Vous êtes sur la page 1sur 12

Available online at www.sciencedirect.

com
R

Virology 309 (2003) 282–293 www.elsevier.com/locate/yviro

A mosaic adenovirus possessing serotype Ad5 and serotype Ad3 knobs


exhibits expanded tropism
Koichi Takayama,a Paul N. Reynolds,a,b Joshua J. Short,a Yosuke Kawakami,a
Yasuo Adachi,a Joel N. Glasgow,a Marianne G. Rots,a,c Victor Krasnykh,a,d
Joanne T. Douglas,a,d and David T. Curiela,d,*
a
Division of Human Gene Therapy, The University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
b
Chest Clinic, Royal Adelaide Hospital, 275 North Terrace, Adelaide, South Australia 5000, Australia
c
Department of Therapeutic Gene Modulation, University Center of Pharmacy, University of Groningen,
Deusinglaan 1, 9713 AV Groningen, The Netherlands
d
Department of Medicine, Pathology, and Surgery, and the Gene Therapy Center, The University of Alabama at Birmingham,
Birmingham, AL 35294-2172, USA

Received 12 July 2002; returned to author for revision 22 November 2002; accepted 2 December 2002

Abstract
The efficiency of cancer gene therapy with recombinant adenoviruses based on serotype 5 (Ad5) has been limited partly because of
variable, and often low, expression by human primary cancer cells of the primary cellular-receptor which recognizes the knob domain of
the fiber protein, the coxsackie and adenovirus receptor (CAR). As a means of circumventing CAR deficiency, Ad vectors have been
retargeted by utilizing chimeric fibers possessing knob domains of alternate Ad serotypes. We have reported that ovarian cancer cells possess
a primary receptor for Ad3 to which the Ad3 knob binds independently of the CAR–Ad5 knob interaction. Furthermore, an Ad5-based
chimeric vector, designated Ad5/3, containing a chimeric fiber proteins possessing the Ad3 knob, demonstrates CAR-independent tropism
by virtue of targeting the Ad3 receptor. Based on these findings, we hypothesized that a mosaic virus possessing both the Ad5 knob and
the Ad3 knob on the same virion could utilize either primary receptor, resulting in expanded tropism. In this study, we generated a dual-knob
mosaic virus by coinfection of 293 cells with Ad5-based and Ad5/3-based vectors. Characterization of the resultant virions confirmed the
incorporation of both Ad5 and Ad3 knobs in the same particle. Furthermore, this mosaic virus was able to utilize either receptor, CAR and
the Ad3 receptor, for virus attachment to cells. Enhanced Ad infectivity with the mosaic virus was shown in a panel of cell lines, with
receptor profiles ranging from CAR-dominant to Ad3 receptor-dominant. Thus, this mosaic virus strategy may offer the potential to improve
Ad-based gene therapy approaches by infectivity enhancement and tropism expansion.
© 2003 Elsevier Science (USA). All rights reserved.

Keywords: Adenovirus; Ad3; Ad5; CAR; Chimeric vector

Introduction of tissue and cell types, its ease of production to high titers,
and its in vivo stability. There are, however, some limita-
Cancer gene therapy using adenoviral vectors is a prom- tions associated with the use of adenovirus serotype 5 (Ad5)
ising treatment alternative (Gomez-Navarro et al., 1999). To vectors for cancer gene therapy. One disadvantage is related
date, most adenoviral vectors have been based on serotype to the expression profile of the primary adenovirus receptor,
5 of species C, because of its ability to infect a wide variety CAR,1 which binds to the knob domain of the Ad5 fiber

1
* Corresponding author. Division of Human Gene Therapy, 901 South Abbreviations used: CAR, coxsackie and adenovirus receptor; Ad5,
19th Street, BMR2 508, University of Alabama at Birmingham, Birming- serotype 5 adenovirus; Ad3, serotype 3 adenovirus; Ad5/3, Ad5 containing
ham, AL 35294-2172. Fax: ⫹1-205-975-7476. a chimeric fiber protein possessing the Ad3 knob; AdMS, mosaic adeno-
E-mail address: david.curiel@ccc.uab.edu (D.T. Curiel). virus; Ad7, serotype 7 adenovirus; Ad5/7, Ad5 containing a chimeric fiber

0042-6822/03/$ – see front matter © 2003 Elsevier Science (USA). All rights reserved.
doi:10.1016/S0042-6822(03)00067-9
K. Takayama et al. / Virology 309 (2003) 282–293 283

Fig. 1. Strategy for mosaic virus generation. The virus genome of Ad5- and Ad5/3-based vectors were transferred into 293 cells by coinfection of both viruses.
The cells infected by both viruses express two types of fibers, the Ad5 wild-type fiber and the Ad5/3 chimeric fiber. Both types of fibers assemble on the
virion at random and generate mosaic viruses.

protein (Bergelson et al., 1997; Henry et al., 1994; Louis et species B in adenovirus classification schemes and binds to
al., 1994; Tomko et al., 1997). Since the efficiency of Ad5 a distinct non-CAR receptor that has not yet been identified
gene transfer primarily depends on the level of CAR, cells (Defer et al., 1990; Gall et al., 1996; Stevenson et al., 1995).
or tissues which express low levels of CAR may be refrac- As expected, the chimeric Ad5/3 adenovirus showed altered
tory to gene therapy with Ad5-based vectors (Cripe et al., tissue tropism and improved infectivity in primary ovarian
2001; Hemmi et al., 1998; Kaner et al., 1999; Kelly et al., cancer cells and various cancer cell lines, characterized by
2000; Li et al., 1999; Miller et al., 1998; Nalbantoglu et al., the levels of CAR, compared to Ad5 (Davidoff et al., 1999;
2001; Pickles et al., 1998; Zabner et al., 1997). Kanerva et al., 2002; Stevenson et al., 1997).
Consequently, strategies to modify Ad tropism to cir- Considering these lines of evidence, we hypothesized
cumvent CAR deficiency have been developed, based on that a mosaic virus displaying both the Ad5 and the Ad3
the use of bispecific retargeting molecules or genetic capsid knobs on the same virion could utilize either the CAR
modifications (Krasnykh et al., 2000). One genetic approach and/or the Ad3 receptor, resulting in enhanced infectivity.
to the modification of Ad tropism involves the incorporation In this study, we have generated such a mosaic adenovirus.
of targeting ligands in capsid proteins, most commonly the Compared to both the Ad5 and the Ad5/3 vectors possessing
fiber. An alternative approach exploits the fact that while knobs with a single receptor specificity, this mosaic Ad has
Ad5 binds to CAR, the fiber knobs of other Ad serotypes significantly enhanced infectivity in various cancer cell
recognize distinct cellular receptors. This has led to the lines and, thus, may offer the potential to improve Ad-based
hypothesis that efficient, CAR-independent gene transfer cancer gene therapy approaches.
could be accomplished by substituting the fiber proteins, or
simply the fiber knob domains, from an Ad5 vector back-
bone with the homologous proteins from an alternative
Results
serotype—a process known as “pseudotyping” (Havenga et
al., 2001; Krasnykh et al., 1996; Miyazawa et al., 1999;
Generation of mosaic virus possessing Ad5 and Ad3
Stevenson et al., 1997; Von Seggern et al., 2000; Zabner et
knobs
al., 1999). In this regard, we have reported the successful
generation of Ad5 virions containing fibers consisting of the
A set of four mosaic adenoviruses designed to display
tail and shaft domains of Ad5 fiber and the knob domain of
both the Ad5 and the Ad3 fiber knobs was generated by
Ad3 (Ad5/3) (Krasnykh et al., 1996). Ad3 is assigned to
coinfection of 293 cells with Ad5luc1 and Ad5/3luc1 at
particle ratios of 90:10, 70:30, 50:50, 30:70 (Fig. 1). DNA
was extracted from each crude virus solution and quantified
protein possessing the Ad7 knob; hCAR, human CAR; VP, viral particle;
FBS, fetal bovine serum; MLP, major late promoter; RLU, relative light
by real-time quantitative PCR. An amount of each mosaic
unit; PVDF, polyvinylidene difluoride; TBST, Tris-buffered saline plus virus preparation, corresponding to 1 ⫻ 108 copies of the
Tween. E4, was used to infect 2 ⫻ 105 HeLa cells. The parental
284 K. Takayama et al. / Virology 309 (2003) 282–293

