2004 - Retargeting of Adenoviral Infection
2004 - Retargeting of Adenoviral Infection
2004 - Retargeting of Adenoviral Infection
Cancer: 108, 136 –145 (2004) Publication of the International Union Against Cancer
Gene therapy is an emerging and promising modality for vectors possess critical properties required for this endeavor. These
the treatment of malignant melanoma and other neoplasms include a highly evolved gene transfer mechanism, the stability of
for which conventional therapies are inadequate. Various virus particles and the ease of virus production at high titers.6 Most
therapeutic genes have shown promise for tumor cell killing.
However, successful gene therapy depends on the develop- importantly, the capsid structure, genome and replication cycle of
ment of efficient and targeted gene transfer vectors. Here we adenoviruses, particularly of the most commonly employed sero-
describe a novel strategy for targeting of adenovirus-medi- type 5, have been extensively characterized, which allows for the
ated gene transfer to melanoma cells. This strategy com- molecular modifications required for their utilization as gene trans-
bines genetic ablation of native adenoviral tropism with re- fer vectors. The necessity of such modifications is predicated by
directed viral binding to melanoma cells via a bispecific the observation that the primary receptor for Ad5 and other ade-
adapter molecule, a bacterially expressed single-chain dia-
body, scDb MelAd, that binds to both the adenoviral fiber novirus serotypes, CAR is widely expressed on normal tissues
protein and to the high molecular weight melanoma-associ- resulting in nonspecific susceptibility to adenoviral infection. In
ated antigen (HMWMAA). This antigen is widely and specif- addition, reduced or absent expression of CAR has been reported
ically expressed on the surface of melanoma cells and its previously for several tumor types, including melanoma, indicat-
expression is associated with tumor development and pro- ing resistance to adenoviral infection by tumor cells in situ.7–9.
gression. Our results showed specific and strong binding of These considerations of adenoviral biology are paralleled by the
the anti-HMWMAA scFv RAFT3 and the bispecific adapter
scDb MelAd to melanoma cells. In adenoviral infection ex- observation of limited efficacy and vector-related toxicity in pre-
periments, we demonstrated i) substantially (>50-fold) re- clinical and clinical adenoviral gene therapy studies. Therefore, the
duced infectivity of capsid mutant adenoviruses, ii) restored development of tropism-modified, tumor-targeted adenoviral gene
(up to 367-fold increase), CAR-independent and HMWMAA- transfer vectors is a key endeavor in current gene therapy research.
mediated infectivity of these mutant viruses by scDb MelAd Towards this goal, the native tropism of adenoviruses needs to be
specifically in melanoma cells, and iii) higher levels of trans-
gene expression in melanoma cells by fiber mutant virus ablated (infectivity ablation) and a new, tumor-specific tropism
complexed with scDbMelAd, relative to a vector with wild- needs to be engineered into viral particles (retargeting).
type fibers. We confirmed the utility of this targeting strat-
egy with human primary melanoma cells that represent clin-
ically relevant substrates. These experiments established
Abbreviations: Ad5, adenovirus serotype 5; bp, base pairs; CAR, cox-
that the retargeting strategy mediates up to 54-fold in- sackie-adenovirus receptor; EGF, epidermal growth factor; FGF, fibroblast
creased adenoviral gene transfer to CAR-negative melanoma growth factor; HMWMAA, high molecular weight melanoma-associated
cells compared to the vector with native tropism. Hence, the antigen; IGF, insulin-like growth factor; IMAC, immobilized metal affinity
HMWMAA-targeted adenoviral vector lacking native tro- chromatography; MAb, monoclonal antibody; PAGE, polyacrylamide gel
pism exhibits both enhanced specificity and augmented in- electrophoresis; pfu, plaque-forming units; scDb, single-chain diabody;
fectivity of gene transfer to melanoma cells, suggesting that scFv single-chain Fv antibody fragment; sCAR, extracellular domain of
it is feasible to use this vector to improve gene therapy for CAR; TNF␣, tumor necrosis factor alpha; vp, viral particles.
malignant melanoma.
© 2003 Wiley-Liss, Inc.
