Imaging characteristics, tissue distribution, and spread of a novel oncolytic vaccinia virus carrying the human sodium iodide symporter

PLoS One. 2012;7(8):e41647. doi: 10.1371/journal.pone.0041647. Epub 2012 Aug 17.

Abstract

Introduction: Oncolytic viruses show promise for treating cancer. However, to assess therapy and potential toxicity, a noninvasive imaging modality is needed. This study aims to determine the in vivo biodistribution, and imaging and timing characteristics of a vaccinia virus, GLV-1h153, encoding the human sodium iodide symporter (hNIS.

Methods: GLV-1h153 was modified from GLV-1h68 to encode the hNIS gene. Timing of cellular uptake of radioiodide (131)I in human pancreatic carcinoma cells PANC-1 was assessed using radiouptake assays. Viral biodistribution was determined in nude mice bearing PANC-1 xenografts, and infection in tumors confirmed histologically and optically via Green Fluorescent Protein (GFP) and bioluminescence. Timing characteristics of enhanced radiouptake in xenografts were assessed via (124)I-positron emission tomography (PET). Detection of systemic administration of virus was investigated with both (124)I-PET and 99m-technecium gamma-scintigraphy.

Results: GLV-1h153 successfully facilitated time-dependent intracellular uptake of (131)I in PANC-1 cells with a maximum uptake at 24 hours postinfection (P<0.05). In vivo, biodistribution profiles revealed persistence of virus in tumors 5 weeks postinjection at 10(9) plaque-forming unit (PFU)/gm tissue, with the virus mainly cleared from all other major organs. Tumor infection by GLV-1h153 was confirmed via optical imaging and histology. GLV-1h153 facilitated imaging virus replication in tumors via PET even at 8 hours post radiotracer injection, with a mean %ID/gm of 3.82 ± 0.46 (P<0.05) 2 days after intratumoral administration of virus, confirmed via tissue radiouptake assays. One week post systemic administration, GLV-1h153-infected tumors were detected via (124)I-PET and 99m-technecium-scintigraphy.

Conclusion: GLV-1h153 is a promising oncolytic agent against pancreatic cancer with a promising biosafety profile. GLV-1h153 facilitated time-dependent hNIS-specific radiouptake in pancreatic cancer cells, facilitating detection by PET with both intratumoral and systemic administration. Therefore, GLV-1h153 is a promising candidate for the noninvasive imaging of virotherapy and warrants further study into longterm monitoring of virotherapy and potential radiocombination therapies with this treatment and imaging modality.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Biological Transport
  • Cell Line, Tumor
  • Cell Transformation, Neoplastic
  • Humans
  • Iodine Radioisotopes / metabolism
  • Male
  • Mice
  • Molecular Imaging*
  • Oncolytic Viruses / genetics*
  • Oncolytic Viruses / physiology*
  • Optical Imaging
  • Pancreatic Neoplasms / diagnostic imaging
  • Pancreatic Neoplasms / metabolism
  • Pancreatic Neoplasms / pathology
  • Pancreatic Neoplasms / virology
  • Positron-Emission Tomography
  • Symporters / genetics*
  • Time Factors
  • Tissue Distribution
  • Vaccinia virus / genetics*
  • Vaccinia virus / physiology*
  • Virus Replication*

Substances

  • Iodine Radioisotopes
  • Symporters
  • sodium-iodide symporter

Grants and funding

This work was supported by grants from Genelux Corporation (Research & Development facility in San Diego, California, United States of America). DH, visitor at Memorial Sloan-Kettering Cancer Center, is a graduate student in AAS's laboratory in the Department of Biochemistry and Graduate School of Life Sciences, University of Würzburg, Germany, and is supported by a graduate stipend from The University of Wuerzburg as well as travel grants from Genelux Corporation and Memorial Sloan-Kettering Cancer Center. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.