You are on page 1of 13

[CANCER RESEARCH 61, 6201– 6212, August 15, 2001]

Eradication of Intraperitoneal and Distant Tumor by Adenovirus-mediated


Interferon-␤ Gene Therapy Is Attributable to Induction of
Systemic Immunity1
Makoto Odaka, Daniel H. Sterman, Rainer Wiewrodt, Yi Zhang, Michael Kiefer, Kunjlata M. Amin, Guan-Ping Gao,
James M. Wilson, James Barsoum, Larry R. Kaiser, and Steven M. Albelda2
Thoracic Oncology Research Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine [D. H. S., R. W., M. K., S. M. A.], Department of Surgery [M. O., K. M. A.,
L. R. K.], Institute for Human Gene Therapy [Y. Z., G-P. G., J. M. W.], University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, and Biogen, Inc.,
Cambridge, Massachusetts 02142 [J. B.]

ABSTRACT Despite repeated observations that mesothelioma is well known to


inhibit host cellular and humoral antitumor immune responses, there
Malignant mesothelioma remains an incurable disease for which im-
may be a role for intrapleural immunotherapy in the treatment of this
mune-modulatory therapies, such as exogenous cytokines, have shown
refractory neoplasm (6, 7). Using animals models that closely mimic
some promise. One such cytokine, IFN-␤, has potent antiproliferative and
immunostimulatory activity in vitro, but its in vivo use has been limited by the human disease (8), positive therapeutic results have been seen with
toxicity. We thus conducted studies evaluating intracavitary delivery of a IFN-␣ (9), interleukin 2 (10), interleukin 12 (11), and antisense
replication-deficient adenoviral (Ad) vector encoding for the murine TGF3-␤ treatment (12). Some of these findings in animal models have
IFN-␤ gene (Ad.muIFN-␤) in mouse models of malignant mesothelioma. been translated into clinical studies. In several Phase I/II clinical trials
In contrast to multiple injections of recombinant protein, a single i.p. conducted over the past decade, some antitumor efficacy has been
injection of Ad.muIFN-␤ into animals with established tumors elicited demonstrated with systemic and local administration of recombinant
remarkable antitumor activity leading to long-term survival in >90% of human interleukin 2 (13, 14) and granulocyte/macrophage-colony
animals bearing either AB12 or AC29 i.p. mesotheliomas. A control stimulating factor (15).
adenovirus vector had minimal antitumor effect in vivo. Significant ther-
An area of especially active interest and promise in the treatment of
apeutic effects were also seen in animals treated with large tumor burdens.
mesothelioma has been the administration of IFNs. IFNs have immu-
Importantly, treatment of i.p. tumor also led to reduction of growth in
tumors established at a distant site (flank). A number of experiments noregulatory effects upon antibody production, NK and T-cell acti-
suggested that these effects were attributable to an acquired CD8ⴙ T-cell- vation, macrophage function, delayed-type hypersensitivity, and
mediated response including: (a) the induction of long-lasting antitumor MHC antigen expression (16 –19). They also have antiangiogenic
immunity; (b) loss of efficacy of Ad.muIFN-␤ in tumor-bearing, immune- properties (20, 21), as well as direct antiproliferative effects (19, 22).
deficient (SCID, SCID/beige) mice; (c) detection of high levels of specific IFNs have been demonstrated to directly inhibit the cellular prolifer-
antitumor cytolytic activity from unstimulated splenocytes harvested from ation of several mesothelioma cell lines in vitro (23, 24), and antitu-
Ad.muIFN-␤-treated animals that was abolished by CD8ⴙ T-cell deple- mor activity has been shown in animal models (9). Accordingly, both
tion; and (d) abrogation of antitumor effects of Ad.muIFN-␤ in tumor- type I IFNs (␣ and ␤) and type II IFN (IFN-␥) have been studied in
bearing CD8ⴙ T-cell-depleted animals. These data show that intracavitary
clinical trials, although the results have not proved as promising as
IFN-␤ gene therapy using an adenoviral vector provides strong CD8ⴙ
T-cell-mediated antitumor effects in murine models of mesothelioma and
hoped. For example, s.c. administration of IFN-␣2a to patients with
suggest that this may be a promising strategy for the treatment of local- mesothelioma yielded overall response rates of ⬍20% (25, 26). The
ized tumors such as mesothelioma or ovarian cancer in humans. Southwest Oncology Study Group reported no clinical responses after
6 weeks of systemic IFN-␤ treatment in 14 patients with pleural
INTRODUCTION mesothelioma and but observed significant toxic side effects (27).
Improved responses were observed when intrapleural natural IFN-␤
Malignant mesothelioma, an asbestos-related neoplasm of the pleu- was studied in 29 patients with malignant pleural effusions, 11 of
ral and peritoneal space, occurs in about 2000 –2500 patients/year in whom had “complete remission of pleural effusion,” including one
the United States and in ⬃10,000 patients worldwide (1, 2). The patient with mesothelioma (28). IFN-␥ has also been studied in the
incidence, especially in Europe, is rising (3). Malignant pleural me- treatment of mesothelioma, especially in the context of intrapleural
sothelioma is unresponsive to chemotherapy and radiotherapy regi- immunotherapy for mesothelioma. Some positive responses have been
mens and typically recurs even after the most aggressive attempts at seen, especially in patients with very early disease (29, 30).
surgical resection (4). Multimodality approaches have been of some One hypothesis for the relatively low response rates of IFNs in
benefit in prolonging survival in very highly selected subgroups of antitumor therapy is that systemic delivery does not engender the
patients but have had a relatively small impact upon the majority of sustained intratumoral cytokine levels necessary for inducing antitu-
the patients diagnosed with this disease (5). As the incidence of mor responses. For example, pharmacokinetic studies after i.v. injec-
pleural mesothelioma peaks over the next 10 –20 years, new thera- tion have shown that the half-life of recombinant IFN-␤ is only ⬃5
peutic measures will be necessary. min (31). A potential method for increasing local tumor cell exposure
to IFNs with minimal systemic side effects could be tumor cell
Received 3/14/01; accepted 6/8/01. transduction with IFN genes. A number of investigators have tested
The costs of publication of this article were defrayed in part by the payment of page this hypothesis for IFN-␤ by stably transducing tumor cells with
charges. This article must therefore be hereby marked advertisement in accordance with
18 U.S.C. Section 1734 solely to indicate this fact. IFN-␤ and showing marked decreases in tumorigenicity (32–34). This
1
This research was supported by Grant NCI PO1 66726 and Specialized Program of idea has been extended recently to show that in vivo delivery of the
Research Excellence Grant P50-CA-83638 from the National Cancer Institute. Support
was also provided by the Benjamin Shein Foundation for Humanity. R. W. is a postdoc-
3
toral fellow of the Mildred Scheel Stiftung für Krebsforschung der Deutschen Krebshilfe The abbreviations used are: TGF, transforming growth factor; Ad, adenoviral; tk,
e.V. (D/98/02288). J. M. W. holds equity in Targeted Genetics. thymidine kinase; RSV, Rous sarcoma virus; pfu, plaque-forming unit; MOI, multiplicity
2
To whom requests for reprints should be addressed, at University of Pennsylvania of infection; NK, natural killer; CMV, cytomegalovirus; MTS, 3-(4,5-dimethylthiazol-2-
Medical Center, Room 856, BRBII/III, 421 Curie Boulevard, Philadelphia, PA 19104. yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfonyl)-2H-tetrazolium; SCID, severe combined
Fax: (215) 573-4469; E-mail: Albelda@mail.med.upenn.edu. immunodeficient.
6201

