You are on page 1of 9

ARTICLE doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.

com on IDEAL

Human and Mouse IFN- Gene Therapy Exhibits Different


Anti-tumor Mechanisms in Mouse Models
Xiao-Qiang Qin,* Carla Beckham, Jennifer L. Brown, Matvey Lukashev, and James Barsoum
Biogen, Inc., 14 Cambridge Center, Cambridge, Massachusetts 02142, USA

*To whom correspondence and reprint requests should be addressed. Fax: (617) 679-3200. E-mail: xiao-qiang_qin@biogen.com.

Previously, we suggested that local human interferon- (IFN-) gene therapy with replication-
defective adenoviral vectors can be an effective cancer treatment. Clinical trials to treat cancers
with adenovirus expressing the human IFN- gene (IFNB1) has been planned. As a continued effort
to explore the mechanisms of action of human IFN- gene therapy that can occur in the clinical
setting, we tested mouse IFN- gene therapy in human xenograft tumors in both ex vivo and in
vivo models. Delivery of the mouse IFN- gene (Ifnb) caused tumor inhibition; this effect was
dependent on the indirect anti-tumor activities of IFN-, notably a stimulation of natural killer
cells. IFN- does not show cross-species activity in its anti-proliferative effect and mouse IFN-
does not cause as significant an anti-proliferative effect on mouse tumor cells as human IFN-
causes on human tumor cells. Therefore, we believe that mouse models using either human IFN-
 or mouse IFN- gene transfer do not capture all aspects of the action of adenovirus-mediated
human IFN- gene therapy that may be present in the clinical setting. Due to its multiple mech-
anisms of action, human IFN- gene therapy may be effective in treating human cancers that are
either sensitive or resistant to the direct anti-proliferative effect of IFN-.

Key Words: IFN- gene delivery, tumor regression, mouse tumor model,
anti-proliferative effect, immune stimulation, NK cells

INTRODUCTION
IFNs can also exhibit an indirect anti-tumor effect
The type 1 interferons (IFNs), the -family and , share through immune stimulation. They induce major histo-
receptor components and have multiple anti-tumor compatibility complex class 1 expression, cause an increase
activities [1–3]. At appropriate concentrations, they in cytotoxic T lymphocyte activity, enhance the genera-
induce the direct anti-proliferative effects that are man- tion of T helper cells, and activate macrophages and nat-
ifested as cytotoxicity/apoptosis and inhibition of cell ural killer (NK) cells both in vitro and in animal models
cycle progression in S phase for solid tumor cells or in [12,13]. Stable gene transfer of IFN- in mouse tumor cells
G1 phase in Daudi Burkitt’s lymphoma cells. It is not blocked the tumorigenic potential and caused the infil-
clear whether IFNs induce cytotoxicity/apoptosis and tration of immunological effector cells such as T lympho-
cell cycle inhibition by activation of the same signal cytes [14–18]. Another indirect anti-tumor activity of IFNs
transduction pathways. However, it seems that these two is the inhibition of angiogenesis. IFN- blocks endothelial
effects are biologically separable because we found that cell migration in vitro and inhibits lymphocyte- and tumor-
IFN- induced the cytotoxicity effect in cells that were induced angiogenesis. IFN- downregulates expression of
insensitive to its cell cycle effect (X.-Q.Q., unpublished the gene encoding angiogenic factor basic fibroblast
data). IFN- and IFN- also were observed to have dif- growth factor and inhibits angiogenesis in vivo [19–24].
ferences in their receptor binding and signaling [4–7]. In Clinical experience with IFN protein therapy of cancer,
certain types of human carcinoma cells, IFN- showed especially in the treatment of solid tumors, has generally
a more marked anti-proliferative effect than IFN- been disappointing. Convincing effect has been observed
[8–10]. Clinically, there are data that suggest a correla- only in limited cancer types such as hairy cell leukemia,
tion between the in vitro sensitivity of malignant cells to chronic myelogenous leukemia, and melanoma [11,25].
the direct anti-proliferative effect induced by IFNs and Pharmacokinetic studies have indicated that IFN- has an
the in vivo clinical effects in patients receiving IFN pro- extremely short half-life in the blood system after systemic
tein therapy [11]. protein administration [26,27]. This suggests that par-

356 MOLECULAR THERAPY Vol. 4, No. 4, October 2001


Copyright © The American Society of Gene Therapy
1525-0016/01 $35.00
doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL
ARTICLE

A B

FIG. 1. Human IFN- gene therapy led to regression of human s.c. xenograft U87 MG tumors in mice and prolonged mouse survival. (A) The mean diameters
of tumors after intratumoral injection with H5.010CMVhIFN- at 1  1011 particles (open triangle), 1  1010 particles (open circle), and 1  109 particles (filled
diamond), or with PBS (filled circle), or with H5.010CMVlacZ at 1  1011 particles (open, inverted triangle), 1  1010 particles (multiplication sign), or 1  109
particles (filled triangle). *P < 0.001 for tumors treated with H5.010CMVhIFN- at 1  1011 particles or H5.010CMVhIFN- at 1  1010 particles versus treated
with PBS; **P < 0.05 for tumors treated with H5.010CMVhIFN- at 1  109 particles versus treated with PBS. (B) Kaplan–Meier plot showing the percentage
survival of mice after the various treatments, indicated by the same symbols as in (A).

