CA2508238C - Reovirus for the treatment of neoplasia - Google Patents

Reovirus for the treatment of neoplasia Download PDF

Info

Publication number
CA2508238C
CA2508238C CA002508238A CA2508238A CA2508238C CA 2508238 C CA2508238 C CA 2508238C CA 002508238 A CA002508238 A CA 002508238A CA 2508238 A CA2508238 A CA 2508238A CA 2508238 C CA2508238 C CA 2508238C
Authority
CA
Canada
Prior art keywords
reovirus
neoplasm
cells
cancer
administration
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA002508238A
Other languages
French (fr)
Other versions
CA2508238A1 (en
Inventor
Patrick W. K. Lee
James Strong
Matthew C. Coffey
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Oncolytics Biotech Inc
Original Assignee
Oncolytics Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/911,383 external-priority patent/US6110461A/en
Application filed by Oncolytics Biotech Inc filed Critical Oncolytics Biotech Inc
Publication of CA2508238A1 publication Critical patent/CA2508238A1/en
Application granted granted Critical
Publication of CA2508238C publication Critical patent/CA2508238C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Landscapes

  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Methods for treating neoplasia, by administering reovirus to a Ras-mediated neoplasm, and use of reovirus for manufacture of a medicament for the treatment of neoplasia, are disclosed. The reovirus is administered so that it ultimately directly contacts cells of the neoplasm. Human reovirus, non-human mammalian reovirus, and/or avian reovirus can be used. If the reovirus is human reovirus, type 1 (e.g., strain Lang), type 2 (e.g., strain Jones), type 3 (e.g., stain Dearing or strain Abney), as well as other serotypes or strains of reovirus can be used. Combinations of more than one type and/or strain of reovirus can be used, as can reovirus from different species of animal. Either solid neoplasms or hematopoietic neoplasms can be treated.

Description

' CA 02508238 1998-08-12 WO 99/08691 , . , PCT/CA98/40774 . ~~
REOVIRUS FOR THE TREATMENT OF NEOPLA.SIA
RELATED APPLICATION
This application is related to US Patent Nos. 6,261,555 and 6,576,234.
S
BACKGROUND OF THE INV~NTiON , Normal cell proliferation is regulated by a balance between growth-promoting proto-oncogenes and growth-constraining tumor-suppressor genes. Tumorigenesis can be caused by genetic alterations to the genome that result in the mutation of those cellular elements that govern the interpretation of cellular signals, such as potentiation of proto-oncogene activity or inactivation of tumor suppression. It ~is believed that the interpretation of these signals ultimately influences the growth and differentiation of a , cell, and that misinterpretation of these signals can result in neoplastic growth , (neoplasia).
Genetic alteration of the proto-oncogene Ras is believed to contribute to approximately 30°,'° of all human tumors (Wiessmuller, L. and Wittinghofer, F. { 1994), Cellular Signaling 6(3):247-267; Barbacid, M. (1987) A Rev. Biochem. 56, 779-827).
The role that Ras plays in the pathogenesis of human tumors is specific to the type of tumor. Activating mutations in Ras itself are found in most types of human malignancies, and are highly represented in pancreatic cancer (80%), sporadic colorectal carcinomas (40-50%), human lung adenocarcinomas (15-24%), thyroid tumors (50%) and myeloid leukemia {30%) (Millis, h'E et al. ( 1995) CA 02508238 1998-08-12 '
2 ~ f PCT/CA98I00774 -2_ Cancer Res. 55:1444; Chaubert, P. et al. (1994), Ant. J. Path. 144:7b7; Bos, J. (1989) Cancer Res. 49:4682). Ras activation is also demonstrated by upstream mitogenic signaling elements, notably by tyrosine receptor kinases (RTKs). These upstream elements, if amplified or overexpressed, ultimately result in elevated Ras activity by the signal transduction activity of Ras. Examples of this include overexpression of PDGI~R
in certain forms of glioblasiomas , as well as in c-erbB-2/neu in breast cancer (Levitzki, A. (1994) Eur. J. Biochem. X26:1; James, P.W., et al. (1994) Oncogene 9:3601;
Bos, J.
( 1989) Cancer Res. 49:4682).
Current methods of treatment for neoplasia include surgery, chemotherapy and radiation. Surgery is typically used as the primary treatment forearly stages of cancer;
however, many tumors cannot be completely removed by surgical means. In addition, metastatic growth of neoplasms may prevent complete cure of cancer by surgery.
Chemotherapy involves administration of compounds having antitumor activity, such as alkylating agents, antimetabolites, and antitumor antibiotics. The efficacy of chemotherapy is often limited by severe side effects, including nausea and vomiting, bone marrow depression, renal damage, and central nervous system depression.
Radiation therapy relies on the greater ability of normal cells, in contrast with neoplastic cells, to repair themselves after treatment with radiation. Radiotherapy cannot be used to treat many neoplasms, however, because of the sensitivity of tissue surrounding the tumor. In addition, certain tumors have demonstrated resistance to radiotherapy and such may be dependent on oncogene or anti-oncogene status of the cell (Lee.
J.M. et al.
(1993) PNAS 90:5742-5746; Lowe. S.W. et al. (i994) Science, ?6tS:807-810;
Raybaud-Diogene. H. et al. (1997) J. Clin. Oncology,15(3):1030-1038). In view of the drawbacks associated with the current means for treating neoplastic growth, the need 2S still exists for improved~methods for the treatment of most types of cancers.
-3- ~ v , SUMMARY OF THE 13~1VENTION
The present invention pertains to methods for treating neoplasia ~ia a~
mammal, using reovirus, and to use of reovirus for manufacture of a medicament for the treatmentof neopiasia.
Reovirus is administered to a neoplasm, in which an element of the Ras signaling pathway (either upstream or downstream) is activated to an extent that results in reovirus-mediated oncolysis of cells of the neoplasm. The reovirus can be administered in a single dose or in multiple doses; furthermore, more than one neoplasm in an individual mammal can be treated concurrently. Both solid neoplasms and hematopoietic neoplasms can be targeted. The reovirus is administered so that it contacts cells of the mammal (e.g., by injection directly into a solid neoplasm, or intravenously into the mammal for a hematopoietic neoplasm). The methods can be used to treat neoplasia in a variety of mammals, including mice, dogs, cats, sheep, goats, cows, horses, pigs, and non-human primates. Preferably, the methods are used to treat neoplasia in humans.
The methods and uses of the invention provide an eff=ective means to treat neoplasia, without the side effects associated with other forms of cancer therapy.
Furthermore, because reovirus is not known to be associated with disease, any safety concerns associated with deliberate administration of a virus are minimized.
In another embodiment, the present invention pertains to a pharmaceutical composition useful for the treatment of a ras-mediated neoplasm in a mammal, the composition comprising an effective amount of at least one reovirus, wherein the amount is effective to result in oncolysis of the ras-mediated neoplasm, and a pharmaceutically acceptable excipient.
In a further embodiment, the present invention pertains to a kit comprising the pharmaceutical composition descn'bed above and means for administering the composition, such as a syringe. The kit may further include a chemotherapeutic agent.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a depiction of the molecular basis of reovirus oncolysis, in which the reovirus usurps the host cell Ras signalling pathway.
Figure 2 is a graphic representation of the effects over time of active (open circles) or inactivated (closed circles) reovirus serotype 3 (strain bearing) on the size of the marine THC-11 tumors grown in severe combined immunodeficiency (SC>D) mice.

