Yonsei Med J.  2008 Oct;49(5):811-818. 10.3349/ymj.2008.49.5.811.

Imaging of Viral Thymidine Kinase Gene Expression by Replicating Oncolytic Adenovirus and Prediction of Therapeutic Efficacy

Affiliations
  • 1Division of Nuclear Medicine, Department of Radiology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea. yunmijin@yuhs.ac
  • 2Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea.

Abstract

PURPOSE
We have used a genetically attenuated adenoviral vector which expresses HSVtk to assess the possible additive role of suicidal gene therapy for enhanced oncolytic effect of the virus. Expression of TK was measured using a radiotracer-based molecular counting and imaging system. MATERIALS AND METHODS: Replication-competent recombinant adenoviral vector (Ad-deltaE1B19/55) was used in this study, whereas replication-incompetent adenovirus (Ad-deltaE1A) was generated as a control. Both Ad-deltaE1B19/55-TK and Ad-deltaE1A-TK comprise the HSVtk gene inserted into the E3 region of the viruses. YCC-2 cells were infected with the viruses and incubated with 2'-deoxy-2'-fluoro-beta-D-arabinofuranosyl-5-iodouracil (I-131 FIAU) to measure amount of radioactivity. The cytotoxicity of the viruses was determined, and gamma ray imaging of HSVtk gene was performed. MTT assay was also performed after GCV treatment. RESULTS: On gamma counter-analyses, counts/minute (cpm)/microgram of protein showed MOIs dependency with deltaE1B19/55-TK infection. On MTT assay, Ad-deltaE1B19/55-TK led to more efficient cell killing than Ad-deltaE1A-TK. On plate imaging by gamma camera, both Ad-deltaE1B19/55-TK and Ad-deltaE1A-TK infected cells showed increased I-131 FIAU uptake in a MOI dependent pattern, and with GCV treatment, cell viability of deltaE1B19/55-TK infection was remarkably reduced compared to that of Ad-deltaE1A-TK infection. CONCLUSION: Replicating Ad-deltaE1B19/55-TK showed more efficient TK expression even in the presence of higher-cancer cell killing effects compared to non-replicating Ad-deltaE1A-TK. Therefore, GCV treatment still possessed an additive role to oncolytic effect of Ad-deltaE1B19/55-TK. The expression of TK by oncolytic viruses could rapidly be screened using a radiotracer-based counting and imaging technique.

Keyword

Oncolysis; adenovirus; thymidine kinase; gene therapy; radiotracer

MeSH Terms

Adenoviridae/*genetics/physiology
Cell Line, Transformed
Cell Line, Tumor
Ganciclovir/pharmacology
Gene Expression
Gene Therapy/methods
Genetic Vectors
Humans
*Oncolytic Virotherapy
Oncolytic Viruses/*genetics/physiology
Simplexvirus/genetics
Tetrazolium Salts/analysis
Thiazoles/analysis
Thymidine Kinase/*genetics/metabolism
Transgenes
Viral Proteins/*genetics/metabolism
Virus Replication

Figure

  • Fig. 1 Characterization of adenoviruses expressing HSVtk gene. (A) Specific primer sets corresponding to E1 and HSVtk gene are shown below the diagram of E1 and E3 region of adenovirus genome, and each expected size of PCR products is 479 bp and 700 bp, respectively. (B) PCR product analysis of Ad-ΔE1A, Ad-ΔE1A-TK, Ad-ΔE1B19/55, and Ad-ΔE1B19/55-TK. The presence of each PCR product verified the expression of the E1A gene and the insertion of HSVtk gene.

  • Fig. 2 Measurement of TK expression by I-131 FIAU and gamma counter. The YCC-2 cells were infected with a range of titers of Ad-ΔE1B19/55-TK (■) or Ad-ΔE1A-TK (▲) for 24 to 72 hours and were incubated with I-131 FIAU. On gamma counter analyses of lysed cells, cpm/µg of protein showed MOIs dependency with a linear increase from 1 to 40 MOI at all time points with ΔE1B19/55-TK infection. With Ad-ΔE1A-TK infection, cpm /µg of protein were unchanged over MOIs after 24 hours of infection and slightly increased after 48 and 72 hours of infection with 40 MOI. MOI, multiplicity of infection.

  • Fig. 3 MTT assay after Ad-ΔE1B19/55-TK (■) or Ad-ΔE1A-TK (▲) infection. The cytotoxicity was determined after infection with a range of titers of Ad-ΔE1B19/55-TK or Ad-ΔE1A-TK for 24 to 72 hours. Ad-ΔE1B19/55-TK led to more efficient cell killing than Ad-ΔE1A-TK after 48 and 72 hours of infection and the amount of viruses needed was lower with Ad-ΔE1B19/55-TK for the same cytotoxicity. MOI, multiplicity of infection.

  • Fig. 4 (A) Gamma ray imaging using I-131 FIAU after Ad-ΔE1B19/55-TK or Ad-ΔE1A-TK infection. The cells were infected with Ad-ΔE1B19/55-TK or Ad-ΔE1A-TK at MOIs of 1, 10, and 40 for 48 hours. Both Ad-ΔE1B19/55-TK and Ad-ΔE1A-TK infected cells showed increased I-131 FIAU uptake in a MOI dependent pattern. (B) The differences in uptakes of I-131 FIAU between the two viral infections were larger with higher viral titers. MOI, multiplicity of infection.