enteral IFN- protein treatment, through intravenous IFN- gene therapy in human cancers. Malignant glioblas-
(i.v.), subcutaneous (s.c.), or intramuscular (i.m.) injec- toma will be the disease target of our first clinical trial
tions, may fail to have an anti-tumor effect due to insuf- because it is a local cancer amenable to direct intratumoral
ficient delivery or a lack of sustained delivery of the pro- therapy and the disease lacks effective therapeutic alter-
tein to the tumor site. natives. We therefore evaluated gene therapy with aden-
We found previously that human IFN- gene therapy ovirus expressing human IFN- in a human xenograft
inhibits tumor formation and causes regression of estab- model. We treated preestablished s.c. U87 MG human
lished tumors in immune-deficient mice, and that the glioma tumors in nude mice with a single, intratumoral
human IFN--mediated anti-proliferative activity alone injection of phosphate-buffered saline (PBS) or adenovirus
can be sufficient to cause tumor regression [28]. We pro- expressing either human IFN- or the negative control
posed that local IFN- gene therapy with replication- gene, lacZ. Human IFN- gene therapy led to tumor regres-
defective adenovirus vectors can be an effective treatment sion and significantly prolonged animal survival (Figs. 1A
for cancer. Others have demonstrated that IFN- gene and 1B). We observed complete tumor regression and
therapy can also abrogate tumorigenicity and eradicate 100% long-term mouse survival at viral doses of 1  1011
established tumors by the collective activation of IFN-- and 1  1010 total particles per mouse.
induced indirect anti-tumor activities [24,29–31]. Here, We next tested the effect of mouse IFN- gene therapy
we further evaluated the effect of both ex vivo and direct in human xenograft tumors. First, we used an ex vivo gene
local mouse IFN- gene therapy on various human delivery model to compare mouse IFN- with human IFN-
xenograft tumors and found that human and mouse IFN-  gene therapy. Previously, a very potent bystander effect
 gene therapy demonstrated two different mechanisms was demonstrated, as adenovirus-mediated human IFN-
of the anti-tumor activity of IFN- gene therapy, both of gene transduction in as few as 1% of human breast carci-
which might be efficacious in the clinical setting. noma MDA-MB-468 cells was sufficient to block tumor
formation [28]. Therefore, we infected these cells in vitro
with an adenovirus that has deletions of the E1 and E3
RESULTS regions and a temperature-sensitive mutation of E2A,
Mouse IFN- Gene Therapy Leads to Tumor encoding mouse Ifnb (H5.110CMVmIFN-) or human
Inhibition in ex Vivo and in Vivo Human IFNB1 (H5.110CMVhIFN-) at a multiplicity such that
Xenograft Tumor Models nearly all cells should be transduced. We collected the
Previously, we demonstrated that local human IFN  gene infected cells and mixed them with uninfected cells imme-
transfer can be an effective treatment for solid tumors in diately before implantation to assess the bystander effect.
mouse models [28]. We are now planning clinical trials to Equal numbers of infected cells, mixtures in which 10%
test the safety and efficacy of adenovirus-mediated human or 1% of cells were infected with H5.110CMVmIFN-, or

MOLECULAR THERAPY Vol. 4, No. 4, October 2001 357


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL

FIG. 2. Ex vivo mouse IFN- gene therapy in


human IFN- gene delivery.
MDA-MB-468 tumors. Uninfected and
infected tumor cells were mixed at various Although human IFN- gene deliv-
ratios immediately before implantation into ery blocked tumor formation, 1%
mice. Uninfected cells (open square), or 100% mouse IFN- gene transduction
H5.110CMVmIFN- transduced cells (open resulted in the formation of tumors
triangle), 10% (open, inverted triangle) or 1%
(filled, inverted triangle) H5.110CMVmIFN-
which subsequently regressed.
transduced cells, or 1% H5.110CMVhIFN- We next examined whether direct
transduced cells (open circle) were injected treatment of tumors with
s.c. into the flanks of nude mice. Mean tumor H5.110CMVmIFN- leads to their
diameter was plotted versus time post-tumor
regression. We injected pre-
cell implantation. *P < 0.01 for mice
implanted with 10% or 1% established s.c. MDA-MB-468 tumors
H5.110CMVmIFN- transduced cells, or 1% in nude mice with H5.110CMVmIFN-
H5.110CMVhIFN- transduced cells, versus , H5.110CMVhIFN-, or
with uninfected cells at day 28. H5.110CMVlacZ, all at the dose of 5
 1010 viral particles/mouse, or PBS.
As in our previous study,
H5.110CMVhIFN- caused rapid
tumor regression, resulting in com-
a mixture in which 1% of cells were infected with plete tumor regression in four of five mice within 1 week
H5.110CMVhIFN- were s.c. implanted into Balb/c nude after the treatment. In this specific experiment, one of five
mice. Consistent with previous observations, human IFN- mice failed to respond to H5.110CMVhIFN- treatment, pos-
 gene transduction of 1% of cells effectively blocked sibly due to a less efficient virus injection. It should be noted
tumor formation (Fig. 2). In contrast, we observed the for- that in several other experiments performed with
mation of small tumors in all mice in the 10% and 1% H5.110CMVhIFN-, we often saw complete tumor regression
H5.110CMVmIFN- treatment groups at end of the first in all mice treated, resulting in 100% animal survival.
week after cell implantation. However, all tumors disap- H5.110CMVmIFN- treatment also caused complete tumor
peared within 30 days (Fig. 2). We also observed that 100% regression (Fig. 3A). However, in contrast to the very rapid
cell infection with H5.110CMVmIFN- effectively blocked regression observed following gene delivery of human IFN-
tumor formation (Fig. 2). This result may not necessarily , the regression was not significant until 2 weeks after the
be due to a mouse IFN- effect because cells that were treatment (Fig. 3A). Once tumors regressed, they did not
100% infected with the control virus (H5.110CMVlacZ) recur over the observation time of 2 months. We further
did not form tumors [28]. Therefore, ex vivo gene delivery tested mouse IFN- gene therapy in established human
of mouse IFN  into as few as 1% of human tumor cells xenograft tumors derived from prostate cancer PC-3 cells.
can inhibit tumorigenesis, albeit less efficiently than Direct intratumoral treatment with H5.110CMVmIFN- also

A B

FIG. 3. Direct in vivo treatment of established tumors derived from human MDA-MB-468 cells (A) or PC-3 cells (B). Preestablished tumors were treated with PBS
(open square), H5.110CMVmIFN- (open circle), H5.110CMVhIFN- (open triangle), or H5.110CMVlacZ (open, inverted triangle) at 5  1010 viral parti-
cles/mouse. Measurements represent mean tumor diameter. P < 0.01 for tumor diameter difference between mouse IFN- or human IFN- gene therapy ver-
sus PBS treatment in both experiments.

358 MOLECULAR THERAPY Vol. 4, No. 4, October 2001


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL
ARTICLE

A B

FIG. 4. Local mouse IFN- and human IFN- gene therapy led to suppression of both local i.p. tumors and metastasis induced by the i.p. injection of MDA-MB-231 cells
in nude mice. (A) A representative mouse that developed large i.p. tumors in the H5.010CMVlacZ treatment group (right) and a mouse that was cured with IFN- gene
therapy. (B) Kaplan–Meier plot showing the percentage survival of mice that were treated with PBS (filled, inverted triangle), H5.010CMVmIFN- (filled circle),
H5.010CMVhIFN- (filled square), or H5.010CMVlacZ (filled triangle) at 2  1010 particles/mouse (2 weeks after i.p. tumor cell injections) over the observation period
of 90 days. P < 0.005, H5.010CMVmIFN- or H5.010CMVhIFN- treatment versus PBS treatment. There is no statistical difference between H5.010CMVlacZ treatment
and PBS treatment.

caused tumor regression (Fig. 3B). In both tumor types, ery leads to a high level of IFN- locally, but with only
tumor regression induced by H5.110CMVmIFN- was slower barely detectable or undetectable levels in circulation.
than H5.110CMVhIFN- mediated regression, even though
delivery of both Ifnb and IFNB1 led to significant long-term The Mechanism of the Mouse IFN- Mediated
animal survival (data not shown). Anti-tumor Effect
To confirm these observations in a different model, we The difference in anti-tumor efficiency led us to speculate
implanted human breast carcinoma MDA-MB-231 cells into that after gene delivery human IFN- and mouse IFN- act
nude mice by intraperitoneal injection. Injection of these through different mechanisms. In these tumor models,
cells led to development of both local (i.p.) tumors and the human IFN- gene therapy mainly executed a direct anti-
subsequent metastasis to liver and lung tissues after 2 to 3 proliferative effect [28]. To investigate whether mouse IFN-
weeks. Mice that received i.p. administration of PBS or the  can cause an anti-proliferative effect in human tumor
control LacZ adenovirus developed large i.p. tumors (Fig. cells, we treated MDA-MB-468 cells (Fig. 5) and human
4A). Within 3 months post treatment, virtually all the mice hepatoma cells (Huh7; data not shown) with equal amounts
were euthanized due to local i.p. tumor burden including of mouse IFN- and human IFN- protein, and examined
ascites and symptoms of metastasis such as jaundice, loss cell viability by both light microscopy and an MTT assay.
of activity, and/or difficulty in breathing (Fig. 4B). However, No anti-proliferative activity was observed with mouse IFN-
i.p. treatment with an adenovirus deleted for the E1 and E3 , even at 30,000 units/ml, whereas human IFN- induced
regions, encoding mouse Ifnb or human IFNB1 a significant decrease in cell viability at 100 units/ml (Fig.
(H5.010CMVmIFN- and H5.010CMVhIFN-, respectively), 5 and data not shown). Because mouse IFN- does not cause
significantly prolonged survival (Fig. 4B). In this experi- any anti-proliferative effect on human cells, it is unlikely
ment, human IFN- gene delivery led to a greater animal that the tumor regression caused by mouse IFN- gene deliv-
survival rate than mouse IFN- gene delivery (Fig. 4B). ery was due to an IFN-mediated direct anti-proliferative
When the human IFN- and mouse IFN- protein levels in effect.
serum were examined by ELISA 3 days after treatment with The modes of IFN- anti-tumor action manifest three
H5.010CMVmIFN- and H5.010CMVhIFN-, no detectable aspects: anti-proliferative effect, immune stimulation, and
mouse or human IFN- was found in these mice. This fur- inhibition of angiogenesis. We investigated whether the
ther supports our previous observation that local gene deliv- mouse IFN- stimulated immune function was critical to the

MOLECULAR THERAPY Vol. 4, No. 4, October 2001 359


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL

FIG. 5. Mouse IFN- failed to cause a significant anti-


proliferative effect on MDA-MB-468 cells at doses rang-
ing from 100 unit/ml to 30,000 units/ml (gray bars),
whereas human IFN- displayed potent anti-proliferative
activity (black bars), as determined in an MTT assay.