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

IFN-␤ gene to flank or brain tumors using various vector approaches 160), which has low levels of MHC class I and is sensitive to NK cell-mediated
resulted in good therapeutic efficacy (35–37). lysis, has been described elsewhere (44). YAC-1 cells were cultured in RPMI
A number of characteristics make pleural malignant mesothelioma 1640 with 10% FBS, 100 units/ml penicillin G, 100 ␮g/ml streptomycin, and
an especially attractive target for gene therapy (38). The location in 2 mM glutamine.
the potential space of the thoracic cavity makes the tumor highly REN cells are a human mesothelioma cell line isolated in our laboratory
from a patient’s tumor specimen (38). I-45 is a human mesothelioma cell line
accessible, facilitating directed administration of gene therapy vectors
obtained as a gift from Dr. Joseph Testa (Fox Chase Cancer Institute, Phila-
and subsequent analysis of treatment effects. In addition, local exten- delphia, PA). These cells were cultured and maintained in RPMI 1640 with
sion of disease, rather than the development of widespread distant 10% FBS, 100 units/ml penicillin G, 100 ␮g/ml streptomycin, and 2 mM
metastases, is responsible for the majority of the morbidity and glutamine.
mortality associated with this neoplasm. Thus, small increments of Recombinant Ad Vectors. Ad vectors encoding the murine and human
improvement in local control could lead to significant improvement in IFN-␤ genes were provided by the Vector Core of the Institute for Human
survival or palliation. Accordingly, our group has successfully com- Gene Therapy (University of Pennsylvania Medical Center). An adenovirus
pleted several Phase I clinical trials of intrapleural gene therapy for transfer vector encoding the murine IFN-␤ gene driven by the CMV early
mesothelioma involving intrapleural instillation of recombinant ad- promoter, pAdCMV-muIFN-␤, was constructed by ligating a cDNA insert
enoviral vectors containing the herpes simplex virus tk (HSVtk) gene encoding murine IFN-␤1a into the plasmid pAd.CMVlink1 (45). pAd.CMV-
muIFN-␤ was cotransfected with the ClaI-restricted H5.010CMVEGP viral
in combination with systemic ganciclovir (39 – 42).
backbone into 293 cells for homologous recombination. The white recombi-
Because of the lack of effective therapy, the potential for response
nant plaques were isolated by a green/white selection process (46). The white
to IFN immunotherapy, and the proven feasibility of local Ad gene plaques were expanded for confirmation in restriction enzyme analysis of the
therapy, malignant mesothelioma represents an ideal tumor to test IFN virus genome and muIFN-␤ expression. A similar virus was prepared by
gene therapy. On the basis of recent successes using an adenovirus ligating a cDNA insert containing the human IFN-␤1a (35) into the same
expressing the human and murine IFN-␤ gene (35, 36), the present shuttle vector.
study was designed to assess the potential of using a recombinant, As a control vector, replication-deficient Ad vector encoding the herpes
replication-deficient adenovirus carrying the murine IFN-␤ gene (Ad- simplex type I tk gene driven by the RSV promoter (Ad.RSVtk) was used as
.muIFN-␤) to treat established i.p. tumors and to explore the mecha- described previously (47). Virus preparations were produced in 293 cells and
nisms of the observed therapeutic effects. purified on CsCl gradients after three rounds of plaque isolation. Vector
preparations were shown to be negative for the presence of wild-type adeno-
We show that Ad.muIFN-␤ efficiently transfects murine mesothe-
virus. The particle:pfu ratio of each preparation was determined in 293 cells
lioma cells both in vitro and in vivo, and that a single i.p. adminis-
and ranged between 50:1 and 150:1.
tration of Ad.muIFN-␤ can effectively treat established malignant Recombinant muIFN-␤. Recombinant muIFN-␤ (41 units/ng) was pro-
mesothelioma tumors in syngeneic, immune-competent mice leading vided by Biogen Inc. The protein was kept at ⫺70°C and did not undergo more
to significantly prolonged survival. The effects were attributable to than one freeze-thaw.
immunological mechanisms because: (a) a single i.p. administration Quantitation of muIFN-␤ Protein Levels by ELISA. One million AB12
of Ad.muIFN-␤ conferred long-lasting, systemic antitumor immunity cells in six-well plates were infected with Ad.muIFN-␤ at a MOI of 50. After
associated with tumor-specific CTL reactivity; and (b) minimal anti- 24 h, the wells were washed and refed with 1 ml of medium containing 1%
tumor efficacy was noted with i.p administration of Ad.muIFN-␤ in FBS. Twenty-four h later, supernatants were collected, and IFN-␤ concentra-
tumor-bearing immune-deficient mice. This systemic immune re- tion was quantified by ELISA. Cell counts per well at the end of the collection
sponse was adequate to inhibit growth of a distant flank tumor upon period were ⬃4 million.
To determine the levels and duration of expression of muIFN-␤ after
treatment of i.p. disease. Thus, intracavitary delivery of the IFN-␤
administration of Ad.muIFN-␤, naı̈ve or tumor-bearing mice received injec-
gene via an Ad vector provides a remarkably effective treatment for tions i.p. with 10 9 pfu of Ad.muIFN-␤. At various time points, animals
locally established murine mesothelioma, as well as generating distant (n ⫽ 3/group) were sacrificed, bled, and subjected to peritoneal lavage with 3
antitumor effects. The mechanism of action in this model appears to ml of PBS. muIFN-␤ levels in blood and peritoneal lavage fluid were then
be primarily dependent on CD8⫹ T lymphocytes rather than on innate determined by ELISA (see below).
immune mechanisms, antiangiogenesis or direct antiproliferative ef- Murine IFN-␤ Assay. Ninety-six-well plates were coated overnight at 4°C
fects. with a monoclonal antibody to mouse IFN-␤ (muIFN-␤ MB-7; Yamasa Shoyu
Co., Ltd., Tokyo, Japan). The antibody was used at a concentration of 10
␮g/ml in the coating buffer containing 50 mM sodium bicarbonate/carbonate,
MATERIALS AND METHODS 0.2 mM MgCl2, and 0.2 mM CaCl2 (pH 9.6). After the plates were blocked with
0.5% nonfat dry milk in PBS for 1 h at room temperature, medium from
Cell Lines. The murine mesothelioma cell lines AB12 and AC29 were infected cells or muIFN-␤ protein standards (Biogen, Inc.) diluted in 0.5%
provided by Dr. Jay K. Kolls of the Louisiana State University School of nonfat dry milk and 0.05% Tween 20 in PBS were added. The plates were then
Medicine (New Orleans, LA). These cell lines were originally generated by Dr. successively incubated at room temperature for 1 h with an anti-muIFN-␤
Bruce Robinson at the Queen Elizabeth II Medical Center (Nedlands, Perth, rabbit sera (produced at Biogen; 1:2000 dilution), 1 h with horseradish per-
Western Australia) by i.p. implantation of asbestos fibers in BALB/c and oxidase-conjugated donkey antirabbit antibody (Jackson ImmunoResearch;
CBA/J mice, respectively, and have been well characterized (8, 43). AB12 and 1:5000 dilution), and the substrate solution (4.2 mM tetramethylbenzidine and
AC29 cells were cultured and maintained in high glucose DMEM (Mediatech, 0.1 M sodium acetate-citric acid, pH 4.9). After the reaction was stopped with
Washington, DC) supplemented with 10% FBS (Georgia Biotechnology, At- 2N H2SO4, absorbance was measured at 450 nm.
lanta, GA), 100 units/ml penicillin, 100 ␮g/ml streptomycin, and 2 mM In Vitro Cytotoxicity Assay. To evaluate the direct antiproliferative effect
glutamine. L1C2 is a murine (BALB/c) bronchoalveolar carcinoma cell line of Ad.muIFN-␤, AB12 and AC29 cells were infected with Ad.muIFN-␤ or
obtained as a gift from Dr. Steven Dubinett (University of California–Los Ad.RSVtk at various concentrations (MOIs of 0.01, 0.1, 1, 10, and 50) and
Angeles, Los Angeles, CA). L1C2 cells were cultured in RPMI 1640 with 10% seeded in 96-well plates at a density of 2 ⫻ 10 3 cells/well. Similar experiments
FBS, 100 units/ml penicillin G, 100 ␮g/ml streptomycin, and 2 mM glutamine. were conducted with Ad.huIFN-␤ using REN or I-45 cells. Survival of infected
The EJ62 line (EJ-6-2-Bam-6a) is a 3T3 mouse fibroblast cell line transformed cells compared with uninfected cells was measured 3 days after infection using
with the H-ras oncogene that forms tumors in BALB/c mice. It was purchased the MTS assay, which is a colorimetric test for the quantification of cell
from the American Type Culture Collection (Manassas, VA) and maintained in viability and proliferation (Cell Titer 96 Aqueous Non-Radioactive MTS Cell
DMEM with 10% FBS, 100 units/ml penicillin G, 100 ␮g/ml streptomycin, Proliferation Assay; Promega Corp., Madison, WI). The percentage of inhibi-
and 2 mM glutamine. The murine lymphoma cell line YAC-1 (ATCC TIB- tion of cell growth was calculated according to the following formula:
6202

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

1 ⫺ A490 of treated group the specificity of the antitumor response. Approximately 11 days after the s.c.
Inhibition 共 % 兲 ⫽ ⫻ 100 challenge, flank tumors were excised and weighed.
A490 of control group To determine the role of T-cell subsets in rechallenge immunity, long-term
survivors from AB12/Ad.muIFN-␤ experiments were rechallenged with AB12
Experimental Animals. Female BALB/c, female CB17-SCID, and female
cells as above. Three groups (each group with 5 mice and 10 flank tumors)
CB17-SCID/beige mice (6 – 8 weeks of age; weight, approximately 20 –25 g)
were studied. One group received i.p. injections of saline, one group was
were obtained from Taconic Laboratory (Germantown, NY). Female CBA/J
depleted of CD4⫹ T cells before and during tumor growth using the schedule
mice (6 – 8 weeks of age; weight, ⬃20 g) were obtained from The Jackson
described below, and one group was depleted of CD8⫹ T cells before and
Laboratory (Bar Harbor, ME). Animals were housed in the animal facility at during tumor growth. After 11 days, animals were sacrificed, and tumor
the Wistar Institute (Philadelphia, PA). All mice were maintained in a patho- weights were determined.
gen-free animal facility for at least 1 week before each experiment. The In Vivo Depletion of CD4ⴙ and CD8ⴙ T Cells. To deplete specific
Animal Use Committees of the Wistar Institute and University of Pennsylvania immune effector cells subsets prior to and during treatment with Ad.muIFN-␤
approved all protocols in compliance with the Guide for the Care and Use of in the AB12 model, BALB/c mice received i.p. injections of 200 ␮g of purified
Laboratory Animals. monoclonal antibodies purified from the anti-CD4⫹ hybridoma GK1.5 and the
Treatment of Tumor-bearing Mice with Ad Vectors. Two independent anti-CD8⫹ hybridoma 53-6.7 (obtained from the American Type Culture
intracavitary tumor models of murine mesothelioma (AB12 and AC29) were Collection). Injections were administered 3 days and 1 day prior to inoculation
established to examine the treatment efficacy of locally administered Ad- with AB12 cells. Thereafter, a maintenance dose of antibody was injected i.p.
.muIFN-␤. Murine mesothelioma (AB12 and AC29) cells were grown to 80% every 7 days throughout the entire experimental period to ensure depletion of
confluence in culture flasks, the medium was aspirated, and cells were har- the targeted cell type. CD4⫹ and CD8⫹ T-cell depletion was confirmed by
vested using 0.05% trypsin-versene. Cells were then pelleted (1000 rpm for 3 flow cytometry of splenic suspensions at the time of tumor injection and
min) and resuspended at 1 ⫻ 105 cells/ml in serum-free DMEM for i.p. weekly afterward. BALB/c mice received i.p. injections of 5 ⫻ 10 5 AB12 cells
administration. AB12 and AC29 cells (5 ⫻ 105 in 0.5 ml) were injected i.p. in 500 ␮l of DMEM on day 0. Four days after tumor inoculation, mice received
into BALB/c and CBA/J mice, respectively, using 25-gauge needles. Four to a single i.p. injection of 500 ␮l of DMEM containing 1 ⫻ 10 9 pfu Ad-
7 days after tumor cell injection, macroscopic (3– 4 mm) tumor nodules could .muIFN-␤. Survival studies were then performed as described above.
be identified on the small bowel mesentery. Later, tumor masses could be Effects of Treatment of i.p. Tumor on Tumors at Distant Sites. To
observed on the diaphragm, peritoneal surface, porta hepatis, lesser sac, and examine the effect of treatment of i.p. treatment of tumor with Ad.muIFN-␤ on
retroperitoneum. Survival studies were performed in lieu of tumor burden the growth of tumor at distant sites, four tumor models of s.c. and/or i.p. AB12
assessment because of the difficulty in harvesting tumor densely adherent to tumors were established. In group 1, 10 mice were injected with 1 ⫻ 106 AB12
the abdominal viscera. Animals were sacrificed when they met predetermined cells in 100 ␮l of serum-free DMEM in each flank. In Group 2, 10 mice were
criteria established for minimizing pain and suffering. Treatment was initiated injected with 1 ⫻ 106 AB12 cells in each flank plus 5 ⫻ 105 AB12 cells and
when approximately 3– 4-mm tumor nodules were identified after tumor cell 500 ␮l of DMEM i.p. In group 3, 10 mice were injected with 1 ⫻ 106 AB12
inoculation. This was usually 4 days after injection of AB12 cells and 7 days cells in each flank and 4 days later received a single i.p. injection of 500 ␮l of
after injection of AC29 cells. DMEM containing 1 ⫻ 109 pfu of Ad.muIFN-␤. This group served as a control
For protein experiments, three groups (n ⫽ 9) of AB12 tumor-bearing mice to see whether systemic circulation of IFN-␤ would affect tumor growth. In
were treated i.p. with saline or with recombinant murine IFN-␤ at a dose of Group 4, 10 mice were injected with 1 ⫻ 106 AB12 cells in each flank plus
3000 units/dose three times weekly (“low dose”) or 30,000 units/dose (“high 5 ⫻ 105 AB12 cells i.p. Four days later, the animals were treated with 1 ⫻ 109
dose”) three times weekly for as long as the mice survived. pfu Ad.muIFN-␤ i.p. This group served as the experimental group to test
whether treatment of i.p. tumor would affect the growth of the flank tumors.
In gene therapy experiments, unless otherwise noted, mice received a single
On day 19 after tumor injection (15 days after treatment with Ad.muIFN-␤),
i.p. administration of Ad.muIFN-␤ at a dose of 1 ⫻ 109 pfu. In control mice,
the flank tumors were harvested and weighed.
Ad.RSVtk was injected at an identical dose of 1 ⫻ 109 pfu without adminis-
To determine the importance of the B- and T-lymphocytes in distant
tration of ganciclovir to control for nonspecific effects of Ad transduction.
response, a similar experiment was performed in three groups of SCID mice
To study the effects of Ad.muIFN-␤ in tumor-bearing immunodeficient
(n ⫽ 10) using the conditions described above for groups 2, 3, and 4.
mice, we used both SCID mice and SCID/beige mice, which underwent i.p
CTL Assay. We performed standard chromium-release assays on murine
injection of murine mesothelioma cell lines (AB12 and AC29) as described
splenocytes to evaluate for the development of tumor-specific CTLs resulting
above.
from Ad.muIFN-␤ treatment of mice bearing i.p. tumors. Splenocytes (two
For the in vivo dose-escalation experiments, BALB/c mice received an i.p. mice/group) were harvested 10 days after i.p. treatment with Ad.muIFN-␤ and
injection of 5 ⫻ 10 5 tumor cells diluted in 500 ␮l of serum-free DMEM. Four used as effector cells. AB12 and EJ62 cells were used as target cells. Impor-
days after tumor implantation, mice received a single i.p. injection of 500 ␮l tantly, the effectors (murine splenocytes from animals post-Ad.muIFN-␤ treat-
of DMEM containing varying doses of Ad.muIFN-␤ (1 ⫻ 10 6, 1 ⫻ 107, ment) were not stimulated in vitro with target cells prior to testing in the
1 ⫻ 10 8, and 1 ⫻ 10 9 pfu). Animals were then observed closely, and survival chromium-release assay. To evaluate NK cell activity, effector cells were
studies were performed. reacted with YAC-1 cells. Target cells (AB12, EJ62, and YAC-1 cells) were
“Bulky” Tumor Model. To examine the effect of Ad.muIFN-␤ instillation labeled with 51Cr by incubating 1 ⫻ 104 cells in a 96-well plate with 3.7 MBq
against bulkier i.p. tumors, we staggered the time of vector instillation (4, 7, of 51Cr (New England Nuclear Life Science Products, Boston, MA) in normal
and 10 days after tumor inoculation) to allow some of the animals to develop growth medium for 1 h and then washed five times with PBS. Varying
larger (5–7 mm) i.p. nodules, as well as allow more tumor involvement in the numbers of effector cells were mixed with target cells to yield E:T ratios of
porta hepatis and small bowel mesentery. BALB/c mice received i.p injections 100:1 to 6.25:1 and were incubated for 5 h in a V-bottomed, 96-well microtiter
of 5 ⫻ 10 5 tumor cells in 500 ␮l of DMEM on day 0; subsequently on day 4, plate. 51Cr release was measured in wells containing effector cells and target
7, or 10, test animals underwent single i.p. instillation of 500 ␮l of DMEM cells (cpm test), wells containing target cells in medium alone (cpm sponta-
containing 1 ⫻ 10 9 pfu of Ad.muIFN-␤. Survival studies were then per- neous), and wells containing target cells plus 10% SDS (cpm total). The
formed. percentage of lysis was calculated using the following formula:
Tumor Rechallenge of Long-Term Survivors. Animals who were long-
term survivors from the above experiments (survival ⬎2 months after primary cpm test ⫺ cpm spontaneous
% lysis ⫽ ⫻ 100
tumor instillation and single Ad.muIFN-␤ treatment) were rechallenged in one cpm total ⫺ cpm spontaneous
flank with s.c. flank injections of 5 ⫻ 10 6 cells of the parental tumor cell line
(AB12 and AC29) in 100 ␮l of serum-free DMEM. Each group contained 10 To deplete CD8⫹ T-cells from the splenocyte population, aliquots of effec-
animals. Long-term survivors in the AB12 model were also challenged s.c. tor cells were incubated for 30 min at 4°C with magnetic MicroBeads conju-
with 5 ⫻ 10 6 cells in the contralateral flank with an additional murine gated to antimouse CD8⫹ T-cell antibodies (Ly-2; Miltenyi Biotec, Inc.,
bronchoalveolar lung cancer cell line, L1C2, syngeneic in BALB/c to evaluate Auburn, CA). Beads and bound CD8⫹ T-cells were magnetically removed
6203