WO 99/08692 I 1 PCT/CA98/0079d The plotted values represent the mean of the measurements with the standard error of the mean also shown.
Figure 3 is a graphic representation of the effects over time of active (open circles) or inactivated (closed circles) reovirus serotype 3 (strain bearing) on the size of human glioblastoma U-87 xenografts grown in SCIf? mice. The plotted values represent the mean of the measurements with the standard error of the mean also shown.
Figure 4 is a graphic representation of the elects over time of active (open circles, open squares) or inactivated (closed circles, closed squares) reovirus serotype 3 (strain bearing) on the size of treated (circles) or untreated (squares) bilateral human glioblastoma U-87 xenografts grown in SCID mice. The platted values represent the mean of the measurements with the standard error of the mean also shown.
The foregoing and other objects, features and advantages of the invention will be apparent from the following more particular description of preferred embodiments of the invention, as illustrated in the accompanying drawings in which like reference 1 S characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating the principles of the invention.
DETAILED DESCRIPTION OF THE INVENTION
The invention pertains to methods of treating a neoplasm in a mammal, by administering reovirus to the neoplasm. The name reovirus (Respiratory and enteric orphan virus) is a descriptive acronym suggesting that these viruses, although not associated with any known disease state in humans, can be isolated from both the respiratory and enteric tracts (Sabin, A.B. (1959), Science 130:966). The mammalian reovirus consists of three serotypes: type 1 (strain Lang or T1L), type 2 (strain Jones, T2J) and type 3 (swain bearing or strain Abney, T3D). The throe semtypes are easily WO 99/08692 ' ~ PCT/CA98/00774 identifiable on the basis of neutralization and hemagg lutinin-inlu'bition assays (Sabin, A.B. (1959), Science 130:966; Fields, B.N. et al. (1996), Fundamental Virology, 3rd Edition, Lippincott-Raven; Rosen, L. (1960) Am. J. Hyg.71:242; Stanley, N.F.
(1967) Br. Med Bull. 23:150).
Although reovirus is not known to be associated with any particular disease (Tyler, K.L. and Fields, B.N., in Fields Virology (Fields, B.N., Knipe, D.M., and Howley, P.M. eds), Lippincott-Raven, Philadelphia, 1996, p. 1597), many people have been exposed to reovirus by the time they reach adulthood {i.e., fewer than 25% in children < 5 years old, to greater than 50% in those 20-30 years old. (Jackson G.G. and Muldoon R.L. ( 1973) J. In~'ect. Dis. 128:811; Stanley N.F. ( 1974) In:
Comparative Diagnosis of Viral Diseases, edited by E. Kurstak and K. Kurstak, 385-421, Academic Press, New York)..
For mammalian reoviruses, the cell surface recognition signal is sialic acid (Armstrong, G.D: et al. (1984), virology 138:37; Gentsch, J.R.K. and Pacitti, A.F.
(1985), J. Yirol. 56:356; Paul R.W. et al. (1989) Virology 17?:382-385). Due to the ubiquitous nature of sialic acid, reovirus binds efficiently to a multitude of cell lines and, as such can potentially target many different tissues; however, there are significant differences in susceptibility to reovirus infection between cell lines.
As described herein, Applicants have discovered that cells which are resistant to reovirus infection became susceptible to reovirus infection when transformed with a gene in the Ras pathway. "Resistance" of cells to reovirus infection indicates that infection of the cells with the virus did not result in significant viral production or yield.
Cells that are "susceptible" are those that demonstrate induction of cytopathic effects, viral protein synthesis, and/or virus production. Resistance to reovirus infection was . found to be at the level of gene translation, rather than at early transcription: while viral transcripts were produced, virus proteins were not expressed. Viral gene transcription WO 99/08692 ' PCT/CA98/00774 in resistant cells correlated with phosphorylation of an approximately 65 kDa cell protein, determined to be double-stranded RNA-activated protein kinase {PKR), that was not observed in transformed cells. Phosphoryladon of PKR lead to inhibition of translation. When phosphorylation was suppressed by 2-aminopurine, a known inhibitor of PKR, drastic enhancement of reovirus protein synthesis occurred in the untransformed cells. Furihennore, in a severe combined immunodeficiency (SCrD) mouse model in which tumors were created on both the right and left hind flanks revealed that reovirus significantly reduced tumor size when injected directly into the right-side tumor; in addition, significant reduction in tumor size was also noted on the left-side tumor which was not directly injected with reovirus, indicating that the oncolytic capacity of the reovirus was systemic as well as local.
These results indicated that reovirus uses the host cell's Ras pathway machinery to downregulate PKR and thus reproduce. Figure I depicts the usurpation of the host cell Ras signalling pathway by reovirus. As shown in Figure 1, for both untransformed (reovirus-resistant) and EGFR-, Sos-, or ras-transformed (reovirus-susceptible) cells, virus binding, internalization, uncoating, and early transcription of viral genes all pmceed normally. In the case of untransformed cells, secondary structures on the early viral transcripts inevitably trigger the phosphorylation of PKR, thereby activating it, leading to the phosphoryIation of the translation initiation factor eIF-2a, and hence the inhibition of viral gene translation. In the case of EGFR-, Sos-, or ras-transformed cells, the PKR phosphorylation step is prevented or reversed by Ras or one of its downstream elements, thereby allowing viral gene translation to ensue. The action of Ras (or a downstream element) can be mimicked by the use of 2-aminopurine (2-AP), which promotes viral gene translation (and hence reovirus infection) in untransformed cells by blocking PKR phosphorylation.

WO 99/08692 ~ ~ PCT/CA98/00774 Based upon these discoveries, Applicants have developed methods for treating neoplasms in mammals. Representative mammals include mice, dogs, cats, sheep, goats, cows, horses, pigs, non-human primates, and humans. In a preferred embodiment, the mammal is a human.
In the methods of the invention, reovirus is administered to a neoplasm in the individual mammal. Representative types of human reovirus that can be used include type 1 (e.g., strain Lang or T1L); type 2 {e.g., strain Jones or T2J); and type 3 {e.g"
strain bearing or strain Abney, T3D or T3A); other strains of reovirus can also be used.
In a preferred embodiment, the reovirus is strain bearing. Alternatively, the reovirus can be a non-.human mammalian reovirus (e.g., non-human primate reovirus, such as baboon reovirus; equine; or canine reovirus), or a non-mammalian reovirus (e.g., avian reovirus). A combination of different semtypes andlor different strains of reovirus, such as r~ovirus from different species of animal, can be used. The reovirus is "naturally-occurring": that is, it can be isolated from a source in nature'and has not been intentionally modified by humans in the laboratory. For example, the reovirus can be from a "field source": that is, from a human patient. If desired, the reovirus can be chemically or biochemically pretreated (e.g., by treatment with a protease, such as chymotrypsin or trypsin) prior to administration to the neoplasm. Such pretreatment removes the outer coat of the virus and may thereby result in better infectivity of the ZO virus.
The neoplasm can be a solid neoplasm (e.g., sarcoma or carcinoma), or a cancerous growth affecting the hematopoietic system {a "hematopoietic neoplasm"; e.g., lymphoma or leukemia). A neoplasm is an abnormal tissue growth, generally forming a distinct mass. that grows by cellular proliferation more rapidly than normal tissue growth. Neoplasms show partial or total lack of structural organization and functional coordination with normal tissue. As used herein, a "neoplasm", also referred to as a CA 02508238 1998-08-12 .
WO 99/08692 ~~ ~ ~ ~ PCTlCA98100774 _$_ "tumor", is intended to encompass hematopoietic neoplasms as well as solid neoplasms.
At least some of the cells of the neoplasm have a mutation in which the Ras gene (or an element of the Ras signaling pathway) is activated, either directly (e.g., by an activating mutation in Ras) or indirectly (e.g., by activation of an upstream element in the Ras pathway). Activation of an upstream element in the Ras pathway includes, for example;
transformation with epidermal growth factor receptor (EGFR) or Sos. A neoplasm that results, at least in part, by the activation of Ras, an upstream element of Ras, or an element in the Ras signalling pathway is referred to herein as a "Ras-mediated neoplasm". One neoplasm that is particularly susceptible to treatment by the methods I O of the invention is pancreatic cancer, because of the prevalence of Ras-mediated neoplasms associated with pancreatic cancer. Other neoplasms that are particularly susceptible to treatment by the methods. of the invention include breast cancer, brain cancer (e.g., glioblastoma), lung cancer, prostate cancer, colorectal cancer, thyroid cancer, renal cancer, adrenal cancer, liver cancer, and leukemia.
The reovirus is typically administered in a physiologically acceptable carrier or vehicle, such as phosphate-buffered saline, to the neoplasm. "Administration to a neoplasm" indicates that the reovirus is administered in a manner so that it contacts the cells of the neoplasm (also referred to herein as "neoplastic cells"). The route by which the reovirus is administered, as well as the formulation, carrier or vehicle, will depend on the location as well as the type of the neoplasm. A wide variety of administration routes can be employed. For example, for a solid neoplasm that is accessible, the reovirus can be administered by injection directly to the neoplasm. For a hematopoietic neoplasm, for example, the reovirus can be administered intravenously or intravascularIy. For neoplasms that are not easily accessible within the body, such as metastases or brain tumors, the reovirus is administered in a manner such that it can be transported systemically through the body of the mammal and thereby reach the WO 99/08692 ~ ~ PCT/CA98100774 neoplasm (e.g., intrathecally, intravenously or intramuscularly).
Alternatively, the reovirus can be administered directly to a single solid neoplasm, where it then is carried systemically through the body to metastases. The reovirus can also be administered subcutaneously, intraperitoneaily, topically (e.g., for melanoma), orally (e.g., for oral or esophageal neoplasm), rectally (e.g., for colorectal neoplasm), vaginally (e.g., for cervical or vaginal neoplasm), nasally or by inhalation spray (e.g., for lung neoplasm).
The reovirus is administered in an amount that is sufEcient to treat the neoplasm (e.g., an "effective amount"). A neoplasm is "treated" when administration of reovirus to cells of the neoplasm effects oncoiysis of the neoplastic cells, resulting in a reduction in size of the neoplasm, or in a complete elimination of the neoplasm. The reduction in size of the neoplasm, or elimination of the neoplasm, is generally caused by lysis of neoplastic cells ("oncolysis") by the reovirus. The effective amount will be determined on ark individual basis and may be based, at least in part, on consideration of the type of reovirus; the individual's size, age, gender; and the size and other characteristics of the neoplasm. For example, for treatment of a human, approximately l Os to 10''-plaque forming units (PFL>] of reovirus can be used, depending on the type, size and number of , tumors present. The reovirus can be administered in a single dose, or multiple doses (i.e., more than one dose). The multiple doses can be administered concurrently, or consecutively (e.g., over a period of days or weeks). The reovirus can also be administered to more than one neoplasm in the same individual.
The invention is further illustrated by the following Exemplification.
EXEMPLIFICATION
MATERIALS AND METHODS
Cells and Virus CA 02508238 1998-08-12 .
WO 99/08692 ~ PCT/CA98/00774 Parental NIH-3T3 cell lines along with NIH-3T3 cells transfornie~d with a number of oncogenes were obtained from a variety of sources. Parental NIH-3T3 and NIH-3T3 cells transfected with the Harvey-ras (H-ras) and E7-ras oncogenes were, a generous gift of Dr. Douglas Falter (Boston University School of Medicine).

