AU2005201079C1 - Treatment of neoplasms with viruses - Google Patents

Treatment of neoplasms with viruses Download PDF

Info

Publication number
AU2005201079C1
AU2005201079C1 AU2005201079A AU2005201079A AU2005201079C1 AU 2005201079 C1 AU2005201079 C1 AU 2005201079C1 AU 2005201079 A AU2005201079 A AU 2005201079A AU 2005201079 A AU2005201079 A AU 2005201079A AU 2005201079 C1 AU2005201079 C1 AU 2005201079C1
Authority
AU
Australia
Prior art keywords
virus
cells
dose
tumor
interferon
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.)
Ceased
Application number
AU2005201079A
Other versions
AU2005201079A1 (en
AU2005201079B2 (en
Inventor
William S. Groene
Robert M. Lorence
Harvey Rabin
Michael S. Roberts
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.)
Wellstat Biologics Corp
Original Assignee
Wellstat Biologics Corp
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 AU42469/00A external-priority patent/AU4246900A/en
Application filed by Wellstat Biologics Corp filed Critical Wellstat Biologics Corp
Priority to AU2005201079A priority Critical patent/AU2005201079C1/en
Publication of AU2005201079A1 publication Critical patent/AU2005201079A1/en
Publication of AU2005201079B2 publication Critical patent/AU2005201079B2/en
Assigned to WELLSTAT BIOLOGICS CORPORATION reassignment WELLSTAT BIOLOGICS CORPORATION Alteration of Name(s) of Applicant(s) under S113 Assignors: PRO-VIRUS, INC.
Application granted granted Critical
Publication of AU2005201079C1 publication Critical patent/AU2005201079C1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16051Methods of production or purification of viral material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2720/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsRNA viruses
    • C12N2720/00011Details
    • C12N2720/12011Reoviridae
    • C12N2720/12051Methods of production or purification of viral material

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Virology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

10/03 '05 17:04 FAX 613 9663 3099 F.B. RICE Co.
o 1A TREATMENT OF NEOPLASMS WITH VIRUSES SField of the Invention This is a divisional of AU 42469/00, the entire contents of which are incorporated herein by reference.
The subject invention relates to viruses that are able to replicate in and cause the death ofneoplastic cells with a deficiency in the interferon(1FN)-mediated antiviral o response. RNA and DNA viruses are useful in this regard. The invention also relates to the use of these viruses for the treatment of neoplastic diseases including cancer and Ci large tumors.
t 10 Background of the Invention 0 Neoplastic disease which includes cancer is one of the leading causes of death among human beings. There are over 1.3 million new cases of cancer diagnose in the United States each year and 550,000 deaths. Detecting cancer early, before it has spread to secondary sites in the body, greatly increases a host's chances of survival.
However, early detection of cancer is not always possible, and even when it is, treatments are unsatisfactory, especially in cases of highly malignant cancers. Cancer treatments, including chemotherapy and radiation, are much less effective in later stages, especially when neoplastic growths are large and/or constitute a high tumor burden. (See Hillard Stanley, Cancer Treat. Reports, Vol. 61, No.l, Jan/Feb 1977, p.
29-36, Tannock, Cancer Research, 42,4921-4926, Dec. 1982).
Tumor regression associated with exposure to various viruses has been reported.
Most of the viruses described are pathogenic in humans, and include mumps and measles. The effect of other specific viruses on particular types of cancer cells has also been described. Smith et al, (1956) Cancer 9,1211 (effect of adenovirus on cervix carcinoma); Holzacpfel et al, (1957) Caner 10,557 (effect ofadenovirus on epithelial tumor); Taylor et al, (1970) J. Natl. Cancer Inst 44,515 (effect of bovine enterovirus-1 onsarcoma-1); Shingu et al, (1991) J General Viroloav. 72,2031 (effect of bovine enterovirus MZ-468 on F-647, a leukemia cells); Suskind et al, (1957) PSEBM, 94,309 (effect of coxsackie B3 virus on HeLa tumor cells); Rukavishnikova et al, (1976) Aa Virol., 20,387 (effect of influenza A strain on ascites tumor).