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

from the incubation medium. The CD8⫹ T-cell-depleted splenocytes were then dose, the percentage of inhibition of Ad.muIFN-␤ was significantly
tested for CTL activity as described above. greater than the corresponding dose of Ad.RSVtk control virus
Statistics. The significance of the in vitro inhibition results were deter- (P ⬍ 0.001). However, these cells were less sensitive to the antipro-
mined by Student’s t test (two-tailed) comparing growth inhibition after liferative effects of the virus, with maximum inhibition being only
administration of Ad.IFN versus Ad.RSVtk. Differences in flank tumor 50 – 60% of control virus at the highest dose of 50 MOI. This could
weights in the animal experiments were determined by one-way ANOVA
represent the fact that these murine lines are less infectable by ade-
adapted for multiple comparisons (Tukey-Kramer test; JMP; SAS Institute
Inc., Cary, NC). Statistical significance was set at P ⬍ 0.05. Kaplan-Meier
novirus than are the human lines, or that murine protein has an
survival curves were analyzed with the Mantel-Cox log-rank test. Results are inherently lower antiproliferative activity than human protein.
expressed as mean ⫾ SEM. These studies show that Ad.IFN-␤ has direct antiproliferative ef-
fects on human and murine mesothelioma cells in vitro. Although the
RESULTS human mesothelioma cells are quite sensitive to the direct toxic
effects of IFN-␤, mouse studies using these lines would have to be
Effects of Ad.IFN-␤ on in Vitro Mesothelioma Cell Growth conducted in immunodeficient animals. Because many of the antitu-
mor effects of IFN-␤ are thought to be attributable to its effects on the
To determine the sensitivity of mesothelioma cells to the direct
immune system (36), we reasoned that such studies would not model
antiproliferative effects of Ad.IFN-␤, we first tested the effect of
the clinical situation in which we would actually use this vector.
IFN-␤ gene transfer on human mesothelioma lines in vitro. Because
Accordingly, we conducted the animal studies using the two murine
there is limited cross-species reactivity, Ad.huIFN-␤ was used. Two
mesothelioma cell lines.
mesothelioma cell lines (REN and I-45) were infected with various
MOIs (MOI ⫽ number of pfu of adenovirus/cell) of Ad.huIFN-␤ or a Treatment of Established i.p. Mesothelioma with Recombinant
control virus, Ad.RSVtk (used without addition of ganciclovir as a Murine IFN-␤ or a Single Administration of Ad.muIFN-␤
control for adenoviral toxicity). After three days, cell number was
determined using an MTS assay. The degree of cell growth inhibition To study a clinically relevant model of mesothelioma, we used two
was calculated by comparison to uninfected mesothelioma cells. As models of murine mesothelioma, both based on injection of mouse
shown in Fig. 1, A and B, doses of Ad.huIFN-␤ as small as 0.01 MOI mesothelioma cells injected into the peritoneal cavity of the appro-
induced significant (30 – 40%) growth-inhibitory effects. At MOIs of priate strain of mice. Cell line AB12 grows in BALB/C mice. Cell line
10, 60 –70% growth inhibition was seen. There was minimal cytotox- AC29 grows in CBA/J mice. Both lines can be injected i.p., where
icity with the control adenovirus. At every dose, the percentage of they form diffuse tumors throughout the peritoneal cavity (43), similar
inhibition induced by Ad.huIFN-␤ was significantly greater than the to the presentation of peritoneal human mesothelioma.
corresponding dose of Ad.RSVtk control virus (P ⬍ 0.001). As a point of comparison, we first tested the efficacy of injection of
To study murine mesothelioma cell lines, an Ad.muIFN-␤ virus IFN-␤ protein in the AB12 model. After visible tumor nodules were
was constructed as described in “Materials and Methods.” The vector confirmed, three groups (n ⫽ 9) of tumor-bearing mice were treated
was then used to infect murine mesothelioma AB12 cells at an MOI i.p. with saline or with recombinant murine IFN-␤ at a dose of 3000
of 50. One day after infection, fresh medium was added and collected units/dose three times weekly (“low dose”) or 30,000 units/dose
for 24 h. Medium supernatants were analyzed by ELISA and found to (“high dose”) three times weekly for as long as the mice survived. The
contain 786 ng/ml ⫾ 71 ng/ml of IFN-␤ per 106 cells. This corre- low dose of 3000 units of IFN-␤ corresponds approximately to the
sponds to ⬃32,000 units of IFN-␤ per 106 cells/24 h. maximally tolerated doses used in a humans (10 million units or
The effects of Ad.muIFN-␤ on the growth of two murine mesothe- ⬃143,000 units/kg body weight). As shown in Fig. 2, administration
lioma cells (AB12 and AC29) were studied as above. As shown in of recombinant IFN-␤ at either dose led to significant (P ⫽ 0.02 by
Fig. 1, C and D, growth was also inhibited by Ad.muIFN-␤. At every log-rank test) increases in median survival from 25 days (control) to

Fig. 1. Effects of Ad.IFN-␤ on in vitro mesothe-


lioma cell growth. Human mesothelioma cells,
REN (A) and I-45 (B) cells were infected with
Ad.huIFN-␤ or Ad.RSVtk at various doses (MOIs
of 0.01, 0.1, 1, 10, and 50) and seeded in 96-well
plates at a density of 2 ⫻ 103 cells/well. After 3
days, cell number was determined using a colori-
metric MTS assay. The percentage of inhibition of
cell growth of Ad.IFN-infected or Ad.RSVtk-in-
fected cells compared with noninfected cells is
plotted. Thus, growth was inhibited by ⬃75% in
REN cells infected with Ad.huIFN-␤ at an MOI of
50. Similar experiments were conducted with Ad-
.muIFN-␤ using the murine mesothelioma AB12
(C) or AC29 (D) cell lines. A–D: bars, SEM.