cells along with their Sos-transformed counterparts (designated TNIH#5) were a generous gift ofDr. Michael Karin (University of California, San Diego). Dr.
Iii.-J.
Kung (Case Western Reserve University) kindly donated parental NTH-3T3 cells along with NIH-3T3 cells transfected with the v-erbB oncogene (designated THC-11).

cells, a derivative of the C3H lOTll2 marine fibroblast line, containing the Harvey-ras gene under the transcriptional control of the mouse metallothionein-1 promoter were obtained from Dr. Nobumichi Hozumi (Mount Sinai Hospital Research Institute).
These 2H 1 cells are conditional ras transformant that express the H-ras oncogene in the presence of 50 pM ZnSO~. All cell lines were grown,in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS).
The NIH-3T3 tet-myc cells were obtained from Dr. R.N. lohnston (University of Calgary) and were grown in DMEM containing 10% heat-inactivated FBS and antibiotics in the presence or absence of 2 pglml tetracycline (Helbing, C.C.
et al., Cancer Res. 57:1255-1258 (1997)). In the presence of tetracycline, expression of the human c-myc gene is repressed. Removal of tetracycline results in the elevation of expression of c-myc by up to 100-fold in these cells, which also display a transformed phenotype.
The PKR ~l" and PICK °I° mouse embryo fibroblasts (MEFs) were obtained from Dr. B.R.G. Williams (the Cleveland Clinic Foundation) and were grown in a-MEM
containing fetal bovine serum and antibiotics as previously described (Yang, Y.L. et al.
EMBO J. 14:609-6106 ( 1995); Dcr, S.D. et al., Proc. Narl. Acad Sci. USA
94:3279-3283 ( 1997)).

WO 99/08692 ' ~ , ~ PCT/CA98/00774 The bearing strain of reovirus serotype 3 used in these studies was propagated in suspension cultures of L cells and purified according to Smith (Smith, R.E.
et al., (1969) Virology, 39:791-800) with the exception that [i-mercaptoethanol {[3-ME) was omitted from the extraction buffer. Reovirus labelled with ['SS]methionine was grown and purified as described by McRae and Joklik (McRae, M.A. and Joklik, W.K., (1978) Virology, 89:578-593). The particle/PFU ratio for purified reovirus was typically 100/1.
Immunofluorescenr analysis of reovirus infection For the immunofluorescent studies the N1H-3T3, ThTIEi#5, H-ras, EJ-ras, 2H1 (+/- ZnSO,,), and THC-11 cells were grown on coverslips, and infected with reovirus at a multiplicity of infection (MOI) of --10 PFU cell or mock-infected by application of the carrier agent (phosphate-buffered saline, PBS) to the cells in an identical fashion as the administration, of virus to the cells. At 48 hours postinfection, cells were fixed in an ethanol/acetic acid (20/1) mixture for 5 minutes, then rehyd~ed by sequential washes in 75%, 50%' and 25% ethanol, followed by four washes with phosphate-buffered saline (PBS). The fixed and rehydrated cells were then exposed to the primary antibody (rabbit polyclonal anti-reovirus type 3 serum diluted 1/100 in PBS) [antiserum prepafed , by injection of rabbits with reovirus serotype 3 in Freund's complete adjuvant, and subsequent bleedings] for 2 hours at room temperature. Following three washes with PBS, the cells were exposed to the secondary antibody [goat anti-rabbit IgG
(whole molecule)-fluorescein isothiocyanate conjugate (FITC) [Sigma IrnmunoChemicals F-0382] diluted 1/100 in PBS containing 10% goat serum and 0.005% Evan's Blue]
for 1 hour at room temperature. Finally, the fixed and treated cells were washed three more times with PBS and then once with double-distilled water, dried and mounted on slides in 90% glycerol containing 0.1 % phenylenediamine, and viewed with a Zeiss Axiophot V

WO 99/08691 ' ~ ~ ~ PCT/CA98/00774 nucroscope on which Carl Zeiss camera was mouated {the magnification for all pictures was 200J~.
Detection of MAP Kinase (ERK) Activity 'The PhosphoPlus p44/42 MAP kinase (Thr202/Tyr204) Anybody kit (New England Biolabs) was used for the detection of MAP kinase in cell lysates according to the manufacturer's instructions. Brie#ly, subconfluent monolayer cultures were lysed with the recommended SDS-containing sample buffer, and subjected to SDS-PAGE, followed by electroblottirtg onto nitrocellulose paper. The membrane was then probed with the primary antibody (anti-total MAPK or anti-phospho-MAPK), followed by the horseradish peroxidase (HRP}-conjugated secondary antibody as described in the manufacturer's instruction manual.
Radiolabelling of reovirus-infected cells and preparation of lysates Confluent monolayers of NIH-3T3, T'N~i#5, H-ras, EJ-ras, 2H 1 (+/- ZnSO,), and THC-11 cells were infected with reovirus (MOI --10 PFU/cell). . At 12 hours postinfection, the media was replaced with methionine-free DMEM containing 10~/°
dialyzed FBS and O.1 rrrCi/ml ['sS)meihionine. After further incubation for 36 hours at 37°C, the cells were washed in phosphate-buffered saline {PBS) and lysed in the same buffer containing 1 % Triton X-100, 0.5% sodium deoxycholate and 1 mM EDTA.
The nuclei were then removed by low speed centrifugation and the supernatants were stored at -70°C until usc.
Preparation of cytoplasmic extracts for in vitro kinase assays Confluent monolayers of the various cell lines were grown on 96 well cell culture plates. At the appropriate time postinfection the media was aspirated off and the WO 99/08692 ' ' , ~ PCT/CA98100774 cells were lysed with a buffer containing 20mM HEPES [pH 7.4),120 mM KCI, 5 mM
MgCIZ, 1 mM dithiothreitol, 0.5% Nonidet P-40, 2 pglml leupeptin, and 50 pglml aprotinin. The nuclei were then removed by low-speed centrifugation and the supernatants were stored at -70°C until use.
Cytoplasmic extracts were normalized for piotein concentrations before use by the Bio-Rad protein microassay method. Each in vitro kinase reaction contained 20 ~1 of cell extract, 7.5 pl of reaction buffer (20 mM H~PES [pH 7.4], 120 mM KCI, 5 mM
MgCh, I mM dithiothreitol, and 10% glycerol) and 7.0 pl ofATP mixture (1.0 pCi[y-'~P]ATP in 7 ul of reaction buffer), and was incubated for 30 minutes at 37 ° C
(Mundschau; L.J.,and Falter, D.V.,J. Biol. Chem., 2673092-23098 (1992)).
Immediately after incubation the labelled extracts were either boiled in Laemmli SDS-sample buffer or were either precipitated with agarose-poly(I)poly{C) beads or imrnunoprecipitated with an anti-PKR antibody.
Agarose poly f~poly(C) precipitation To each in vitro kinase reaction mixture, 30 pl of a 50'/° Ag poly(I)poly(C) Type 6 slurry (Pharmacia LKB Biotechnology) was added, and the mixture was incubated at ,
4°C for 1 h. The Ag poly(I)poly(C) beads with the absorbed, labelled proteins were then washed four times with was buffer (20 mM HEPES [7.5 pH], 90 mM KCI, 0.1 mM .
EDTA, 2 mM dithiothreitol, 10% glycerol) at room temperature and mixed with 2X
Laemmli SDS sample bu#fer. The beads were then boiled for 5 min, and the released proteins were analyzed by SDS-PAGE.
Polymerase chain reaction Cells at various times postinfection were harvested and resuspended in ice cold THE {10 mM Tris [pH 7.8], 150 mM NaCl, 1 mM EDTA) to which NP-40 was then WO 99/08691 - ~ ~ PCT/CA98/00774 added to a final concentration of 1 %. A.Rer 5 minutes, the nuclei were pelkted and RNA was extracted from the supernatant using the phenol:chloroform procedure.
Equal amounts of total cellular RNA from each sample were then subjected to RT-PCR
(along, H., er al., (1994).Anal. Biochem., 223:251-258) using random hexanucleotide primers (Pharmacia) and reverse transcriptase (G1BC0-BRL) according to the manufacturers' protocol. The cDNA's from the RT-PCR step was then subjected to selective amplification of reovirus sl cDNA using the primer 5'- , AATTCGATTTAGGTGACACTATAGCTATTGGTCGGATG-3' (SEQ ID NO:1 ) and
5'-CCCTT'TTGACAGTGATGCTCCGTTATCACTCG-3' (SEQ ID N0:2) that amplify a predicted 1 I 6 by fragment. These primer sequences were derived from the S