The earliest references described partial tumor regression in patients treated with live attenuated viral vaccine with the aim to vaccinate them against smallpox or rabies.
See DePace, N. G. (1912) Ginecoloia 9,82-88; Salmon, P. Baix (1922) Comnt Rend. Soc. Biol., 86,819-820. Partial regression of tumors and regression of leukemias have also been COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:03 FAX 013 9603 3099 F.B. RICE Co. 2006 10/03 '05 17:05 FAX 813 9683 3099 F.B. RICE Co. I~iooo 2 noted during naturally occurring measles infections. See Pasquinucci, G. (1971) Lan.i 1, 136; Gross, S. (197 1) Lance%. 1, 397-398; SliWnng, AZ and Ziegler, ItL (197 1) Lanicet. 2, 105-106. In one study of 90 cancer patients intentionally infected with five mumps vinis, partial tumor regresion was noted in 79 cases. SM Asada (1994) Cacr 34,1907-1928.
While the side effects of these viruses were temporary, serious sequelaE of infection with these human pathogens is of major concer.
Viruses are caxc-gouized as follows [see Murphy A and Kingsbury DW, 1990, In.- Virologv. 2 u"d Edition (Ed. Fields, sN.,Raven Press, New York, pp 9-35]: Dividing Chairacteristics Virus Family Names RNA viruses se RNA, positive-sense, Is nonsegmented, nonenveloped, saRNA, positive-sense, nonsegrnentd enveloped, sRNA, negative-sense, nonsegmenced, enveloped, ssRNA, gavesne segmented, enveloped sENA, ambrisense, Segmented, enveloped ds bRNA, positive-scns segmented, nonenvelaped saRNA, DNA step in replication, positivesense, nonsegmned., enveloped Picornaviridae. Calciviridas Togaviridat. Fkrviviridae, Coronoviridac Rhabdovirida, Fitoviridae, Paranyoviiidne Onbhornyxoviride Ewzyaviridae, Areneviridae Reoviridee. Binwirfdae Retroviridae SBMI-01 158839 Received by IP Australia: Time (I-tm) 17:19 Date 2005-03-10 10/03 '05 17:05 FAX 613 9863 3099 F.B. RICE Co. 1007 0 3 c0 DNA viruses ss/daDNA, nonenveloped Hepadnaviridae ssDNA, nonenveloped Parvoviridae dsDNA, nonenveloped Papovaviridae. Adenoviridae dsDNA, enveloped Herpesviridae, Poxviridae, SIrldovirdae O ss single stranded o ds double-stranded itn o Included among the family Herpesviridae (or Herpesviruses), are the subfamilies Alphaherpsvirus (including Genus Varicellavirus and Genus Simpexvirus), Betaherpesvirus, and Gammaherpesvirus.
Newcastle disease virus Is a member of the Paramyxoviridae (or Paramyxoviruses). The natural hosts for NDV are chickens and other birds. NDV typically binds to certain molecules on the surface of animal host cells, fuses with the cell surface, and injects its genetic material into the host. NDV is a cytocidal virus. Once inside the cell, the viral genes direct the host cell to make copies of the virus leading to death of the host cell, releasing the copies of NDV which infect other cells. Unlike some viruses, NDV is not known to cause any serious human disease. Unlike other kinds of viruses 1TLV-1, Hepatitis Paramyxoviruses are not known to be carcinogenic.
Temporary regression of tumors has been reported in a small number of patients exposed to NDV See, Csatary, L.K. (1971) Lancet, 2. 825. Csatary noted the regression of a gastrointestinal cancer in a chicken fanner during an epidemic of Newcastle disease in his chickens. In a similar anecdotal report, Cassel, W.A. and Garrett. R.E. (1965) Cancer, 18, 863-868, noted regression of primary cervical cancer, which had spread to the lymph nodes, in a patient following injection of NDV into the cervical tumor. Since the mechanism of tumoricidal activity was thought to be immunologic, no work was carried out to address direct tumor cytotoxicity of the virus. Instead, efforts focused upon the immuno-modulating effects of NDV. ee, for example. Murray, Cassel, Torbin, A.H, Olkowski, Moore, M.E. (1977) Canr. 40, 680; Casel, Murray, Phillips, H.S.