primers amplified only Ad5/3luc1 and AdMSluc1. The use


of the MLP-specific primers indicated that equal amounts of
viral DNA were employed as template. These results there-
fore confirm that the population of mosaic vectors,
AdMSluc1, contained two separate viral genomes, derived
from Ad5luc1 and Ad5/3luc1, which were encapsidated by
the mosaic capsids.

Mosaic virions incorporate both Ad5 and Ad5/3 fibers

Since the two viral genomes incorporated into the mosaic


virus population differ only in the knob region of the fiber,
we tested our prediction that AdMSluc1 would contain two
fiber proteins, the wild-type Ad5 fiber and the chimeric Ad5
fiber with the Ad3 knob domain, Ad5/3. To examine fiber
incorporation during mosaic virus assembly, we performed
Fig. 2. Mosaic virus generation by coinfection of Ad5 and Ad5/3 at various Western blotting analysis using the monoclonal antibody
ratios. HeLa cells were infected by crude virus lysate of Ad51uc1, Ad5/
(MoAb) 4D2 that recognizes the common Ad5 shaft tail
31uc1, mixtures of Ad51uc1⫹Ad5/3luc1 at various ratios (open bar), or
four kinds of mosaic viruses, AdMSluc1 (closed bar). Luciferase activity domain of both types of fiber. As shown in Fig. 4, this
was measured after 48 h incubation and is expressed as relative light units analysis of Ad5luc1 and Ad5/3luc1 revealed a single band
(RLU) per milligram of protein. Each point represents the mean of three corresponding to each fiber monomer protein (lanes 1 and
experiments ⫾ SD. 7). Fibers from Ad5luc1 and Ad5/3luc1 exhibited different
electrophoretic mobility via SDS–PAGE and were identi-
fied easily by MoAb 4D2. In contrast, AdMSluc1, and a
vectors, Ad5luc1 and Ad5/3luc1, were employed as con-
70:30 mixture of Ad5luc1⫹Ad5/3luc1, displayed two bands
trols, together with various mixtures of these two vectors.
corresponding to the Ad5 wild-type fiber and the Ad5/3
The luciferase activities were measured after 48 h, as shown
chimeric fiber (lanes 3 and 5). In both cases, the intensity of
in Fig. 2. Ad5/3luc1 showed 4.2 times higher luciferase
the upper band, corresponding to the Ad5/3 chimeric fiber,
activity in HeLa cells than Ad5luc1, while the luciferase
activities of the various mixtures of Ad5luc1 ⫹ Ad5/3luc1
ranged between those of 100% Ad5luc1 and 100% Ad5/
3luc1, increasing with the Ad5/3luc1 content of the mixture.
Surprisingly, each of four mosaic viruses showed a higher
luciferase activity than an equal number of particles of
Ad5/3luc1 alone. The mosaic virus generated by coinfecting
293 cells at an Ad5:Ad5/3 ratio of 70:30 showed the highest
luciferase activity in HeLa cells, 2.8 times and 12 times
higher than Ad5/3luc1 and Ad5luc1, respectively. This ob-
servation encouraged us to perform quantitative analysis
using the purified mosaic virus.

The mosaic virus population contains two separate viral


genomes

Based on the preliminary results, we selected the ratio of


70% Ad5luc1 and 30% Ad5/3luc1 for a large-scale produc-
tion of the mosaic virus (AdMSluc1). The mosaic
AdMSluc1, Ad5luc1, and Ad5/3luc1 were purified by two
rounds of cesium chloride density centrifugation. The virus Fig. 3. Amplication of viral DNA by PCR. DNA extracted from 1 ⫻ 108
particle number (VP) of each purified virus was determined VP of each virus was amplified by PCR with primer pairs corresponding to
from the absorbance of 260 nm. PCR was performed with major late promoter region, Ad5 knob region, and Ad3 knob region. Each
primers specific for either the Ad5 or the Ad3 knob, while region was amplified in 25 or 28 cycles of PCR. The template DNA was
primers specific for the major late promoter (MLP) were extracted from Ad5luc1 (lane 1), a mixture of 70% Ad5luc1 ⫹ 30%
Ad5/31uc1 (lane 2), AdMSluc1 (lane 3), or Ad5/3luc1 (lane 4). All PCR
used to quantify the viral genome. As shown in Fig. 3, the products showed the correct size fragment confirmed by size marker (lane
Ad5 knob-specific primers amplified only the Ad5luc1 and M) and PCR product from each backbone plasmid as a positive control
AdMSluc1 viral genomes, while the Ad3 knob-specific (lane C).
K. Takayama et al. / Virology 309 (2003) 282–293 285

Fig. 4. Western blotting for fiber protein. 2 ⫻ 108 VP of each purified virus sample were dissociated in SDS sample buffer and heated at 96°C for 3 min.
Viruses, bound to immobilized MoAb 1D6.14 on a 96-well plate, were also solubilized in SDS buffer after the binding reaction. All samples were separated
on a 4 to 12% gradient polyacrylamide gel followed by electrotransfer onto PVDF membrane. After being blocked with TBST-casein, the membrane was
treated with MoAb 4D2, followed by treatment with goat anti-mouse IgG-HRP conjugate. The color reaction was developed by incubation of the membrane
with Sigma Fast diaminobenzidine. Lane 1, Ad5luc1 without treatment; 2, Ad5luc1 with MoAb treatment; 3, Ad5luc1⫹Ad5/31uc1 mixture without treatment;
4, Ad51uc1⫹Ad5/31uc1 mixture with treatment; 5, AdMS1uc1 without treatment; 6, AdMSluc1 with treatment; 7, Ad5/3luc1 without treatment; 8,
Ad5/3luc1 with treatment.