sequence determined previously (Nagata, L., et al.,(1984) Nucleic Acids Res., 12:8699~
8710). The GAPDH primers (along, H., et al., (1994) Anal. Biochem., 223:351-358), S'-CGGAGTCAACGGATTTGGTCGTAT-3' (SEQ >D N0:3) and 5'-AGCCTTCTCCATGGTGGTGAAGAC-3'(SEQ ID N0:4) were used to amplify a predicted 306 by GAPDH ~agment which served as a PCR and gel Loading control.
Selective amplification of the s 1 and GAPDH eDNA's was performed using Taq DNA
polymerase (GIHCO-BRL) according to the manufacturers' protocol using a Perkin Elmer Gene Amp PCR system 9600. PCR was carried out for 28 cycles with each consisting of a denaturing step for 30 seconds at 97°C, annealing step for 45 seconds at 55 °C, and polymerization step for 60 seconds at 72 °C. PCR
products were analyzed by electrophoresis through an ethidium bromide-impregnated TAE-2% agarose gel and photographed under ultra-violet illumination with Polaroid 57 film.
Immunoprecipitation and SDS PAGE analysis Immunoprecipitation of 3sS-labelled reovirus-infected cell lysates with anti-reovirus semtype 3 serum was carried out as previously described (Lee, P.W.K.
et al.

WO 99/08692 ' ; ~ PCTICA98I00774 (1981) Yirology,108:134-146). Immunoprecipitation of'zP-labelled cell lysates with an anti-PKR antibody (from Dr. Michael Mathews, Cold Spring Harbor) was similarly carried out. Immunoprecipitates were analyzed by discontinuous SDS-PAGE
according to the protocol of Laemmli (Laemmli, U.K., (1970) Nature, 227:680-685).
EXAMPLE 1. Activated Intermediates in the Ras Signalling Pathway Augment Reovirus Infection Efficiency it was previously shown that 3T3 cells and their derivatives lacking epidermal growth factor receptors (EGFR) are poorly infectible by reovirus, whereas the same cells transformed with either EGFR~or v-erb B are highly susceptible as determined by cytopathic effects, viral protein synthesis, and virus output (Strong, J.E. et aL,( 1993) Yirology,197:405-411; Strong, J.E. and Lee, P.W.K., (1996) J. Yirol., 70:612-616).
To determine whether downstream mediators of the EGFR signal transduction pathway may be involved, a number of different NIH 3T3-derived, transformed with constitutively activated oncogenes downstream of the EGFR, were assayed for relative 1 S susceptibility to reovirus infection. Of particular interest were intermediates in the ras signalling pathway (reviewed by Barbacid, M., Annu. Rev. Biochem., 56:779-827 (1987); Cahill, M.A., et al., Curr. Biol., 6:16-19 (1996)). To investigate the Ras signalling pathway, IVIH 3T3 parental cell lines and NgI 3T3 lines transfected with activated versions of Sos (Aronheim, A., et al.,(1994) Cell, 78:949-961) or ras (Mundschau, L.J. and Faller, D.V., (1992) J. Biol. Chem., 267:23092-23098) oncogenes were exposed to reovirus, and their capacity to pmmote viral protein synthesis was compared.
Detection of viral proteins was initially carried out using indirect immunofluorescent microscopy as described above. The results indicated that whereas the N!H 3T3 cells adopted a typically flattened, spread-out morphology with marked WO 99/08692 ' ' ~ PCT/C~98/00774 -16-.
contact inhibition, the transformed cells all grew as spindle-shaped cells with much less contact inhibition. On comparing the uninfected parental cell lines with the various transformed cell lines, it was apparent that the morphology of the cells was quite .
distinct upon transformation. Upon challenge with reovirus, it became clear that parental 1VIH 3T3 line was poorly infectible (<5%), regardless of the source of the parental IvTIH 3T3 line. In contrast, the transfected cell lines each demonstrated' relatively pronounced immunofluorescence by 48 hours postinfecdon (data not shown).
To demonstrate that viral protein synthesis was more effcient in the Sos- or Rae-transformed cell lines, cells were continuously labeled with [35S]-methionine from 12 to 48 hr postinfection and the proteins were analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), as described above.
The results showed clearly that the levels of viral protein synthesis were significantly higher in the Sos- or Ras-transformed cells than in parental NIFi 3T3 cells.
The identities of the viral bands were confirmed by immunoprecipitation of the labeled proteins with polyclonal anti-reovirus antibodies. Since the uninfected ~ 3T3 cells and their transformed counterparts displayed comparable levels of cellular protein synthesis and doubling times (data not shown), the observed difference in the level~of viral protein synthesis could not~be due to intrinsic differences in growth rates or translation efficiencies for these cell lines:
The long-term fate of infected NtH-3T3 cells was followed by passaging these cells for at least 4 weeks. They grew normally and appeared healthy, with no sign of lyric or persistent infection; no virus could be detected in the medium after this time (data not shown).

WO 99/08692 ~ ~ PCT/CA98/00774 EXAMPLE 2. Enhanced Infecti'bility Conferred by Activated Oncogenes is Not Due to Long-term Transformation or the Generalized Transformed State of the Cell To determine whether the differences in susceptibility may be the result of long-term effects of transformation, or the result of the activated oncogene itself, a cell line expressing a zinc-inducible cellular Harvey-ras (c-H-ras) gene was tested for susceptibility to reovirus infectibility, as described above. 'These cells, called 2H 1, were derived from the C3H l OTl/2 cell line which is poorly infectible by reovirus (data not shown), and cant' the c-H-ras gene under the control of the mouse metallathionine-I
promoter (Trimble, W.S. et al. ( 1986) Nature, 311:782-784).
Cells were either mock-treated or pretreated with 50 ~.~M ZaSO,18 hours prior to infection or mock-infection (administration of ca~crier agent), followed by indirect immqnofluorescent analysis of these cells at 48 hours postinfection or mock-infection.
The results (not shown) demonstrated that uninduced cells were poorly infectible (<5°,'°) whereas those induced for only 18 hours were much more susceptible (>40%).
Enhanced viral protein synthesis in the Zn-induced 2H1 cells was further confirmed by , metabolic labeling of the cells with ['SS]methionine followed by SDS-PAGE
analysis of virus-specific proteins (not shown).
Based on these observations, the augmentation of reovirus infection efficiency in the transformed cells is a direct result of the activated oncogene product(s), and not due to other factors such as aneuploidy often associated with long-term transformation, or other accumulated mutations that may be acquired under a chronically transformed state (e.g., p53 or myc activation).
To show further that susceptibility to reovirus infection is not a result of transformation per se (i.e., a result of the transformed state of the host cell), IVIH-3T3 cells containing a tetracycline-controlled human c-myc gene (tet-myc cells) were WO 99/08692 ~ ~ PCT/CA98/00774 -1$-.
examined (HeIbing, C.C. et al., CancerRes. 57:1255-1258 (1997)). These cells normally are maintained in tetracycline (2 pglml) which represses the expression of c-myc. Removal of tetracycline under normal growth conditions (10% fetal bovine serum) leads to accumulation of the c-Myc protein and the cells display a transformed phenotype. We found that these cells were unable to support virus growth either in the presence or in the absence of tetracycline (data not shown), suggesting that susceptibility to reovirus infection is not due to the general, transformed state of the host cell, but rather requires specific transformation by elements of the Ras signaling pathway.
A good indicator of activation of the Ras signaling pathway is the activation of the MAP kinases ERK 1 and ERKZ (for a review, see Robinson, M.J. and Cobb, M.H., Curr. Opin. Cell. BioL 9:180-186 (1997)). In this regard, it was found that, compared with unrransformed cells, Ras-transformed cells have a significantly higher activity (data not shown). Furthermore, an examination of a number of human cancer cell Lines has revealed an excellent correlation between the level of ERK1/2 activity and susceptibility to reovirus infection (data not shown), although ERK1/2 itself does not appear to play any role in it. Mouse L cells and human HeLa cells, in which reovirus grows very well, both manifest high ERK1/2 activity (data not shown).
EXAMPLE 3. Viral Transcripts are Generated but Not Translated in Reovirus-Resistant hlIH 3T3 Cells The step at which reovirus infection is blocked in nonsusceptible NiH 3T3 cells was also identified. Because virus binding and virus internalization for nonsusceptible cells were comparable to those observed for susceptible cells (Strong, J.E. er al., (1993) Virology,197:405-411 ), the transcription of viral genes was investi Bated.