6204

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

were not able to detect activity in the serum at any time point.
Peritoneal lavage levels were 43.2 ⫾ 14 ng/ml at 5 min after injection,
15.5 ⫾ 2.4 ng/ml at 30 min after injection, and undetectable after 4 h.
After one injection of Ad.muIFN-␤, 1 day after i.p. injection, serum
levels averaged 2.7 ⫾ 0.7 ng/ml. At the same time, peritoneal lavage
levels averaged 12.5 ⫾ 4.5 ng/ml. By days 4 and 14, however,
muIFN-␤ levels were below the level of detection in both serum and
lavage fluid. Similar results were seen when 10 9 pfu of Ad.muIFN-␤
were injected into tumor-bearing animals; clearly detectable levels
were present in peritoneal lavage fluid 1 and 2 days after injection but
were undetectable by 4 days. Thus, as expected, peritoneal levels were
very short lived after injection of recombinant protein (⬍4 h), whereas
Fig. 2. Effects of recombinant IFN-␤ on survival in a malignant mesothelioma tumor detectable levels of IFN-␤ were present up to 2 days after the single
model. Three groups (n ⫽ 9) of mice bearing i.p. AB12 tumors were treated i.p. with
saline (✳) or with recombinant murine IFN-␤ at a dose of 3000 units/dose three times
injection of Ad.muIFN-␤.
weekly (“low dose”; F) or 30,000 units/dose (“high dose”; E) three times weekly for as
long as the mice survived. Administration of recombinant IFN-␤ at either dose led to Dose-Response Experiments
significant (P ⫽ 0.02 by log-rank test) increases in median survival from 25 days (control)
to 37 days; however, there was only one long-term (⬎60 days) survivor. There was no To examine the dose-response characteristics of Ad.muIFN-␤, two
difference between the low- and high-dose groups.
sets of experiments were performed using the AB12 model. In the first
experiment, groups of 10 mice were injected with 5 ⫻ 105 AB12
37 days; however, there was only one long-term (⬎60 days) survivor. tumor cells on day 0 and, after confirmation of visible tumor nodules
There was no difference between the low- and high-dose groups. on day 4, were injected with one dose of virus or vehicle. Doses of
Fig. 3 shows the Kaplan-Meier survival curves of groups of tumor- Ad.muIFN-␤ were 109, 108, 107, or 106 pfu. Animals were then
bearing mice (10/group) treated with one dose of Ad.muIFN-␤ (109 followed for survival. As shown in the Kaplan-Meier survival curve
pfu), a control adenoviral vector Ad.RSVtk (109 pfu) given without (Fig. 4A), all control animals died by day 27. Compared with controls,
ganciclovir, or vehicle alone. Vector was injected after confirmation a clear dose-response effect of vector was seen with long-term sur-
of the presence of visible tumor nodules (3 mm in size) in the vival rates of 90% in the animals injected with 109 pfu (P ⬍ 0.0001)
peritoneum. This occurred either 4 days (AB12) or 7 days (AC29) and 30% in the animals injected with 108 pfu (P ⬍ 0.0001). Survival
after injection of tumor cells. was slightly but significantly increased in the 107 pfu animals
All animals injected with adenoviral vectors showed slight ruffling (P ⬍ 0.03). There was no increase on the survival rate in animals
of their fur for 2–3 days after injection; however, no differences were treated with 106 pfu of Ad.muIFN-␤.
noted between control and muAd.IFN-␤-injected animals. Similarly,
no side effects were observed in non-tumor-bearing animals injected
with the vector. In the AB12-injected animals (Fig. 3A), all control
animals (saline and Ad.RSVtk) died by day 31 with a median survival
of 22 days. In contrast, animals injected with one dose of Ad-
.muIFN-␤ had a 100% survival rate (P ⬍ 0.0001). In the AC29-
injected animals (Fig. 3B), the vehicle-injected control animals were
all dead by 60 days with a median survival of 40 days. Animals
injected with the control adenovirus had a slight prolongation of
median survival (40 days versus 54 days), and there were two long-
term survivors. This difference did not quite reach significance
(P ⫽ 0.065 via log-rank test) compared with controls. However,
animals injected with one dose of Ad.muIFN-␤ had a 100% survival
rate (P ⬍ 0.0001), similar to the results with the AB12 cell line. These
experiments have been repeated multiple times with virtually identical
results (see below).
These studies demonstrate that although multiple doses of recom-
binant muIFN-␤ result in a small survival advantage, a single dose of
Ad.muIFN-␤ is remarkably effective in treating two separate models
of i.p. established murine mesothelioma. In both experiments, all of
the animals survived after Ad.muIFN-␤ treatment.

Evaluation of Serum and i.p. Levels of IFN-␤ after i.p.


Injection of muIFN-␤ and Ad.muIFN-␤

To determine the local and systemic levels of IFN-␤ after i.p.


injection of muIFN-␤ or Ad.muIFN-␤, animals were given a single
dose of 30,000 units of recombinant protein or one dose of 10 9 pfu
Fig. 3. Effects of Ad.muIFN-␤ on survival in two malignant mesothelioma tumor
of Ad.muIFN-␤ via an i.p. injection. At various time points after models. A single i.p. dose (arrows) of 1 ⫻ 109 pfu of Ad.muIFN-␤ (E) or Ad.RSVtk (Œ)
injection, animals were sacrificed, and the levels of IFN-␤ in serum or was administered to BALB/c mice bearing AB12 tumors 4 days after tumor injection (A)
peritoneal lavage fluid were determined by ELISA. Levels in serum or to CBA/J mice bearing AC29 tumors 7 days after injection (B). Control mice received
500 ␮l of saline i.p. (✳). Animals (n ⫽ 10/group) were then followed for survival.
and after a 3-ml peritoneal lavage at baseline were below detection Significant long-term survival (100%) was observed in treated mice receiving Ad-
levels (⬍0.4 ng/ml). After one injection of recombinant muIFN-␤, we .muIFN-␤ in both models (P ⬍ 0.0001).
6205

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

Induction of Long-Term Immunity after Ad.muIFN-␤ Therapy

To determine whether treatment with Ad.muIFN-␤ therapy induced


an acquired immune response against the tumor cells, treated animals
“cured” of their tumors were rechallenged with a s.c. injection of
tumor. Groups of 10 mice “cured” of AB12 tumors (⬎60 days after
tumor injection and treatment with Ad.muIFN-␤) or naı̈ve, control
BALB/c mice were injected in both flanks with either 5 ⫻ 106 AB12
cells or L1C2 cells. L1C2 cells are a mouse lung adenocarcinoma cell
line that also grows in BALB/c mice.
After 11 days, the flank tumors were removed and weighed. As
shown in Fig. 5, there were statistically significant differences
(P ⬍ 0.0001) in the size of the tumors induced by injection of AB12
cells into “cured” mice versus previously untreated mice. Specifically,
tumors in previously treated mice did not grow, in contrast to the
marked growth of the same cells injected into naı̈ve BALB/C mice.
Interestingly, there appeared to be some “cross-sensitization” to the
L1C2 cell line, because these cells did not grow well in the “cured”
mice compared with control mice (P ⬍ 0.0001). Similar results were
seen with the AC29 model, where none (0 of 10) of the “cured” mice
injected with AC29 cells developed flank tumors, whereas all of the
control CBA/J mice (10 of 10 tumors) developed large flank tumors
(data not shown).
These data indicate that animals successfully treated with Ad-
.muIFN-␤ develop long-lasting protective immunity upon rechallenge
Fig. 4. Dose-response experiments. A, BALB/c mice bearing AB12 tumors were with the same tumor. In one case, this antitumor response appeared to
treated with a single dose of Ad.muIFN- ␤ via the i.p. route at varying doses, i.e., 109 (E),
10 (F), 107 (䡺), 106 (f) pfu at day 4 after tumor inoculation. Control mice received 500
8 extend to a second tumor type.
␮l saline i.p. (✳). Animals were then followed for survival. Significant increases in
survival were seen in animals given 109, 108, and 107 pfu of virus. B, BALB/c mice Effects of Ad.muIFN-␤ in Immunodeficient Animals
(n ⫽ 9/group) received i.p injections of AB12 tumor cells on day 0. Subsequently, on day
4 (E), day 7 (F), or day 10 (䡺), test animals were administered a single i.p. dose of Although our rechallenge experiments indicated development of
1 ⫻ 109 pfu of Ad.muIFN-␤ and were followed for survival. Control animals received a
dose of saline on day 4 (✳). Significant increases in survival were seen in all treated long-term immunity to tumor cells, these studies did not define how
groups. tumor cells were eliminated after i.p. injection. IFN-␤ has been shown
to work by a number of mechanisms, including direct cytotoxicity,
In the second experiment, we varied the time of vector injection inhibition of angiogenesis, NK and macrophage activation, as well as
after tumor inoculation to determine whether Ad.muIFN-␤ would be induction of an acquired (T-cell-mediated) immune response (see
effective against larger tumors. Groups of 11 mice were injected with “Discussion”). To determine which of these mechanisms were oper-
5 ⫻ 105 AB12 tumor cells on day 0. On days 4, 7, and 10 after tumor ative in our model, we tested the efficacy of Ad.muIFN-␤ after
injection, two mice were sacrificed to confirm the size of visible injection into tumor-bearing immunodeficient mice.
tumor nodules. At day 4, multiple 3– 4-mm tumor nodules were noted Groups of 10 SCID/beige mice were injected with 5 ⫻ 105 AB12
on the small bowel and mesentery. One group of animals received an
i.p. injection of medium alone (control), and a second group of
animals (group 1) were injected i.p. with one dose of Ad.muIFN-␤
(109 pfu). On day 7, the nodules had increased to 5– 6 mm in size. At
this time, group 2 animals were injected i.p. with one dose of Ad-
.muIFN-␤ (109 pfu). By day 10, there was extensive bulky disease
with large irregularly shaped 7– 8-mm nodules throughout the perito-
neal cavity, as well as smaller nodules on the bowels, mesentery, and
diaphragm. At this time, group 3 animals were injected i.p. with one
dose of Ad.muIFN-␤ (109 pfu).
As shown in Fig. 4B, control animals died rapidly with a median
survival of 17 days and with all animals dead by 28 days. All treated
groups had significant increases in survival compared with control
animals (P ⬍ 0.0001) with a clear dose-response effect noted. Ani-
mals injected on day 7 had a 33% long-term survival rate with an
increase in median survival to 44 days (P ⬍ 0.0001). Even those
animals with large tumors treated on day 10 had a significant
(P ⫽ 0.0003) increase in median survival (31 days versus 17 days) Fig. 5. Existence of long-term antitumor immunity. Long-term survivors in the AB12
with one long-term survivor. Consistent with our previous study (Fig. model (survival ⬎2 months; see Fig. 3A) were rechallenged with the s.c. parental AB12
tumor cell line (䡺) or with the syngeneic L1C2 tumor cell line (f). Previously untreated
3), animals injected at day 4 had 100% survival. control animals were injected with the same number of tumor cells. Eleven days after the
Thus, Ad.muIFN-␤ successfully treats tumors in a dose-responsive s.c. challenge, flank tumors were excised and weighed. In the control mice, both tumor
fashion. The dose inducing complete survival was 109 pfu. Significant cell lines produced tumors that grew to large size. In the previously “cured” animals,
AB12 tumor growth was completely inhibited. Interestingly, growth of L1C2 cells was
therapeutic effects were also seen in animals with large tumor bur- also markedly diminished. These data demonstrate the induction of long-term antitumor
dens. immunity. Bars, SE.
6206