(1983) Cancer, 52, 856; Bohle, Schlag, PJ., Liebrich, Hohenberger, Manasterski, Miller, and Schirrmacher, V. (1990) Qmer, 66, 1517-1523.
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:05 FAX 613 9663 3099 FB. RICE Co. 0oo8 S4 SThe selection of a specific virus for tumor regression was based on serendipity or trial and eror in the above citations. Only recently, have rational, mechanism-based approaches for Sviia use in cancer treatment been developed using DNA viruses, Examples of this type of o approach are found in the development of recombinant adenoviral vectors that replicate only in tumors of specific tissue origin (Rodriguez, R. et al, 1997 Cancer Res., 57:2559-2563), or those that lack certain key regulatory proteins (Bischoff, JR, et al, 1996 Science, 274:373- 376). Another recent approach has been the use of a replication-incompetent recombinant 0 adenoviral vector to restore a critical protein function lost In some tumor cells (Zhang, WW, Set al, 1994 Cancer ene therapy 1:5-13). Finally, herpes simplex virus has also been S0 engineered to replicate preferentially in the rapidly dividing cells that characterize tumors S(Minet, ct al, 1994 Cancer Res.. 54:3963-3966).
U.S. Application Ser. No. 08/260,536. hereby incorporated by reference in its entirety, discloses the use of NDV or other Paramyxovirus in the treatment of cancer.
Viral IFN transaene expression One common approach to the treatment of cancer with viral therapeutics has been the use of virus vectors for the delivery of certain genes to the tumor mass.
Recombinant adenovirus, adeno-associated virus, vaccinia virus and retroviruses have all been modified to express an Interferon gene alone or in combination with other cycokine 3 genes.
In Zhang et al. ((1996) Proc. Nat. Acad. Se., USA 93:45134518), a recombinant adenovirus expressing a human interferon consensus synthetic) gene was used to treat human breast cancer (and other) xenografts in nude mice. The authors concluded combination of viral oncolysis with a virus of low pathogenicity, itself resistanr to the effects of IFN and IFN gene therapy, might be a fruitful approach to the treatment of a variety of different tumors, in particular breast cancer." In contrast to subject invention which relates to interferon-sensitive viruses, Zhang et al. (1996) teach the use of an interferon-resistant adenovirus in the treatment of tumors.
In Zhang et al. ((1996) Cancer Gene Ther., 331-38), adeno-associated virus (AAV) 3 expressing consensus IFN was used to transduce human tumor cells in vitro followed by injection into nude mice. The transduced tumors either did not form tumors or grew slower than the non-transduced controls. Also, injection of one transduced human tumor cell into the tumor mass of another, non-transduced tumor resulted in a small decrease in size.
COMS ID No: SBMI-01158839 Received by IP Australia; Time 17:19 Date 2005-03-10 10/03 '05 17:06 FAX 613 9663 3099 F.B. RICE Co. a009 SIn Peplinski et al. ((1996) Arm. Sure. Oc., 3:15-23), IFN gamma (and other cytokines, Sexpressed either alone, or in combination) were tested in a mouse breast cancer model. Mice Swere immunized with tumor cells virally modified with recombinant vaccinia virus. When o re-challenged with tumor cells, the mice immunized with virally modified cells had statistical improvement in the disease-free survival time.
Gastl, et al. ((1992) Cancer Res., 52:6229-6236), used IFN gamma-expressing retroviral r vectors to transduce renal carcinoma calls in vitro. These cells were shown to produce higher Samounts of a number of proteins important for the function of the immune system.
SRestifo et al. ((1992) Exp. fed. 175:1423-1431), used IFN gamma-expressing retroviral vector to transduce a munrine sarcoma cell line allowing the tumor cell line to more 0efficiently present viral antigens to CDS8 T cells.