WO 99/0869 ' ' ~ . ~ PCT/CA98J00774 ,, The relative amounts of reovirus S 1 transcripts generated in N!H 3T3 cells and the Ras-transformed cells during the first 12 hours of infection were compazed after amplification of these transcripts by polymerase chain reaction (PCR), as described above. The results demonstrated that the rates of accumulation of S 1 transcripts in the two cell lines were similar, at least up to 12 hours postinfection. Similar data were obtained when rates of accumulation of other reovirus transcripts were compared (data not shown). These results demonstrate that infection block in nonsusceptible cells is not at the level of transcription of viral genes, but rather, at the level of translation of the transcripts.
At later times, the level of viral transcripts present in untransformed NIH-cells decreased significantly whereas transcripts in transformed cells continued to accumulate (data not shown). The inability of these transcripts to be translated in NIH-3T3 sells probably contributed to their degradation. As expected, the level of viral transcripts in infected L~ cells was at least comparable with that in infected Ras-transformed cells (data not shown).
EXAMPLE 4. A 65 kDa Protein is Phosphorylated in Reovirus-treated NIH 3T3 Cells but Not in Reovirus-in:ected Transformed Cells Because viral transcripts were generated, but not translated, in NIH 3T3 cells, it was investigated whether the double-stranded RNA (dsRNA}- activated kinase, PKR, is activated (phosphorylated) in these cells (for example, by S 1 mRNA
transcripts which have been shown to be potent activators of PKR {{Bischoff, J.R. and Samuel, C.E., (1989) Virology,172:106-115), which in tum leads to inhibition of translation of viral genes. The corollary of such a scenario would be that in the case of the transformed cells, this activation is prevented, allowing viral protein synthesis to ensue.

WO 99/08692 ' ~ ' PCT/CA98I00774 -20. .
1~IH 3T3 cells aad v-erbB- or Ras- transformed cells (designated THC-11 and H-ras, respectively) were treated with reovirus (i.e., infected) or mock-infected (as above), and at 48 hours post treatment, subjected to in vitro kinase reactions, followed by autoradiographic analysis as described above.
The results clearly demonstrated that there was a distinct phosphoprotein migrating at approximately 65 kDa, the expected size of PKR, only in the I~IIH
3T3 cells and only after exposure to reovirus. This protein was not labeled in the lysates of either the uninfected transformed cell lines or the infected transformed cell lines.
Instead, a protein migrating at approximately 100 kDa was found to be labeled in the transformed cell lines after viral infection. This protein was absent in either the preinfection or the postinfection lysates of the NIH 3T3 cell line, and was not a reovirus protein because it did not react with an anti-reovirus serum that precipitated all reovirus proteins (data not shown). A similar 100 kDa protein was also found to be 32P-labeled in in vitro kinase reactions of postinfection lysates of the Sos-transformed cell lines (data not shown).
That intermediates in the Ras signalling pathway were responsible for the lack of phosphorylation of the 55 kDa protein was further confirmed by the use of the 2H1 cells which contain a Zn-inducible Ras oncogene. Unindueed 2H1 cells(relatively resistant to reovirus infection, as shown above), were capable of producing the 65 kDa phosphoprotein only after exposure to virus. However, 2H1 cells subjected to Zn-induction of the H-Ras oncogene showed significant impairment of the production of this phosphoprotein. This impairment coincided with the enhancement of viral synthesis. These results therefore eliminated the possibility that the induction of the 65 kDa phosphoprotein was an NIH 3T3-specific event, and clearly established the role of Ras in preventing (or reversing) induction of the production of this phosphoprotein.
The Zn-induced 2H1 cells did not produce the I00 kDa phosphoprotein seen in the infected, chronically transformed H-Ras cells.

