4.5 Article

Polymer stealthing and mucin-1 retargeting for enhanced pharmacokinetics of an oncolytic vaccinia virus

Journal

MOLECULAR THERAPY-ONCOLYTICS
Volume 21, Issue -, Pages 47-61

Publisher

CELL PRESS
DOI: 10.1016/j.omto.2021.03.011

Keywords

-

Funding

  1. Engineering and Physical Sciences Research Council (EPSRC) [EP/L024012/1]
  2. Linacre College Brewer Street Scholarship
  3. Clarendon Scholarship
  4. All Souls Hugh Springer Scholarship
  5. EPSRC [EP/L024012/1] Funding Source: UKRI

Ask authors/readers for more resources

Vaccinia virus (VV) shows potential for cancer treatment due to its tumor tropism, but its clinical utility is limited by poor penetration into tumors. Chemical stealthing techniques improve the pharmacokinetics of VV. Experiments demonstrate that coating VV with amphiphilic polymer and attaching targeting antibodies can enhance its infectivity and circulation time in vivo, potentially improving its therapeutic effectiveness.
Vaccinia virus (VV) is a powerful tool for cancer treatment with the potential for tumor tropism, efficient cell-to-cell spread, rapid replication in cancer cells, and stimulation of anti-tumor immunity. It has a well-defined safety profile and is being assessed in late-stage clinical trials. However, VV clinical utility is limited by rapid bloodstream neutralization and poor penetration into tumors. These factors have often restricted its route of delivery to intratumoral or intrahepatic artery injection and may impede repeat dosing. Chemical stealthing improves the pharmacokinetics of non-enveloped viruses, but it has not yet been applied to enveloped viruses such as VV. In the present study, amphiphilic polymer was used to coat VV, leading to reduced binding of a neutralizing anti-VV antibody (81.8% of polymer-coated VV [PCVV] staining positive versus 97.1% of VV [p = 0.0038]). Attachment of anti-mucin-1 (aMUC1) targeting antibody, to give aMUC1-PCVV, enabled binding of the construct to MUC1. In high MUC1 expressing CAPAN-2 cells, infection with PCVV was reduced compared to VV, while infection was restored with aMUC1 -PCVV. Pharmacokinetics of aMUC1 -PCVV, PCVV, and VV were evaluated. After intravenous (i.v.) injection of 1 x 10(8) viral genomes (VG) or 5 x 10(8) VG, circulation time for PCVV and aMUC1 -PCVV was increased, with similar to 5-fold higher circulating dose at 5 min versus VV.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available