Howard, et al. ((1994) Ann. NY Acad Sci., 716:167-187), used IFN gamma-expressing retrovira] vector to transduce murine and human melanoma tumor cells. These cells were observed to increase the expression of proteins important to immune function. These cells were also less tumorigenic in mice as compared to the non-transduced parent line, and resulted in activation of a tumor-specific CTL response in vivo.
Use of Therapeutic Doses of Interferon as an Adjuvnt to Viral Cancer Theray Because of the known immune-enhancing properties of lN, several studies have examined the use of IFN protein in combination with other viral cancer vaccine therapies.
In Kirchner et al. ((1995) World J. T ol., 13:171-173), 208 patients were immunized with autologous, NDV-modified, and lethally irradiated renal-cell carcinoma tumor cells, and were co-treated with low dose IL-2 or IFN alpha. The authors stated that this treatment regime results in an improvement over the natural course in patients with locally-advanced renal-cell carcinoma. The dose was approximately 3.3 x 10i to 2.2 x 10 PFU/kg. This was a local therapy, as opposed to a systemic approach, with the goal of inducing an anti-tumor immune response.
Tanaka et al. ((1994) J. Immunother. Emphasis Tumor Immunol. 16:283-293), coadministered IFN alpha with a recombinant vaccinia virus as a cancer vaccine therapy model in mice. This study showed a statistical improvement in survivability in mice receiving IFN as compared to those that did not. The authors attributed efficacy of IFN to the induction of CDS-positive T cells in those animals.
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 00 6
O
O
Arroyo et al. ((1990) Cancer Immunol. Immunother., 31:305-311) used a mouse model Sof colon cancer to test the effect of IFN alpha and/or IL-2 co-therapy on the efficacy of a vaccinia virus colon oncolysate (VCO) cancer treatment. They found that the triple O treatment of VCO+IL-2+IFN was most efficacious in this murine model. This approach relies on immunization as the mechanism of anti-tumor activity.
SIFN was used in these studies to augment the ability of the cancer cells to be Srecognized by the immune system.
SIn a preferred embodiment there is provided viruses for the treatment of diseases including cancer.
C, In a further preferred embodiment there is provided viruses for the treatment of neoplastic diseases including cancer.
In a further preferred embodiment there is provided a means by which candidate viruses are selected and/or screened for use in the therapy of neoplastic diseases.
In a further preferred embodiment there is provided guidance in the genetic engineering of viruses in order to enhance their therapeutic utility in the treatment of neoplastic diseases.
In a further preferred embodiment there is provided a means with which to screen potential target cells for viral therapy with the goal of assessing the sensitivity of the candidate target cells to viral killing.
In a further preferred embodiment there is provided guidance in the management of viral therapy.
In a further preferred embodiment there is provided a method for treating large tumors.
In a further preferred embodiment there is provided purified virus and methods for obtaining same.
Summary of the Invention This invention relates to a method of infecting a neoplasm in a mammal with a virus comprising administering an interferon-sensitive, replication-competent clonal virus, selected from the group consisting of RNA viruses and the DNA virus families of Adenovirus, Parvovirus, Papovavirus, Iridovirus, and Herpesvirus, to the mammal.
10/03 '05 17:06 FAX 613 9663 3099 F.B. RICE Co. 0i1 7 o This invention also relates to a method of infecting a neoplasm in a mammal with a virus comprising systemically administering an interferon-sensitive, replication-competent clonal Svirus to the mammal.
This invention also relates to a method of treating a neoplasm including cancer in a mammal comprising administering to the mammal a therapeutically effective amount of an interferon-sensitive, replication-competent, clonal virus selected from the group consisting of 0 RNA viruses, and the DNA virus families of Adenovirus, Parvovirus, Papovavirus, o Iridovirus, and Herpesvirus.
SThis invention also relates to a method of infecting a neoplasm in a mammal with a virus a comprising administering an interferon-sensitive, replication-competem clonal vaccinia virus, Shaving one or more mutations in one or more viral genes involved with blocking intferfon's antiviral activity selected from the group of genes consisting of K3L, E3L and B 18R, to the mammal.