  • Fig. 5 Combination treatment with Ad-ΔE1B19/55-TK (■) or Ad-ΔE1A-TK (▲) and GCV. GCV was added at final concentration of 100 µg/mL at 48 hours postinfection. After additional 3 days of incubation, MTT assay was performed. Cell viability of ΔE1B19/55-TK infection was remarkably reduced compared to that of Ad-ΔE1A-TK infection. MOI, multiplicity of infection; GCV, ganciclovir.


Reference

1. Glasgow JN, Bauerschmitz GJ, Curiel DT, Hemminki A. Transductional and transcriptional targeting of adenovirus for clinical applications. Curr Gene Ther. 2004. 4:1–14.
Article
2. Barnett BG, Crews CJ, Douglas JT. Targeted adenoviral vectors. Biochim Biophys Acta. 2002. 1575:1–14.
Article
3. Kay MA, Glorioso JC, Naldini L. Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nat Med. 2001. 7:33–40.
Article
4. Hermiston T. Gene delivery from replication-selective viruses: arming guided missiles in the war against cancer. J Clin Invest. 2000. 105:1169–1172.
Article
5. Kirn D. Clinical research results with dl1520 (Onyx-015), a replication-selective adenovirus for the treatment of cancer: what have we learned? Gene Ther. 2001. 8:89–98.
Article
6. Wildner O, Morris JC, Vahanian NN, Ford H Jr, Ramsey WJ, Blaese RM. Adenoviral vectors capable of replication improve the efficacy of HSVtk/GCV suicide gene therapy of cancer. Gene Ther. 1999. 6:57–62.
Article
7. Raki M, Hakkarainen T, Bauerschmitz GJ, Särkioja M, Desmond RA, Kanerva A, et al. Utility of TK/GCV in the context of highly effective oncolysis mediated by a serotype 3 receptor targeted oncolytic adenovirus. Gene Ther. 2007. 14:1380–1388.
Article
8. Kim J, Cho JY, Kim JH, Jung KC, Yun CO. Evaluation of E1B gene-attenuated replicating adenoviruses for cancer gene therapy. Cancer Gene Ther. 2002. 9:725–736.
Article
9. Bischoff JR, Kirn DH, Williams A, Heise C, Horn S, Muna M, et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science. 1996. 274:373–376.
Article
10. Lee H, Kim J, Lee B, Chang JW, Ahn J, Park JO, et al. Oncolytic potential of E1B 55 kDa-deleted YKL-1 recombinant adenovirus: correlation with p53 functional status. Int J Cancer. 2000. 88:454–463.
Article
11. Dix BR, O'Carroll SJ, Myers CJ, Edwards SJ, Braithwaite AW. Efficient induction of cell death by adenoviruses requires binding of E1B55k and p53. Cancer Res. 2000. 60:2666–2672.
12. Braithwaite AW, Russell IA. Induction of cell death by adenoviruses. Apoptosis. 2001. 6:359–370.
13. Nanda D, Vogels R, Havenga M, Avezaat CJ, Bout A, Smitt PS. Treatment of malignant gliomas with a replicating adenoviral vector expressing herpes simplex virus-thymidine kinase. Cancer Res. 2001. 61:8743–8750.
14. Haviv YS, Takayama K, Glasgow JN, Blackwell JL, Wang M, Lei X, et al. A model system for the design of armed replicating adenoviruses using p53 as a candidate transgene. Mol Cancer Ther. 2002. 1:321–328.
15. Edholm D, Molin M, Bajak E, Akusjärvi G. Adenovirus vector designed for expression of toxic proteins. J Virol. 2001. 75:9579–9584.
Article
16. Lambright ES, Amin K, Wiewrodt R, Force SD, Lanuti M, Propert KJ, et al. Inclusion of the herpes simplex thymidine kinase gene in a replicating adenovirus does not augment antitumor efficacy. Gene Ther. 2001. 8:946–953.
Article
17. Zinn KR, Chaudhuri TR, Buchsbaum DJ, Mountz JM, Rogers BE. Simultaneous evaluation of dual gene transfer to adherent cells by gamma-ray imaging. Nucl Med Biol. 2001. 28:135–144.
Article
18. Johnson M, Sato M, Burton J, Gambhir SS, Carey M, Wu L. Micro-PET/CT monitoring of herpes thymidine kinase suicide gene therapy in a prostate cancer xenograft: the advantage of a cell-specific transcriptional targeting approach. Mol Imaging. 2005. 4:463–472.
19. Jacobs A, Voges J, Reszka R, Lercher M, Gossmann A, Kracht L, et al. Positron-emission tomography of vector-mediated gene expression in gene therapy for gliomas. Lancet. 2001. 358:727–729.
20. Cascante A, Abate-Daga D, Garcia-Rodríguez L, González JR, Alemany R, Fillat C. GCV modulates the antitumoural efficacy of a replicative adenovirus expressing the Tat8-TK as a late gene in a pancreatic tumour model. Gene Ther. 2007. 14:1471–1480.
Article
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr