You are on page 1of 11

Oncogene (2000) 19, 2 ± 12

ã 2000 Macmillan Publishers Ltd All rights reserved 0950 ± 9232/00 $15.00
www.nature.com/onc

A mutant oncolytic adenovirus targeting the Rb pathway produces


anti-glioma e€ect in vivo

Juan Fueyo*,1, Candelaria Gomez-Manzano1, Ramon Alemany1, Polly SY Lee1,


Timothy J McDonnell2, Paraskevi Mitlianga1, Yue-Xi Shi1, VA Levin1, WK Alfred Yung1
and Athanassios P Kyritsis1
1
Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas,
TX 77030, USA; 2Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe
Blvd., Houston, Texas, TX 77030, USA

E€ective anti cancer strategies necessitate the use of otherwise quiescent cells (Nevins, 1992). Two segments
agents that target tumor cells rather than normal tissues. of E1A are important for binding Rb; one includes
In this study, we constructed a tumor-selective adeno- amino acids 30 ± 60 and the other amino acids 120 ± 127
virus, D24, that carries a 24-bp deletion in the E1A (Whyte et al., 1988, 1989). Deletion of either region
region responsible for binding Rb protein. Immunopreci- prevented the formation of detectable E1A/Rb
pitation analyses veri®ed that this deletion rendered D24 complexes in vitro and in vivo (Whyte et al., 1989). In
unable to bind the Rb protein. However, titration the present study, we used these observations to
experiments in 293 cells demonstrated that the D24 develope a mutant adenovirus which encodes a E1A
adenovirus could replicate in and lyse cancer cells with protein with deletion of amino acids 120 ± 127 that
great eciency. Lysis of most human glioma cells was selectively targets cells with abnormal Rb control.
observed within 10 ± 14 days after infection with D24 at Targeting the Rb pathway is important in treating
10 PFU/cell. In vivo, a single dose of the D24 virus gliomas because abnormalities of the p16/Rb/E2F
induced a 66.3% inhibition (P50.005) and multiple pathway are present in most gliomas (Fueyo et al.,
injections, an 83.8% inhibition (P50.01) of tumor 1998a; Gomez-Manzano et al., 1998). Targeting this
growth in nude mice. However, normal ®broblasts or pathway by replacing of lost tumor suppressor activity
cancer cells with restored Rb activity were resistant to through the transfer of p16 and Rb genes has produced
the D24 adenovirus. These results suggest that the E1A- cytostatic e€ects (Fueyo et al., 1998a; Gomez-Manzano
mutant D24 adenovirus may be clinically and therapeu- et al., 1998). Transfer of E2F-1 resulted in powerful
tically useful against gliomas and possibly other cancers anti-cancer e€ect since the exogenous wild-type E2F-1
with disrupted Rb pathway. Oncogene (2000) 19, 2 ± 12. induced apoptosis and inhibited tumor growth in vivo
(Fueyo et al., 1998b). However, treating human glioma
Keywords: oncolytic virus; gene therapy; Rb; E2F; E1A tumors with these adenovirus constructs realistically
cannot a€ect signi®cant portions of the tumor, mainly
because replication-de®cient adenoviral vectors are
unable to transfer the exogenous gene to sucient
Introduction numbers of cancer cells (Puumalainen et al., 1998).
Thus, although targeting the p16/Rb/E2F pathway
The properties of adenoviruses to rely on host cells for produces an anti-cancer e€ect in vitro, this imperfection
their own propagation can be utilized to obtain anti- of the vector system limits the therapeutic e€ect of the
neoplastic e€ects on human tumors. In this regard, gene in vivo. To overcome these diculties, we
adenoviruses synthesize proteins that compel infected combined the selectivity of the anti-cancer e€ect with
cells to replicate the viral DNA. The E1A proteins are a better delivery system in constructing a mutant
the ®rst virus-speci®c polypeptides synthesized after adenovirus with a deletion of eight amino acids in the
adenoviral infection, and are required for viral Rb-binding region of the E1A protein. We hypothe-
replication to occur (Dyson and Harlow, 1992; Flint sized that this mutant virus would be selective for
and Shenk, 1997). The targets of E1A proteins, such as tumors because most normal brain cells are arrested in
Rb, seem to modulate the cell cycle by regulating cell G0, which halts the replication of the mutant
progression from G0 and G1 into S phase. In the case adenovirus. However, this adenovirus would also be
of Rb, binding between the Rb protein and the E1A able to replicate in cancer cells that have disrupted Rb
proteins results in release of E2F from preexisting function. Thus, this virus would kill target cells, and
cellular E2F-Rb complexes. The E2F is then free to amplify and spread the e€ect from cell to cell within
activate both the E2 promoter of the adenovirus and the tumor, but not the surrounding di€erentiated cells.
several cell cycle-regulatory genes. The transcriptional
activation of these cellular genes in turn helps to create
an environment suitable for viral DNA synthesis in
Results