that received intratumoral


H5.110CMVmIFN- treatment and exam-
ined NK cell function. Increased NK
cytolysis was observed in these mice com-
pared with control mice (Fig. 7). In the
same experiment, anti-asialo GM1 antis-
era pretreatment blocked NK cell stimu-
lation (Fig. 7). In addition, we also tested
the splenocytes of some mice in the
MDA-MB-468 ex vivo mouse IFN- gene
delivery experiment. Enhanced NK cell
activity was observed following implan-
tation of H5.110CMVmIFN- transduced
anti-tumor effect in these models. Mice bearing s.c. MDA- cells (data not shown).
MB-468 tumors were pretreated with PBS or anti-asialo GM1 We next tested whether mouse IFN- gene delivery
antiserum, which depletes NK cells. We then treated these might have an effect on an untreated tumor at a distinct
mice with intratumoral injection of PBS, H5.110CMVmIFN- site. We implanted MDA-MB-468 cells on both flanks of
, H5.110CMVhIFN-, or H5.110CMVlacZ at 5  1010 par- nude mice. Mice bearing tumors of roughly equivalent size
ticles/mouse. Treatment with PBS or H5.110CMVlacZ did on each side were selected for the experiment. We then
not cause tumor regression in either the PBS or anti-asialo treated tumors on the right flank with H5.110CMVmIFN-
GM1 antisera pretreatment groups. Treatment with  or H5.110CMVlacZ at 1  1011 particles/mouse or PBS
H5.110CMVmIFN- caused tumor regression and signifi- and monitored tumor growth on both flanks. We found
cantly prolonged animal survival in the PBS pretreatment that mouse IFN- gene therapy caused the regression of
group. However, anti-asialo GM1 antisera pretreatment sig- both the treated tumor and the untreated tumor on the
nificantly reduced the H5.110CMVmIFN- anti-tumor effect opposite flank (Fig. 8).
in terms of both tumor size (Fig. 6A) and mouse survival Type 1 IFNs including IFN- have been shown to be
(Fig. 6B). In contrast, human IFN- gene delivery led to inhibitors of angiogenesis and can inhibit tumor growth
complete tumor regression in both the presence and the by blocking tumor vascularization [19–24]. We investi-
absence of the anti-asialo GM1 antisera pretreatment (Figs. gated whether mouse IFN- gene transfer caused an
6A and 6B). These data indicate that NK cell activity had a observable effect on the blood vasculature in tumors. At 2
role in the H5.110CMVmIFN--mediated tumor regression. weeks after treatment with H5.110CMVmIFN- or
It should be noted that the anti-asialo GM1 antisera pre- H5.110CMVlacZ, we harvested tumors to examine the
treatment did not completely abolish the anti-tumor effect microvascular density and morphology. No significant dif-
of H5.110CMVmIFN-. The lack of complete abolition of ference was noted in the vasculature following treatment
the anti-tumor effect might indicate that NK cell depletion with these two viruses (data not shown). Recently, mouse
was not complete or that the mouse IFN- induced anti- IFN- gene therapy was also examined in a mouse col-
tumor effect did not rely solely on the stimulation of NK orectal cancer liver metastases model. No significant reduc-
cells. Similar results were observed in tumor modeling per- tion in tumor blood vessels was observed after mouse Ifnb
formed in SCID/beige mice, which lack NK cells. Human gene delivery [32].
IFN- gene therapy was still effective in SCID/beige mice;
however, the anti-tumor activity of mouse IFN- gene ther-
apy was reduced, although not completely eliminated, in DISCUSSION
these mice (data not shown). Although the differential anti- We previously demonstrated that local human IFN- gene
tumor effect of H5.110CMVmIFN- in SCID/beige and nude therapy with adenovirus vectors led to tumor regression [28].
mice may result from other genetic differences of these two Our study supports the testing of local human IFN- gene
mouse species, it is also consistent with a model in which therapy in cancer patients. Clinical trials testing adenovirus
the anti-tumor effect of mouse IFN- gene therapy is due to expressing human IFN- in cancers will be initiated soon.
an indirect activity mediated, at least in part, by NK cells. Here, we have examined ex vivo and direct in vivo mouse
To confirm that mouse IFN- gene delivery enhanced IFN- gene therapy in human xenograft tumor models. The
the NK cell activity, we collected splenocytes from mice growth of these human tumor cells in vitro is completely

360 MOLECULAR THERAPY Vol. 4, No. 4, October 2001


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL
ARTICLE

FIG. 6. NK cell depletion caused an inhibitory effect on the anti-tumor activ-


ity of mouse IFN-, but not human IFN- gene therapy. (A) The mean diam-
A
eters of MDA-MB-468 tumors in mice that had tail vein PBS pretreatment and
intratumoral treatment with PBS (multiplication sign), H5.110CMVmIFN-
(open square), H5.110CMVhIFN- (open triangle), H5.110CMV-lacZ (open cir-
cle), or in the mice that had anti-asialo GM1 antibody pretreatment and then
intra-tumor treatment with PBS (filled triangle), H5.110CMVmIFN- (filled
square), H5.110CMVhIFN- (filled, inverted triangle), or H5.110CMV-lacZ
(open circle). *The P value for the diameter difference of tumors treated with
H5.110CMVmIFN- in the presence or absence of anti-asialo GM1 antibody
is < 0.05 as calculated with the t test. (B) Kaplan-Meier plot showing the per-
centage survival of mice over the observation period of 95 days. Treatments
are indicated with the same symbols as in (A).