Grant sponsor: Deutsche Forschungsgemeinschaft; Grant numbers;
Key words: adenovirus targeting; melanoma, single-chain diabody; NE832/1; Grant sponsor: National Institutes of Health; Grant numbers:
high molecular weight melanoma-associated antigen; CAR/integrin- R01 HL67962, P50 CA89019, R01 CA86881, U19 DK57958; Grant spon-
binding ablation sor: CFAR AIDS; Grant number: P30AI27767; Grant sponsor: UAB
Center for AIDS Research; Grant number: P30 A1-27767
FIGURE 6 – Expression of CAR and HMWMAA on primary melanoma cells and scDb MelAd-mediated infection of primary melanoma cells:
transductional targeting and infectivity enhancement. (a) Detection of CAR expression on established melanoma cell lines (SK-MEL-28, A375M
and Mel888) and primary melanocytes isolated from 2 different patients (Pat. 1 and 2) by flow cytometry after staining of cells with the anti-CAR
monoclonal antibody RmcB (thick line) or isotype control (gray). (b) Analysis of HMWMAA expression on primary melanoma cells by flow
cytometry after staining with scFv RAFT3 (thick line) or control (gray). (c) Luciferase assay after transduction of the melanoma cell line
SK-MEL-28 and of primary melanoma cells by AdGL (wild-type capsid), or by AdGLmF (fiber mutant) with or without scDb MelAd. Numbers
indicate ratios of mean RLUs of AdGLmF with scDb to AdGLmF without scDb (upper numbers) or of AdGLmF with scDb to AdGL without
scDb (lower numbers). Mean RLUs and standard deviations (bars) are shown.
mutation of the penton base RGD motif, because melanoma cells a result of the artificial cell culture environment. For this reason,
strongly, but not specifically, express integrins. we examined the developed strategy for retargeting of adenoviral
Tropism-modification of adenoviral vectors for gene therapy infection with freshly purified, primary melanoma cells that rep-
and viral oncolysis is mandated whenever expression of the ade- resent clinically more relevant substrates. Indeed, flow cytometry
novirus receptor CAR is variable, low or absent on tumor cells as analysis of 2 isolates from different patients showed a complete
described for several tumor types including melanoma.8 In fact, paucity of CAR expression in 1 and intermediate CAR expression
recent studies describe CAR as a tumor suppressor gene.54 There- in the other specimen. These results are in accord with a previous
fore, strong expression of CAR in established cell lines might be study of Hemmi et al,8 in which a large number of freshly purified
ScDb FOR RETARGETING OF ADENOVIRUS INFECTION TO MELANOMA 143
tumor cells and tissue-sections was analyzed for CAR expression. The strategy for retargeting of adenoviral infection established
In contrast, all established melanoma cell lines showed strong in our study embodies a platform for further therapeutic develop-
CAR expression. Importantly, we could clearly show with these ment. Clearly, scFvs represent attractive tools for vector targeting
primary cell cultures, representing CAR-negative or intermediate because novel scFvs that bind to a target of interest can be derived
CAR expressing melanoma cells, that the targeted adenovirus by existing technologies such as the selection of phage libraries or
results in specific and substantially enhanced adenoviral infectiv- cloning from monoclonal antibodies. Given that the scDb-targeting
ity. Melanoma cells from both patients showed strong expression adapter is derived from a cell-binding scFv antibody fragment, the
of HMWMAA and AdGLmF complexed to scDb MelAd resulted targeting strategy investigated in our study is flexible and can be
in 54-fold increased transduction of CAR-negative melanoma cells applied to different tumors or targets by switching this scFv
relative to the vector with wild-type capsid. component. Furthermore, to avoid immunogenicity it might be
necessary to replace nonhuman components of the scDb with fully
Our results are in accord with recent reports51,52 on retargeting
human sequences. In this regard, the Vchain of the HMWMAA-
of adenovirus infection with capsid mutant viruses and a distinct
binding scFv was already replaced with a human sequence and a
adapter molecule, the tandem-scFv, indicating a broad applicabil- similar procedure could be applied to the residual domains of the
ity and underlining the potential of the capsid mutant/adapter scDb. Notably, the scDb MelAd can be employed for gene therapy