was weaker than the lower band, corresponding to the Ad5 cells, based on their expression of the primary receptors
wild-type fiber. AdMSluc1 also seemed to have more Ad5 for Ad5 (CAR) and Ad3. After infection with each virus,
wild-type fiber than chimeric fiber, similar to the mixture of the luciferase activities in this panel of cell lines were
Ad5⫹Ad5/3. Following densitometric analysis of band in- measured. In our previous work, we reported that
tensity, lanes 1, 3, 5, and 7 were similar, indicating that SKOV3.ip1 and OV-4 express the Ad3 receptor domi-
equivalent numbers of VPs contained a similar total amount nantly, while 293HEK cells express CAR as the domi-
of fiber protein. nant receptor (Kanerva et al., 2002). Consistent with our
These viruses were also applied to an ELISA plate for previous results, SKOV3.ip1 and OV-4 showed signifi-
specific virus binding to an immobilized anti-Ad5 knob cantly higher luciferase activity with Ad5/3luc1 than
MoAb, 1D6.14. After washing, virus still bound to the wells Ad5luc1, as shown in Fig. 5. Conversely, some cell lines
was solubilized in SDS buffer and analyzed by SDS–PAGE such as C33A, H157, and 293HEK cells were more
with each intact virus as a control. The lysate of Ad5luc1 susceptible to Ad5luc1 infection, presumably due to their
bound to the immobilized MoAb showed the same band as dominant CAR expression. The luciferase activity of the
intact Ad5luc1 (lanes 1 and 2) and the lysate from the well Ad5luc1 and Ad5/3luc1 mixture was also examined in
incubated with Ad5/3luc1 showed no band (lanes 7 and 8), each cell line and showed intermediate values between
indicating that MoAb 1D6.14 is able to bind the Ad5 knob those for Ad5luc1 and Ad5/3luc1. No significant en-
specifically without any cross-reaction with the Ad5/3 chi- hancement of infectivity was shown by this virus mixture.
meric fiber. We obtained additional evidence about the However, the luciferase activity with AdMSluc1 was superior
specificity of MoAb 1D6.14 from the lysate of the Ad5luc1 to levels noted with Ad5luc1 and Ad5/3luc1 in all cell lines
⫹ Ad5/3luc1 mixture. After the binding reaction with the tested. AdMSluc1 showed a 300-fold increase in luciferase
MoAb 1D6.14, only the Ad5 fiber protein was detected in activity compared to Ad5luc1, in OV-4 cells. Likewise,
the well incubated with Ad5luc1 ⫹ Ad5/3luc1 mixture AdMSluc1 showed 5.9 times higher luciferase activity than
(lanes 3 and 4). Finally, we treated the AdMSluc1 in the that of Ad5/3luc1 in C33A cells. These data revealed that
same way and detected both fiber bands in the mosaic virus the mosaic virus infected cells more efficiently than
lysates following binding to the ELISA plate (lanes 5 and Ad5luc1 and Ad5/3luc1, regardless of the dominant knob
6). Based on these results, we concluded that the majority of receptor. Furthermore, the mosaic virus luciferase activity
the virions in the AdMSluc1 population contain both the was higher than the sum total of luciferase activities with
Ad5 wild-type fiber and the Ad5/3 chimeric fiber within the Ad5luc1 and Ad5/3luc1 in all cells tested, indicating that the
same particle. presence of Ad5 and Ad3 knobs on the same virion creates
a synergistic increase in Ad infectivity: we term this phe-
Infectivity profile of mosaic adenovirus in various cell nomenon the “mosaic effect.”
lines
Analysis of mosaic Ad binding using purified Ad3 and
The mosaic virus demonstrated enhanced infectivity Ad5 knob protein
on HeLa cells compared with Ad5luc1 and Ad5/3luc1, as
shown in Fig. 2. However, it was unclear whether the Although we showed an infectivity enhancement with
infectivity enhancement was specific for this cell line. To the mosaic virus in various cell lines, we wished to
address this question, we examined the infectivity profile determine the manner in which the mosaic Ad utilized
of AdMSluc1 in various cell lines, as compared to CAR and/or the Ad3 receptor during infection. To clarify
Ad5luc1 and Ad5/3luc1. We selected nine cell lines in- this point, we performed infection-blocking experiments
cluding ovarian cancer, non-small-cell lung cancer, pan- with purified Ad5 knob and/or Ad3 knob. For these
creatic cancer, glioma, cervical cancer, and 293HEK experiments, we selected two cell lines: C33A as a CAR-
286 K. Takayama et al. / Virology 309 (2003) 282–293

Fig. 5. Infectivity profile of Ad5, Ad5/3, and mosaic virus in various cell lines. Ovarian cancer cells, SKOV3.ipl and OV-4, pancreas cancer cells, Panc-I,
lung cancer cells, NCI-H157 and NCI-H358, uterine cervical cancer cells, HeLa and C33A, glioma cells, U118, and 293HEK cells were infected by Ad5luc1
(open bar), Ad5/3luc1 (closed bar), a mixture of Ad5luc1⫹Ad5/3luc1 (gray bar), or AdMSluc1 (checked bar) at 100 VP/cell. The luciferase activity was
measured after 48 h incubation and expressed as relative light units (RLU). Each point represents the mean of three experiments ⫾ SD.

dominant cell line and SKOV3.ip1 as an Ad3 receptor-


dominant cell line. In fact, a higher percentage of C33A
cells than SKOV3.ip1 cells are CAR-positive by FACS
analysis, as shown in Fig. 6, and a rough correlation
was shown between CAR expression and luciferase ac-
tivity with Ad5luc1 in these cell lines. With C33A cells,
purified Ad5 knob protein blocked gene transfer of mo-
saic Ad significantly at the lowest concentration of 0.1
␮g/ml, and this blocking effect increased in a dose-
dependent manner, as shown in Fig. 7A. Purified Ad3
knob protein did not show any suppressive effect even at
the highest concentration of 100 ␮g/ml, but the combi-
nation of Ad5 and Ad3 knob protein blocked the mosaic
virus infection more strongly than Ad5 knob alone. On
the other hand, purified Ad3 knob blocked the mosaic
virus infection in SKOV3.ip1 cells in a dose-dependent
manner, as shown in Fig. 7B. However, infectivity of the
mosaic virus was not blocked by Ad5 knob, a finding
Fig. 6. FACS analysis for CAR expression in tumor cell lines. 2 ⫻ 105 of consistent with our previous results (Kanerva et al.,
SKOV3.ipl, HeLa, and C33A cells were incubated with primary MoAb 2002). Further, the addition of Ad5 knob to Ad3 knob
RmcB to human CAR. The cells were then washed with 1 ml PBS/BSA/ showed no additional effect. These data demonstrated
azide and incubated with the secondary FITC-labeled goat anti-mouse IgG.
Then 1 ⫻ 104 cells were analyzed by flow cytometry. Expression levels of
that the mosaic virus is able to bind to CAR-dominant
CAR and negative control are shown as areas filled in gray and black, cells with Ad5 wild-type fiber and to Ad3 receptor-
respectively. dominant cells with Ad5/3 chimeric fiber.
K. Takayama et al. / Virology 309 (2003) 282–293 287

Fig. 7. Mosaic virus infectivity in the presence of purified Ad3 and/or Ad5 knob protein. 1 ⫻ 105 (A) C33A cells and (B) SKOV3.ipl cells were preincubated
with the indicated concentrations of each recombinant knob protein for 10 min followed by the AdMSluc1 infection at 100 VP/cell. The knob proteins used
for competition were as follows: closed square: Ad3 knob; closed diamond: Ad5 knob; closed triangle: Ad3 ⫹ Ad5 knob containing each knob protein at
the indicated concentration. A luciferase assay was performed after 48 h incubation, and the luciferase activity is expressed as relative light units (RLU). Each
point represents the mean of three experiments ⫾ SD.