The E1A-mutant D24 adenovirus produced anti-cancer


*Correspondence: J Fueyo, Department of Neuro-Oncology, Box e€ect in vitro
100, University of Texas M.D. Anderson Cancer Center, 1515
Holcombe Boulevard, Houston, Texas, TX 77030, USA The replication-competent D24 virus is a human
Received 7 June 1999; revised 21 September 1999; accepted 28 adenovirus 5 that contains a 24-bp deletion in its
September 1999 E1A region (Figure 1). Since this deletion did not
Oncolytic adenovirus for glioma treatment
J Fueyo et al
3
produce a stop codon, and the D24 could express a
mutant E1A protein (Figure 1), this suggested that the
a
mutant adenovirus can replicate. Immunoprecipitation
experiments con®rmed however that the mutant E1A
protein could not form complexes with the Rb protein
(Whyte et al., 1988, 1989) (Figure 1).
The infectivity of the adenovirus has been previously
tested in a variety of glioma and sarcoma cell lines that
have disruptions in the p16/Rb pathway (Fueyo et al.,
1996a, 1998c). The D24 adenovirus had similar e€ects
on the viability of U-251 MG, D-54 MG, U-87 MG,
U-373 glioma cells and Saos-2 osteosarcoma cells.
Thus, treating any of these cells with the mutant
adenovirus at a multiplicity of the infection (MOI) of
ten produced complete monolayer cytolysis (85% or
b more) within 14 days, with most of the glioma cell
lines, and the Saos-2 osteosarcoma line, showing
cytopathic e€ect within 7 days. These results were
con®rmed by both Trypan blue and crystal violet
viability assays (Figure 2). Dose-dependence experi-
ments revealed that ®ve MOIs of the D24 adenovirus
were enough to induce a noticeable cytopathic e€ect
(Figure 2), although the timing and intensity of the
oncolytic e€ect varied slightly among the cell lines
tested. For example, by 7 days after treatment with ®ve
MOI of D24 adenovirus the oncolytic e€ect was
complete in U-251-MG. In contrast, D-54 MG cells
required 10 MOI to obtain a complete monolayer lysis,
and U-87 MG cells required ®ve MOI and 14 days of
infection.

The replication of D24 is responsible for the anti-cancer


e€ect
Since wild-type adenoviruses can infect and lyse
quiescent ®broblasts (Bischo€ et al., 1996; Heise et
c al., 1997; Rothman et al., 1998; Hay et al., 1999), we
were interested to ascertain whether the D24 had lost
its ability to replicate in and kill normal cells. For these
experiments CCD32-Lu lung ®broblasts were arrested
in G1 phase after seeded at a low density of 56103
cells per well in six-well plates, and cultured with
medium plus 1% serum, and infected with ten plaque-
forming units (PFU) per cell (the maximum dose used
to treat cancer cells) of D24 or UV-D24 (control)
adenovirus. Eight days after the infection no differ-
ences were found in the numbers of D24- and control-
infected cells (3.36103+0.11; 2.56103+0.07, respec-
tively). Moreover, D24 infection did not result in
changes in the morphology of ®broblasts (Figure 3).
These results did not re¯ect an inability of the
adenovirus to infect normal cells because these lung
®broblasts could be infected with 50 MOI (470% of
the cells) of Ad5CMV-GFP (Figure 3). Next, we
infected U-251 MG and U-373 MG cells plated at
Figure 1 (a) Schematic representation of the D24 adenovirus. 56103 cells per well in six-well plates and cultured with
Shown are the two Rb-binding regions of the E1A sequence medium plus 1% serum, with ten PFU/cell of D24 or
(hatched boxes). The 24 nucleotides that have been deleted and UV- D24 adenovirus. Seven days after the infection, we
the corresponding amino acid translation are indicated. (b) observed a massive cytopathic e€ect, with a
Detection of the E1A protein by Western blotting. Twenty-four
hours after infection, total protein from U-251 MG cells infected 94.5+4.6% and 90.1+4.1% decrease in viability of
with Ad5CMV-pA, dl1520, or D24 at a dose of 100 MOI, were D24-infected-U-251 MG and U-373 MG glioma cells,
analysed with anti-E1A antibody as described in Materials and respectively, with respect to the UV-D24-infected cells.
methods. (c) Immunoprecipitation of E1A/host protein com- To con®rm that the D24 adenovirus has di€erent
plexes. Total lysates from U-251 MG cells infected with D24,
dl1520, or Ad5CMV-pA at a dose of 100 MOI were
e€ects on the viability of normal and cancer cells, we
immunoprecipitated using an antibody agarose-conjugated anti- next performed replication assays in which 293 cells
E1A were incubated with cell lysates of either normal or
Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
4
a cancer cells. To obtain the cell lysates from either
CCD32-Lu, D-54 MG or U-251 MG cells were plated
at the same density (104 cells per well in six-well plate),
infected with one MOIs of either the D24 or the UV-
D24 adenovirus, and allowed to grow for 6 days. As
soon as 2 days after the infection with D-54 MG or U-
251 MG cell lysates, 293 cells showed destruction of
the monolayer cultures (Figure 4). In contrast, 293 cell
did not die when the lysates were from normal cells
(Figure 4). These results suggested that the D24
produced progeny virus more eciently in cancer cells
than in normal cells.
Next we used plaque assays to quantify the
cytopathic e€ect in terms of viral titers. Virus yields
were determined by titer in 293 cells at 6 days after the
infection of D-54 MG, U-251 MG and CCD32-Lu with
one MOI of either the D24 adenovirus or the UV-D24
control adenovirus. The D24 titer was 60 and 80 times
higher than the initial dose in two independent
experiments with the D-54 MG lysates and 200 and
400 times higher in the U-251 MG lysate experiments.
By contrast, the initial titer did not increase when the
lysates were from CCD32-Lu cells (Figure 4). The UV-
D24 infected cells did not induce production of new
virus. The di€erences in the viral titers observed in the
experiments with D-54 MG and U-251 MG agreed with
the di€erences in cytopathic e€ect observed in the
crystal violet assays. These experiments showed that the
D24 adenovirus replicated eciently in glioma cells and
that normal ®broblasts restricted the spread of the D24.

Transfer of Rb restricts the D24-adenovirus mediated


cytolysis
To further con®rm the hypothesis that the D24
adenovirus replicates in a cell cycle restricted manner
when the Rb pathway is functionally normal, we next
b
evaluated the ability of the mutant adenovirus to
replicate in cells arrested and expressing wild-type Rb.
For these experiments we used the osteosarcoma cell
line Saos-2, which has a well-characterized disruption
of the Rb pathway (Huang et al., 1988) can be easily
infected with adenovirus (Fueyo et al., 1998c), and has
a well-characterized response to the transfer of an
exogenous Rb (Huang et al., 1988; Fueyo et al., 1998c).
Saos-2 cells were infected with 100 MOI of an
adenoviral vector carrying the exogenous wild-type
Rb cDNA, or the Ad5CMV-pA adenovirus, and 72 h
later were infected with ten MOIs of either the D24 or
the UV-D24 adenovirus. As expected, cells that had
been pretreated with a vector control, Ad5CMV-pA,
were sensitive to the lytic e€ect of the D24 adenovirus
with a complete cytopathic e€ect (90.5+2.6% decrease
in viability) observed within 5 days after the viral
infection. By contrast, cells infected with the
Figure 2 Anti-cancer e€ects of the D24 adenovirus in vitro. (a)
Ad5CMV-Rb acquired an oncolytic-resistant pheno-
Dose-dependence assay. Plated glioma cells were infected with type with an 88.7+3.1% increase in viability, that
D24 adenovirus at indicated MOI. Seven days (U-251 MG), 9 persisted for at least 10 days after infection with the
days (D-54 MG), or 14 days (U-87 MG) after infection, the plates D24 adenovirus (Figure 5).
were stained with crystal violet. (b) Viability assay. Glioma cells
were seeded in six-well culture plates for 20 h, and then infected
Since, the e€ect of D24 is theoretically restricted by
with either D24 or UV-D24 adenovirus at ten PFU/cell. Cells were cell cycle factors, we monitored the changes in the cell
counted in triplicate dishes of each treatment 7 (U-251 MG, U- cycle pro®le of Saos-2 cells in parallel with the
373 MG), 9 (D-54 MG) or 14 (U-87 MG) days after infection, experiments described above. Saos-2 cells infected
and the viability assessed by Trypan blue exclusion. Shown is the
growth of cells treated with D24 (hatched bars) compared with
with the Ad5CMV-Rb were arrested in G1 phase by
control cells (100%). The number of viable cells is the the third day after infection (77.2+3% of Rb-infected
mean+standard deviation (s.d.) of three di€erent wells cells versus 59.7+3% of control-infected cells). The
Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
5
accumulation of cells in G1 correlated with the ®fth day after D24 infection showed that the number of
observed growth inhibition (71.9+3.2%) relative to cells in S phase was 2.4- and 5.5-fold lower in the Rb-
cells infected with an adenovirus control. Flow- pretreated cells, in comparison with adenovirus
cytometric analyses of the cell cycle performed on the control-pretreated cells, in two independent experi-

Figure 3 E€ect of D24 on the viability of normal ®broblasts. (a) Phase-contrast photomicrographs of D-54 MG glioma cells (top)
and CCD32-Lu lung ®broblasts (bottom) 7 days after infection with ten MOI of UV-D24 (left) or D24 adenovirus (right).
Magni®cation, 6100. (b) Infectivity of CCD32-Lu cells. Fluorescence (top) and light microscopic images (bottom) of the same ®eld
of lung ®broblasts 3 days after the transfer of GFP cDNA using 50 MOI of Ad5CMV-GFP. (6100)

Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
6
ments. These results suggest that restoration of Rb To further con®rm the virus-suppressive e€ect of the
rendered D24 unable to induce eciently entry of cells Rb protein, we next examined the ability of the cyclin-
into S phase, and therefore precluding viral replication. dependent-kinase inhibitor p21, a regulator of Rb

a b

Figure 4 Replication assays. (a) Two days after infection, 293 cells treated with supernatant of D24-infected CCD32-Lu (top), D-54
MG (center), and U-251 MG cells (bottom), were photomicrographed. (b) Viral titration. Normal lung ®broblasts and D-54 MG
and U-251 MG glioma cells were seeded at 104 cells per well in six-well plates and cultured with medium with 1% fetal bovine
serum for 24 h and then infected with one PFU/cell of D24 adenovirus (empty bar). Six days after infection, the virus was released
from the cells by three freeze/thaw cycles, and the resulting supernatant was titered on 293 cells. Virus yields are expressed as PFU/
ml. Shown are the results from two independent experiments (solid and hatched bars). (c) Electron microscopy of a cancer cell
treated with ten MOI of D24. Four days after infection, numerous viral particles were observed in the cancer cells (left). An area of
the cell has been enlarged to show the viral structures (right)

Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
7
function, to reduce the e€ect of the D24 on the viability subcutaneously in nude mice, and when tumors
of wild-type Rb cells. D-54 MG cells were infected with reached at least 5 mm in diameter, tumors were treated
100 MOI of an adenoviral vector carrying the with either D24 (n=6) or UV-D24 (n=6) at a dose of
exogenous wild-type p21 cDNA or Ad5CMV-pA, and 56108 PFU per animal every 5 days for a total of ®ve
3 days later were infected with the D24 virus at ten times. D24-treated tumors showed an 83.8% growth
MOI. For cells that had not been infected with inhibition (P50.01, t-student, double sided), with
Ad5CMV-p21 vector, a total cytopathic e€ect tumor regression in two of the six mice tested. Tumors
(74.2+2.9% decrease of viability) was observed 5 showing regression have been followed for over 2
days after the infection with D24. In contrast, p21- months without evidence of regrowth. Hematoxylin-
pretreatment provided partial protection against the eosin staining revealed that, after D24 treatment, the
D24 adenovirus as re¯ected by a 64.2+4% increase in remaining tumor mass consisted largely in necrotic
viability. As was true for the Rb-transfer experiment, tissue. Similar results were obtained in tumors formed
growth inhibition (40+1.9%) was apparent 3 days after the subcutaneous injection of 107 D54 MG cells in
after infection with Ad5CMV-p21. Thus, p21-treated the right ¯ank of nude mice. These experiments
cells showed an overrepresentation in the G1 phase of involved the intratumoral injection of 109 PFU of
the cell cycle in comparison with cells treated with the either D24 (n=5) or UV-D24 (n=5) every 5 ± 7 days for
adenovirus control (91.4+0.4% and 75.3+0.9%, four doses. With these doses, we observed an 85.7%
respectively). reduction in the size of treated tumors relative to the
size of the control tumors (P50.05, t-student, double
sided). As true for the previous group, two of the ®ve
Treatment with D24 induces entry of cells into S phase
D24-treated mice showed tumor regression.
and cell death
In addition to the multiple injection experiments, we
Flow cytometric analysis of the DNA content of the investigated the e€ect of a single low dose of the D24
D24-infected cells showed an accumulation of cells in adenovirus on tumor growth. Tumors in six mice were
the S phase by the third day after the infection. Thus, injected with 107 PFU of the adenovirus control, and
81+10.1% of D24-infected-U-251 MG cells were in the tumors in ®ve mice were treated with a similar dose of
S phase of the cell cycle versus 10+2.3% of the UV- D24. At the end of the experiment, tumors in the D24-
D24-infected cells. Similar results were observed in D- treated group were 66.3% smaller than those in the
54 MG cells, and 77.4% and 77.3% of D-24-infected- UV-D24-treated animals (P50.005, t-student, double
cells were in S phase (in comparison with 14.5% and sided). Despite the low viral dose, all of the D24-
15.3% of the control-infected cells) in two independent treated tumors displayed growth inhibition (Figure 6).
experiments. As mentioned above, accumulation of None of the mice used in these experiments died before
cells in the S phase was also observed in D-24-infected the ending of the experiment. In addition, we did not
Saos-2 cells. At 7 days after infection, cytopathic e€ect observe any sign of systemic toxicity including weight
was widespread. At that moment, the majority of the loss, abdominal swelling, hunched posture, or diarrhea.
cells detached from the dish and cellular debris were
numerous. However, there was only a small population
of cells in the sub-G1 area as assessed by ¯ow Discussion
cytometric analyses. Thus, 7.72+3.8% of D-24-
infected -U-251 MG were in the sub-G1 region Our results indicate that the mutant adenovirus D24 is a
(versus 1.7+0.1 of control infected cells). The potent cytolytic agent against glioma in vitro and in vivo
discrepancy between the morphological and ¯ow- and that the presence of a functionally active Rb protein
cytometric data suggested that the virus-mediated cell in quiescent normal cells could halt this cytopathic e€ect
death was probably due to both cell lysis and of the mutant D24 adenovirus. Thus the E1A-mutant
apoptosis. Consistent with this, electron microscopy protein was unable to deregulate the cell cycle and kill
showed necrosis and apoptosis features in the infected non-cycling wild-type Rb cells. Transfer of exogenous Rb
cells (data not shown). Interestingly, D24 adenovirus to Rb-null cells resulted in resistance to the adenoviral
induced cell death even in mutant-p53 cells such as U- e€ect in previously virus-sensitive cancer cells, thus
251 MG glioma cells (Gomez-Manzano et al., 1996) con®rming the ability of the wild-type Rb protein to
and Saos-2 (Masuda et al., 1987). These results block D24 replication. These results are important for
indicated that although p53 can cooperate with other future therapeutic applications of the D24 adenovirus
viral and cell proteins to enhance adenovirally- because the existence of a cell cycle-mediated mechanism
mediated cell death (Ridgway et al., 1997), wild-type of resistance to the virus suggests that the anticipated
p53 status was not a major determinant of ecient D24 toxicity of an adenovirus expressing a handicapped E1A
replication and thus its oncolytic e€ect. protein will be limited to the very few, occasionally
dividing, reactive glial cells in the adult human brain.
The anti-cancer e€ect of the adenovirus was
The D24 adenovirus suppresses tumor growth in vivo
potent. In vitro, cell destruction was massive even
To study the antitumor e€ect of D24 in vivo, the growth with very low doses of the virus. In vivo a single
of D-54 MG tumors treated with D24 was compared local injection of a low dose of the adenovirus
with the growth of tumors those treated with an UV- resulted in tumor inhibition. Multiple injections
inactivated D24 control virus in a nude mice model. produced tumor regression in four of 11 animals.
Tumor size was measured periodically until tumors in Regression of human gliomas established subcuta-
the control animals achieved the maximum acceptable neously in mice is a strong indication of the power of
burden. In the ®rst group of experiments, pre- the replication-competent strategy. In fact, the
established D-54 MG xenografts were implanted inability of current vectors to transduce sucient
Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
8
numbers of tumor cells in vivo may be the major Ontario, Canada) as follows. The shuttle plasmid pXC1,
hurdle to successful gene therapy of cancer (Roth which contains the adenovirus 5 sequences from 22 ± 5790
and Cristiano, 1997), especially in cases in which the nucleotides, was modi®ed by using phosphorylated muta-
bystander e€ect on nontransduced cells is limited. genic primers to loop out and delete 24 bases, from 919 to
943, of the adenovirus type 5 corresponding to amino acid
Thus, replication-de®cient adenovirus could transfer
residues L, T, C, H, E, A, C, and F of the E1A.
an exogenous gene to less than 15% of glioma cells
in human patients (Puumalainen et al., 1998). Several
factors, however, favor the use of oncolytic Con®rmation of the D24-E1A by sequencing the genome of the
adenoviruses for the treatment of brain tumors. adenovirus
First, gliomas do not metastasize, and therefore an A 1-kb fragment of the E1A region of the adenoviral
ecient local approach should be enough to cure the construct was ampli®ed by polymerase chain reaction (PCR)
disease. Second, every glioma harbors several using sense (5'-GATTGGCCACCATGAGACATAT-
populations of cells expressing di€erent genetic TATCTGC-3') and antisense (5'-CTGTGGCCATTTAA-
abnormalities (Sidransky et al., 1992; Collins and CACGCCATGCA-3') primers. Subsequently, 1 mg of
concentrated PCR sample was added to 1 ml of each primer
James, 1993; Furnari et al., 1995; Kyritsis et al.,
(primer dilution 1 : 3 of 0.1 mg/ml) and the volume was
1996b). Thus, the spectrum of tumors sensitive to the brought to 9.5 ml with distilled water. This mixture of PCR
transfer of a single gene to cancer cells may be fragments and primers was added to a ¯uorescein-labeled
limited. Third, replication competent adenoviruses can reaction mix (Applied Biosystems, Foster City, CA, USA)
infect and destroy cancer cells that are arrested in and sequenced in a 373A automated DNA sequencer
G0. Since gliomas invariably include non-cycling cells, (Applied Biosystems).
this property is important. Finally, the p16-Rb
pathway is abnormal in the majority of gliomas Construction of the D24 by homologous recombination in 293
(Hamel et al., 1993; Henson et al., 1994; Hirvonen et cells
al., 1994; Jen et al., 1994; Schmidt et al., 1994;
To generate the mutant adenovirus, the shuttle vectors pXC1-
Costello et al., 1996, 1997; Fueyo et al., 1996b;
D24, which carries the mutant E1A region of the adenovirus,
Kyritsis et al., 1996a; Ueki et al., 1996), thus making and pBHG10 (Microbix Biosystem Inc.), which contains the
the D24 strategy appropriate for most of these rest of the adenoviral genome except for the E3 region, were
tumors. cotransfected by liposome-mediated method into 293 cells to
A tumor-selective mutant virus seems a promising allow homologous recombination. To generate infectious
solution to the problem of gene delivery that is virions, individual plaques were isolated and ampli®ed, and
hindering the complete success of conventional gene the viral titers were determined by plaque assays.
therapy. Recent advances in the knowledge of how
viruses replicate are being used to design tumor- Adenoviruses
selective oncolytic viruses. In gliomas, three kinds of
Other vectors used included the UV-D24 adenovirus, a D24
viruses have been shown to be useful in animal virus inactivated by ultraviolet light; Ad5CMV-pA, a E1-
models: reoviruses that can replicate selectively in deleted adenovirus that carries no exogenous genes (Alemany
tumors with an activated ras pathway (Co€ey et al., et al., 1996); Ad5CMV-Rb, a E1-deleted adenovirus that
1998); genetically altered herpes simplex viruses carries the Rb cDNA (Fueyo et al., 1998c); Ad5CMV-GFP, a
(Martuza et al., 1991; Mineta et al., 1995; vector that can express the green ¯uorescent protein
Andreanski et al., 1997), including those that can (Alemany et al., 1997); Ad5CMV-p21, that carries the p21
be activated by the di€erent expression of proteins cDNA (Gomez-Manzano et al., 1997); dl1520, a replication
in normal and cancer cells (Chase et al., 1998); and competent adenovirus with a partial deletion of the E1B gene
mutant adenoviruses that are unable to express the (Barker and Berk, 1987).
55-kDa E1B protein and are used to treat p53-
mutant tumors (Bischof et al., 1996; Heise et al., Cell lines and infection conditions
1997; Freytag et al., 1998; Kirn et al., 1998). Taken The human glioma cell lines U-251 MG (homozygous
together, these reports con®rm the relevance of deletion of p16 locus, Arap et al., 1995), U-373 MG (lack
oncolytic viruses as anti-cancer agents. In all three of expression of p16 protein, Fueyo et al., 1998b), and U-87
systems, the goal is the intratumoral spread of the MG (homozygous deletion of p16 locus, Arap et al., 1995);
e€ect and the ability to di€erentiate cancer from the human sarcoma cell line Saos-2 (lack of wild-type Rb
normal cells. Our own studies on the D24 expression, Huang et al., 1998); and the normal lung
adenovirus support the tenet that genetically mod- ®broblast line CCD32-Lu were obtained from the American
i®ed adenoviruses that target cellular pathways at Type Culture Collection (Manassas, VA, USA). The D-54
key points may have both potent and selective anti- MG human glioma cells (lack of expression of p16 protein.
Fueyo et al., 1996a) were a generous gift from Dr Bigner
cancer e€ects in gliomas. Of further importance is (Duke University, Durham, NC, USA). The cell lines were
the presence of functional inactivation of the Rb infected as described previously (Gomez-Manzano et al.,
protein in most human cancers (Sherr, 1996), an 1996; Fueyo et al., 1998b).
observation that suggests that this new therapeutic
approach may be applicable to many malignancies.
Western blot analysis

Materials and methods Total cell lysates were prepared by incubating cells for 1 h at
48C in RIPA bu€er (150 mM NaCl, 1% Triton X-100, 1%
sodium deoxycholate, 0.1% sodium dodecyl sulfate [SDS],
In vitro site-directed mutagenesis
20 mM EDTA, and 50 mM Tris, pH 7.4) at di€erent times
The adenovirus D24 was constructed from a modi®ed after the infection. Thirty micrograms of protein from each
adenovirus type 5 genome (Microbix Biosystem Inc., sample was then subjected to 7% SDS-Tris-glycine gel

Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
9
electrophoresis. The membrane was blocked with 3% nonfat anti-adenovirus-2 E1A protein 13 S-5 (Santa Cruz Biotech-
milk, 0.05% Tween 20, 0.9% NaCl, and 50 mM Tris, pH 7.5, nology Inc., Santa Cruz, CA, USA), and mouse anti-human
and incubated with the following primary antibodies: rabbit actin IgG (Amersham Corp., Arlington Heights, IL, USA).

a
b

Figure 5 Rescue of cancer cells from the oncolytic e€ect of D24 by Rb. (a) Saos-2 cells were infected ®rst with 100 MOI of
Ad5CMV-pA (top) or Ad5CMV-Rb (bottom). Three days later cells were infected with ten MOI of D24, and allowed to grow for 5
days. Magni®cation, 6100. (b) Saos-2 cells were infected with 100 MOI of Ad5CMV-pA (top) or Ad5CMV-Rb (bottom), and 3
days later with D24 at the indicated MOI. Five days after the infection with D24, the plates were stained with crystal violet. (c) Saos-
2 cells were infected with 100 MOI of Ad5CMV-pA or Ad5CMV-Rb and 3 days later with ten MOI of the D24 (solid bars) or UV-
D24 (hatched bars). Five days after infection viability was assessed by Trypan blue exclusion. Shown is the relative growth of the
control cells (100%) with respect to the D24 treated cells

Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
10
The secondary antibodies were horse-radish peroxidase-
a conjugated donkey anti-rabbit IgG (Amersham) and horse-
radish peroxidase-conjugated donkey anti-mouse IgG
(Amersham). The membranes were developed according to
Amersham's electrochemi-luminiscence protocol.

Immunoprecipitation
Ten mg of antibody agarose conjugated rabbit anti-adeno-
virus-2 E1A protein 13 S-5 (Santa Cruz Biotechnology Inc.),
was added to 500 mg of total cell proteins from cells infected
with D24, dl1520, or Ad5CMV-pA, and incubated at 48C
overnight. The pellet was collected by centrifugation, washed
with RIPA, and subjected to 7% SDS polyacrylamide gel
electrophoresis followed by immunoblotting with goat anti-
human Rb C-15 antibody (Santa Cruz Biotechnology, Inc.)
(Fueyo et al., 1998c).

Viability assay
Cells were seeded at 105 cells per well in six-well plates and
allowed to grow for 20 h. Cells were infected with D24,
b
Ad5CMV-pA, Ad5CMV-Rb, or UV-D24 at the indicated
MOI. After 7 ± 14 days of incubation the cell monolayers
were ®xed with methanol and stained with 0.2% crystal
violet. The experiment was performed three times for each
cell line. Trypan blue experiments were performed as
described elsewhere (Fueyo et al., 1998b).

Flow cytometric analyses


These assays were performed as described elsewhere (Gomez-
Manzano et al., 1996; Fueyo et al., 1998c).

Transfer of exogenous wild-type Rb or p21


Characterization of the Rb- and p21-adenovirus and its
infection into cells were described elsewhere (Gomez-
Manzano et al., 1997; Fueyo et al., 1998c).

Infectivity experiments in normal cells


CCD32-Lu lung ®broblasts were infected with 10 ± 100 MOI
c of the Ad5CMV-GFP; and 3 days later, the cells were
examined in terms of both interference and ¯uorescent
contrast. The percentage of infected cells was determined by
counting the number of green ¯uorescent cells among 500
examined cells.

Viral replication experiments


Viral production was quanti®ed by plaque assay. Six days
after infection, cells were scraped into culture medium and
lysed by three cycles of freezing and thawing. Cell lysates
were clari®ed by centrifugation and the supernatant was
serially diluted in medium for the infection of 293 cells in 6-
well dishes. After 1 h of incubation at 378C, the infected cells
were overlaid with 3 ml of 1.25% SeaPlaque agarose (FMC,
Rockland, ME, USA) in DMEM/F12 (10% fetal bovine
serum). Additional agarose was added to each dish 4 days
later. Plaques were visualized at 7 days after infection.