approach. The study of van Beusechem and co-workers51 exploited of HMWMAA-positive tumors other than melanoma, such as
an EGFR-binding adapter for transduction of brain tumors. Hei- leukemias, osteosarcomas or gliomas. In addition, the specificity of
demann et al. 52 targeted gastric cancers with an adapter binding to our approach can be further increased by incorporation of selective
epithelial cell adhesion molecule. In contrast to our strategy for promoters for transcriptional targeting of gene expression.58 – 60
HMWMAA-targeted adenoviral gene transfer, the former study This concept of combined transductional and transcriptional tar-
shows increased selectivity and efficacy of EGFR-targeted adeno- geting has been described previously with bispecific chemical
viral gene transfer after ablation of the penton-integrin interaction. conjugates targeted to pulmonary endothelium or osteosarco-
These observations indicate that the role of the penton-integrin ma.61,62 Finally, transductional targeting of adenoviral infection
interaction in targeted gene transfer depends on the targeted cell will be pivotal also for our recently described approach of mela-
type (i.e., its integrin expression) and/or the target molecule. noma cell killing by adenoviral oncolysis.63 In this regard, we
Based on the proof-of-concept provided by our study, future observed that CAR-negative primary melanoma cells were resis-
investigations will have to determine the feasibility of the de- tant to cell killing by melanoma-targeted conditionally replicating
scribed adenovirus retargeting strategy in vivo. In previous reports, adenoviruses or even to wild-type adenoviruses (DMN, AAR,
targeting of adenoviral vectors to tumors has been demonstrated unpublished observations).
after loco-regional application of the virus.31,55 However, for tar- In conclusion, we describe in this report a novel strategy for
geting of gene transfer to metastatic melanoma adenoviruses need retargeting of adenoviral infection to malignant melanoma via a
to be applied systemically. Whereas adenoviral gene transfer has tumor antigen and independent of the adenovirus receptor CAR.
been successfully targeted to pulmonary endothelium after sys- Importantly, we demonstrate specific transduction of melanoma
temic application32 the analysis of tumor-targeted vectors in a cells with an efficacy up to 54-fold superior to wild-type adeno-
systemic context is more complex. In this regard, xenograft tumor virus. Thus, the adenovirus retargeting strategy developed in this
models have shown recently that the efficient transduction of report offers a new means to improve efficiency and safety of gene
tumor cells in vivo not only requires tumor-targeted vector tropism therapy for malignant melanoma and other cancers.
but also a means to overcome physical barriers, such as the
extracellular matrix surrounding tumor nests.56 Moreover, mouse ACKNOWLEDGEMENTS
models of spontaneous tumorigenesis, which are currently being We are grateful to Dr. T.J. Eberlein, Dr. I. Hart and Dr. J.
developed for malignant melanoma,57 might be the preferred Schlom for cell lines. Special thanks to Dr. J. Blackwell, Dr. A.
means for in vivo analysis of tumor-targeted gene transfer. As well, Kanerva, T. Korn, Dr. A. Pereboev, Dr. L. Rivera, Dr. T. Seki, Dr.
in vivo studies are required to analyze the stability and immuno- A. Steinkasserer, W. Stöhr, Dr. T. Strong, Dr. K. Suzuki and Dr.
genicity of adapter-targeted adenoviruses after systemic injection A. Volk for their valuable contributions. We would like to ac-
and would furthermore allow for comparison of the different knowledge the Center For Aids Research Flow Cytometry Facility
adapter formats, such as sCAR-ligand fusion proteins, tandem (support provided by CFAR AIDS Core Grant #P30AI27767) and
scFvs and scDbs in this respect. However, analysis of targeted Marion Spell for providing flow cytometry equipment and exper-
adenoviruses in mouse models requires targeting ligands that are tise. DNA sequencing was performed in the DNA Sequencing and
cross-reactive with the corresponding mouse antigen, unlike the Analysis Core of the UAB Center for AIDS Research, supported
scDb MelAd described in this report. by grant P30 A1-27767.
REFERENCES
1. Gomez-Navarro J, Curiel DT, Douglas JT. Gene therapy for cancer. with efficient adenovirus-mediated transgene expression in human
Eur J Cancer 1999;35:2039 –57. melanoma cell cultures. Hum Gene Ther 1998;9:2363–73.