Virus infectivity is not enhanced by free fiber protein Likewise, Ad5/3luc1 infectivity was also not affected by
Ad5 knob protein. We repeated the same experiment with
We next sought to determine if the engagement of one each virus at 1000 VP/cell and obtained similar results (data
Ad receptor serotype by its cognate knob was sufficient to not shown). Thus, these studies confirmed that each knob
improve the infectivity of the virus bearing the other sero- protein alone could not induce the infectivity enhancement.
type knob, resulting in an improvement in infectivity. We Furthermore, this was also consistent with the previous
therefore examined the infectivity enhancement of Ad5luc1 result that the mixture of both viruses did not show any
and Ad5/3luc1 in the presence of the respective free knob infectivity enhancement regardless of the mixture ratio, as
proteins. HeLa cells were infected with Ad5luc1 or Ad5/ shown in Fig. 2.
3luc1 in the presence of various concentrations of Ad5 or
Ad3 knob protein. As shown in Fig. 8, the luciferase activ- Cytoplasmic tail domain of CAR is not involved in the
ities with Ad5luc1 and Ad5/3luc1 infection were suppressed mosaic effect
by recombinant Ad5 knob and Ad3 knob, respectively, in a
dose-dependent manner. At a concentration of 100 ␮g/ml of We investigated the possibility of a contribution of
each knob protein, the luciferase activities of the relevant CAR–Ad3 receptor interaction to mosaic Ad infectivity
adenovirus were below 5% of those without knob protein. enhancement. We hypothesized that any receptor interac-
Importantly, the luciferase activity of Ad5luc1 showed no tion resulting in positive cooperativity of mosaic infectivity
change at the highest concentration of Ad3 knob protein. would require the intracellular domain of CAR. To directly

Fig. 8. Infectivity of Ad5 and Ad5/3 in the presence of purified Ad3 or Ad5 knob protein. 1 ⫻ 105 HeLa cells were preincubated with the indicated
concentrations of (A) purified Ad5 knob protein or (B) purified Ad3 knob protein for 10 min followed by Ad5luc1 (closed diamond) or Ad5/3luc1 (closed
square) infection at 100 VP/cell. Luciferase assay was performed after 48 h incubation, and the luciferase activity is expressed as relative light units (RLU).
Each point represents the mean of three experiments ⫾ SD.
288 K. Takayama et al. / Virology 309 (2003) 282–293

Fig. 9. Correlation between mosaic virus infectivity and CAR expression level. 1 ⫻ 105 parental U118 cells (open bar), modified U118-hCAR cells (gray
bar), and U118-hCAR-tailless cells (closed bar) were infected by Ad5luc1, Ad5/3luc1, and AdMSluc1 at 100 VP/cell. Luciferase assay was performed after
48 h incubation. Each luciferase activity with Ad5luc1 or AdMSluc1 is expressed as the ratio to the Ad5/3 luciferase activity.

address this issue, we utilized the CAR-negative U118 hu- 1999; Miller et al., 1998). Moreover, it was also reported
man glioma cell line and two genetic variants expressing that CAR protein is down regulated in highly tumorigenic
either human CAR (U118-hCAR) or an hCAR deletion prostate cancer cell lines (Okegawa et al., 2000). Due to
mutant lacking the intracellular domain (U118-hCAR-tail- variable expression of CAR on human primary cancer cells,
less). Importantly, parental U118 cells and the two hCAR the utility of Ad5 as a cancer gene therapy vector is com-
derivatives appear to express the Ad3 receptor equally, as promised, limiting overall efficiency of cancer gene therapy.
evidenced by similar luciferase activities following Ad5/ On this basis, systems to circumvent intervening tumor-
3luc1 infection (data not shown). Initially, we compared the associated CAR deficiency are required.
luciferase activities of Ad5luc1 and Ad5MSluc1 in all three In this regard, native Ad5 tropism can be modified to
cell lines, expressed as a percentage of Ad5/3luc1 luciferase circumvent CAR deficiency and to enhance the adenovirus
activity for purposes of normalization. As expected, infectivity. One approach is pseudotyping, i.e., retargeting
Ad5luc1 transgene activity in the hCAR and hCAR-tailless Ad by creating chimeric fibers possessing knob domains
expressing cell lines was increased 11-fold and 16-fold, derived from alternate serotypes which bind to receptors
respectively, compared to the CAR-negative parental line as other than CAR. To this end, we have constructed nonrep-
shown in Fig. 9. Likewise, mosaic virus luciferase activity licating Ads containing chimeric fibers with the tail and
increased 6-fold in U118-hCAR cells and 7.5-fold in the shaft domains of Ad serotype 5 and the knob domain of
U118-hCAR-tailless line versus parental U118 cells. Impor- serotype 3 (Krasnykh et al., 1996). Our previous work has
tantly, we observed no reduction in AdMSluc1 infectivity in revealed that a distinct Ad3 receptor exists in ovarian cancer
the U118-hCAR-tailless cells versus those expressing full- cells based on a novel knob binding assay and that the
length CAR. These findings suggest that the CAR cytoplas- Ad5/3 chimeric vector is retargeted to the Ad3 receptor
mic domain does not contribute to the infectivity enhance- (Kanerva et al., 2002). Furthermore, competition assays
ment of mosaic virus. In other words, CAR and the Ad3 with purified Ad5 and Ad3 knob protein suggested that the
receptors do not interact with each other through their cy- knob regions of Ad5 and Ad5/3 interact with their cognate
toplasmic tails to enhance the mosaic virus infectivity. receptor independently. These observations led us to make
a dual-knob mosaic virus possessing both Ad5 and Ad3
knob regions. In this regard, coinfection of 293 cells with
Discussion Ad5 and Ad5/3 gave rise to a population of viruses pos-
sessing both the Ad5 wild-type fiber and the Ad5/3 chimeric
A major obstacle to be overcome in Ad5-based cancer fiber on the same virion.
gene therapy has been the paucity of the primary receptor, From competition assays with purified knob, we con-
CAR, on human primary tumor cells. Variable expression of firmed that the mosaic virus can attach to either cellular
CAR has been documented in many cancer types including receptor, CAR or Ad3 receptor. As expected, this mosaic
glioma, melanoma, bladder cancer, rhabdomyosarcoma, virus enhanced the Ad infectivity in various cell lines. The
neuroblastoma, and ovarian cancer (Cripe et al., 2001; lucifease activity mediated by the mosaic virus exceeded the
Davidoff et al., 1999; Fechner et al., 2000; Hemmi et al., sum total of that with Ad5luc1 and Ad5/3luc1 in all cells
1998; Kanerva et al., 2002; Kelly et al., 2000; Li et al., tested, suggesting synergistic effect on infection. Interest-
K. Takayama et al. / Virology 309 (2003) 282–293 289