Electronic microscopy
Figure 6 Anti-cancer e€ect in vivo. (a) Mice with subcutaneous
D-54 MG tumors (derived from a pre-established tumor line)
Four days after infection, pellets from U-251 MG cells
were treated with 56108 PFU of the D24 (solid bars) or UV-D24 infected with UV-D24, or D24 were ®xed in 2% glutaralde-
(hatched bars) given every 5 days a total of ®ve times. Results are
reported as the mean+s.d. *D24 produced an 83.8% growth
inhibition (P50.01). (b) Mice with D-54 MG tumors (derived
from inoculation of glioma cells) were treated with 109 PFU of with tumors of D-54 MG glioma cells were treated with a single
the D24 (solid bars) or UV-D24 (hatched bars) given every 5 ± 7 intratumoral injection of 107 PFU of D24 (solid bars) or UV-D24
days a total of four times. Results are reported as the mean+s.d. (hatched bars). Results are reported as the mean+ s.d. *Growth
*D24 produced an 85.7% growth inhibition (P50.05). (c) Mice inhibition by D24 was 66% (P50.005)

Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
11
hyde and post-®xed for 1 h in 1% osmium tetroxide. Samples measured, and tumor volume was calculated using the
were counterstained with uranyl acetate and lead citrate and formula a6b260.4 (Attia and Weiss, 1996). When the
examined under a transmission electron microscope (Gomez- tumors had reached the maximum acceptable size, the mice
Manzano et al., 1996) were killed, and their tumors were excised and analysed by
histological ®xation and staining with hematoxylin and eosin.
Each experiment was performed separately on at least ®ve
Animal studies
di€erent animals from each group. Tumor responses to each
The animal studies were carried out at the animal facility of treatment were compared by the two-tailed t-student test.
The University of Texas M.D. Anderson Cancer Center in
accordance with institutional guidelines. Mice were accli-
mated and caged in groups of ®ve or less. All mice were fed
animal chow and water ad libitum. Animals were anesthetized
with methoxy¯urane before all procedures and were observed
until fully recovered from the e€ect of the anesthesic. Pieces Acknowledgments
of approximately 3 mm in diameter of D-54 MG pre- We gratefully acknowledge Christine Wogan (Department
established tumors, or 107 D-54MG parental cells were of Scienti®c Publications, The University of Texas M.D.
inoculated subcutaneously into the right ¯anks of 6- to 8- Anderson Cancer Center (UTMDACC)) for her editorial
week-old athymic BALB/c-nu/nu (nude) mice (Harlan assistance, Karen Ramirez (Department of Immunology,
Sprague-Dawley Co., Houston, TX, USA). When tumors UTMDACC) for her contribution to the ¯ow cytometry
reached at least 5 mm in diameter, they were injected with studies, and Jim Lemoine (Department of Medical
either D24 or UV-D24 at one of the indicated doses. The Graphics, (UTMDACC) for his help in the design of the
largest (a) and smallest (b) diameters of each tumor were ®gures.