2. Alemany R, Balague C, Curiel DT. Replicative adenoviruses for 9. Barnett BG, Crews CJ, Douglas JT. Targeted adenoviral vectors.
cancer therapy. Nat Biotechnol 2000;18:723–727. Biochim Biophys Acta 2002;1575:1–14.
3. Nabel GJ. Development of optimized vectors for gene therapy. Proc 10. Sotomayor MG, Hu H, Antonia S, Sotomayor EM, Pardoll DM.
Natl Acad Sci U S A 1999;96:324 – 6. Advances in gene therapy for malignant melanoma. Cancer Control
4. Verma IM, Somia N. Gene therapy: promises, problems and pros- 2002;9:39 – 48.
pects. Nature 1997;389:239 – 42. 11. Wilson BS, Imai K, Natali PG, Ferrone S. Distribution and molecular
5. Vile RG, Russell SJ, Lemoine NR. Cancer gene therapy: hard lessons characterization of a cell-surface and a cytoplasmic antigen detectable
and new courses. Gene Ther 2000;7:2– 8. in human melanoma cells with monoclonal antibodies. Int J Cancer
6. Zhang WW. Development and application of adenoviral vectors for 1981;28:293–300.
gene therapy of cancer, Cancer Gene Ther 1999;6:113–38. 12. Natali PG, Imai K, Wilson BS, Bigotti A, Cavaliere R, Pellegrino
7. Miller CR, Buchsbaum DJ, Reynolds PN, Douglas JT, Gillespie GY, MA, Ferrone S. Structural properties and tissue distribution of the
Mayo MS, Raben D, Curiel DT. Differential susceptibility of primary antigen recognized by the monoclonal antibody 653.40S to human
and established human glioma cells to adenovirus infection: targeting melanoma cells. J Natl Cancer Inst 1981;67:591– 601.
via the epidermal growth factor receptor achieves fiber receptor- 13. Harper JR, Reisfeld RA. Inhibition of anchorage-independent growth
independent gene transfer. Cancer Res 1998;58:5738 – 48. of human melanoma cells by a monoclonal antibody to a chondroitin
8. Hemmi S, Geertsen R, Mezzacasa A, Peter I, Dummer R. The pres- sulfate proteoglycan. J Natl Cancer Inst 1983;71:259 – 63.
ence of human coxsackievirus and adenovirus receptor is associated 14. Berd D, Herlyn M, Koprowski H, Mastrangelo MJ. Flow cytometric
144 NETTELBECK ET AL.
determination of the frequency and heterogeneity of expression of Ryan PC, Kaleko M, Stevenson SC. Targeting adenoviral vectors by
human melanoma-associated antigens. Cancer Res 1989;49:6840 – 4. using the extracellular domain of the coxsackie-adenovirus receptor:
15. Burg MA, Grako KA, Stallcup WB. Expression of the NG2 proteo- improved potency via trimerization. J Virol 2002;76:1892–903.
glycan enhances the growth and metastatic properties of melanoma 36. Ebbinghaus C, Al-Jaibaji A, Operschall E, Schoffel A, Peter I, Greber
cells. J Cell Physiol 1998;177:299 –312. UF, Hemmi S. Functional and selective targeting of adenovirus to
16. Kageshita T, Kuriya N, Ono T, Horikoshi T, Takahashi M, Wong GY, high-affinity Fcgamma receptor I-positive cells by using a bispecific
Ferrone S. Association of high molecular weight melanoma-associ- hybrid adapter. J Virol 2001;75:480 – 489.
ated antigen expression in primary acral lentiginous melanoma lesions 37. Haisma HJ, Grill J, Curiel DT, Hoogeland S, van Beusechem VW,
with poor prognosis. Cancer Res 1993;53:2830 –3. Pinedo HM, Gerritsen WR. Targeting of adenoviral vectors through a
17. Kupsch JM, Tidman NH, Kang NV, Truman H, Hamilton S, Patel N, bispecific single-chain antibody. Cancer Gene Ther 2000;7:901– 4.
Newton Bishop JA, Leigh IM, Crowe JS. Isolation of human tumor- 38. Nettelbeck DM, Miller DW, Jérôme V, Zuzarte M, Watkins SJ,
specific antibodies by selection of an antibody phage library on Hawkins RE, Müller R, Kontermann RE. Targeting of adenovirus to
melanoma cells. Clin Cancer Res 1999;5:925–31. endothelial cells by a bispecific single-chain diabody directed against
18. Siccardi AG, Buraggi GL, Callegaro L, Mariani G, Natali PG, Abbati, the adenovirus fiber knob domain and human endoglin (CD105). Mol
A, Bestagno M, Caputo V, Mansi L, Masi R, et al. Multicenter study Ther 2001;3:882–91.
of immunoscintigraphy with radiolabeled monoclonal antibodies in 39. Kang N, Hamilton S, Odili J, Wilson G, Kupsch J. In vivo targeting
patients with melanoma. Cancer Res 1986;46:4817–22. of malignant melanoma by 125Iodine- and 99mTechnetium-labeled
19. Spitler LE, del Rio M, Khentigan A, Wedel NI, Brophy NA, Miller single-chain Fv fragments against high molecular weight melanoma-
LL, Harkonen WS, Rosendorf LL, Lee HM, Mischak RP, et al. associated antigen. Clin Cancer Res 2000;6:4921–31.
Therapy of patients with malignant melanoma using a monoclonal 40. Kupsch JM, Tidman N, Bishop JA, McKay I, Leigh I, Crowe JS.
antimelanoma antibody-ricin A chain immunotoxin. Cancer Res Generation and selection of monoclonal antibodies, single-chain Fv
1987;47:1717–23. and antibody fusion phage specific for human melanoma-associated
20. Yang, HM, Reisfeld RA. Pharmacokinetics and mechanism of action antigens. Melanoma Res 1995;5:403–11.
of a doxorubicin-monoclonal antibody 9.2.27 conjugate directed to a 41. Neri D, Natali PG, Petrul H, Soldani P, Nicotra MR, Vola R, Rivella
human melanoma proteoglycan. J Natl Cancer Inst 1988;80:1154 –9. A, Creighton AM, Neri P, Mariani M. Recombinant anti-human
21. Eary JF, Schroff RW, Abrams PG, Fritzberg AR, Morgan AC, Kasina melanoma antibodies are versatile molecules. J Invest Dermatol 1996;
S, Reno JM, Srinivasan A, Woodhouse CS, Wilbur DS. Successful 107:164 –70.
imaging of malignant melanoma with technetium-99m-labeled mono- 42. Noronha EJ, Wang X, Desai SA, Kageshita T, Ferrone S. Limited
clonal antibodies. J Nucl Med 1989;30:25–32. diversity of human scFv fragments isolated by panning a synthetic
22. Lamki LM, Zukiwski AA, Shanken LJ, Legha SS, Benjamin RS, phage-display scFv library with cultured human melanoma cells.
Plager CE, Salk DF, Schroff RW, Murray JL. Radioimaging of J Immunol 1998;161:2968 –76.
melanoma using 99mTc-labeled Fab fragment reactive with a high 43. Wang B, Chen YB, Ayalon O, Bender J, Garen A. Human single-
molecular weight melanoma antigen. Cancer Res 1990;50:904s–908s. chain Fv immunoconjugates targeted to a melanoma-associated chon-
23. Ghose T, Ferrone S, Blair AH, Kralovec Y, Temponi M, Singh M, droitin sulfate proteoglycan mediate specific lysis of human mela-
Mammen M. Regression of human melanoma xenografts in nude mice noma cells by natural killer cells and complement. Proc Natl Acad Sci
injected with methotrexate linked to monoclonal antibody 225.28 to U S A 1999;96:1627–32.
human high molecular weight-melanoma associated antigen. Cancer 44. Abken H, Hombach A, Heuser C, Reinhold U. A novel strategy in the
Immunol Immunother 1991;34:90 – 6. elimination of disseminated melanoma cells: chimeric receptors en-
24. Fodstad O, Faye R, Hoifodt HK, Skovlund E, Aamdal S. Immuno- dow T cells with tumor specificity, Recent Results Cancer Res 2001;
bead-based detection and characterization of circulating tumor cells in 158:249 – 64.
melanoma patients. Recent Results Cancer Res 2001;158:40 –50. 45. Tordsson JM, Ohlsson LG, Abrahmsen LB, Karlstrom PJ, Lando PA,
25. Bergelson, JM, Cunningham JA, Droguett G, Kurt-Jones EA, Krithi- Brodin TN. Phage-selected primate antibodies fused to superantigens
vas A, Hong JS, Horwitz MS, Crowell RL, Finberg RW. Isolation of for immunotherapy of malignant melanoma, Cancer Immunol Immu-
a common receptor for Coxsackie B viruses and adenoviruses 2 and 5. nother 2000;48:691–702.