The invention also relates to a method of treating a neoplasm including cancer in a mammal administering to the mammal a therapeutically effective amount of an interferonsensitive, replication-competent vaccinia virus having one or more mutations in one or more viral genes involved with blocking interferon's antiviral activity selected from the group of genes consisting of K3L, E3L and B I R.
The invention also relates to a method of infecting a neoplasm at least 1 cm in size with a 0 virus in a mammal comprising administering a clonal virus, selected from the group consisting of RNA viruses; Hepadenavirus; Parvovirus; Papovavirus; Herpesvirus; Poxvirus; and Iridovirus, to the mammal.
The invention also relates to a method of treating a neoplasm in a mammal, comprising administering to the mammal a therapeutically effective amount of a clonal virus, selected from the group consisting of RNA viruses; Hepadenavirus; Parvovirus; (4) Papovavirus; Herpesvirus; Poxvirus; and Iridovirus, whirein the neoplasm is at least 1 centimeter in size.
The invention also relates to a method of treating a tumor in a mammal, comprising administering to the mammal a therapeutically effective amount of an RNA virus cytocidal to 0 the tumor, wherein the mammal has a tumor burden comprising at least 1.5% of the total body weight.
The invention also relates to a method of screening tumor cells or tissue freshly removed from the patient to determine the sensitivity of the cells or tissue to killing by a virus COMS ID No: S8MI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:07 FAX 613 9683 3099 F.B. RICE Co. 012 0 8 0 comprising subjecting the cells or tissue to a differential cyrotxicity assay using an S interferon-sensitive virus.
The invention also relates to a method for identifying a virus with antineoplastic activity O in a mammal comprising a) using the test virus to infect i) cells deficient in LFN-mediated antiviral activity, and ii) cells competent in IFN-mediated antiviral activity, and b) determining whether the test virus kills the cells deficient in IFN-mediated antiviral activity preferentially to the cells competent in interferon-mediated antiviral activity.
o The invention also relates to a method of making viruses for use in antineoplastic therapy O comprising: a) modifying an existing vims by diminishing or ablating a viral mechanism for in 0 the inactivation of the antiviral effects of FN, and optionally b) creating an attenuating o mutation that results in lower virulence than said existing virus.
The invention also relates to a method of controlling viral replication in a mammal treated with a virus selected from the group consisting of RNA viruses, Adenoviruses, Poxviruses, Iridoviruses, Parvoviruses, Hepadnaviruses, Varicellaviruses, Betaherpesviruses.
and Gammaherpesviruses comprising administering an antiviral compound.
The invention also relates to a method of screening tumor cells, tumor tissue, or tissue sections to determine which tumor cells or tissue allow a virus to bind comprising subjecting the cells, tissues, or tissue sections to an immunoassay or immunostain for the amount of virus receptor present on the tumor cells or tumor tissue.
:0 The invention also relates to a method of Infecting a neoplasm in a mammal with a virus comprising systemically administering a desensitizing dose of an inerferon-sensitive, replication-competent clonal virus to the mammal before administering at least one subsequent higher dose of a virus.
The invention also relates to a method of infecting a neoplasm in a mammal with a virus comprising administering an interferon-sensitive, replication-competent clonal virus to the mammal over a course of at least 4 minutes.
This invention also relates to a method of infecting a neoplasm in a mammal with a virus comprising administering a replication-competent clonal virus selected from the group consisting of the Newcastle disease virus strain MKI07, Newcastle disease virus strain NJ o Roakin, Sindbis virus, and Vesicular stomatids virus.