ingly, recombinant Ad5 knob protein did not block the Concerning the mosaic variation, the ratio of Ad5 and
mosaic virus infectivity in HeLa cells despite the fact that a Ad3 knob number on the virion seems to be important, as
significant suppression effect was observed with anti-Ad5 shown in Fig. 2. Since the mosaic virus population is het-
knob antibody. Taken together, the mosaic effect seemed to erogeneous, a part of the population may not contribute to
be synergistic rather than additive. Since the affinity of each the infectivity enhancement due to the deviated ratio of
knob for its cognate receptor is unchanged, the coexistence knob. This means that the data in this study may represent
of both knobs on the same virion likely contributes to the the total effect of a heterogeneous virus population with
synergistic infectivity enhancement. various knob ratios. Therefore, selection of the mosaic virus
One explanation for the synergistic infectivity enhance- with optimal knob ratio could potentially improve the mo-
ment is that binding of the Ad3 knob to the Ad3 receptor saic effect more intensely. If the spatial relationship be-
triggers an alteration in the CAR–Ad5 knob affinity, with tween both knobs and their receptors is involved in the
the interaction between the two receptors inducing confor- mosaic effect, alignment of each knob on the virion is
mational or functional alterations of CAR. Previous studies responsible for it.
with mutated CAR have reported that the cytoplamic tail In conclusion, we have described a mosaic adenovirus
functions as a sorting domain for basolateral targeting in generated by coinfection of 293 cells of Ad5 and Ad5/3
epithelial cells (Cohen et al., 2001) and is not essential for chimeric virus. This mosaic virus displays expanded tro-
adenoviral infection (Van’t Hof and Crystal, 2001). How- pism and enhances the Ad infectivity in various cancer cell
ever, other recent reports have suggested that overexpressed lines. Thus, mosaic virus strategy has a possibility to im-
CAR acts as a tumor inhibitor in prostate cancer (Okegawa prove the effect of cancer gene therapy in various contexts.
et al., 2000) and might have an unknown function other than
sorting. In this study, the comparison between the U118-
hCAR cells and U118-hCAR-tailless cells indicated that Materials and methods
CAR and the Ad3 receptor do not seem to interact with each
other via cytoplasmic domain. Cell culture
Another possible explanation of the mosaic effect is a
positional advantage of the Ad5 knob on the mosaic virus Human cervical cancer cell lines HeLa and C33A, hu-
anchored to the Ad3 receptor. If the Ad3 receptor and CAR man non-small-cell lung cancer cell lines NCI-H157 and
colocalize on the cell surface, the Ad5 knob may easily NCI-H358, human pancreatic cancer cell line Panc-1, hu-
access to CAR close to the Ad3 receptor, allowing compe- man breast cancer cell line AU-565, and the human glioma
tition with recombinant Ad5 knob to be overcome. cell line U118 were obtained from the American Type
The pathway of intracellular trafficking of Ad5 differs Culture Collection (Manassas, VA). The 293 human trans-
from that of Ad3 (Defer et al., 1990). Ad5 uses the cyto- formed embryonal kidney cell line was purchased from
plasmic route preferentially after internalization, while Ad3 Microbix (Toronto, Ontario, Canada). Human ovarian ade-
is sequestered within phagosomes. A study with an Ad5/7 nocarcinoma cell line SKOV3.ip1 was obtained from Dr.
chimeric vector possessing the Ad7 fiber on the Ad5 capsid Janet Price (M.D. Anderson Cancer Center, Houston, TX).
reported that this Ad5/7 chimera traffics via the lysosomal Human ovarian adenocarcinoma cell line OV-4 was ob-
pathway as well as Ad7 (Miyazawa et al., 1999). If the fiber tained from Dr. Timothy J. Eberlein (Harvard Medical
protein modulates the virus trafficking pathway, the mosaic School, Boston, MA). All cell lines were cultured in the
virus may utilize both pathways to the nucleus. This is recommended media at 37°C in a humidified atmosphere of
another possible reason for synergistic enhancement of 5% CO2.
transgene expression. The precise mechanism for the mo-
saic effect observed in this study will be elucidated by the Construction of cell lines expressing hCAR and an hCAR
detailed analysis of Ad3 receptor once this has been cloned. truncation mutant that lacks the cytoplasmic domain
As the result of the presence of both the Ad5 and the Ad3
knobs on the same virion, the mosaic virus displays ex- To assess the role of the cytoplasmic domain of CAR in
panded tropism covering the infectivity spectrum of both infection by the mosaic Ad, we engineered a truncated form
Ad5 and Ad3 and may be promising for cancer gene therapy of human CAR (hCAR) lacking this domain. The 2.4-kb
approaches involving gene transfer to cells or tissues refrac- BamHI-NotI fragment carrying the hCAR cDNA was first
tory to Ad5 and Ad3 infection. Transgene expression from subcloned from pcDNAI.hCAR (obtained from Robert W.
various preexisting Ad5-based vectors should be enhanced Finberg, Harvard Medical School, Boston, MA) into the
by this mosaic formation strategy in combination with Ad5/ mammalian expression vector pcDNA3 (Invitrogen, Carls-
3-based vector. An Ad5/7 chimeric vector may also be bad, CA). A truncation mutant designated hCAR-tailless, in
available for mosaic virus formation since, similar to Ad3, which a stop codon was inserted into the cDNA at the
Ad7 belongs to subgroup B. This mosaic virus strategy is an position corresponding to amino acid residue 261 (Freimuth
easy and feasible method for infectivity enhancement and et al., 1999), was constructed by PCR mutagenesis using
tropism expansion. pcDNA3-hCAR as the template. The PCR product was
290 K. Takayama et al. / Virology 309 (2003) 282–293

inserted into the BamHI and NotI sites of pcDNA3 to give PCR amplification of viral genome
pcDNA3-hCARtailless. The integrity of the construct was
verified by DNA sequencing. For 20 min 1 ⫻ 108 VP of Ad5luc1, Ad5/3luc1, or
Human U118 glioma cells, which are refractory to Ad AdMSluc1 were incubated in 0.1% SDS and 1 mM EDTA
infection due to a paucity of CAR although they express the at 56°C to extract the viral DNA. The DNA was diluted
␣v integrins necessary for virus internalization (Miller et al., 1/1000 in TE and 1 ␮l of this solution was used as a
1998), were stably transfected with pcDNA3-hCARtailless template for PCR. Each viral genome was amplified in a
or with pcDNA3-hCAR as a control. Individual single-cell PCR reaction mixture (Qiagen, Valencia, CA) containing 50
clones of U118-hCAR-tailless and U118-hCAR were iso- nM primer pairs with 25 or 28 cycles of denaturation (94°C,
lated and expanded by selection in the presence of 400 1 min), annealing (50°C, 1 min), and extension (72°C, 1
min). The sequences of the primers were as follows:
␮g/ml G418.
Ad5knob-sense: 5⬘ AGTGCTCATCTTATTATAAGA3⬘;
Ad3knob-sense: 5⬘CGCACATCCTATGTTATG3⬘; knob-
Recombinant adenoviruses antisense: 5⬘CACCACCGCCCTATCCTGAT3⬘; MLP-sense:
5⬘GGTTAATTAAGCATGTCCCTGACTCGCAT3⬘; MLP-
Two replication-incompetent Ad vectors containing a antisense: 5⬘TTGCGCGTGCACCTGGTGCCCGACGA3⬘.
firefly luciferase transgene cassette in place of the deleted The Ad5luc1 and Ad5/3luc1 backbone plasmids were used as
E1 region were used. Ad5luc1 was generated in our labo- positive control templates.
ratory and has been described previously (Krasnykh et al., To quantify the viral genome DNA in the crude virus
2001). The genome of Ad5/3luc1 (Ad containing chimeric solutions, the extracted DNA was amplified by real-time
fibers with the tail and shaft domains of Ad serotype 5 and PCR as reported previously (Adachi et al., 2001). The viral
the knob domain of serotype 3) was constructed by homol- DNA was isolated from 200 ␮l of each crude viral solution
ogous DNA recombination in Escherichia coli using the using a Blood DNA kit (Qiagen). Viral DNA was eluted
previously described plasmid pNEB.PKF5/3 in a scheme with 100 ml of elution buffer [10 mM Tris–Cl (pH 8.5)].
described by Dmitriev et al. (1998). The vector of interest One microliter of eluted sample was analyzed by real-time
PCR using a Light Cycler (Roche) to evaluate the Ad5 E4
was rescued by transfecting 293 cells with the resultant Ad
copy number. Oligonucleotides corresponding to the sense
genome. The viruses were propagated on 293 cells and
strand of the Ad5 E4 region (5⬘-TGACACGCATACTCG-
purified by two rounds of cesium chloride density centrif-
GAGCTA-3⬘: 34,885–34,905 nt), the antisense strand of the
ugation. The VP concentration was determined spectropho-
E4 region (5⬘-TTTGAGCAGCACCTTGCATT-3⬘: 34,977–
tometrically, using a conversion factor of 1.1 ⫻ 1012 viral 34,958 nt), and a TaqMan probe (5⬘-CGCCGCCCATG-
particles per absorbance unit at 260 nm (Maizel et al., CAACAAGCTT-3⬘: 34,930 – 34,951 nt) were synthesized
1968), and standard plaque assays on 293 cells were per- and used as primers and probe for real-time PCR analysis.
formed to determine infectious particles (Mittereder et al., The PCR conditions were as follows: 35 cycles of denatur-
1996). The ratio of VP:infectious particles was 20, 61, and ation (94°C, 20 s), annealing (55°C, 20 s), and extension
15 for Ad5luc1, Ad5/3luc1, and AdMSluc1, respectively. (72°C, 30 s). An Ad backbone vector pTG3602 (Transgene,
Strasbourg, France) was used to generate a standard curve
Generation of mosaic adenovirus for Ad E4 DNA copy number.