insensitive to mouse IFN-. Both ex vivo and in vivo mouse


IFN- gene delivery caused significant anti-tumor effects.
These effects were dependent on indirect anti-tumor activi-
ties, notably but not exclusively a stimulation of NK cells.
This is consistent with a previous study in which NK cell acti-
vation was implicated in the rejection of PC-3 cells stably
expressing mouse IFN- in mice [24]. However, our data fur-
ther indicate that direct local IFN- gene therapy can also B
lead to an activation of the innate immune system, result-
ing in tumor regression. This underscores the notion that
direct local IFN- gene therapy with adenovirus vectors
might be an effective treatment for human cancers that are
either sensitive or resistant to the IFN- induced direct anti-
proliferative activity.
Available animal tumor models for exploring IFN-
gene therapy generally use human tumor cells in immune-
deficient mice or mouse tumor cells in immune-competent
mice. We believe that animal modeling with immune-
competent mice may not be representative of human IFN-
 gene therapy in the clinical setting. Because only tumors
of mouse origin grow in the immune-competent mice and
IFN- does not show cross-species activity, only mouse
IFN- gene therapy can be tested in these mice. Human
IFN- and mouse IFN- seem to have biological differences
in terms of anti-proliferative activity. We noticed that
mouse IFN- does not cause as significant an anti-prolif-
erative effect on the mouse tumor cells as human IFN- vated systemically through immune organs such as spleen.
does on human tumor cells, as measured by MTT assay and The NK activation in spleen can result from vector dis-
cell cycle analysis in multiple mouse cell lines (X.-Q.Q., semination into the spleen or the circulation of a low level
C.B., and Amie Dergay, unpublished data). Clearly, animal of IFN- that is released by the vector-transduced tumor
modeling with immune-competent mice can reveal the cells. Therefore, the mechanisms of action of IFN- gene
indirect anti-tumor mechanisms of IFN- gene therapy therapy in a clinical setting will be very likely to include
such as T-cell-mediated immune responses. On the other both IFN--induced direct and indirect effects. We think
hand, our previous modeling with human xenograft that human IFN- gene therapy could generate a combi-
tumors in immune-deficient mice directly tested human natorial effect resulting from both the direct and the indi-
IFN- gene therapy in tumors of human origin. It did not rect anti-tumor activities, which could be highly effica-
focus on its indirect anti-tumor mechanisms of action. cious and have a long-lasting anti-tumor effect.
Other results [24,29–31] have indicated that IFN- gene Although we did not find differences in the microvas-
therapy can also cause an effective anti-tumor effect by cular density or morphology between H5.110CMVmIFN-
means of stimulation of both innate and adoptive immu-  treated tumors and control tumors, we cannot rule out
nity. Our current study further confirms that local IFN- the possibility that inhibition of angiogenesis is involved
gene delivery can lead to the activation of NK cells. The in the anti-tumor effect. It is possible that mouse IFN- did
NK cells can be activated locally in the tumors and/or acti- not cause a direct disruption of blood microvasculatures

MOLECULAR THERAPY Vol. 4, No. 4, October 2001 361


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL

FIG. 8. Mouse IFN- gene delivery into tumors on one flank caused regres-
FIG. 7. NK cell cytotoxicity assay showing that mouse IFN- gene delivery led sion of tumors in both flanks. Mice bearing established MDA-MB-468 tumors
to the activation of NK cells and that the activation can be blocked by the anti- on both flanks were treated with an intratumoral injection of PBS,
asialo GM1 antibody treatment. NK cell cytotoxicity was measured as the spe- H5.110CMVmIFN-, or H5.110CMVlacZ (both at 1  1011 particles/mouse)
cific release of the carboxyfluorescein molecule BCECF as described [35]. Each in the right flank tumor. The sizes of tumors in both flanks were measured.
treatment group contained three mice. The effector cells were acquired from The mean tumor diameters of the left and right flanks in the PBS group (open
the mixed spleen tissues of the three mice. Effector to target ratios used were and filled squares, respectively), in the H5.110CMVmIFN- group (open and
100:1, 50:1, 25:1, and 12.5:1 in triplicate. The data shown were derived from filled circles, respectively), and in the H5.110CMV-lacZ group (open and filled
the 100:1 samples. The lower effector to target ratios were not observed to triangles, respectively) were plotted versus time post virus treatment. There is
have cytotoxic activity. The error bars are SE, which were calculated from the a significant difference between tumors in each flank of the mice that were
triplicate samples. treated with H5.110CMVmIFN- versus PBS-treated tumors (P < 0.05).