Science 1997;275:1320 –3. 46. Martin F, Kupsch J, Takeuchi Y, Russell S, Cosset FL, Collins M.
26. Wickham, TJ, Mathias P, Cheresh DA, Nemerow GR. Integrins alpha Retroviral vector targeting to melanoma cells by single-chain anti-
v beta 3 and alpha v beta 5 promote adenovirus internalization but not body incorporation in envelope, Hum Gene Ther 1998;9:737– 46.
virus attachment. Cell 1993;73:309 –19. 47. Douglas JT, Miller CR, Kim M, Dmitriev I, Mikheeva G, Krasnykh
27. Roelvink PW, Mi Lee G, Einfeld DA, Kovesdi I, Wickham TJ. V, Curiel DT. A system for the propagation of adenoviral vectors with
Identification of a conserved receptor-binding site on the fiber proteins genetically modified receptor specificities. Nat Biotechnol 1999;17:
of CAR-recognizing adenoviridae. Science 1999;286:1568 –71. 470 –5.
28. Kirby I, Davison E, Beavil AJ, Soh CP, Wickham TJ, Roelvink PW, 48. Alemany R, Curiel DT. CAR-binding ablation does not change bio-
Kovesdi I, Sutton BJ, Santis G. Mutations in the DG loop of adeno- distribution and toxicity of adenoviral vectors. Gene Ther 2001;8:
virus type 5 fiber knob protein abolish high-affinity binding to its 1347–53.
cellular receptor CAR. J Virol 1999;73:9508 –14. 49. Krasnykh VN, Mikheeva GV, Douglas JT, Curiel DT. Generation of
29. Kirby I, Davison E, Beavil AJ, Soh CP, Wickham TJ, Roelvink PW, recombinant adenovirus vectors with modified fibers for altering viral
Kovesdi I, Sutton BJ, Santis G. Identification of contact residues and tropism. J Virol 1996;70:6839 – 46.
definition of the CAR-binding site of adenovirus type 5 fiber protein. 50. Einfeld DA, Schroeder R, Roelvink PW, Lizonova A, King CR,
J Virol 2000;74:2804 –13. Kovesdi I, Wickham TJ. Reducing the native tropism of adenovirus
30. Douglas JT, Rogers BE, Rosenfeld ME, Michael SI, Feng M, Curiel vectors requires removal of both CAR and integrin interactions.
DT. Targeted gene delivery by tropism-modified adenoviral vectors. J Virol 2001;75:11284 –91.
Nat Biotechnol 1996;14:1574 – 8. 51. van Beusechem VW, Grill J, Mastenbroek DC, Wickham TJ, Ro-
31. Gu DL, Gonzalez AM, Printz MA, Doukas J, Ying W, D’Andrea M, elvink, PW, Haisma HJ, Lamfers ML, Dirven CM, Pinedo HM,
Hoganson DK, Curiel DT, Douglas JT, Sosnowski BA, Baird A, Gerritsen WR. Efficient and selective gene transfer into primary
Aukerman SL, Pierce GF. Fibroblast growth factor 2 retargeted ade- human brain tumors by using single-chain antibody-targeted adeno-
novirus has redirected cellular tropism: evidence for reduced toxicity viral vectors with native tropism abolished. J Virol 2002;76:2753– 62.