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

tumor cells on day 0. On day 4, two mice were sacrificed to confirm


the presence of visible tumor nodules. After confirmation, animals
were injected with one dose of Ad.muIFN-␤ (109 pfu) or vehicle.
Animals were then followed for survival. As shown in Fig. 6A, in
contrast to the immunocompetent BALB/c mice injected with AB12
cells (see Fig. 3), the therapeutic efficacy of Ad.muIFN-␤ was almost
completely lost, with no increase in median survival and no survival
advantage. Because SCID/beige mice are deficient in B cells, T cells,
and NK cells, we conducted a similar experiment in SCID mice that
lack B and T cells but have intact NK function. As shown in Fig. 6B,
virtually identical results were obtained; the therapeutic efficacy of
Ad.muIFN-␤ was almost completely lost, with virtually no increase in
median survival and no survival advantage. Fig. 7. Effect of in Ad.muIFN-␤ on BALB/c mice bearing AB12 tumor cells after
depletion of CD4⫹ or CD8⫹ T cells. A single i.p. dose of 1 ⫻ 109 pfu of was administered
The SCID mouse experiment was repeated using the AC29 cells. to BALB/c mice who had been injected i.p. with AB12 cells 4 days earlier. Antibodies
Seven days after tumor inoculation and confirmation of tumor growth, specific for CD8⫹ and CD4⫹ T cells were used to deplete these cell types before and
during treatment with Ad.muIFN-␤. There were no differences in survival between the
animals were injected with one dose of Ad.muIFN-␤ (109 pfu) or untreated animals (✳) and the CD8⫹ T-cell-depleted animals treated with Ad.muIFN-␤
vehicle. As shown in Fig. 6C, in this model, there was a slight increase (f). The animals depleted of CD4⫹ T cells (Œ) showed a significant increase in survival
in median survival from 26 to 32 days (P ⬍ 0.0001); however, all of but with no long-term survivors. Fully immunocompetent mice treated with Ad.muIFN-␤
(E) had an 80% survival rate in this experiment.
the treated animals were dead by day 39.
These data show that in the AB12 model, almost all of the thera-
peutic effects of Ad.muIFN-␤ are attributable to factors related to
acquired immunity. In the AC29 model, there appears to be a small Determination of the Immune Mechanism of Tumor Killing
increase in survival (which may be attributable to Ad or IFN-␤
Depletion of CD4 and CD8 Cells. The experiments described
effects), but these effects do not lead to long-term survival.
above indicate that acquired immune responses are the primary mech-
anism of the effects of Ad.muIFN-␤ in our models. To more precisely
define this mechanism, groups of mice were treated with anti-CD4 or
anti-CD8 antibodies to deplete these populations of cells. After ade-
quacy of depletion was confirmed by performing fluorescence-acti-
vated cell sorter analysis of spleen cells (data not shown), groups of
10 BALB/c mice received injections of 5 ⫻ 105 AB12 tumor cells on
day 0. On day 4, after confirmation of the presence of visible tumor
nodules, animals were injected with one dose of Ad.muIFN-␤ (109
pfu) or vehicle and followed for survival. Depletion of the appropriate
T-cell population was confirmed weekly by analysis of spleen cells by
flow cytometry (data not shown).
As shown in Fig. 7, there were no significant differences in survival
between the untreated animals and the CD8⫹ T-cell-depleted animals
treated with Ad.muIFN-␤. In contrast, the animals depleted of CD4⫹
T cells showed a marked increase in survival, although not to the level
of intact animals (P ⬍ 0.0004 versus the saline group). This experi-
ment indicates that, in this model, CD8⫹ T cells are central to the
therapeutic effects of Ad.muIFN-␤. CD4⫹ T cells also play a role in
the generation of antitumor immunity, but even in their absence,
significant antitumor effects are still observed.
To determine the role of CD4⫹ and CD8⫹ T cells in established
tumor immunity, groups (n ⫽ 5) of naı̈ve mice or mice cured of AB12
tumors by previous injection of Ad.IFN-␤ were reinjected with AB12
cells in both flanks (Fig. 8). In one group, CD4⫹ T cells were depleted
using antibody injections at the time of tumor rechallenge of “cured”
animals, and in a second group, CD8⫹ T cells were depleted by
antibody injections. Tumor growth was then assessed at 11 days.
Consistent with previous data (Fig. 5), AB12 cells injected into the
flanks of naı̈ve animals grew to large size, whereas tumor growth was
completely inhibited in previously treated, “cured” mice (Fig. 8).
Depletion of CD8⫹ T cells completely blocked this antitumor effect,
allowing tumors to grow to near control size in these animals. How-
ever, depletion of CD4⫹ T cells had very little effect on tumor growth
Fig. 6. Effect of Ad.muIFN-␤ treatment in immunodeficient mouse strains. Four days
after the injection of AB12 cells, a single i.p. dose of 1 ⫻ 109 pfu of Ad.muIFN-␤ (E;
(tumor size was not significantly different from that seen in nonde-
arrows) was administered to SCID/beige (A) or SCID (B) mice. The same dose of virus pleted “cured” mice). This experiment demonstrates that CD8⫹ T
was administered to SCID mice 7 days after i.p. injection of AC29 cells (C). Control mice cells are absolutely required for antitumor immunity. CD4⫹ T cells
received 500 ␮l saline i.p. (✳). Animals (n ⫽ 10/group) were then followed for survival.
The therapeutic efficacy of Ad.muIFN-␤ was markedly diminished in the immunodefi- not directly involved in the recall antitumor effects but seem to be
cient mice. more important in helping to generate CD8⫹ T cells.
6207

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

2, 10 mice were injected with 1 ⫻ 106 AB12 cells in each flank plus
5 ⫻ 105 AB12 cells i.p. This group served as a control group to rule
out inhibitory effects on flank tumor growth by the concurrent growth
of i.p. tumor. In group 3, 10 mice were injected with 1 ⫻ 106 AB12
cells in each flank and 4 days later were treated i.p. with 1 ⫻ 109 pfu
of Ad.muIFN-␤. This group served as a control to see whether
systemic circulation of IFN-␤ after i.p. injection would affect tumor
growth. In group 4, 10 mice were injected with 1 ⫻ 106 AB12 cells
in each flank plus 5 ⫻ 105 AB12 cells i.p. Four days later, the animals
were treated with 1 ⫻ 109 pfu of Ad.muIFN-␤ i.p. This group served
as the experimental group to test whether treatment of i.p. tumor
would affect the growth of the flank tumors.
Fig. 8. Importance of CD4⫹ or CD8⫹ T cells for long-term antitumor immunity. On day 19 after tumor injection (15 days after Ad.muIFN-␤), the
Long-term (cured) survivors in the AB12 model (survival ⬎2 months) were rechallenged
with bilateral flank injections of parental AB12 tumor cell line. Previously untreated flank tumors were harvested and weighed. As shown in Fig. 10A,
control (naı̈ve) animals (n ⫽ 5) were injected with the same number of tumor cells. In one AB12 flank tumors grew to large size in the control animals (group 1),
cured group (n ⫽ 5), no T-cell depletion was performed. In one cured group (n ⫽ 5),
CD4⫹ T cells were depleted using antibody injections at the time of tumor rechallenge of
“cured” animals. In the third cured group (n ⫽ 5), CD8⫹ T cells were depleted by
antibody injections. Tumor growth was then assessed by weight at 11 days after tumor
injection. AB12 cells injected into the flanks of naı̈ve animals grew to a large size,
whereas tumor growth was completely inhibited in previously treated, “cured” mice.
Depletion of CD8⫹ T cells completely blocked this antitumor effect (tumors grew to a
large size and were not significantly different in size than naı̈ve controls). Depletion of
CD4⫹ T cells did not prevent the inhibition of tumor growth [tumor size was not
significantly different from that seen in nondepleted “cured” mice; both groups were
significantly different from naı̈ve mice (P ⬍ 0.05)]. This experiment demonstrates that
CD8⫹ T cells, but not CD4⫹ T cells, are required for antitumor immunity. Bars, SE.

Evaluation of Cytotoxic T Cells. To confirm the importance of


cytotoxic CD8⫹ T cells, control BALB/c mice, AB12 tumor-bearing
mice treated with a control Ad.RSVtk adenovirus, and AB12 tumor-
bearing mice treated with Ad.muIFN-␤ were sacrificed 10 days after
vector treatment (14 days after tumor instillation). Splenocytes were
isolated and assessed for the presence of CTLs using chromium-
labeled AB12 or EJ62 (a murine fibrosarcoma that grows in BALB/c
mice) cell targets. Of note, the splenocytes were not stimulated in
vitro. Fig. 9A shows that there was significant cytolytic activity of
splenocytes from the Ad.muIFN-␤-treated animals against AB12 tar-
gets. In contrast, there was no cytolytic activity seen in control
splenocytes or splenocytes from the animals treated with the control
Ad.RSVtk vector against the AB12 targets. No cytolytic activity was
seen against EJ62 targets using splenocytes from the Ad.muIFN-␤-
treated animals or any treatment group (Fig. 9B).
To confirm that the cytolytic activity was attributable to CD8⫹ T
cells, splenocytes from control or Ad.muIFN-␤-treated animals were
depleted of CD8⫹ T cells using anti-CD8-coated beads prior to use in
a CTL assay. This treatment completely eliminated cytolytic activity
of the cells against the chromium-labeled AB12 targets (Fig. 9C).
Splenocytes from control or Ad.muIFN-␤-treated animals were also
tested for cytolytic effects against labeled YAC-1 target cells. No
significant lysis was seen at E:T ratios of up to 100 (data not shown),
indicating minimal NK cell activity.
These experiments demonstrate that treatment of i.p. AB12 tumor
with Ad.muIFN-␤ generates tumor-specific CD8⫹ cytotoxic T cells
and that these cells account for almost all of the in vitro antitumor
activity. No evidence for active NK cell lysis was seen. Fig. 9. In vitro cytotoxic activity of splenocytes from tumor-bearing BALB/c mice
treated with i.p. Ad.muIFN-␤. A single i.p. dose of 1 ⫻ 109 pfu of Ad.muIFN-␤ or
Ad.RSVtk was administered to BALB/c mice bearing AB12 tumors 4 days after tumor
Effects of Treatment of i.p. Tumor on Tumors at Distant Sites injection. Ten days after i.p. treatment, splenocytes were harvested and used as effector
cells. AB12 (A and C) or syngeneic EJ62 (B) cells were chromium labeled and used as
The demonstration of systemic cytotoxic CD8⫹ T lymphocytes target cells. The effector cells were not stimulated in vitro with target cells prior to testing
suggested that antitumor effects might not be limited to the local (i.p.) in the chromium-release assay. As shown in A, there was significant cytolytic activity of
splenocytes from the Ad.muIFN-␤ treated animals (E) against AB12 targets. In contrast,
tumor site. To determine whether successful treatment of local (i.p.) there was no cytolytic activity against the AB12 targets using control splenocytes (✳) or
disease could lead to antitumor effects at distant (flank) sites, the splenocytes from the animals treated with the control Ad.RSVtk vector (Œ). As shown in
following four groups of animals were established. In group 1, 10 B, no cytolytic activity was seen against EJ62 targets using splenocytes from any
treatment group. In C, splenocytes depleted of CD8⫹ T cells (⽧) by the use of magnetic
mice were injected with 1 ⫻ 106 AB12 cells in each flank. This group beads completely lost their cytotoxic activity against the parental AB12 cells when
served as the control group for the growth of flank tumors. In group compared with nondepleted splenocytes (E).
6208