Mosaic adenoviruses, designated AdMSluc1, were gen- Western blotting


erated by coinfection of 293 cells with both Ad5luc1 and
Western blotting for fiber protein was performed as de-
Ad5/3luc1. Monolayers of 293 cells in T75 flasks were
scribed previously (Pereboev et al., 2001; Seki et al., 2002).
coinfected with a total of 2 ⫻ 109 particles of Ad5luc1 and
Briefly, 2 ⫻ 108 purified Ad virions (Ad5luc1, Ad5/3luc1,
Ad5/3luc1 in mixtures containing 100, 90, 70, 50, 30, 10, or
or AdMSluc1) were diluted in 2⫻ SDS sample buffer (Bio-
0% of Ad5luc1. After infection for 3 h at 37°C, the infection Rad, Hercules, CA) and heated at 96°C for 3 min. Virus
medium was replaced with fresh complete medium, and the samples were separated on a 4 to 12% gradient polyacryl-
293 cells were incubated for 72 h. The 293 cells were amide gel followed by electrotransfer onto a polyvinylidene
harvested and resuspended in 1 ml of DMEM:F12 medium difluoride (PVDF) membrane. After blocking with TBST-
containing 2% FBS and subjected to four freeze-thaw cy- casein [Tris-buffered saline (10 mM Tris–HCl, pH 7.4, 150
cles. The crude viral lysates were used in the initial exper- mM NaCl) plus Tween 20 to 0.05% and casein to 0.5%],
iments. Based on the results of these preliminary experi- membranes were treated with mouse monoclonal antibody,
ments, a ratio of Ad5luc1:Ad5/3luc1 of 7:3 was chosen for MoAb 4D2 (1:2000 dilution). Then, the membrane was
coninfection of 293 cells for the large-scale preparation of incubated with goat anti-mouse IgG-HRP conjugate
the mosaic adenovirus, AdMSluc1. Purification and titering (DAKO, Carpinteria, CA). The color reaction was devel-
were performed as described above. oped by incubation of the membrane with Sigma Fast dia-
K. Takayama et al. / Virology 309 (2003) 282–293 291

minobenzidine (Sigma). MoAb 4D2 directed against the tail were solubilized in 1⫻ SDS buffer and applied to a gradient
domain of Ad5 fiber protein was generated at the University polyacrylamide gel. Western blotting was performed as
of Alabama at Birmingham Hybridoma Core Facility (Dmi- described above.
triev et al., 2000; Karayan et al., 1994).
Competitive binding assay
Recombinant fiber knob protein production
To investigate the ability of recombinant Ad5 and Ad3
Recombinant Ad5 and Ad3 fiber knob proteins with knobs to block adenovirus infection, Ad5luc1 and Ad5/
N-terminal 6⫻ His tags were expressed in E. coli using the 3luc1 were added to cells in the presence of purified knob
pQE30 expression vector (Qiagen). To increase protein proteins. Monolayers of C33A, SKOV3.ip1, and HeLa cells
yields, the expressed Ad3 fiber knob protein was purified in 12-well plates were preincubated with increasing concen-
from inclusion bodies using the BugBuster Protein Extrac- trations of Ad5 or Ad3 knob in 500 ␮l DMEM:F12 con-
tion Reagent (Novagen, Madison, WI) inclusion body pro- taining 2% FBS for 10 min at room temperature. Ad5luc1,
tocol as recommended by the manufacturer. The protein was Ad5/3luc1, or AdMSluc1 were added at 100 or 1000 VP/
further purified using the Talon metal affinity resin (Clon- cell, diluted in 100 ␮l of DMEM:F12 containing 2% FBS,
tech, Palo Alto, CA) as recommended by the manufacturer. followed by 1 h incubation at room temperature. The cells
The concentration of the purified proteins was determined were washed once with DMEM:F12 containing 2% FBS
by Bio-Rad DC protein assay. The ability of each knob and maintained in complete media. After 48 h of incubation
protein to form a homotrimer was verified by Western blot at 37°C, the cells were lysed and luciferase assays per-
of unboiled samples. The primary antibody used for detec- formed as described below. In the experiment with the
tion was the monoclonal anti-polyhistidine clone HIS-1 anti-Ad5 knob monoclonal antibody, various amounts of
antibody (Sigma) and the secondary antibody was peroxi- 1D6.14 were preincubated with 1 ⫻ 108 VP of each virus at
dase-conjugated goat antimouse IgG (DAKO). RT for 30 min in a total volume of 20 ␮l HBS. Then, the
virus–antibody complexes were diluted to 1 ml in DMEM:
Virus binding to immobilized monoclonal anti-Ad5 knob F12 containing 2% FBS, and 100 ␮l aliquots were added to
antibody 1 ⫻ 105 cells in 12-well plates.