and enhanced antitumor activity in mice., Cancer Res 1999;59:2608 – 52. Heideman DA, Van Beusechem VW, Offerhaus GJ, Wickham TJ,
14. Roelvink PW, Craanen ME, Pinedo HM, Meijer CJ, Gerritsen WR.
32. Reynolds PN, Zinn KR, Gavrilyuk VD, Balyasnikova IV, Rogers BE, Selective gene transfer into primary human gastric tumors using
Buchsbaum DJ, Wang MH, Miletich DJ, Grizzle WE, Douglas JT, epithelial cell adhesion molecule-targeted adenoviral vectors with
Danilov SM, Curiel DT. A targetable, injectable adenoviral vector for ablated native tropism, Hum Gene Ther 2002 13:1677– 85.
selective gene delivery to pulmonary endothelium in vivo. Mol Ther 53. Iida J, Meijne AM, Oegema TR, Jr., Yednock TA, Kovach NL, Furcht
2000;2:562–78. LT, McCarthy JB. A role of chondroitin sulfate glycosaminoglycan
33. Dmitriev I, Kashentseva E, Rogers BE, Krasnykh V, Curiel DT. binding site in alpha4beta1 integrin-mediated melanoma cell adhe-
Ectodomain of coxsackievirus and adenovirus receptor genetically sion. J Biol Chem 1998;273:5955– 62.
fused to epidermal growth factor mediates adenovirus targeting to 54. Okegawa T, Pong RC, Li Y, Bergelson JM, Sagalowsky AI, Hsieh JT.
epidermal growth factor receptor-positive cells. J Virol 2000 74: The mechanism of the growth-inhibitory effect of coxsackie and
6875– 84. adenovirus receptor (CAR) on human bladder cancer: a functional
34. Kashentseva EA, Seki T, Curiel DT, Dmitriev IP. Adenovirus target- analysis of car protein structure. Cancer Res 2001;61:6592– 600.
ing to c-erbB-2 oncoprotein by single-chain antibody fused to trimeric 55. Rancourt C, Rogers BE, Sosnowski BA, Wang M, Piche A, Pierce
form of adenovirus receptor ectodomain. Cancer Res 2002;62:609 – GF, Alvarez RD, Siegal GP, Douglas JT, Curiel DT. Basic fibroblast
16. growth factor enhancement of adenovirus-mediated delivery of the
35. Kim J, Smith T, Idamakanti N, Mulgrew K, Kaloss M, Kylefjord H, herpes simplex virus thymidine kinase gene results in augmented
ScDb FOR RETARGETING OF ADENOVIRUS INFECTION TO MELANOMA 145
therapeutic benefit in a murine model of ovarian cancer. Clin Cancer 60. McCart J, Wang Z, Xu H, Hu Y, Park B, Alexander H, Bartlett D.
Res 1998;4:2455– 61. Development of a melanoma-specific adenovirus, Mol Ther 2002;6:471.
56. Shayakhmetov DM, Li ZY, Ni S, Lieber A. Targeting of adenovirus 61. Reynolds PN, Nicklin SA, Kaliberova L, Boatman BG, Grizzle WE,
vectors to tumor cells does not enable efficient transduction of breast Balyasnikova IV, Baker AH, Danilov SM, Curiel DT. Combined
cancer metastases, Cancer Res 2002;62:1063– 8. transductional and transcriptional targeting improves the specificity of
57. Tietze MK, Chin L. Murine models of malignant melanoma, Mol Med transgene expression in vivo. Nat Biotechnol 2001;19:838 – 42.
Today 2000;6:408 –10. 62. Barnett B, Tillman B, Curiel D, Douglas J. Dual targeting of adenoviral
58. Vile RG, Hart IR. Use of tissue-specific expression of the herpes vectors at the levels of transduction and transcription enhances the spec-
simplex virus thymidine kinase gene to inhibit growth of established ificity of gene expression in cancer cells. Mol Ther 2002;6:377.
murine melanomas following direct intratumoral injection of DNA. 63. Nettelbeck DM, Rivera AA, Balague C, Alemany R, Curiel DT.
Cancer Res 1993;53:3860 – 4. Novel oncolytic adenoviruses targeted to melanoma: specific viral
59. Nettelbeck DM, Jérôme V, Müller R. Gene therapy: designer promot- replication and cytolysis by expression of E1A mutants from the
ers for tumour targeting.Trends Genet 2000;16:174 – 81. tyrosinase enhancer/promoter. Cancer Res 2002;62:4663–70.