but caused a blockage of tumor vascularization. This block- through the activation of NK cells. We further argued that
age effect could then cause an inhibition of tumor growth mouse models with either human IFN- or mouse IFN-
and, therefore, an anti-angiogenesis effect could occur gene delivery could not capture all aspects of the action
despite the lack of observable differences in the microvas- of IFN- gene therapy in the clinical situation. One con-
cular density between tumors treated with the mouse IFN- cern in the clinical usefulness of IFN- gene therapy is
 virus and the controls. Likewise, we could not conclude that cancer cells resistant to the direct anti-proliferative
that other IFN- effects such as activation of macrophages activity of IFN- may arise. This work suggests that IFN--
were not involved in the tumor regression observed in our resistant cancer cells may be eradicated by the indirect
study. The fact that NK cell deletion did not completely activities of IFN- on the host immune system, even in the
abolish the tumor regression effect mediated by mouse absence of direct anti-proliferative activity.
IFN- gene therapy may suggest the involvement of other
effector cells such as macrophages. However, we can con-
clude that NK cell activation had a major role in our mod- MATERIALS AND METHODS
els as mouse IFN- gene therapy activated NK cells and NK Recombinant adenoviruses. The production and purification of the sec-
cell depletion substantially reduced the anti-tumor effect. ond-generation adenoviruses H5.110CMVhIFN- and H5.110CMVlacZ
were described previously [28]. These viruses have deletions of the E1 and
We also conclude that another major indirect IFN- anti-
E3 genes and a temperature-sensitive mutation of E2a. The second-gener-
tumor activity, the T-cell-mediated immune response, was ation adenovirus encoding mouse Ifnb (H5.110CMVmIFN- [32]) and the
not necessary for an overt anti-tumor effect because we first-generation adenovirus (having deletions only in the E1 and E3 regions)
used immune-deficient mice that lack T cells. encoding lacZ, human IFNB, and mouse Ifnb (H5.010CMVlacZ [33],
We have tested the effect of mouse IFN- gene therapy H5.010CMVhIFN-, and H5.010CMVmIFN-, respectively) were used.
H5.110CMVhIFN- and H5.110CMVlacZ were previously described as
in s.c. human xenograft tumor models and demonstrated H5.110hIFN- and H5.110lacZ, respectively [28].
tumor inhibition that was independent of the IFN-induced
Cell culture. Human breast carcinoma cells MDA-MB-468 and MDA-MB-
direct anti-proliferative effect as well as indirect activities 231, prostate carcinoma cells PC-3, and glioblastoma cells U87 MG were
involving T cells. By depleting NK cells, we found that obtained from the American Type Culture Collection (Rockville, MD).
IFN- gene therapy causes tumor regression at least in part Huh7, a line of human hepatoma cells, has been described [34]. MDA-MB-

362 MOLECULAR THERAPY Vol. 4, No. 4, October 2001


Copyright © The American Society of Gene Therapy
doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL
ARTICLE