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

effects were seen in animals treated with various Ad.muIFN-␤ doses


(Fig. 4A) and with very large tumor burdens (Fig. 4B). A dose of
1 ⫻ 10 9 pfu of Ad.muIFN-␤ led to almost 100% survival. Taking into
account size differences (⬃3000-fold), this dose would be the human
“equivalent” of 3 ⫻ 10 12 pfu of virus, a dose similar to the highest
dose of Ad.HSVtk (1 ⫻ 10 12 pfu) that we administered in our
previous mesothelioma clinical trial (40). Even a single dose of vector
one log lower, 1 ⫻ 10 8 pfu, significantly prolonged survival com-
pared with control vector and resulted in complete tumor regression in
⬃30% of the treated animals. A single i.p injection of Ad.muIFN-␤ in
mice bearing bulky intra-abdominal mesothelioma tumors (7 or 10
days after instillation) also resulted in prolongation of survival com-
pared with controls, although with less antitumor efficacy than mice
treated at earlier tumor stages (day 4). These results suggest that
treatment of mesothelioma at an early stage or in association with a
surgical “debulking” procedure to remove gross disease may be
successful strategies for Ad.muIFN-␤ gene therapy in future human
clinical trials. In addition to effective treatment of local i.p. tumors,
we were also able to demonstrate that treatment of i.p. tumor led to
inhibition of growth of tumor cells at a distant (flank) site (Fig. 10).
This finding raises the exciting potential of using i.p. (or intrapleural)
therapy as a way to treat systemic disease.
The type I IFNs, IFN-␣ and IFN-␤, are a family of closely related
cytokines that possess potent antiviral and immunoregulatory activi-
ties (17, 18, 48). IFN-␣ and IFN-␤ are produced by both immune and
nonimmune cell types and can be induced to high systemic levels in
response to viral infections. Type I IFNs can inhibit tumor growth
Fig. 10. Effects of treatment of i.p. tumor on tumors at distant sites. A, to examine the
antitumor effects on distant sites after i.p. treatment, four treatment groups were estab-
directly by suppressing cell replication and inducing differentiation or
lished. In group 1, 10 mice were injected with 1 ⫻ 106 AB12 cells in each flank. In group apoptosis (19, 22, 49) and indirectly by activating NK-cell mediated
2, 10 mice were injected with 1 ⫻ 106 AB12 cells in each flank plus 5 ⫻ 105 AB12 cells lysis (48) and tumoricidal properties of macrophages (32, 33, 50).
i.p. In group 3, 10 mice were injected with 1 ⫻ 106 AB12 cells in each flank and 4 days
later were treated i.p. with 1 ⫻ 109 pfu of Ad.muIFN-␤. In group 4, 10 mice were injected Type I IFNs can also promote tumor regression by suppression of
with 1 ⫻ 106 AB12 cells in each flank plus 5 ⫻ 105 AB12 cells i.p., and 4 days later, the angiogenesis (21) and stimulation of specific cellular immune re-
animals were treated with 1 ⫻ 109 pfu of Ad.muIFN-␤ i.p. On day 19 after tumor sponses. Additionally, IFN-␤ also has the potential to induce NK cell
injection, the flank tumors were harvested and weighed. Flank tumors grew to large size
in groups 1–3. In contrast, flank tumor growth was markedly inhibited in group 4 proliferation (51) and to promote antitumor T-cell responses by in-
(P ⬍ 0.05, all comparisons). Bars, SE. B, to determine the importance of T and B cells ducing proliferation of memory phenotype subsets and prolonging
in this distant tumor effect, groups 2, 3, and 4 were established as described above using
immunodeficient SCID mice. Flank tumors grew to a large size in all groups. In contrast
survival of activated populations (52, 53). Finally, type I IFNs can
with the experiment above, group 4 tumors were not inhibited. Bars, SE. up-regulate MHC class I expression (54).
Taking advantage of these multiple potential mechanisms for anti-
tumor effects, a number of groups have shown that expression of
and this growth was not affected by the presence of tumor growing i.p. IFN-␤ by tumor cells (through ex vivo or in vivo transfection) results
(group 2) or by the injection of Ad.muIFN-␤ into the peritoneal cavity in impressive antitumor effects in animal models. In 1994, Yagi et al.
(group 3; P ⬎ 0.05 for all comparisons). However, flank tumor (55) showed antitumor effects of cationic liposome encapsulated
growth was markedly inhibited in the group 4 animals (P ⬍ 0.05, all huIFN-␤ in human gliomas that had been transplanted into nude mice.
comparisons). More recent work by the same group has shown antitumor effects and
To confirm that this antitumor response was dependent on acquired the generation of cellular immunity by liposomal-mediated transfer of
immune responses, the experiment was repeated using immunodefi- murine IFN-␤ into mouse gliomas (37, 56). Studies performed by Xie
cient SCID mice instead of BALB/c animals (in this experiment, et al. (32) and Xu et al. (33) of injecting human and murine cancer
group 1 animals were not studied). In contrast to the results in cells transfected with murine IFN-␤ into nude mice demonstrated that
immunocompetent animals, the growth of flank tumors in the group 4 cell growth of transduced tumor cells was markedly inhibited, with the
SCID animals was not inhibited (Fig. 10B). These experiments show antitumor effects being attributed to up-regulation of inducible nitric
that treatment of i.p. tumor also results in “distant” antitumor effects oxide synthase in murine macrophages. Qin et al. (35) showed that an
that require an intact immune system. adenovirus expressing the human IFN-␤ gene (Ad.huIFN-␤) could be
injected into established human tumors (breast, colon, cervical, and
DISCUSSION hepatoma) growing as flank tumors in SCID or nude mice and induce
significant inhibition of tumor growth. Because these latter studies
The data presented here demonstrate remarkable in vivo antitumor were done in immunodeficient animals, effects were likely attributa-
activity of murine IFN-␤ gene delivery using an adenoviral vector ble primarily to innate immune responses, direct antiproliferative
(Ad.muIFN-␤) in two murine models of mesothelioma. More than effects, or antiangiogenic activity.
90% of animals bearing either AB12 (Fig. 3A) or AC29 (Fig. 3B) i.p. In experiments with perhaps more relevance to an actual clinical
mesotheliomas tumors showed complete tumor regression. A control scenario, Lu et al. (36) showed that an Ad.muIFN-␤ vector multiply
adenovirus vector had minimal antitumor effect in vivo. In contrast, injected into a murine sarcoma line growing in the flanks of immu-
multiple i.p. injections of high doses of recombinant mu.IFN-␤ had nocompetent C3H mice led to tumor growth inhibition. In these
only minimal therapeutic effects (Fig. 2). Significant therapeutic studies, an antitumor immune response was postulated because: (a)
6209

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

injection of Ad.muIFN-␤ induced a prominent T-cell infiltration into performed, the study of Lu et al. (36) was similar to ours in that
the tumors; (b) reinjected tumors did not grow in previously treated immunity to re-injection of tumor cells was observed and that the
animals; and (c) the vector was ineffective when injected into tumors therapeutic effects of Ad.muIFN-␤ were largely lost in SCID mice,
growing in immunodeficient mice. thus suggesting minimal effects of NK cells. Important differences
Given this multiplicity of effects, it was thus of interest to define the between the studies included: (a) that multiple doses of vector were
mechanisms by which i.p. injection of Ad.muIFN-␤ led to its antitu- administered to flank tumors by Lu et al. (Ref. 36; compared with a
mor effects in our mesothelioma models. Our studies show that single i.p. dose in our study); (b) no cross sensitization was observed;
acquired, CD8⫹ T-cell-mediated immune responses are primarily and (c) an “immunogenic” tumor (UV-2237m) versus “non-immuno-
responsible for the therapeutic efficacy observed. There appear to be genic” tumor (AC29; Ref. 57) was used. Experiments showing that
relatively small contributions from other mechanisms, such as macro- local treatment could lead to distant therapeutic effects were not
phage or NK cell activation, direct antiproliferative effects, or anti- performed in either of the other studies.
angiogenesis. This conclusion is based on the following data: It is interesting to speculate on why administration of Ad.muIFN-␤
(a) Successful treatment led to antitumor immunity, evidenced by was so effective in our models of mesothelioma. An exceptionally
the failure of AB12 cells to grow in the flanks of previously treated powerful T-cell response was generated because treatment with only
animals (Fig. 5). This immunity was completely abolished by deple- a single dose of Ad.muIFN-␤ not only produced a high percentage of
tion of CD8⫹ T cells at the time of rechallenge (Fig. 8). Interestingly long-term tumor-free survivors but also resulted in inhibitory effects
(see below), this antitumor immunity extended to a second, unrelated on tumors cells growing at a distant site. In addition, we saw CTL
lung cancer tumor. activity in splenic cells without the need for in vitro stimulation. In
(b) The therapeutic effects of Ad.muIFN-␤ were almost completely previous immunotherapy experiments using liposomes containing
attenuated in SCID/beige and SCID mice (Fig. 6). The complete lack DNA and using the same cell lines, but with multiple treatments, we
of efficacy in SCID/beige mice, who lack B, T, and NK cells, shows found that at least one round of in vitro stimulation was needed to
that the antiproliferative activity of IFN-␤ on the tumor cells and the detect CTL activity (43). Treatment in the current study led to the
antiangiogenic properties were minimal. This is in marked contrast to generation of systemic immunity to challenge with parental tumor
the effects of IFN-␤ injected into human tumors growing on SCID/ cells; however, somewhat surprisingly, we also observed immunity to
beige mice reported by Qin et al. (35). The experiments in SCID mice, another syngeneic, nonmesothelioma tumor, the L1C2 murine lung
which lack B and T cells but have functional macrophages, neutro- adenocarcinoma. The appearance of such cross-sensitization has been
phils, and NK cells, were particularly useful in showing that the reported (58) but is unusual.
There are a number of possible explanations for the strength of
IFN-␤-dependent macrophage and NK cell activation that appear
these effects. For example, the route of administration may be impor-
prominent in other models were of only minor significance important
tant. The peritoneal cavity is an anatomical site where immune re-
in our system. It is interesting that in at least one other syngeneic
sponses are vigorous, and thus the i.p. route has been used often for
murine tumor model (the growth of liver metastases after injection of
in vivo immunization (59, 60). One reason for this is the large surface
the CT26 colon carcinoma line), NK cell activation appears to be very
area available. Another advantage could be the presence of large
important in the antitumor effects.4
numbers of free floating macrophages and dendritic cells in this cavity
(c) We were able to demonstrate that splenocytes isolated from
(61) that have free access to the tumors growing at the surface of the
Ad.muIFN-␤-treated animals (but not control animals or animals
peritoneum. In addition, it has been recognized recently that normal
treated with control adenoviral vector) had high specific cytolytic
mesothelial cells also have antigen-presenting capability and may be
activity against AB12 tumor cells (Fig. 9). This lytic activity was
participating in the generation of antitumor immune responses (60, 62,
completely abolished by depletion of CD8⫹ T cells (Fig. 9C) showing
63). Experiments are ongoing to test the effects of Ad.muIFN-␤
that CD8⫹ T cells, rather than NK cells, were of primary importance.
injected into flank tumors compared with i.p. tumors. The extent of
Of note is the fact that CTL activity was elicited in cells without the gene transfer after vector administration may also be important. We
need for in vitro stimulation, suggesting very strong CTL responses. have shown previously that large numbers of tumor cells and normal
(d) We were able to abrogate completely the antitumor effects of mesothelial cells are transduced after i.p. injection of adenovirus (38).
Ad.muIFN-␤ by depleting animals of CD8⫹ T cells using injection of There may also be gene transfer into leukocytes within the peritoneal
anti-CD8 antibodies (Fig. 7). A significant, but lesser, inhibitory cavity, although we have not specifically examined this issue. Future
effect was seen after depletion of CD4⫹ T cells, suggesting that both studies are aimed at exploring some of these questions.
arms of the acquired immune system play important roles in gener- There are also some factors specific to mesothelioma that may also
ating antitumor immunity. However, depletion of CD4⫹ T cells did be important. Unlike many tumors, mesotheliomas express high levels
not block the ability of “cured” mice to reject a rechallenge of tumor of MHC class I antigens (64) that may make them more resistant to
cells (Fig. 8). NK cell activity and more susceptible to CD8⫹ T-cell lysis. Both
These findings share similarities and differences with the previous AB12 and AC29 cells express high levels of MHC class I molecules
studies of Natusme et al. (56) and Lu et al. (36). Similar to our (data not shown). Mesothelioma cells make large amounts of TGF-␤
findings, Natusme et al. (56), who injected liposomally encapsulated that may induce an immunosuppressive environment (12). Bielefeldt-
IFN-␤ into murine brain tumors, found immunity to re-injected tumor Ohmann et al. (9) showed that IFN-␣ suppressed TGF-␤ mRNA
cells and detected splenic CTLs, the specific antitumor cytolytic expression in mesothelioma tumors. Although not examined in our
activity of which could be abrogated completely by CD8⫹ T-cell study, it is possible that IFN-␤ inhibited the production of TGF-␤ in
depletion. They were also able to block antitumor effects in animals a similar fashion, thus improving immune responses. Finally, the fact
by administration of anti-CD8 monoclonal antibodies. In contrast to that mesothelioma cells are sensitive to the antiproliferative and
our results, however, detection of CTL activity required two to three apoptotic effects of Ad.muIFN-␤ (Fig. 1) could play a key role in
rounds of in vitro stimulation, and no cross-sensitization responses generating appropriate “danger signals” needed to drive effective
were noted. Although a more limited immunological analysis was antitumor responses (65, 66). The importance of having the tumor
cells transduced is suggested in preliminary experiments, where i.p.
4
F. Spitz, University of Pennsylvania, personal communication. injection of Ad.muIFN-␤ followed by injection of tumor cells is
6210