WO 99/08692 ' , ~ PCT/CA98/00774 EXAMPLE 5. Induction of Phosphorylation of the 65 kDa.Protein Requires Active Viral Transcription Since production of the 65 kDa phosphoprotein occurred only in cells that were resistant to reovirus infection, and only after the cells were exposed to reovirus, it was investigated whether active viral transcription was required for production of the 68 kDa phosphoprotein. Reovirus was UV-treated to inactivate its genome prior to administration of the reovirus to NI~i 3T3 cells. For UV-treatment, reovirus was suspended in DMEM to a concentration of approximately 4 x 10$ PFU/mL and exposed to short-wave (254 nm) LJV light for 20 minutes. W-inactivated virus were non-infectious as detenmined by lack of cytopathic effects on mouse L-929 fibroblasts and lack of viral protein synthesis by methods of [3sSJ-methionine labelling as previously described. Such IJV treatment abolished viral gene transcription, as analyzed by PCR, and hence viral infectivity (data not shown). The cells were then incubated for 48 hours, and lysates were prepared and subjected to in visro'ZP-labeling as before. The results showed that NIfi 3T3 cells infected with untreated reovirus produced a prominent 65 kDa ~ZP-labelled band not found in uninfected cells. Cells exposed to , UV-treated reovirus behaved similarly to the uninfected control cells, manifesting little phosphorylation of the 65 kDa protein. Thus, induction of the phosphorylation of the 65 kDa phosphoprotein is not due to dsRNA present in the input reovirus;
rather, it requires de novo transcription of the viral genes, consistent with the identification of the 65 kDa phosphoprotein as PKR.
EXAMPLE 6. Identification of the 65 kDa Phosphoprotein as PKR
To determine whether the 65 kDa phosphoprotein was PKR, a dsRNA binding experiment was carried out in which poly(1)-poly(c) agarose beads were added to'ZP
labeled lysates , as described above. After incubation for 30 minutes at 4°C, the beads CA 02508238 1998-08-12 , .
WO 99/08b92 - PCT/CA98/00774 -22- .
were washed, and bound proteins were released and analyzed by SDS-PAGE. The results showed that the 65 kDa phosphoprotein produced in the postinfection iVl~i 3T3 cell lysates was capable of binding to dsRNA;~ such binding is a well-recognized characteristic of PKR. In contrast, the 100 kDa phosphoprotein detected in the infected H-ras-transformed cell line did not bind to the Poly(I)-poly(c) agarose. The 65 kDa phosphoprotein was also immunoprecipitable with a PKR-specific antibody (provided by Dr. Miice Mathews, Cold Spring Harbor Laboratory), confirming that it was indeed Pi~R.
EXAMPLE 7. PKR Inactivation or Deletion Results in Enhanced Infectibility of Untransformed Cells If PKR phosphorylation is responsible for the shut-off of viral gene translation in NIH-3T3 cells, and one of the functions of the activated oncogene products) in the transformed cells is the prevention of this phosphorylation event, then inhibition of PKR
phosphorylation in IVIH-3T3 cells by other means (e.g. drugs) should result in the enhancement of viral protein synthesis, and hence infection, in these cells.
To test this idea, 2-aminopurine was used. This drug has been shown to possess relatively specific inhibitory activity towards PKR autophosphorylation (Samuel, C.E. and Brody, M., (1990) Virology, 176:106-113; Hu, Y. and Conway, T.W. (1993), J. Interferon Res., 13:323-328). Accordingly, htIH 3T3 cells were exposed to 5 mM 2-aminopurine concurrently with exposure to reovirus. The cells were labeled with [35S]methionine from 12 to 48 h postinfection, and lysates were harvested and analyzed by SDS-PAGE.
The results demonstrated that exposure to 2-aminopurine resulted in a significantly higher level of viral protein synthesis in hIIH 3T3 cells {not shown). The enhancement was particularly pronounced after immunoprecipitating the lysates with an anti-reovirus serum. These results demonstrate that PKR phosphorylation leads to WO 99/08692 ' ~ PCT/CA98/00774 inhibition of viral gene translation, and that inhibition of this.phosphorylation event releases the translation block. Therefore, intermediates in the Ras signalling pathway negatively regulate PKR, leading to enhanced infectibility of Ras-transformed cells.
Interferon Vii, known to induce PKR expression, was found to significantly reduce reovirus replication in Ras-transformed cells (data not shown).
A more direct approach to defining the role of PKR in reovirus infection is through the use of cells that are devoid of PKR. Accordingly, primary embryo fibroblasts from wild-type PKR +~" and PKR °/° mice (Yang, Y.L.
et al. EMBO J.
14:6095-6106 (1995)) were compared in terms of susceptibility to reovirus infection.
i 0 The results clearly showed that reov~rus proteins were synthesized at a significantly higher level in the PKR °/° cells than in the PKR'/' cells.
These experiments demonstrated that PKR inactivation or deletion enhanced host cell susceptibility to reovisus infection in the same way as does transformation by Ras or elements of the Ras signaling pathway, thereby providing strong support of the role of elements of the Ras signaling pathway in negatively regulating PKR.
EXAMPLE 8 Inactivation of PKR in Transformed Cells Does Not Involve MEK
Receptor tyrosine kinases such as EGFRs are known to stimulate the mitogen-activated or extracellular signal-regulated kinases (ERK1/2) via Ras (see Robinson, M.J. and Cobb, M.H., Curr. Opin. Cell. Biol. 9:180-186 (1997)). This stimulation requires the phosphorylation of ERK1/2 by the mitogen-activated extracellular signal-regulated kinase, kinase MEK, which itself is activated (phosphorylated) by Raf, a serine-threonine kinase downstream of Ras. To determine if MEK activity was required for PKR inactivation in transformed cells, the effect of the recently identified EK
inhibitor PD98059 (Dudley, D.T. et al., Proc. Natl. Acad Sci. USA 92:7686-7689 WO 99/08692 ~ PCT/CA98/00774 (1995); Waters, S.D. et al., J. Biol. Chem. 270:20883-20886 (1995)) on infected Ras-transformed cells was studied.
H-Ras-transformed cells were grown to 80% confluency and infected with reovirus at an m.o.i. of approximately 10 p.fu.lcell. PD98059 (Calbiochem), dissolved in dimethylsulfoxide (DMSO), was applied to the cells at the same time as the virus (final concentration of PD98059 was 50 pM). The control cells received an equivalent volume of DMSO. The cells were labeled with 35S-methionine from 12 to 48 hours post-infection. Lysates were then prepared, immunoprecipitat~ with the polyclona3 anti-reovirus serotype 3 serum end analyzed by SDS-PAGE.
The results (data not shown) showed that PD98059, at a~concentration that effectively inhibited ERKl/2 phosphorylation, did not inhibit reovirus protein synthesis in the transformed cells. On the contrary, PD98039 treatment consistently caused a slight enhancemenx of viral protein synthesis in these cells; the reason for this is under investigation. Consistent with the lack of inhibition of vital protein synthesis in the presence of PD98059, the PKR in these cells remained unphosphorylated (data not shown). As expected, PD98059 had no effect on reovirus-induced PKR
phosphorylation in untransformed N1H-3T3 cells (data not shown). These results indicated that MEK and ERK1/2 are not involved in PKR activation.
EXAMPLE 9. In Vivo Oncolytic Capability of Reovirus A severe combined immunodeficiency (SCm) host tumor model was used to assess the efficacy of utilizing reovirus for tumor reduction. Male and female mice (Charles River, Canada) were injected with v-erbB-transformed NIH 3T3 mouse fibroblasts (designated THC-11 cells) in two subcutaneous sites overlying the hind flanks. In a first trial, an injection bolus of 2.3 X 105 cells in 100 p.l of sterile PBS was WO 99/08693 ~ ~ ~. PCT/CA98/00774 used. In a second trial, an injection bolus of 4.8 X 106 cells in 100 p.l PBS
was used.
Palpable tumors were evident approximately two to three weeks post injection.
Reovirus serotype three (strain bearing) was injected into the right-side tumor mass (the "treated tumor mass") in a volume of 20 ~tl at a concentration of 1.0 X 109 plaque forming units (PF>.)]/ml. The left-side .tumor mass (the "untreated tumor mass") was left untreated. The mice were observed for a period of seven days following injection with reovirus, measurements of tumor size were taken every two days using calipers, and weight of tumors was measured after sacrifice of the animals.
All mice were sacrificed on the seventh day. Results are shown in Table 1.
Table 1 Tumor Mass after Treatment with Reovirus Trial 1 (n=8) mean untreated tumor mass fi02 mg mean treated tumor mass 284 mg Tria12 (n =12) mean control tumor mass 1523.5 mg mean untreated tumor mass 720.9 mg mean treated tumor mass 228.0 mg The treated tumor mass was 47% of that of the untreated tumor mass in trial 1, and 31.b% of the untreated tumor mass in trial 2. These results indicated that the virus-treated tumors were substantially smaller than the untreated tumors, and that there may be an additional systemic effect of the virus on the untreated tumor mass.
Similar experiments were also conducted using unilateral introduction of tumor cells. SCID mice were injected subcutaneously and uniiaterally_in the hind flank with v-erbB-transformed IVIla 3T3 mouse fibroblasts (THC-11 cells). Palpable tumors (mean area 0.31 cm2) were established after two weeks. Eight animals were then given CA 02508238 1998-08-12 . , WO 99/08692 ' PCT/CA98/00774 _2~
a single intratumoral injection of 1.0 x 10' PFUs of reovirus serotype 3 (strain bearing) in phosphate-buffered saline (PBD). Control tumors (n=10) were injected with equivalent amounts of W-inactivated virus. Tumor growth was followed for 12 days, during which time no additional reovirus treatment was administered.
Results, shown in Figure 2, demonstrated that treatment of these tumors with a single dose of active reovirus (open circles) resulted in dramatic repression of tumor growth by the thirteenth day (endpoint), when tumors in the control animals injected with a single dose of inactivated reovirus (closed circles) exceeded the acceptable tumor burden. This experiment was repeated several times and found to be highly reproducible, thus further demonstrating the efficacy of reovirus in repressing tumor growth.
Example 10: In Vivo Oncolytic Capability of Reovirus Against Human Breast Cancer-Derived Cell Lines ' In vivo studies were also carried out using human breast carcinoma cells in a SCm mouse model. Female SCID mice were injected with 1 x 109 MDAMB468 cells in two subcutaneous sites, overlying both hind flanks. Palpable tumors were evident approximately two to four weeks post injection. Undiluted reovirus serotype three (strain bearing) was injected into the right side tumor mass in a volume of 20 pl at a concentration of 1.0 x 10'° PFU/ml. The results are shown in Table 2.

WO 99/08692 ~ ' PCT/CA98/00774 _27_ , ,, Table 2 Tumor Mass After Treatment with Reovirus TREATMENT mean untreated tumor mean treated tumor mass mass (right (kft side) ' side) Reovirus (N=8) 29.02 g ' 38.33 g Contml (N=8) 171.8 g 128.54 g *Note: One of the control mice died early during the treatment phase. None of the reovirus-treated mice died.
Although these studies were preliminary, it was clear that the size of the tumors in the reovirus-treated animals was substantially lower than that in the untreated animals. However, the size of the tumors on the right (treated) side of the reovirus-treated animals was slightly larger on average than the left (untreated) side.
This was unexpected but may be explained by the composition of the mass being taken up by inflammatory cells with subsequent fibrosis, as well as by the fact that these tumors were originally larger on the right side on average than the left. The histologic composition of the tumor masses is being investigated. These results also support the systemic effect the reovirus has on the size of the untreated tumor on the contralateral slide of reovirus injection.
E~~AMPLE 11. Susceptibility of Additional Human Tumors to Reovirus Oncolysis In view of the in vivo results presented above, the oncolytic capability observed in marine cells was investigated in cell lines derived from additional human tumors.
A. Materials and Methods WO 99/08692 ~ ' PCT/CA98/00774 -fig- .
Cells and Vir us All cell lines were grown in Dulbecco's modified Eagles medium (DMEIVI) containing 10% fetal bovine serum (FBS).
The bearing strain of reovirus serotype 3 used in these studies was propagated in suspension cultures of L cells and purified according to Smith (Smith, R.E.
et a1, (1969) Virology, 39:791-800) with the exception that ~i-mercaptoethanol (~i-ME) was omitted from the extraction buffer. Reovirus labelled with [3sS]methionine was grown and purified as descn'bed by McRae and Joklik (McRae, M.A. and Joklik, W.K., (1978) Virolo~~, 89:578-593). The partiele/PFU ration for purified reovirus was typically 100/1.
Cytopathic e, fj"ects of reovinrs on cells ' Confluent monolayers of cells were infected with reovirus serotype 3 (strain bearing) at a multiplicity of infection (MO>] of approximately 4d plaque forming units (PFLT) per cell. Pictures were taken at 36 hour postinfection for both reovirus-infected and mock-infected cells.
ImmunoJluorescent analysis of reovirus infection For the immunofluorescent studies the cells were grown on coverslips, and infected with reovirus at a multiplicity of infection (MOl] of -10 PFU/cell or mock-infected as described above. At various times postinfection, cells were fixed in an ethanoUacetic acid (20/1) mixture for 5 minutes, then rchydrated by subsequential washes in 75%, 50% and 25% ethanol, followed by 4 washes with phosphate-buffered saline (PBS). The fixed and rehydrated cells were then exposed to the primary antibody (rabbit polyclonal anti-reovirus type 3 serum:diluted 1/100 in PBS) for 2 hr at room temperature. Following 3 washes with PBS, the cells were exposed to the secondary WO 99/08692 ~ ' PCT/CA98/00774 antibody [goat anti-rabbit IgG (whole molecule) fluotescein isothiocyanate (FITC) conjugate diluted 1/100 in PBS containing 10% goat serum and O.a05% Evan's Blue counterstain] for 1 hour at room temperature. Finally, the fixed and treated cells were washed 3 more times with PBS, followed by 1 leash with double-distilled water, dried and mounted on slides in 90% glycerol containing 0:1 % phenylenediamine, and viewed with a Zeiss Axiophot microscope mounted with a Carl Zeiss camera (magnification for all pictures was 200 x).
Infection of cells and quantitation of virus Confluent monolayers of cells grown in 24-well plates were infected with reovirus at an estimated multiplicity of 10 PFU/cell. After 1 hour incubation at 37°C, the monolayers were washed with warm DMEM-10% FBS, and then incubated in the same medium. At various times postinfection, a mixture of NP-40 end sodium deo~: vcholate was added directly to the medium on the infected monolayers to final concentrations of 1 % and 0.5%, respectively. The lysaxes were then harvested and virus yields were determined by plaque titration on L-929 cells.
Radiolabelling of reovirus-Infected cells and preparation of Ivsates Confluent monolayers of cells were infected with reovirus (MOI ~10 PFU/cell).
At various times postinfection, the media was replaced with methionine-free DMEM
containing 10% dialyzed PBS and 0.1 mCi/ml ['sS]methionine. After further incubation for 1 hour at 37°C, the cells were washed in phosphate-buffered saline (PBS) and lysed in the same buffer containing 1 % Triton X-100, 0.5% sodium deoxycholate and 1 mM
EDTA. The nuclei were then removed by low speed centrifugation and the supernatants was stored at 70°C until use.