468, PC-3, and Huh7 cells were maintained as adherent cultures in MEM l of PBS containing various doses of adenoviral vectors was injected
containing 10% fetal bovine serum, 2 mM L-glutamine, 100 units/ml peni- directly into the center of the 4- to 7-mm diameter tumors in a single injec-
cillin, and 100 g/ml streptomycin. U87 MG cells were maintained as tion. The tumor size was measured in length and width using calipers and
adherent culture in Eagle’s minimal essential medium containing 2 mM L- presented as the average tumor diameter (mm). Mean tumor diameter is
glutamine and Earle’s BSS adjusted to contain 1.5 g/l sodium bicarbonate, the average of the tumor width plus length, averaged for all mice in a
0.1 mM nonessential amino acids, 1.0 mM sodium pyruvate, and 10% fetal group. There were usually five mice per group. For NK cell depletion exper-
bovine serum. Mouse Lymphoma Yac-1 cells were purchased from iments, anti-asialo GM1 antibody administration was started approximately
American Type Culture Collection (Rockville, MD). These cells were main- 1 week after tumor cell implantation. We administered 50 l anti-asialo
tained as nonadherent cultures at 37C and 5% CO2 in RPMI 1640 sup- GM1 antibody (30 mg/ml) to mice via tail vein injections as suggested by
plemented with 10% fetal bovine serum, 2 mM L-glutamine, 100 units/ml the manufacturer’s instructions (Wako BioProducts, Richmond, VA). The
penicillin, and 100 g/ml streptomycin. animals were treated every 4 to 5 days for four times before the tumors were
MTT assay. To assess the effect of IFN- on cell survival, we carried out treated with various viruses or PBS controls. For generating i.p. tumors and
an MTT assay using the MTT-based In Vitro Toxicology Assay kit (Sigma). metastasis, 100 l of PBS containing 5  106 human breast carcinoma cells
Human or mouse IFN- was diluted in DMEM medium without phenol red MDA-MB-231 was i.p. injected into nude mice. Subsequently, the mice
containing 10% fetal bovine serum, 2 mM glutamine, 100 units/ml peni- were subjected to i.p. treatments with PBS, H5.010CMVmIFN-,
cillin, and 100 g/ml streptomycin, and was aliquoted in a 96-well plate. H5.010CMVhIFN-, or H5.010CMVlacZ at 2 weeks after the tumor cell
The human breast carcinoma cells MDA-MB-468 were then added to the injections. We treated 10 mice in each group. Animal death was defined
plate at 1000 cells/well. The final concentrations of human or mouse IFN- by euthanizing mice due to local tumor burden including severe ascites
 was 0, 100, 300, 1000, 3000, 10,000, and 30,000 units/ml. The cells were and/or symptoms of metastasis such as loss of activity and/or difficulty in
incubated with IFN- for 5 days at 37C, 5% CO2, after which the MTT breathing. For testing the effect of mouse IFN- gene delivery into a tumor
reagent was added per the manufacturer’s protocol. The MTT formazan on one side on the growth of tumors at both sides, a large number of mice
crystals were allowed to completely dissolve. The O.D. of each well was then was implanted with MDA-MB-468 cells on both flanks and only mice bear-
read at 570 nm. Eight data points for each condition were used for acquir- ing a tumor on each side were selected for the experiment.
ing the average and the error bar.
Quantitation of IFN- proteins by ELISA. Sera were collected through ACKNOWLEDGMENTS
retro-orbital bleeding 3 days after i.p. injection of H5.010CMVmIFN- and We thank James Wilson and Guang-Ping Gao (Institute for Human Gene Therapy,
H5.010CMVhIFN- in mice. ELISA for human IFN- protein was carried out University of Pennsylvania) for providing adenovirus vectors, and Paula Hochman
as described [28]. Quantitation of mouse IFN- protein by ELISA was per- and Gerry Majeau (both from Biogen) for providing mouse IFN- protein.
formed essentially the same as that for the human IFN- ELISA, with the
following exceptions. We coated 96-well plates overnight at 4C with 10 RECEIVED FOR PUBLICATION MAY 8; ACCEPTED AUGUST 7, 2001.
g/ml anti-mouse IFN- antibody (MA-MB-7, a monoclonal antibody pre-
pared in Biogen) in coating buffer containing 50 mM sodium bicarbon- REFERENCES
ate/carbonate and 0.2 mM MgCl2 (pH 9.6). The plates were blocked with 1. Lengyel, P. (1982). Biochemistry of interferons and their actions. Annu. Rev. Biochem. 51:
0.5% nonfat dry milk in PBS. The standards for mouse IFN- were pur- 251–282.
chased from Access Biomedical (San Diego, CA). Serum samples were 2. Gresser, I. (1989). Antitumor effects of interferon. Acta Oncol. 28: 347–353.
diluted in 0.5% nonfat dry milk with 0.05% Tween-20 in PBS. All the suc- 3. Belardeli, F., and Gresser, I. (1996). The neglected role of type 1 interferon in the T-cell
response: implications for its clinical use. Immunol. Today 17: 369–372.
cessive steps are as in the human IFN- ELISA.
4. Gordon, I., Stevenson, D., Tumas, V., and Natham, C. (1983). Mouse interferon recep-
NK cell assay. Mice were treated with anti-asialo GM1 antibody starting tors: difference in their response to A and  interferons. J. Gen. Virol. 64: 2777–2780.
approximately 1.5 weeks after s.c. implantation of MDA-MB-468 cells. We 5. Abramovich, C., et al. (1994). Differential tyrosine phosphorylation of the IFNAR chain
administered 50 l anti-asialo GM1 antibody (30 mg/ml) to mice via tail of the type I interferon receptor and of an associated surface protein in response to IFN-
vein injections as suggested by the manufacturer’s instructions (Wako  and IFN-. EMBO J. 13: 5871–5877.
6. Abramovich, C., Chebath, J., and Revel, M. (1994). The human interferon -receptor
BioProducts, Richmond, VA). The animals were injected with the anti-
protein confers differential responses to human interferon- versus interferon- sub-
asialo GM1 antibody every 4 to 5 days for four times. Mice were euthanized types in mouse and hamster cell transfectants. Cytokine 6: 414–424.
for measuring NK activity 7 days after the intratumoral injections of PBS 7. Erlandsson, L., et al. (1998). Interferon- is required for interferon- production in mouse
or viruses. Each treatment group included three mice. NK activity was fibroblasts. Curr. Biol. 8: 223–226.
measured based on a described assay [35] with slight modifications. Briefly, 8. Sica, G., Fabbroni, L., Castagnetta, L., Cacciatore, M., and Pavone-Macaluso, M. (1989).
the effector cells were collected by breaking apart mouse spleens with Antiproliferative effect of interferons on human prostate carcinoma cell lines. Urol. Res.
tweezers in RPMI-1640 medium and passing them through 20-G needles 17: 111–117.
several times. The cells were then washed with PBS and resuspended in 9. Rosenblum, M. G., et al. (1990). Growth inhibitory effects of interferon- but not inter-
feron- on human glioma cells: correlation of receptor binding, 2,5-oligoadenylate syn-
RPMI-1640 media at 2.0  107 cells/ml. Yac-1 cells were used as targets and
thetase and protein kinase activity. J. Interferon Res. 10: 141–151.
their medium was changed 18 h before NK assay to assure that the cells 10. Johns, T. G., et al. (1992). Antiproliferative potencies of interferons on melanoma cell
were in log growth. The labeling of the target cells was performed as lines and xenografts: higher efficiency of interferon-. J. Natl. Cancer Inst. 84: 1185–1190.
described [35]. After incubating the effector and target cells for 2.5 h at 37C 11. Einhorn, S., and Grandér, D. (1996). Why do so many cancer patients fail to respond
and 5% CO2, 100 l of each assay well were transferred to a multiscreen to interferon therapy? J. Interferon Cytokine Res. 16: 275–281.
96-well filtration plate (Millipore, Bedford, MA). Cells were further sepa- 12. Tough, D. F., Borrow, P., and Sprent, J. (1996). Induction of bystander T cell prolifera-
rated from medium by vacuum filtration. Fluorescence was measured at ton by viruses and type 1 interferon in vivo. Science 272: 1947–1950.
485/530 excitation/emission on the Cytofluor 4000 (PerSeptive Biosystems, 13. Rogge, L., et al. (1997). Selective expression of an interleukin-12 receptor component
by human T helper cells. J. Exp. Med. 185: 825–831.
Framingham, MA).
14. Ferrantini, M., et al. (1993). 1-Interferon gene transfer into metastatic friend leukemia
In vitro viral transduction. The viral transduction was carried out as cells abrogated tumorigenicity in immunocompetent mice: antitumor therapy by means
described [28]. After the transduction, cells were collected with of interferon-producing cells. Cancer Res. 53: 1107–1112.
trypsin/EDTA solution and washed twice with PBS. 15. Ferrantini, M., et al. (1994). IFN-1 gene expression into metastatic murine adenocar-
cinoma(TS/A) results in CD8+ T cell-mediated tumor rejection and development of anti-
Animal models. Female BALB/c nu/nu from Charles River Laboratory were tumor immunity: Comparative studies with IFN- producing TS/A cells. J. Immunol. 153:
used in both the ex vivo and in vivo s.c. tumor model experiments. All mice 4604–4615.
were maintained in the pathogen-free Biogen animal facility. A total of 16. Kaido, T., et al. (1995). IFN-1 gene transfection completely abolishes the tumorigenicity
approximately 2  106 cells in 100 l of PBS were implanted s.c. into the of murine B16 melanoma cells in allogeneic DBA/2 mice and decreases their tumori-
right flank or both right and left flanks of mice to generate tumors for both genicity in syngeneic C57BL/6 mice. Int. J. Cancer 60: 221–229.
ex vivo and in vivo experiments. For the direct intratumoral treatment, 100 17. Sarkar S., et al. (1995). Injection of irradiated B16 melanoma genetically modified to