References

Alemany R, Ruan S, Kataoka M, Koch PE, Mukhopadhyay Gomez-Manzano C, Fueyo J, Kyritsis AP, McDonnell TJ,
T, Cristiano RJ, Roth JA and Zhang WW. (1996). Cancer Steck PA, Roth JA, Steck KD, Levin VA and Yung WKA.
Gene. Ther., 3, 296 ± 301. (1996). Cancer Res., 56, 694 ± 699.
Alemany R, Dai Y, Lou YC, Sethi E, Prokopenko E, Josephs Gomez-Manzano C, Fueyo J, Kyritsis AP, McDonnell TJ,
SF and Zhang WW. (1997). J. Virol. Methods, 68, 147 ± 159. Steck PA, Levin VA and Yung WKA. (1997). J. Natl.
Andreanski S, Soroceanu L, Flotte ER, Chou J, Market JM, Cancer Inst., 89, 1036 ± 1044.
Gillespie GY, Roizman B and Whitley RJ. (1997). Cancer Gomez-Manzano C, Fueyo J, Kyritsis AP and Yung WKA.
Res., 57, 1502 ± 1509. (1998). Gene Transfer and Therapy for Neurological
Arap W, Nishikawa R, Furnari FB, Cavenee WK and Huang Disorders. Chiocca AE and Breake®eld X (eds). Human
HJ. (1995). Cancer Res., 55, 1351 ± 1354. Press Inc.: Totowa, NJ, pp. 201 ± 225.
Attia MA and Weiss DW. (1996). Cancer Res., 26, 1787 ± Hamel W, Westphal M and Shepard HM. (1993). J.
1800. Neurooncol., 16, 159 ± 165.
Barker DD and Berk AJ. (1987). Virology, 156, 107 ± 121. Hay JG, Shapiro N, Sautho€ H, Heitner S, Phupakdi W and
Bischo€ JR, Kirn DH, Williams A, Heise C, Horn S, Muna Rom WN. (1999). Hum. Gene. Ther., 10, 579 ± 590.
M, Ng L, Nye JA, Sampson-Johannes A, Fattaey A, Heise C, Sampson-Johannes A, Williams A, McCormick F,
McCormick F. (1996). Science, 274, 373 ± 376. Von Ho€ DD and Kirn DH. (1997). Nat. Med., 3, 639 ±
Chase M, Chung RY and Chiocca EA. (1998). Nat. 645.
Biotechnol., 16, 444 ± 448. Henson JW, Schnitker BL, Correa KM, vom Deimling A,
Co€ey M, Strong JE, Forsyth PA and Lee PWK. (1998). Fassbender F, Xu HJ, Benedict WF, Yandell DW and
Science, 282, 1332 ± 1334. Louis DN. (1994). Ann. Neurol., 36, 714 ± 721.
Collins VP and James CD. (1993). FASEB J., 7, 926 ± 930. Hirvonen HE, Salonen R, Sandberg MM, Vuorio E, Vastrik
Costello JF, Berger MS, Huang H-JS, Cavenee WK. (1996). I, Kotilainen E and Kalimo H. (1994). Br. J. Cancer, 69,
Cancer Res., 56, 2405 ± 2410. 16 ± 25.
Costello JF, Plass C, Arap W, Chapman VM, Held WA, Huang HJ, Yee JK, Shew JY, Chen PL, Bookstein R,
Berger MS, Huang H-JS and Cavenee WK. (1997). Cancer Friedmann T, Lee EY and Lee WH. (1988). Science, 242,
Res., 57, 1250 ± 1254. 1563 ± 1566.
Dyson N and Harlow E. (1992). Cancer Surv., 12, 161 ± 195. Jen J, Harper JW, Bigner SH, Bigner DD, Papadopuolos N,
Flint J and Shenk T. (1997). Annu. Rev. Genet., 31, 177 ± 212. Markowitz S, Wilson JKV, Kinzler W and Vogelstein B.
Freytag SO, Rogulski KR, Paielii DL, Gilbert JF and Kim (1994). Cancer Res., 54, 6353 ± 6358.
JK. (1998). Hum. Gene. Ther., 9, 1323 ± 1333. Kirn D, Herminston T and McCormick F. (1998). Nat. Med.,
Fueyo J, Gomez-Manzano C, Yung WKA, Clayman GL, 4, 1341 ± 1342.
Liu TJ, Bruner Jm, Levin VA and Kyritsis AP. (1996a). Kyritsis AP, Zhang B, Zhang W, Xiao M, Takeshima H,
Oncogene, 12, 103 ± 110. Bondy ML, Cunningham JE, Levin VA and Bruner J.
Fueyo J, Gomez-Manzano C, Bruner JM, Saito Y, Zhang B, (1996a). Oncogene, 12, 63 ± 67.
Zhang W, Levin VA, Yung WKA and Kyritsis AP. Kyritsis AP, Xu R, Bondy ML, Levin VA and Bruner JM.
(1996b). Oncogene, 13, 1615 ± 1619. (1996b). Mol. Carcinog., 15, 1 ± 4.
Fueyo J, Gomez-Manzano C, Yung WKA and Kyritsis AP. Martuza RL, Malick A, Markert JM, Ru€ner KL and Coen
(1998a). Neurology, 51, 1250 ± 1255. DM. (1991). Science, 10, 854 ± 856.
Fueyo J, Gomez-Manzano C, Yung WKA, Liu TJ, Alemany Masuda H, Miller C, Koe‚er HP, Battifora H and Cline MJ.
R, McDonnell TJ, Shi X, Rao JS, Levin VA and Kyritsis (1987). Proc. Natl. Acad. Sci. USA., 84, 7716 ± 7719.
AP. (1998b). Nat. Med., 4, 685 ± 690. Mineta T, Rabkin SD, Yazaki T, Hunter WD and Martuza
Fueyo J, Gomez-Manzano C, Yung WKA, Liu TJ, Alemany RL. (1995). Nat. Med., 9, 938 ± 943.
R, Bruner JM, Chintala SK, Rao JS, Levin VA and Nevins JR. (1992). Science, 258, 424 ± 429.
Kyritsis AP. (1998c). Neurology, 50, 1307 ± 1315.
Furnari FB, Huang HJ and Cavenee WK. (1995). Cancer
Surv., 25, 233 ± 275.

Oncogene
Oncolytic adenovirus for glioma treatment
J Fueyo et al
12
Puumalainen AM, Vapalahti M, Agrawal RS, Kossila M, Sherr CJ. (1996). Science, 274, 1672 ± 1677.
Laukkanen J, Lehtolainen P, Viita H, Paljarvi L, Sidransky D, Mikkelsen T, Schwechheimer K, Rosemblum
Vanninen R and Yla-Herttuala S. (1998). Hum. Gene. ML, Cavanee W and Volgestein B. (1992). Nature, 355,
Ther., 9, 1769 ± 1774. 846 ± 847.
Ridgway P, Hall AR, Myers CJ and Braithwaite AW. (1997). Ueki K, Ono Y, Henson JW, E®rd JT, von Deimiling A and
Virology, 237, 404 ± 413. Louis DN. (1996). Cancer Res., 56, 150 ± 153.
Roth JA and Cristiano RJ. (1997). J. Natl. Cancer Inst., 89, Whyte P, Ruley HE and Harlow E. (1988). J. Virol., 62, 257 ±
21 ± 39. 265.
Rothman T, Hengstermann A, Whitaker NJ, Sche€ner M Whyte P, Williamsom NM and Harlow E. (1989). Cell, 56,
and zur Hausen H. (1998). J. Virol., 72, 9470 ± 9478. 67 ± 75.
Schmidt EE, Ichimura K, Reifenberger G, Collins VP.
(1994). Cancer Res., 54, 6321 ± 6324.

Oncogene

You might also like