Monoclonal antibody, MoAb 1D6.14, which specifically Ad-mediated gene transfer assays
recognizes the trimeric Ad5 fiber knob protein, has been
described previously (Douglas et al., 1996). Briefly, anti- In 12-well plates 1 ⫻ 105 cells were infected for 3 h at
knob MoAbs were generated by established methods after 37°C at 10, 100, and 1000 VP/cell by adding Ad5luc1,
immunization of BALB/c mice with Ad5, followed by two Ad5/3luc1, or AdMSluc1 diluted in 500 ␮l of DMEM:F12
rounds of immunization with purified recombinant Ad5 containing 2% FCS. Cells were washed once with DMEM:
knob. Sensitized lymphocytes were fused with P3-X63- F12 containing 2% FCS and incubated for 48 h in complete
Ag8.653 cells. The reactivity of the hybridoma supernatants medium. The luciferase activity in the cell lysate was de-
with trimeric Ad5 knob was determined in an ELISA. The termined with a luciferase assay kit (Luciferase Assay Sys-
ability of the hybridoma supernatants to neutralize Ad5 tem, Promega) and a FB12 luminometer (Zylux Corp.).
infection was determined in an endpoint cytopathic effect Background luciferase activities were subtracted from the
assay. Reactivity with the Ad fiber shaft can therefore be readings.
excluded.
MoAb 1D6.14 or control mouse IgG (Sigma) were di- Determination of receptor expression by flow cytometry
luted in 50 mM NaHCO3 (pH 9.6) at a concentration of 5
␮g/ml and immobilized on a Nunc-Maxisorp ELISA plate Cells grown in T75 flasks were washed with PBS, har-
overnight. After unbound MoAb was removed, each well vested by incubating with 0.53 mM EDTA in PBS, and
was washed three times with TBS-T (50 mM Tris–HCl, pH resuspended in PBS containing 0.1% sodium azide and 1%
7.6, in normal saline). Each well was then filled with 200 ␮l BSA Cells (2 ⫻ 105) were incubated with 1 ml of a 1:50
of DMEM:F12 media containing 10% FBS to block non- dilution of primary MoAb RmcB to human CAR for 1 h at
specific binding at room temperature for 2 h. After blocking, 4°C. The MoAb RmcB was produced using a hybridoma
each well was washed three times with TBS and used for purchased from ATCC (Fechner et al., 2000). An isotype-
virus binding. Placed in the well and incubated at RT for 2 h matched normal mouse IgG1 was used as negative control.
were 8 ⫻ 107 or 8 ⫻ 108 VP of Ad5luc1, Ad5/3luc1, The cells were washed once with 1 ml of PBS/BSA/azide
AdMSluc1, or a mixture of Ad5luc1 ⫹ Ad5/3luc1 in 80 ␮l and incubated with 1 ml of 1:100 dilution of the secondary
of DMEM:F12 containing 2% FBS. Then, 10 ␮l of the virus FITC-labeled goat anti-mouse IgG (Jackson Immunore-
solution in each well was used to infect 1 ⫻ 105 HeLa cells search Laboratories, West Grove, PA). Ten thousand cells
to examine the luciferase activity. The wells were then were analyzed immediately by flow cytometry at the Uni-
washed with TBS three times and viruses bound to the well versity of Alabama at Birmingham FACS Core Facility.
292 K. Takayama et al. / Virology 309 (2003) 282–293

The FITC-positive cell population for each cell line was Gall, J., Kass-Eisler, A., Leinwand, L., Falck-Pedersen, E., 1996. Adeno-
determined by gating cells incubated with buffer only (neg- virus type 5 and 7 capsid chimera: fiber replacement alters receptor
tropism without affecting primary immune neutralization epitopes.
ative control) at 1%. J. Virol. 70, 2116 –2123.
Gomez-Navarro, J., Curiel, D.T., Douglas, J.T., 1999. Gene therapy for
cancer. Eur. J. Cancer 35, 867– 885.
Acknowledgments Havenga, M.J., Lemckert, A.A., Grimbergen, J.M., Vogels, R., Huisman,
L.G., Valerio, D., Bout, A., Quax, P.H., 2001. Improved adenovirus
We thank Aleksandr Pereboev, Larisa Pereboeva, vectors for infection of cardiovascular tissues. J. Virol. 75, 3335–3342.
Hemmi, S., Geertsen, R., Mezzacasa, A., Peter, I., Dummer, R., 1998. The
Mikhali Shakhmatov, and Lioudmila Kaliberova for excel- presence of human coxsackievirus and adenovirus receptor is associ-
lent technical support. This work was supported by National ated with efficient adenovirus-mediated transgene expression in human
Cancer Institute Grant CA83821, The CapCURE Founda- melanoma cell cultures. Hum. Gene Ther. 9, 2363–2373.
tion, The Lustgarten Foundation, United States Department Henry, L.J., Xia, D., Wilke, M.E., Deisenhofer, J., Gerald, R.D., 1994.
of Defense Grant 991018, and the American Cancer Soci- Characterization of the knob domain of the adenovirus type 5 fiber
protein expressed in Escherichia coli. J. Virol. 68, 5239 –5246.
ety.
Kaner, R.J., Worgall, S., Leopold, P.L., Stolze, E., Milano, E., Hidaka, C.,
Ramalingam, R., Hackett, N.R., Singh, R., Bergelson, J., Finberg, R.,
Falck-Pedersen, E., Crystal, R.G., 1999. Modification of the genetic
References program of human alveolar macrophages by adenovirus vectors in vitro
is feasible but inefficient, limited in part by the low level of expression
Adachi, Y., Reynolds, P.N, Yamamoto, M., Wang, M., Takayama, K., of the coxsackie/adenovirus receptor. Am. J. Respir. Cell Mol. Biol. 20,
Matsubara, S., Muramatsu, T., Curiel, D.T., 2001. A midkine promoter- 361–370.
based conditionally replicative adenovirus for treatment of pediatric Kanerva, A., Mikheeva, G.V., Krasnykh, V., Coolidge, C.J., Lam, J.T.,
solid tumors and bone marrow tumor purging. Cancer Res. 61, 7882– Mahasreshti, P.J., Baker, S.D., Straughn, M., Barnes, M.N., Alvarez,
7888. R.D., Hemminki, A., Curiel, D.T., 2002. Targeting adenovirus to the
Bergelson, J.M., Cunningham, J.A., Droguett, G., Kurt-Jones, E.A., Krithi- serotype 3 receptor increases gene transfer efficiency to ovarian cancer
vas, A., Hong, J.S., Horwitz, M.S., Crowell, R.L., Finberg, R.W., 1997. cells. Clin. Cancer Res. 8, 275–280.
Isolation of a common receptor for coxsackie B viruses and adenovi- Karayan, L., Gay, B., Gerfaux, J., Boulanger, P.A., 1994. Oligomerization
ruses 2 and 5. Science 275, 1320 –1323. of recombinant penton base of adenovirus type 2 and its assembly with
Cohen, C.J., Gaetz, J., Ohman, T., Bergelson, J.M., 2001. Multiple regions fiber in baculovirus-infected cells. Virology 202, 782–795.
within the coxsackieviurs and adenovirus receptor cytoplasmic domain Kelly, F.J., Miller, C.R., Buchsbaum, D.J., Gomez-Navarro, J., Barnes,
are required for basolateral sorting. J. Biol. Chem. 276, 25392–25398. M.N., Alvarez, R.D., Curiel, D.T., 2000. Selectivity of TAG-72-tar-
Cripe, T.P., Dunphy, E.J., Holub, A., Vasi, N.H., Mahller, Y.Y., Collins, geted adenovirus gene transfer to primary ovarian carcinoma cells
M.H., Snyder, J.D., Krasnykh, V., Curiel, D.T., Wickham, T., DeGre- versus autologous mesothelial cells in vitro. Clin. Cancer Res. 6,
gori, J., Bergelson, J.M., Currier, M.A., 2001. Fiber knob modifications 4323– 4333.
overcome low, heterogeneous expression of the coxsackievirus-adeno- Krasnykh, V., Belousova, N., Korokhov, N., Mikheeva, G., Curiel, D.T.,
virus receptor that limits adenovirus gene transfer and oncolysis for 2001. Genetic targeting of an adenovirus vector via replacement of the
human rhabdomyosarcoma cells. Cancer Res. 61, 2951–2960. fiber protein with the phage T4 fibritin. J. Virol. 75, 4176 – 4183.
Davidoff, A.M., Stevenson, S.C., McClelland, A., Shochat, S.J., Vanin, Krasnykh, V., Dmitriev, I., Navarro, J.G., Belousova, N., Kashentseva, E.,
E.F., 1999. Enhanced neuroblastoma transduction for an improved Xiang, J., Douglas, J.T., Curiel, D.T., 2000. Advanced generation
antitumor vaccine. J. Surg. Res. 83, 95–99. adenoviral vectors possess augmented gene transfer efficiency based
Defer, C., Belin, M.-T., Cailet-boudin, M.-L., Boulanger, P., 1990. Human upon coxsackie adenovirus receptor-independent cellular entry capac-
adenovirus-host cell interactions: comparative study with members of ity. Cancer Res. 60, 6784 – 6787.
subgroups B and C. J. Virol. 64, 3661–3673. Krasnykh, V.N., Mikheeva, G.V., Douglas, J.T., Curiel, D.T., 1996. Gen-
Dmitriev, I., Kashentseva, E., Rogers, B.E., Krasnykh, V., Curiel, D.T., eration of recombinant adenovirus vectors with modified fibers for
2000. Ectodomain of coxsackievirus and adenovirus receptor geneti- altering viral tropism. J. Virol. 70, 6839 – 6846.
cally fused to epidermal growth factor mediates adenovirus targeting to Li, Y., Pong, R.C., Bergelson, J.M., Hall, M.C., Sagalowsky, A.I., Tseng,
epidermal growth factor receptor-positive cells. J. Virol. 74, 6875– C.P., Wang, Z., Hsieh, J.T., 1999. Loss of adenoviral receptor expres-
6884. sion in human bladder cancer cells: a potential impact on the efficacy
Dmitriev, I., Krasnykh, V., Miller, C.R., Wang, M., Kashentseva, E., of gene therapy. Cancer Res. 59, 325–330.
Mikheeva, G., Belousova, N., Curiel, D.T., 1998. An adenovirus vector Louis, N., Fender, P., Barge, A., Kitts, P., Chroboczek, J., 1994. Cell-
with genetically modified fibers demonstrates expanded tropism via binding domain of adenovirus serotype 2 fiber. J. Virol. 68, 4104 –
utilization of a coxsackievirus and adenovirus receptor-independent 4106.
cell entry mechanism. J. Virol. 72, 9706 –9713. Maizel, J.V.J., White, O., Scharff, M.D., 1968. The polypeptides of ade-
Douglas, J.T., Rogers, B.E., Rosenfeld, M.E., Michael, S.I., Feng, M., novirus. I. Evidence for multiple protein components in the virion and
Curiel, D.T., 1996. Targeted gene delivery by tropism-modified adeno- a comparison of types 2, 7A, and 12. Virology 36, 115–125.
viral vectors. Nat. Biotechnol. 14, 1574 –1578. Miller, C.R., Buchsbaum, D.J., Reynolds, P.N., Douglas, J.T., Gillespie,
Fechner, H., Wang, X., Wang, H., Jansen, A., Pauschinger, M., Scherubl, G.Y., Mayo, M.S., Raben, D., Curiel, D.T., 1998. Differential suscep-
H., Bergelson, J.M., Schultheiss, H.P., Poller, W., 2000. Trans-comple- tibility of primary and established human glioma cells to adenovirus
mentation of vector replication versus Coxsackie-adenovirus-receptor infection: targeting via the epidermal growth factor receptor achieves
overexpression to improve transgene expression in poorly permissive fiber receptor-independent gene transfer. Cancer Res. 58, 5738 –5748.
cancer cells. Gene Ther. 7, 1954 –1968. Mittereder, N., March, K.L., Trapnell, B.C., 1996. Evaluation of the con-
Freimuth, P., Springer, K., Berard, C., Hainfeld, J., Bewley, M., Flanagan, centration and bioactivity of adenovirus vectors for gene therapy.
J., 1999. Coxsackievirus and adenovirus receptor amino-terminal im- J. Virol. 70, 7498 –7509.
munoglobulin V-related domain binds adenovirus type 2 and fiber knob Miyazawa, N., Leopold, P.L., Hackett, N.R., Ferris, B., Worgall, S., Falck-
from adenovirus type 12. J. Virol. 73, 1392–1398. Pedersen, E., Crystal, R.G., 1999. Fiber swap between adenovirus
K. Takayama et al. / Virology 309 (2003) 282–293 293