MOLECULAR THERAPY Vol. 4, No. 4, October 2001 363


Copyright © The American Society of Gene Therapy
ARTICLE doi:10.1006/mthe.2001.0464, available online at http://www.idealibrary.com on IDEAL

secrete IFN- causes regression of an established tumor. Int. J. Oncol. 7: 17–24. 27. Fierlbeck, G., et al. (1996). Pharmacodynamics of recombinant IFN- during long-term
18. Tüting, T., et al. (1997). Interferon- gene therapy for cancer: retroviral transduction of treatment of malignant melanoma. J. Interferon Cytokine Res. 16: 777–781.
fibroblasts and particle-mediated transfection of tumor cells are both effective strategies 28. Qin, X.-Q., et al. (1998). Interferon- gene therapy inhibits tumor formation and causes
for gene delivery in murine tumor models. Gene Ther. 4: 1053–1060. regression of established tumors in immune-deficient mice. Proc. Natl. Acad. Sci. USA 95:
19. D’Amore, P. A. (1992). Mechanisms of endothelial growth control. Am. J. Respir. Cell Mol. 14411–14416.
Biol. 6: 1–8. 29. Dong, Z., et al. (1998). Expression of interferon- in UV2237m mouse fibrosarcoma cells
20. Sidky, Y., and Borden, E. (1987). Inhibition of angiogenesis by interferons: effects on suppressed their tumorigenicity and metastasis in syngeneic mice. Cancer Immunol.
tumor- and lymphocyte-induced vascular responses. Cancer Res. 47: 5155–5161. Immunother. 46: 137–146.
21. Dvorak, H. F., and Gresser, I. (1989). Microvascular injury in pathogenesis of interferon- 30. Lu, W., Fidler, I. J., and Dong, Z. (1999). Eradication of primary murine fibrosarcomas
induced necrosis of subcutaneous tumors in mice. J. Natl. Cancer Inst. 81: 497–502. and induction of systemic immunity by adenovirus-mediated interferon  gene therapy.
22. Dinney, C. P. N., et al. (1998). Inhibition of basic fibroblast growth factor expression, Cancer Res. 59: 5202–5208.
angiogenesis, and growth of human bladder carcinoma in mice by systemic interferon- 31. Xie, K., et al. (1997). Abrogation of tumorigenicity and metastasis of murine and human
 administration. Cancer Res. 58: 808–814. tumor cells by transfection with murine interferon  gene: possible role of nitric oxide.
23. Singh, R. K., et al. (1995). Interferons  and  down-regulate the expression of basic Clin. Cancer Res. 3: 2283–2294.
fibroblast growth factor in human carcinomas. Proc. Natl. Acad. Sci. USA 92: 4562–4566. 32. Tada, H., et al. (2001). Systemic IFN- gene therapy results in long-term survival in mice
24. Dong, Z., et al. (1999). Suppression of angiogenesis, tumorigenicity, and metastasis by with established colorectal liver metastases. J. Clin. Invest. 108: 83–95.
human prostate cancer cells engineered to produce interferon-. Cancer Res. 59: 33. Gao, G. P., Yang, Y., and Wilson, J. M. (1996). Biology of adenovirus vectors with E1
872–879. and E4 deletions for liver-directed gene therapy. J. Virol. 70: 8934–8943.
25. Gutterman, J. U. (1994). Cytokine therapeutics: lessons from interferon-. Proc. Natl. 34. Nakabayashi, H., Taketa, K., Yamane, T., Oda, M., and Sato, J. (1985). Hormonal con-
Acad. Sci. USA 91: 1198–1205. trol of -fetoprotein secretion in human hepatoma cell lines proliferating in chemically
26. Salmon, P., Le Cotonnec, J. Y., Galazka, A., Abdul-Ahad, A., and Darragh, A. (1996). defined medium. Cancer Res. 45: 6379–6383.
Pharmacokinetics and pharmacodynamics of recombinant human interferon- in healthy 35. Wierda, W.G., Mehr, D.S., and Kim, Y. B. (1989). Comparison of fluorochrome-labeled
male volunteers. J. Interferon Cytokine Res. 16: 759–764. and 51-Cr- labeled targets for natural killer cytotoxicity assay. J. Immunol. Methods 122:
15–24.

364 MOLECULAR THERAPY Vol. 4, No. 4, October 2001


Copyright © The American Society of Gene Therapy

You might also like