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

ineffective whereas ex vivo transduction of tumor cells prior to i.p. 16. Sen, G. C., and Lengyel, P. The interferon system. J. Biol. Chem., 267: 5017–5020,
1992.
injection leads to complete cure in immunocompetent mice but not in 17. Pfeffer, L. M., Dinarello, C. A., Herberman, R. B., Williams, B. R., Borden, E. C.,
SCID mice. Bordens, R., Walter, M. R., Nagabhushan, T. L., Trotta, P. P., and Pestka, S.
In summary, we report that adenovirus-mediated muIFN-␤ gene Biological properties of recombinant ␣-interferons: 40th anniversary of the discovery
of interferons. Cancer Res., 58: 2489 –2499, 1998.
therapy can exert potent antitumor activities in in vivo models of 18. Biron, C. A. Role of early cytokines, including ␣ and ␤ interferons (IFN-␣/␤), in
murine malignant mesothelioma. A single i.p. administration of Ad- innate and adaptive immune responses to viral infections. Semin. Immunol., 10:
.muIFN-␤ was sufficient to cause complete regression of i.p. tumors 383–390, 1998.
19. Qin, X. Q., Runkel, L., Deck, C., DeDios, C., and Barsoum, J. Interferon-␤ induces
in immunocompetent animals and induce long-lasting immune re- S phase accumulation selectively in human transformed cells. J. Interferon Cytokine
sponses that protected them from s.c. challenge with parental tumor Res., 17: 355–367, 1997.
cells and at least one other syngeneic tumor. Effects on distant tumor 20. Brem, H., Gresser, I., Grosfeld, J., and Folkman, J. The combination of antiangio-
genic agents to inhibit primary tumor growth and metastasis. J. Pediatr. Surg., 28:
were also seen. This potent antitumor effect, likely resulting from the 1253–1257, 1993.
local immune stimulatory effects of muIFN-␤, could be a critical 21. Singh, R. K., Gutman, M., Bucana, C. D., Sanchez, R., Llansa, N., and Fidler, I. J.
Interferons ␣ and ␤ down-regulate the expression of basic fibroblast growth factor in
factor in cancer gene therapy trials in which the degree of gene human carcinomas. Proc. Natl. Acad. Sci. USA, 92: 4562– 4566, 1995.
delivery to tumor cells is likely to be limited and a significant 22. Lokshin, A., Mayotte, J. E., and Levitt, M. L. Mechanism of interferon ␤-induced
bystander effect will be required. Although not prominent in our squamous differentiation and programmed cell death in human non-small-cell lung
cancer cell lines. J. Natl. Cancer Inst., 87: 206 –212, 1995.
animal models, it is possible that in clinical trials, the increased 23. Zeng, L., Buard, A., Monnet, I., Boutin, C., Fleury, J., Saint-Etienne, L., Brochard, P.,
sensitivity of human mesothelioma cells to the antiproliferative effects Bignon, J., and Jaurand, M. C. In vitro effects of recombinant human interferon-␥ on
of human IFN-␤, plus the potent antiangiogenic and immunostimula- human mesothelial cells. Int. J. Cancer, 55: 515–520, 1993.
24. Bouard, A., Vivo, C., Monnet, I., Boutin, C., Pilatte, Y., and Jaurand, M. C. Human
tory effects on macrophages and NK cells, could further contribute to malignant mesothelioma cell growth: activation of Janus kinase 2 and signal trans-
antitumor activity. Therefore, intracavitary IFN-␤ gene therapy pro- ducer and activator of transcription 1␣ for inhibition by interferon-␥. Cancer Res., 58:
840 – 847, 1998.
vides a promising strategy for the treatment of some solid tumors such 25. Christmas, T. I., Manning, L. S., Garlepp, M. J., Musk, A. W., and Robinson, B. W.
as mesothelioma or ovarian cancer in humans. Effect of interferon-␣2a on malignant mesothelioma. J. Interferon Res., 13: 9 –12,
1993.
26. Ardizzoni, A., Pennucci, M. C., Castagneto, B., Mariani, G. L., Cinquegrana, A.,
ACKNOWLEDGMENTS Magri, D., Verna, A., Salvati, F., and Rosso, R. Recombinant interferon ␣2b in the
treatment of diffuse malignant pleural mesothelioma. Am. J. Clin. Oncol., 17: 80 – 82,
1994.
We thank Patina Zarcone for performing the IFN-␤ ELISAs and Veena
27. Von Hoff, D. D., Metch, B., Lucas, J. G., Balcerzak, S. P., Grunberg, S. M., and
Kapoor for technical assistance. We also recognize Dr. Stephen Eck and the Rivkin, S. E. Phase II evaluation of recombinant interferon-␤ (IFN-␤ser) in patients
other members of the Thoracic Oncology Laboratory for input and useful with diffuse mesothelioma: a Southwest Oncology Group study. J. Interferon Res.,
discussion. 10: 531–534, 1990.
28. Rosso, R., Rimoldi, R., Salvati, F., De Palma, M., Cinquegrana, A., Nicolo, G.,
Ardizzoni, A., Fusco, U., Capaccio, A., Centofanti, R., Neri, M., Cruciani, A. R., and
Maisto, L. Intrapleural natural ␤ interferon in the treatment of malignant pleural
REFERENCES
effusions. Oncology, 45: 253–256, 1988.
1. Antman, K. H. Natural history and epidemiology of malignant mesothelioma. Chest, 29. Boutin, C., Viallat, J., Van Zandwijk, N., Douillard, J. T., Paillard, J. C., Guerin, J. C.,
103: 373S–376S, 1993. Mignot, P., Migueres, J., Varlet, F., Jehan, A., et al. Activity of intrapleural recom-
2. Sterman, D. H., Kaiser, L. R., and Albelda, S. M. Advances in the treatment of binant ␥-interferon in malignant mesothelioma. Cancer (Phila.), 67: 2033–2037,
malignant pleural mesothelioma. Chest, 116: 504 –520, 1999. 1991.
3. Peto, J., Decarli, A., La Vecchia, C., Levi, F., and Negri, E. The European mesothe- 30. Boutin, C., Nussbaum, E., Monnet, I., Bignon, J., Vanderschueren, R., Guerin, J. C.,
lioma epidemic. Br. J. Cancer, 79: 666 – 672, 1999. Menard, O., Mignot, P., Dabouis, G., and Douillard, J. Y. Intrapleural treatment with
4. Rusch, V. W. Pleurectomy/decortication and adjuvant therapy for malignant mesothe- recombinant ␥-interferon in early stage malignant pleural mesothelioma. Cancer
lioma. Chest, 103: 382S–384S, 1993. (Phila.), 74: 2460 –2467, 1994.
5. Sugarbaker, D. J., Heher, E. C., Lee, T. H., Couper, G., Mentzer, S., Corson, J. M., 31. Salmon, P., Le Cotonnec, J. Y., Galazka, A., Abdul-Ahad, A., and Darragh, A.
Collins, J. J., Jr., Shemin, R., Pugatch, R., and Weissman, L. Extrapleural pneumo- Pharmacokinetics and pharmacodynamics of recombinant human interferon-␤ in
nectomy, chemotherapy and radiotherapy in the treatment of diffuse malignant pleural healthy male volunteers. J. Interferon Cytokine Res., 16: 759 –764, 1996.
mesothelioma. J. Thorac. Cardiovasc. Surg., 102: 10 –15, 1991. 32. Xie, K., Bielenberg, D., and Huang, S. Abrogation of tumorigenicity and metastasis
6. Fitzpatrick, D. R., Peroni, D. J., and Bielefeldt-Ohmann, H. The role of growth factors of murine and human tumor cells by transfection with the murine IFN-␤ gene:
and cytokines in the tumorigenesis and immunobiology of malignant mesothelioma. possible role of nitric oxide. Clin. Cancer Res., 3: 2283–2294, 1997.
Am. J. Respir. Cell. Mol. Biol., 12: 455– 460, 1995. 33. Xu, L., Xie, K., and Fidler, I. J. Therapy of human ovarian cancer by transfection with
7. Bielefeldt-Ohmann, H., Jarnicki. A. G., and Fitzpatrick, D. R. Molecular pathobiol- the murine interferon-␤ gene: role of macrophage-inducible nitric oxide synthase.
ogy and immunology of malignant mesothelioma. J. Pathol., 178: 369 –378, 1996. Hum. Gene Ther., 9: 2699 –2708, 1998.
8. Davis, M. R., Manning, L. S., Whataker, D., Garlepp, M. J., and Robinson, B. W. S. 34. Rozera, C., Carlei, D., Lollini, P. L., De Giovanni, C., Musiani, P., Di Carlo, E.,
Establishment of a murine model of malignant mesothelioma. Int. J. Cancer, 52: Belardelli, F., and Ferrantini, M. Interferon (IFN)–␤ gene transfer into TS/A adeno-
881– 886, 1992. carcinoma cells and comparison with IFN-␣. Am. J. Pathol., 154: 1211–1222, 1999.
9. Bielefeldt-Ohmann, H., Fitzpatrick, D. R., Marzo, A. L., Jarnicki, A. G., Musk, 35. Qin, X. Q., Tao, N., Dergay, A., Moy, P., Fawell, S., Davis, A., Wilson, J. M., and
A. W., and Robinson, B. W. Potential for interferon-␣-based therapy in mesotheli- Barsoum, J. Interferon-␤ gene therapy inhibits tumor formation and causes regression
oma: assessment in a murine model. J. Interferon Cytokine Res., 15: 213–223, 1995. of established tumors in immune-deficient mice. Proc. Natl. Acad. Sci. USA, 95:
10. Robinson, B., Bowman, R., Manning, L., Musk, A., and Van Hazel, G. Interleukin-2 14411–14416, 1998.
and lymphokine-activated killer cells in malignant mesothelioma. Eur. Respir. Rev., 36. Lu, W., Fidler, I. J., and Dong, Z. Eradication of primary murine fibrosarcomas and
3: 220 –222, 1993. induction of systemic immunity by adenovirus-mediated interferon ␤ gene therapy.
11. Caminschi, I., Venetsanakos, E., Leong, C. C., Garlepp, M. J., Robinson, B. W., and Cancer Res., 59: 5202–5208, 1999.
Scott, B. Cytokine gene therapy of mesothelioma. Immune and antitumor effects of 37. Natsume, A., Mizuno, M., Ryuke, Y., and Yoshida, J. Antitumor effect and cellular
transfected interleukin-12. Am. J. Respir. Cell Mol. Biol., 21: 347–356, 1999. immunity activation by murine interferon-␤ gene transfer against intracerebral glioma
12. Fitzpatrick, D. R., Bielefeldt-Ohmann, H., Himbeck, R. P., Jarnicki, A. G., Marzo, in mouse. Gene Ther., 6: 1626 –1633, 1999.
A. L., and Robinson, B. W. Transforming growth factor-␤: antisense RNA-mediated 38. Smythe, W. R., Kaiser, L. R., Hwang, H. C., Amin, K. M., Pilewski, J. M., Eck, S. J.,
inhibition affects anchorage-independent growth, tumorigenicity and tumor-infiltrat- Wilson, J. M., and Albelda, S. M. Successful adenovirus-mediated gene transfer in an
ing T-cells in malignant mesothelioma. Growth Factors, 11: 29 – 44, 1994. in vivo model of human malignant mesothelioma. Ann. Thorac. Surg., 57: 1395–
13. Goey, S. H., Eggermont, A. M., Punt, C. J., Slingerland, R., Gratama, J. W., 1401, 1994.
Oosterom, R., Oskam, R., Bolhuis, R. L., and Stoter, G. Intrapleural administration of 39. Treat, J., Kaiser, L. R., Sterman, D. H., Litzky, L., Davis, A., Wilson, J. M., and
interleukin 2 in pleural mesothelioma: a Phase I-II study. Br. J. Cancer, 72: 1283– Albelda, S. M. Treatment of advanced mesothelioma with the recombinant adenovi-
1288, 1995. rus H5.010RSVTK: a Phase I trial (BB-IND 6274). Hum. Gene Ther., 7: 2047–2057,
14. Astoul, P., Picat-Joossen, D., Viallat, J., and Boutin, C. Intrapleural administration of 1996.
interleukin-2 for the treatment of patients with malignant pleural mesothelioma: a 40. Sterman, D. H., Treat, J., Litzky, L. A., Amin, K. M., Coonrod, L., Molnar-Kimber,
Phase II study. Cancer (Phila.), 83: 2099 –2104, 1998. K., Recio, A., Knox, L., Wilson, J. M., Albelda, S. M., and Kaiser, L. R. Adenovirus-
15. Davidson, J. A., Musk, A. W., Wood, B. R., Morey, S., Ilton, M., Yu, L. L., Drury, mediated herpes simplex virus thymidine kinase/ganciclovir gene delivery in patients
P., Shilkin, K., and Robinson, B. W. Intralesional cytokine therapy in cancer: a pilot with localized malignancy: results of a Phase I clinical trial in malignant mesotheli-
study of GM-CSF infusion in mesothelioma. J. Immunother., 21: 389 –398, 1998. oma. Hum. Gene Ther., 9: 1083–1092, 1998.
6211

