Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Oncoimmunology ; 8(1): e1512329, 2019.
Article in English | MEDLINE | ID: mdl-30546947

ABSTRACT

Multiple immunotherapeutics have been approved for cancer patients, however advanced solid tumors are frequently refractory to treatment. We evaluated the safety and immunogenicity of a vaccination approach with multimodal oncolytic potential in non-human primates (NHP) (Macaca fascicularis). Primates received a replication-deficient adenoviral prime, boosted by the oncolytic Maraba MG1 rhabdovirus. Both vectors expressed the human MAGE-A3. No severe adverse events were observed. Boosting with MG1-MAGEA3 induced an expansion of hMAGE-A3-specific CD4+ and CD8+ T-cells with the latter peaking at remarkable levels and persisting for several months. T-cells reacting against epitopes fully conserved between simian and human MAGE-A3 were identified. Humoral immunity was demonstrated by the detection of circulating MAGE-A3 antibodies. These preclinical data establish the capacity for the Ad:MG1 vaccination to engage multiple effector immune cell populations without causing significant toxicity in outbred NHPs. Clinical investigations utilizing this program for the treatment of MAGE-A3-positive solid malignancies are underway (NCT02285816, NCT02879760).

2.
Mol Ther ; 23(6): 1066-1076, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25807289

ABSTRACT

Oncolytic viruses (OVs) have shown promising clinical activity when administered by direct intratumoral injection. However, natural barriers in the blood, including antibodies and complement, are likely to limit the ability to repeatedly administer OVs by the intravenous route. We demonstrate here that for a prototype of the clinical vaccinia virus based product Pexa-Vec, the neutralizing activity of antibodies elicited by smallpox vaccination, as well as the anamnestic response in hyperimmune virus treated cancer patients, is strictly dependent on the activation of complement. In immunized rats, complement depletion stabilized vaccinia virus in the blood and led to improved delivery to tumors. Complement depletion also enhanced tumor infection when virus was directly injected into tumors in immunized animals. The feasibility and safety of using a complement inhibitor, CP40, in combination with vaccinia virus was tested in cynomolgus macaques. CP40 pretreatment elicited an average 10-fold increase in infectious titer in the blood early after the infusion and prolonged the time during which infectious virus was detectable in the blood of animals with preexisting immunity. Capitalizing on the complement dependence of antivaccinia antibody with adjunct complement inhibitors may increase the infectious dose of oncolytic vaccinia virus delivered to tumors in virus in immune hosts.


Subject(s)
Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Vaccinia virus/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cell Line, Tumor , Chlorocebus aethiops , Disease Models, Animal , Drug Delivery Systems , Feasibility Studies , Female , HeLa Cells , Humans , Injections, Intralesional , Macaca fascicularis/immunology , Male , Neoplasms/blood , Neoplasms/therapy , Neutralization Tests , Pyridones/immunology , Pyridones/pharmacology , Rats , Rats, Inbred F344 , Smallpox Vaccine/blood , Smallpox Vaccine/immunology , Vaccination , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...