WO 99/08692 ~ PCT/CA98/00774 Immunoprecipitation and SDS PAGE analysis Iymunoprecipitation of [~sS]-labelled reovirus-infected cell lysates with anti-reovirus serotype 3 serum was carried out as previously described (Lee, P.W.K.
et al.
( 198 / ) Virology,108:134-146). Immunoprecipitates were analyzed by discontinuous SDS-PAGE according to the protocol of Laemmli (I:aemmli, U.K., (1970) Nature, 227:680-685).
B. Breast Cancer The c-erbB-2/neu gene encodes a transmembrane protein with extensive homology to the EGFR that is overexpressed in 20-30% of patients with breast cancer (Yu, D. et al. (1996) Oncogene 13:1359).Since it has been established herein that Ras activation, either through point mutations or through augmented signaling cascade elements upstream of Ras (including the c-erbB-2/neu homologue EGFR) ultimately creates a hospitable environment for reovirus replication, an array of cell lines derived from human breast cancers were assayed far reovirus susceptibility. The cell Iines included MDA-MD-435SD (ATCC deposit HTB-129), MCF-7 (ATCC deposit HTB-22), T-27-D (ATCC deposit HTB-133), BT-20 (ATCC deposit HTB-19), HBL-100 (ATCC deposit HTB-124), MDA-MB-468 (ATCC deposit HTB-132), and SKBR-3 (ATCC deposit HTB-30).
Based upon induction of cytopathic effects, and viral protein synthesis as measured by radioactive metabolic labeling and immunofluorescence as described above, it was found that five out of seven of the tested breast cancers were susceptible to reovirus infection: MDA-MB-435S, MCF-7, T-27-D, MDA MB-468, and SKBR-3 were exquisitely sensitive to infection, while BT-20 and HBL-100 demonstrated no infectibility.

WO 99/08692 ~ ' PCT1CA98I00774 C. Brain Glioblastoma Next a variety of cell lines derived from human brain glioblastomas was investigated. The cell lines included A-172, U-' 118, U-178, U-563, U-251, U-87 and U-373 (cells were a generous gift from Dr. Wee Yong, University of Calgary).
Six out of seven glioblastoma cell lines demonstrated susceptibility to reovirus infection, including U-118, U-178, U-563, U-251, U-87 and U-373, while A-172 did not demonstrate any infectibility, as measured by cytopathic effects, immunofluorescence and (35S]-methionine labeling of reovirus proteins.
The U-8? glioblastoma cell line was investigated further. To assess the sensitivity of U-87 cells to reovirus, U-87 cells (obtained from Dr. Wee Yong, University of Calgary) were grown to 80% confluency and were then challenged with reovinis at a multiplicity of infection (MOn of 10. Within a period'of 48 hours there was a dramatic, widespread cytopathic effect (data not shown). To demonstrate further that the lysis of these cells was due to replication of reovirus, the cells were then pulse-labeled with (~SSJmethionine for three hour periods at various times postinfection and proteins were analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis , (SDS-PAGE) as described above. The results (not shown) clearly demonstrated effective reovirus replication within these cells with resultant shutoff' of host protein synthesis by 24 hours posdnfection.
The U-87 cells were also introduced as human tumor xenografts into the hind flank of 10 SC1D mice. U 87 cells were grown in Dulbecco's modified Eagle's medium containing 10% fetal bovine serum, as described above. Cells were harvested, washed, and resuspended in sterile PBS; 2.0 x 106 cells in 100 pl were injected subcutaneousiy at a site overlying the hind flank in five- to eight-week old male SCID
mice (Charles River, Canada): Tumor gmwth was measured twice weekly for a period WO 99108692 ~ PCT/CA98/00774 _32_ .
of four weeks. Results, shown in Figwe 3, demonstrated that treatment of U-87 tumors with a single intratumoral injcction of 1.0 x 10' PFUs of live reovirus (open circles, n=5) resulted in drastic repression of tumor growth, including tumor regression by the fourth week post-treatment (P=0.008), in comparison with treatment of tumors with a single intratumoral injection of the same amount of LTV-inactivated reovirus (closed circles, n=5).
Hematoxylin/eosin (HE)-staining of the remaining microfoci of the tumors.
treated with active virus, performed as described (H. Lyon, Cell Biology, A
Laboratory Handbook, J.E. Cells, ed., Acade~pnic Press, 1994, p. 232) revealed that the remaining tumor mass consisted largely of normal stroma without appreciable numbers of viable tumor cells, nor was there any eevidence of infiltration of tumor cells into the underlying sceletal muscle (data not shown). Necrosis of tumor cells was due to direct lysis by the virus, the same mechanism of cell killing as by reovirus in vitro.
To. determine if there was viral spread beyond the tumor mass, immunofluorescent microscopy using antibodies directed against total reovirus proteins was conducted as described above, on sections of the tumor and adjoining tissue. It was found that reovirus-specific proteins were confined to the tumor mass; no viral staining was detected in the underlying skeletal muscle {data not shown). As expected, viral proteins ~~ere not present in tumors injected with the UV-inactivated virus (data not shown). These results demonstrated that reovirus replication in these animals was highly tumor specific with viral amplification only in the target U-87 cells.
Since most tumors are highly vascularized, it was likely that some virus could enter the blood stream following the lysis of the infected tumor cells. To determine if there was systemic spread of the virus, blood was harvested from the treated and control animals, and serially diluted for subsequent plaque titration. Infectious virus was found to be present in the blood at a concentration of 1 x 105 PFLTs/ml (data not shown).