subgroups B and C alters intracellular trafficking of adenovirus gene Stevenson, S.C., Rollence, M., Marshall-Neff, J., McClelland, A., 1997.
transfer vectors. J. Virol. 73, 6056 – 6065. Selective targeting of human cells by a chimeric adenovirus vector
Nalbantoglu, J., Larochelle, N., Wolf, E., Karpati, G., Lochmuller, H., containing a modified fiber protein. J. Virol. 71, 4782– 4790.
Holland, P.C., 2001. Muscle-specific overexpression of the adenovirus Stevenson, S.C., Rollence, M., White, B., Weaver, L., McClelland, A.,
primary receptor car overcomes low efficiency of gene transfer to 1995. Human adenovirus serotypes 3 and 5 bind to two different
mature skeletal muscle. J. Virol. 75, 4276 – 4282. cellular receptors via the fiber head domain. J. Virol. 69, 2850 –
Okegawa, T., Li, Y., Pong, R.C., Bergelson, J.M., Zhou, J., Hsieh, J.T., 2857.
2000. The dual impact of coxsackie and adenovirus receptor expression Tomko, R.P., Xu, R., Philipson, L., 1997. HCAR and MCAR, the human
on human prostate cancer gene therapy. Cancer Res. 60, 5031–5036. and mouse cellular receptors for subgroup C adenoviruses and group B
Pereboev, A., Pereboeva, L., Curiel, D.T., 2001. Phage display of adeno- coxsackieviruses. Proc. Natl. Acad. Sci. USA 94, 3352–3356.
Van’t Hof, W., Crystal, R.G., 2001. Manipulation of the cytoplasmic and
virus type 5 fiber knob as a tool for specific selection and validation.
transmembrane domains alters cell surface levels of the coxsackie-
J. Virol. 75, 7107–7113.
adenovirus receptor and changes the efficiency of adenovirus infection.
Pickles, R.J., McCarty, D., Matsui, H., Hart, P.J., Randell, S.H., Boucher,
Hum. Gene Ther. 12, 25–34.
R.C., 1998. Limited entry of adenovirus vectors into well-differentiated
Von Seggern, D.J., Huang, S., Fleck, S.K., Stevenson, S.C., Nemerow,
airway epithelium is responsible for inefficient gene transfer. J. Virol.
G.R., 2000. Adenovirus vector pseudotyping in fiber-expressing cell
72, 6014 – 6023. lines, improved transduction of Epstein-Barr virus-transformed B cells.
Roelvink, P.W., Lizonova, A., Lee, J.G., Li, Y., Bergelson, J.M., Finberg, J. Virol. 74, 354 –362.
R.W., Brough, D.E., Kovesdi, I., Wickham, T.J., 1998. The coxsack- Zabner, J., Chillon, M., Grunst, T., Moninger, T.O., Davidson, B.L.,
ievirus-adenovirus receptor protein can function as a cellular attach- Gregory, R., Armentano, D., 1999. A chimeric type 2 adenovirus vector
ment protein for adenovirus serotypes from subgroups A, C, D, E, and with a type 17 fiber enhances gene transfer to human airway epithelia.
F. J. Virol. 72, 7909 –7915. J. Virol. 73, 8689 – 8695.
Seki, T., Dmitriev, I., Kashentseva, E., Takayama, K., Rots, M.G., Suzuki, Zabner, J., Freimuth, P., Puga, A., Fabrega, A., Welsh, M.J., 1997. Lack of
K., Curiel, D.T., 2002. Artificial extension of the adenovirus fiber shaft high affinity fiber receptor activity explains the resistance of ciliated
inhibits infectivity in coxsackievirus and adenovirus receptor-positive airway epithelia to adenovirus infection. J. Clin. Invest. 100, 1144 –
cell lines. J. Virol. 76, 1100 –1108. 1149.

Vous aimerez peut-être aussi