae Infections/metabolism , Reoviridae Infections/virology , Signal Transduction , Time Factors
3.
J Immunol ; 194(9): 4397-412, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25825443

ABSTRACT

Tumor-associated immunosuppression aids cancer cells to escape immune-mediated attack and subsequent elimination. Recently, however, many oncolytic viruses, including reovirus, have been reported to overturn such immunosuppression and promote the development of a clinically desired antitumor immunity, which is known to promote favorable patient outcomes. Contrary to this existing paradigm, in this article we demonstrate that reovirus augments tumor-associated immunosuppression immediately following its therapeutic administration. Our data show that reovirus induces preferential differentiation of highly suppressive CD11b(+), Gr-1(+), Ly6C(high) myeloid cells from bone marrow hematopoietic progenitor cells. Furthermore, reovirus administration in tumor-bearing hosts drives time-dependent recruitment of CD11b(+), Gr-1(+), Ly6C(high) myeloid cells in the tumor milieu, which is further supported by virus-induced increased expression of numerous immune factors involved in myeloid-derived suppressor cell survival and trafficking. Most importantly, CD11b(+), Gr-1(+), Ly6C(high) myeloid cells specifically potentiate the suppression of T cell proliferation and are associated with the absence of IFN-γ response in the tumor microenvironment early during oncotherapy. Considering that the qualitative traits of a specific antitumor immunity are largely dictated by the immunological events that precede its development, our findings are of critical importance and must be considered while devising complementary interventions aimed at promoting the optimum efficacy of oncolytic virus-based anticancer immunotherapies.


Subject(s)
Genetic Vectors , Immunomodulation , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/immunology , Oncolytic Viruses , Phenotype , Animals , Antigens, Ly/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , CD11b Antigen/metabolism , Cell Differentiation , Chemotaxis/immunology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Mammalian orthoreovirus 3/genetics , Mammalian orthoreovirus 3/immunology , Mice , Myeloid Cells/cytology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Receptors, Chemokine/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tumor Microenvironment/immunology
4.
Oncolytic Virother ; 3: 69-82, 2014.
Article in English | MEDLINE | ID: mdl-27512664

ABSTRACT

Reovirus, a double-stranded ribonucleic acid virus and benign human pathogen, preferentially infects and kills cancer cells in its unmodified form, and is one of the leading oncolytic viruses currently undergoing clinical trials internationally. With 32 clinical trials completed or ongoing thus far, reovirus has demonstrated clinical therapeutic applicability against a multitude of cancers, including but not limited to breast cancer, prostate cancer, pancreatic cancer, malignant gliomas, advanced head and neck cancers, and metastatic ovarian cancers. Phase I trials have demonstrated that reovirus is safe to use via both intralesional/intratumoral and systemic routes of administration, with the most common adverse reactions being grade I/II toxicities, such as flu-like illness (fatigue, nausea, vomiting, headache, fever/chills, dizziness), diarrhea, and lymphopenia. In subsequent Phase II trials, reovirus administration was demonstrated to successfully decrease tumor size and promote tumor necrosis, thereby complementing compelling preclinical evidence of tumor destruction by the virus. Importantly, reovirus has been shown to be effective as a monotherapy, as well as in combination with other anticancer options, including radiation and chemotherapeutic agents, such as gemcitabine, docetaxel, paclitaxel, and carboplatin. Of note, the first Phase III clinical trial using reovirus in combination with paclitaxel and carboplatin for the treatment of head and neck cancers is under way. Based on the evidence from clinical trials, we comprehensively review the use of reovirus as an anticancer agent, acknowledge key obstacles, and suggest future directions to ultimately potentiate the efficacy of reovirus oncotherapy.

5.
Mol Ther ; 21(2): 338-47, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23299799

ABSTRACT

Immunosuppression associated with ovarian cancer (OC) and resultant peritoneal carcinomatosis (PC) hampers the efficacy of many promising treatment options, including immunotherapies. It is hypothesized that oncolytic virus-based therapies can simultaneously kill OC and mitigate immunosuppression. Currently, reovirus-based anticancer therapy is undergoing phase I/II clinical trials for the treatment of OC. Hence, this study was focused on characterizing the effects of reovirus therapy on OC and associated immune microenvironment. Our data shows that reovirus efficiently killed OC cells and induced higher expression of the molecules involved in antigen presentation including major histocompatibility complex (MHC) class I, ß2-microglobulin (ß2M), TAP-1, and TAP-2. In addition, in the presence of reovirus, dendritic cells (DCs) overcame the OC-mediated phenotypic suppression and successfully stimulated tumor-specific CD8+ T cells. In animal studies, reovirus targeted local and distal OC, alleviated the severity of PC and significantly prolonged survival. These therapeutic effects were accompanied by decreased frequency of suppressive cells, e.g., Gr1.1+, CD11b+ myeloid derived suppressor cells (MDSCs), and CD4+, CD25+, FOXP3+ Tregs, tumor-infiltration of CD3+ cells and higher expression of Th1 cytokines. Finally, reovirus therapy during early stages of OC also resulted in the postponement of PC development. This report elucidates timely information on a therapeutic approach that can target OC through clinically desired multifaceted mechanisms to better the outcomes.


Subject(s)
Carcinoma/therapy , Immunomodulation , Oncolytic Virotherapy/methods , Ovarian Neoplasms/therapy , Peritoneal Neoplasms/therapy , Reoviridae/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP Binding Cassette Transporter, Subfamily B, Member 3 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cellular Microenvironment , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Female , Genetic Vectors , Humans , Immunotherapy , Mice , Mice, Inbred C57BL , Phenotype , Real-Time Polymerase Chain Reaction , Reoviridae/immunology
SELECTION OF CITATIONS
SEARCH DETAIL