WO 998692 ' ~ ' PCT/CA98100774 The high degree of tumor specificity of the virus, combined with systemic spread, suggested that reovirus could be able to replicate in glioblastoma tumors remote from the initially infected tumor, as demonstrated above with regard to breast cancer cells.. To verify this hypothesis, SCID mice were implanted bilaterally with U-human tumor xenografts on sites overlying each hind flank of the animals.
These tumors were allowed to grow until they measured 0.5 x 0.5 cm. The left-side tumors were then administered a single infection of reovirus in treated animals (n=S); control animals (n=7) were mock-treated with UV-inactivated virus. Tumors were again measured twice weekly for a period of four weeks.
Results, shown in Figure 4, demonstrated that inhibition and eventual regression of both the treated (circles) and untreated tumor masses (squares) occurred only in the live reovirus-treated animals (open circles and squares), in contrast with the inactivated reovirus-treated animals (closed circles and squares). Subsequent immunofluorescent analysis revealed that reovirus proteins were present in both the ipsilateral (treated) as well as the contralateral (untreated) tumor, indicating that regression on the untreated side was a result of reovirus oncolysis (data not shown).
D. Pancreatic Carcinoma Cell lines derived from pancreatic cancer were investigated for their susceptibility to reovirus infection. The cell lines included Capan-1 (ATCC
deposit HTB-79), BxPC3 (ATCC deposit CRL-1687), MIAPACA-2 (ATCC deposit CRL-1420), PANC-1 (ATCC deposit CRL-1469), AsPC-1 (ATCC deposit CRL-1682) and Hs766T (ATCC deposit HTB-I34).
Five of these six cell lines demonstrated suscept'bility to reovirus infection including Capan-I, MIAPACA-2, PANC-1, AsPC-1 and Hs766T, whereas BxPC3 demonstrated little infectability as assayed by virus-induced cytopathological effects, -34. .
immunofluorescence and {3SS]-labelling. Interestingly, four of the five cell lines demonstrating susceptibility to reovirus oncolysis have been shown to possess transforming mutations in codon 12 of the K-ras gene (Capon-1, MIAPACA-2, PANC-and AsPC-1) whereas the one lacking such susceptibility (BxPC3) has been shown to lack such a mutation (Berrozpe, G., et al. (1994), Int. J. Cancer, 58:185-191). The status of the other K-ras codons is currently unknown for the Hs766T cell line.
E3~LE 12. Use of Reovirus as an Cmcolytic Agent in Immune-Competent Animals A syngeneic mouse model was developed to investigate use of reovirus in immune-competent animals rather than in SCID mice as described above. C3H mice (Charles River) were implanted subcutaneously with 1.0 x 10' PFUs ras-transformed C3H cells (a gift of D. Edwards, University of Calgary). Following tumor establishment, mice were treated with a series of injections of either live reovirus (1.0 x 10$ PFUs) or UV-inactivated reovirus. Following an initial series (six injections over a nine-day course), test animals received a treatment of dilute reovirus (1.0 x 10' PFUs) every second day. Mock-treated animals received an equivalent amount of UV-inactivated virus.
Results demonstrated that.reovirus was an effective oncolytic agent in these immune competent animals. All of the test animals showed regression of tumors;
5 of the 9 test animals exhibited complete tumor regression after 22 days, a point at which the control animals exceeded acceptable tumor burden. Furthermore, there were no identifiable side effects in the animals treated with reovirus.
To assess the e#fects of previous reovirus exposure on tumor repression and regression, one-half of a test group was challenged with reoviivs (intramuscular injection of 1.0 x 10g PFUs, type 3 bearing) prior to tumor establishment. Two weeks CA 02508238 1998-08-12 , WO 99/08692 . ~ ' PCT/CA98/0077A
_ ~ 1 a#~er challenge, neutralizing antibodies could be detected in all exposed animals.
following tumor establishment, animals were treated with a series of either live or UV-inactivated reovirus, as described above.
Results (data not shown) demonstrated that animals with circulating neutralizing antibodies to reovirus (i.e., those challenged with reovirus prior to tumor establishment) exhibited tumor repression and regression similar to those animals in which there was no prior exposure to reovirus. Thus, reovirus can serve as an effective oncolytic agent even in immune-competent animals with previous exposure to reovitus.
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention as defined by the appended claims.

Claims (30)

What is claimed is:
1. Use of a reovirus for the manufacture of a medicament for treating a neoplasm in a mammal, wherein PKR is inactivated or deleted in the neoplasm.
2. The use of claim 1, wherein phosphorylation of PKR in the neoplasm is decreased or inhibited.
3. The use of claim 1 or 2, wherein the reovirus is a human reovirus.
4. The use of claim 3, wherein the human reovirus is selected from the group consisting of type 1 reovirus, type 2 reovirus, and type 3 reovirus.
5. The use of claim 1 or 2, wherein the reovirus is a nonhuman reovirus.
6. The use of claim 5, wherein the nonhuman reovirus is selected from the group consisting of mammalian reovirus and avian reovirus.
7. The use of any one of the claims 1 to 6, wherein the reovirus comprises more than one type of reovirus.
S. The use of any one of claims 1 to 6, wherein the reovirus comprises more than one strain of reovirus.
9. The use of any one of claims 1 to 8, wherein the reovirus is a field isolate.
10. The use of any one of claims 1 to 9, wherein the reovirus is treated with a protease prior to administration.
11. The use of any one of claims 1 to 10, wherein the neoplasm is a solid neoplasm.
12. The use of any one of claims 1 to 11, wherein the neoplasm is selected from the group consisting of pancreatic cancer, breast cancer, brain cancer, lung cancer, prostate cancer, colorectal cancer, thyroid cancer, renal cancer, adrenal cancer and liver cancer.
13. The use of any one of claims 1 to 10, wherein the neoplasm is a hematopoietic neoplasm.
14. The use of claim 13, wherein the hematopoietic neoplasm is selected from the group consisting of lymphoma and leukemia.
15. The use of any one of claims 1 to 14, wherein the neoplasm is metastatic.
16. The use of any one of claims 1 to 15, wherein the mammal is selected from the group consisting of mice, dogs, cats, sheep, goats, cows, horses, pigs, and nonhuman primates.
17. The use of any one of claims 1 to 15, wherein the mammal is a human.
18. The use of any one of claims 1 to 17, wherein the medicament is adapted for administration by a route selected from the group consisting of intrathecal, intravenous, intramuscular and intraperitoneal administration.
19. The use of any one of claims 1 to 18, wherein the reovirus comprises approximately 3 to 10 12 plaque forming units of reovirus.
20. The use of any of claims 1 to 19, wherein the medicament is adapted for administration in a single dose.
21. The use of any one of claims 1 to 19, wherein the medicament is adapted for administration in more than one dose.
22. The use of any one of claims 1 to 21, wherein the medicament is adapted for administration to more than one neoplasm in the mammal.
23. A pharmaceutical composition useful for the treatment of a neoplasm, wherein PKR
is inactivated or deleted in the neoplasm, the composition comprising:

(a) an effective amount of at least one reovirus, wherein the amount is effective to result in oncolysis of the neoplasm; and (b) a pharmaceutically acceptable excipient.
24. The composition of claim 23, wherein phosphorylation of PKR in the neoplasm is decreased or inhibited.
25. The composition of claim 23 or 24 which is adapted for parenteral administration.
26. The composition of claim 25, wherein the parenteral administration is selected from the group consisting of intravenous administration and subcutaneous administration.
27. The composition of claim 23 or 24 which is adapted for administration directly into the neoplasm.
28. The composition of claim 23 or 24 which is adapted for topical application.
29. A kit comprising a pharmaceutical composition according to any one of claims 23 to 28 and means for administering the composition.
30. The kit of claim 29, wherein the means for administering is a syringe.
CA002508238A 1997-08-13 1998-08-12 Reovirus for the treatment of neoplasia Expired - Lifetime CA2508238C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/911,383 1997-08-13
US08/911,383 US6110461A (en) 1997-08-13 1997-08-13 Reovirus for the treatment of neoplasia
CA002283280A CA2283280C (en) 1997-08-13 1998-08-12 Reovirus for the treatment of neoplasia

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA002283280A Division CA2283280C (en) 1997-08-13 1998-08-12 Reovirus for the treatment of neoplasia

Publications (2)

Publication Number Publication Date
CA2508238A1 CA2508238A1 (en) 1999-02-25
CA2508238C true CA2508238C (en) 2008-01-15

Family

ID=35005612

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002508238A Expired - Lifetime CA2508238C (en) 1997-08-13 1998-08-12 Reovirus for the treatment of neoplasia

Country Status (1)

Country Link
CA (1) CA2508238C (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013163727A1 (en) * 2012-04-30 2013-11-07 Oncolytics Biotech Inc. Protecting modified viruses from neutralizing antibodies using the reovirus sigma 1 protein

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7306902B2 (en) 2002-06-28 2007-12-11 Oncolyties Biotech Inc. Oncolytic viruses as phenotyping agents for neoplasms
US8491884B2 (en) 2000-05-03 2013-07-23 Oncolytics Biotech Inc. Virus clearance of neoplastic cells from mixed cellular compositions

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013163727A1 (en) * 2012-04-30 2013-11-07 Oncolytics Biotech Inc. Protecting modified viruses from neutralizing antibodies using the reovirus sigma 1 protein
WO2013163724A1 (en) * 2012-04-30 2013-11-07 Oncolytics Biotech Inc. Protecting modified viruses from neutralizing antibodies using the reovirus sigma 1 protein

Also Published As

Publication number Publication date
CA2508238A1 (en) 1999-02-25

Similar Documents

Publication Publication Date Title
CA2283280C (en) Reovirus for the treatment of neoplasia
US8709443B2 (en) Reovirus for the treatment of cellular proliferative disorders
CA2508238C (en) Reovirus for the treatment of neoplasia
CA2615684C (en) Reovirus for the treatment of cellular proliferative disorders
AU2004201630B2 (en) Reovirus for the treatment of cellular proliferative disorders
LEE et al. Patent 2283280 Summary
MXPA99010176A (en) Reovirus for the treatment of neoplasia
AU2007202448A1 (en) Reovirus for the treatment of cellular proliferative disorders

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20180813