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 Virol ; 92(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-29997212

ABSTRACT

We are pursuing cancer immunotherapy with a neuro-attenuated recombinant poliovirus, PVSRIPO. PVSRIPO is the live attenuated type 1 (Sabin) poliovirus vaccine carrying a heterologous internal ribosomal entry site (IRES) of human rhinovirus type 2 (HRV2). Intratumoral infusion of PVSRIPO is showing promise in the therapy of recurrent WHO grade IV malignant glioma (glioblastoma), a notoriously treatment-refractory cancer with dismal prognosis. PVSRIPO exhibits profound cytotoxicity in infected neoplastic cells expressing the poliovirus receptor CD155. In addition, it elicits intriguing persistent translation and replication, giving rise to sustained type I interferon (IFN)-dominant proinflammatory stimulation of antigen-presenting cells. A key determinant of the inflammatory footprint generated by neoplastic cell infection and its role in shaping the adaptive response after PVSRIPO tumor infection is the virus's inherent relationship to the host's innate antiviral response. In this report, we define subversion of innate host immunity by PVSRIPO, enabling productive viral translation and cytopathogenicity with extremely low multiplicities of infection in the presence of an active innate antiviral IFN response.IMPORTANCE Engaging innate antiviral responses is considered key for instigating tumor-antigen-specific antitumor immunity with cancer immunotherapy approaches. However, they are a double-edged sword for attempts to enlist viruses in such approaches. In addition to their role in the transition from innate to adaptive immunity, innate antiviral IFN responses may intercept the viral life cycle in cancerous cells, prevent viral cytopathogenicity, and restrict viral spread. This has been shown to reduce overall antitumor efficacy of several proposed oncolytic virus prototypes, presumably by limiting direct cell killing and the ensuing inflammatory profile within the infected tumor. In this report, we outline how an unusual recalcitrance of polioviruses toward innate antiviral responses permits viral cytotoxicity and propagation in neoplastic cells, combined with engaging active innate antiviral IFN responses.


Subject(s)
Glioblastoma/immunology , Immunity, Innate , Interferon-Induced Helicase, IFIH1/immunology , Oncolytic Viruses/immunology , Poliovirus/immunology , Cell Line, Tumor , Glioblastoma/pathology , Glioblastoma/therapy , Glioblastoma/virology , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/genetics , Oncolytic Viruses/genetics , Poliovirus/genetics , Receptors, Virus/genetics , Receptors, Virus/immunology
2.
Cancer ; 120(21): 3277-86, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24939611

ABSTRACT

Recently, the century-old idea of targeting cancer with viruses (oncolytic viruses) has come of age, and promise has been documented in early stage and several late-stage clinical trials in a variety of cancers. Although originally prized for their direct tumor cytotoxicity (oncolytic virotherapy), recently, the proinflammatory and immunogenic effects of viral tumor infection (oncolytic immunotherapy) have come into focus. Indeed, a capacity for eliciting broad, sustained antineoplastic effects stemming from combined direct viral cytotoxicity, innate antiviral activation, stromal proinflammatory stimulation, and recruitment of adaptive immune effector responses is the greatest asset of oncolytic viruses. However, it also is the source for enormous mechanistic complexity that must be considered for successful clinical translation. Because of fundamentally different relationships with their hosts (malignant or not), diverse replication strategies, and distinct modes of tumor cytotoxicity/killing, oncolytic viruses should not be referred to collectively. These agents must be evaluated based on their individual merits. In this review, the authors highlight key mechanistic principles of cancer treatment with the polio:rhinovirus chimera PVSRIPO and their implications for oncolytic immunotherapy in the clinic.


Subject(s)
Immunity, Innate/genetics , Neoplasms/genetics , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Humans , Immunotherapy , Neoplasms/virology , Poliomyelitis/genetics , Receptors, Virus/immunology , Virus Internalization
3.
J Struct Biol ; 180(1): 249-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22584152

ABSTRACT

Chemical biotinylation of protein complexes followed by binding to two-dimensional (monolayer) crystals of streptavidin is shown to be an effective way to prepare cryo-EM specimens from samples at low protein concentration. Three different multiprotein complexes are used to demonstrate the generality of this method. In addition, native thermosomes, purified from Sulfolobus solfataricus P2, are used to demonstrate that a uniform distribution of Euler angles is produced, even though this particle is known to adopt a preferred orientation when other methods of cryo-EM specimen preparation are used.


Subject(s)
Biotin/chemistry , Cryoelectron Microscopy/methods , Streptavidin/chemistry , Adsorption , Animals , Apoferritins/chemistry , Apoferritins/ultrastructure , Bacterial Proteins/chemistry , Biotinylation , Crystallization , Desulfovibrio vulgaris , Horses , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/ultrastructure , Protein Binding , Protein Structure, Quaternary , Sulfolobus solfataricus , Thermosomes/chemistry , Thermosomes/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...