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Ad.IFN-␤ FOR MESOTHELIOMA

41. Molnar-Kimber, K. L., Sterman, D. H., Chang, M., Elbash, M., Elshami, A., Roberts, 53. Marrack, P., Kappler, J., and Mitchell, T. Type I interferons keep activated T cells
J. R., Treat, J., Wilson, J. M., Kaiser, L. R., and Albelda, S. M. Humoral and cellular alive. J. Exp. Med., 189: 521–530, 1999.
immune responses induced by adenoviral-based gene therapy for localized malig- 54. Spear, G. T., Paulnock, D. M., Jordan, R. L., Meltzer, D. M., Meritt, J. A., and
nancy: results of a Phase I clinical trial for malignant mesothelioma. Hum. Gene Borden, E. C. Enhancement of monocyte class I and II histocompatibility antigen
Ther., 9: 2121–2133, 1998. expression in man by in vivo ␤-interferon. Clin. Exp. Immunol., 69: 107–115, 1987.
42. Sterman, D. H., Molnar-Kimber, K., Iyengar, T., Chang, M., Lanuti, M., Amin, K., 55. Yagi, K., Hayashi, Y., Ishida, N., Ohbayashi, M., Ohishi, N., Mizuno, M., and
Pierce, B., Kang, E., Treat, J., Recio, A., Litzky, L., Wilson, J. M., Kaiser, L. R., and Yoshida, J. Interferon-␤ endogenously produced by intratumoral injection of cationic
Albelda, S. M. A pilot study of systemic corticosteroid administration in conjunction liposome-encapsulated gene: cytocidal effect on glioma transplanted into nude mouse
with intrapleural adenoviral vector administration in patients with malignant pleural brain. Biochem. Mol. Biol. Int., 32: 167–171, 1994.
mesothelioma. Cancer Gene Ther., 7: 1511–1518, 2000. 56. Natsume, A., Tsujimura, K., Mizuno, M., Takahashi, T., and Yoshida, J. IFN-␤ gene
43. Lanuti, M., Rudginsky, S., Force, S., Lambright, E. S., Chang, M. Y., Amin, K., therapy induces systemic antitumor immunity against malignant glioma. J. Neuro-
Kaiser, L. R., Scheule, R. K., and Albelda, S. M. Cationic lipid:bacterial DNA Oncol., 47: 117–124, 2000.
complexes elicit anti-tumor effects and adaptive immunity in murine intraperitoneal 57. Manning, L. S., Davis, M. R., Bielefeldt-Ohmann, H., Marzo, A. L., Garlepp, M. J.,
tumor models. Cancer Res., 60: 2955–2963, 2000. Whitaker, D., and Robinson, B. W. S. Evaluation of immunogenicity of murine
44. Kiessling, R., Klein. E., and Wigzell, H. “Natural” killer cells in the mouse. I. mesothelioma cells by immunization. Eur. Respir. Rev., 3: 234 –237, 1993.
Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and 58. Adris, S., Chuluyan, E., Bravo, A., Berenstein, M., Klein, S., Jasnis, M., Carbone, C.,
distribution according to genotype. Eur. J. Immunol., 5: 112–117, 1975. Chernajovsky, Y., and Podhajcer, O. L. Mice vaccination with interleukin 12-
45. Davis, A. R., and Wilson, J. M. Adenoviral vectors. In: N. C. Dracopoli, J. L. Haines, transduced colon cancer cells potentiates rejection of syngeneic non-organ-related
tumor cells. Cancer Res., 60: 6696 – 6703, 2000.
B. R. Korf, D. T. Muir, C. C. Morton, C. E. Seidman, J. G. Seidman, and D. R. Smith
59. Heel, K. A., and Hall, J. C. Peritoneal defences and peritoneum-associated lymphoid
(eds.), Current Protocols in Human Genetics, pp. 12.4 –12.4 –16. New York: Wiley,
tissue. Br. J. Surg., 83: 1031–1036, 1996.
1996.
60. Valle, M. T., Degl’Innocenti, M. L., Bertelli, R., Facchetti, P., Perfumo, F., Fenoglio,
46. Davis, A. R., Meyers, K., and Wilson, J. M. High Throughput Method for Creating
D., Kunkl, A., Gusmano, R., and Manca, F. Antigen-presenting function of human
and Screening Recombinant Adenoviruses. Gene Ther., 5: 1148 –1152, 1998.
peritoneum mesothelial cells. Clin. Exp. Immunol., 101: 172–176, 1995.
47. Smythe, W. R., Hwang, H. C., Amin, K. M., Eck, S. L., Davidson, B. L., Wilson, 61. Melichar, B., Savary, C., Kudelka, A. P., Verschraegen, C., Kavanagh, J. J., Edwards,
J. M., Kaiser, L. R., and Albelda, S. M. Use of recombinant adenovirus to transfer the C. L., Platsoucas, C. D., and Freedman, R. S. Lineage negative HLA-DR⫹ cells with
herpes simplex virus thymidine kinase (HSVtk) gene to thoracic neoplasms: an the phenotype of undifferentiated dendritic cells in patients with carcinoma of the
effective in vitro drug sensitization system. Cancer Res., 54: 2055–2059, 1994. abdomen and pelvis. Clin. Cancer Res., 4: 799 – 809, 1998.
48. Biron, C. A., Nguyen, K. B., Pien, G. C., Cousens, L. P., and Salazar-Mather, T. P. 62. Gjersten, M. K., Saeterdal, I., Beiske, K., and Gaudernack, G. Antigen-presenting
Natural killer cells in antiviral defense: function and regulation by innate cytokines. function of human peritoneum mesothelial cells isolated from a pancreatic carcinoma
Annu. Rev. Immunol., 17: 189 –220, 1999. patient after mutant Ras peptide vaccination. Cancer Immunol. Immunother., 43:
49. Belhumeur, P., Lanoix, J., Blais, Y., Forget, D., Steyaert, A., and Skup, D. Action of 262–268, 1996.
spontaneously produced ␤ interferon in differentiation of embryonal carcinoma cells 63. Hausmann, M. J., Rogachev, B., Weiler, M., Chaimovitz, C., and Douvdevani, A.
through an autoinduction mechanism. Mol. Cell. Biol., 13: 2846 –2857, 1993. Accessory role of human peritoneal mesothelial cells in antigen presentation and
50. Fujihara, M., Ito, N., Pace, J. L., Watanabe, Y., Russell, S. W., and Suzuki, T. Role T-cell growth. Kidney Int., 57: 476 – 486, 2000.
of endogenous interferon-␤ in lipopolysaccharide-triggered activation of the inducible 64. Garlepp, M. J., Christmas, T. I., Manning, L. S., Davis, M. R., and Robinson, B. W. S.
nitric-oxide synthase gene in a mouse macrophage cell line, J774. J. Biol. Chem., 269: Constitutive and inducible HLA antigen expression in malignant mesothelioma. Eur.
12773–12778, 1994. Respir. Rev., 3: 231–233, 1993.
51. Orange, J. S., and Biron, C. A. Characterization of early IL-12, IFN-␣␤, and TNF 65. Fuchs, E. J., and Matzinger, P. Is cancer dangerous to the immune system? Semin.
effects on antiviral state and NK cell responses during murine cytomegalovirus Immunol., 8: 271–280, 1996.
infection. J. Immunol., 156: 4746 – 4756, 1996. 66. Melcher, A., Todryk, S., Hardwick, N., Ford, M., Jacobson, M., and Vile, R. G.
52. Tough, D. F., Borrow, P., and Sprent, J. Induction of bystander T cell proliferation by Tumor immunogenicity is determined by the mechanism of cell death via induction
viruses and type I interferon in vivo. Science (Wash. DC), 272: 1947–1950, 1996. of heat shock protein expression. Nat. Med., 4: 581–587, 1988.

6212

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.
Eradication of Intraperitoneal and Distant Tumor by
Adenovirus-mediated Interferon- β Gene Therapy Is Attributable
to Induction of Systemic Immunity
Makoto Odaka, Daniel H. Sterman, Rainer Wiewrodt, et al.

Cancer Res 2001;61:6201-6212.

Updated version Access the most recent version of this article at:
http://cancerres.aacrjournals.org/content/61/16/6201

Cited articles This article cites 61 articles, 16 of which you can access for free at:
http://cancerres.aacrjournals.org/content/61/16/6201.full#ref-list-1

Citing articles This article has been cited by 18 HighWire-hosted articles. Access the articles at:
http://cancerres.aacrjournals.org/content/61/16/6201.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://cancerres.aacrjournals.org/content/61/16/6201.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.

Downloaded from cancerres.aacrjournals.org on December 14, 2020. © 2001 American Association for Cancer
Research.

You might also like