Included in the invention are: i) a clonal Paramyxovirus purified by ultracentrifugation without pelleting; ii) a clonal Paramyxovirus purified td a level of at least 2 x 109 PFU per mg of protein; COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:07 FAX 613 9663 3099 F.B. RICE Co. 1A013 9 O iii) a clonal Paramyxovirs purified to a level of at least I x I 0 PFU per mg of N protein; protein; S 5 v) a clonal RNA virus purified to a level of at least 2 x 10 PFU per mg ofprotein; vi) a clonal RNA virus purified to a level of at least 1 x 10' 0 PFU per mg of protein: N vii) a clonal RNA virus purified to a level of at least 6 x 1010 PFU per mg of protein; 0 viii) a clonal cytocidal DNA virus which is interferon-sensitive and purified to a level Sof at least 2 x 10 PFU/mg protein; t 0 ix) a replication-competent vaccinia virus having a) one or more mutations in one or o more of the K3L, E3L and B18R genes, and b) an attenuating mutation in one or more of the genes encoding thymidine kinase, ribonucleotide reductase. vaccinia growth factor, thymidylate kinase, DNA ligase, dUTPase; x) a rcplication-competent vaccinia virus having one or more mutations in two or more genes selected from the group consisting of K3L, E3L, and B18R; xi) a Herpesvirus having a modification in the expression of the analog causing the Herpesvirus to have increased interferon sensitivity; and xii) a Reovirus having an attenuating mutation at omega 3 causing said virus to become interferon-sensitive.
a xiii) a replication competent cytocidal virus which is interferon sensitive and purified to a level of at least 2 x 10 9 PFU/mg protein.
xiv) a reovirus purified to a level of at least 2 x 10' PFU/mg protein.
Also included in the invention are the following methods; i) a method of purifying an RNA virus comprising the steps of a) generating a clonal virus; and b) purifying said clonal virus by ultracentrifugation without pelleting; or c) purifying said clonal virus by tangential flow filtration with or without subsequent gel perneation chromatography, and ii) a method of purifying a Paramyxovirus comprising purifying the virus by ulracentrifugation without pelleting, or by tangential flow filtration with or without 0 subsequent gel permeation chromatography.
The invention also relates to a method of treating a disease in a mammal, in which the diseased cells have defects in an interferon-mediated antiviral response, comprising COMS ID No; SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 00 administering to the mammal a therapeutically effective amount of an interferonsensitive, replication-competent, clonal virus.
The invention also relates to a method of infecting a neoplasm in a mammal with a virus comprising administering an interferon-responsive, replication-competent clonal RNA virus to the mammal.
The invention a method of treating a disease of viral etiology in a mammal comprising administering to the mammal a therapeutically effective amount of a replication-competent clonal virus.
I The invention also relates to a method of infecting a neoplasm in a mammal comprising administering a virus selected from the group of families consisting of C Paramyxoviridae, Orthomyxoviridae, Rhabdoviridae, Togaviridae, Flaviviridae, Picornaviridae, Coronaviridae, Reoviridae, Poxviridae, Herpesviridae, and Parvoviridae.
The invention also relates to a method of treating tumor ascites comprising administering an interferon-sensitive, replication-competent clonal virus.
The invention also relates to a method of reducing pain in a mammal comprising administering an interferon-sensitive, replication competent clonal virus.
The invention also relates to a method of treating a neoplasm in a mammal comprising subjecting a sample from said mammal to an immunoassay to detect the amount of virus receptor present, and if the receptor is present, administering an interferon-sensitive, replication competent clonal virus, which bind the receptor, to the mammal.
According to the invention there is also provided a method of infecting a neoplasm in a mammal comprising administering an interferon-sensitive, replicationcompetent clonal RNA virus, wherein said virus is to be administered over a period of at least 20 minutes.
According to the invention there is also provided use of an interferon-sensitive, replication-competent clonal RNA virus in the manufacture of a medicament for infecting a neoplasm in a mammal, wherein said virus is to be administered over a period of at least 20 minutes.
Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps.
Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a 00 IUA context for the present invention. It is not to be taken as an admission that any or all of Sthese matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application.
Brief Description of the Drawings Figure 1 shows the effect of anti-interferon-beta antibody on viral antigen expression and infectious titer in NHEK (normal human epithelial keratinocytes) cells.
Figure 2 shows the effect of interferon-beta on viral antigen expression in different cells (normal human skin fibroblasts CCD922-sk and two types of head and ,IC neck carcinoma cells (KB and Hep2 cells).
Figure 3A shows the effect of interferon on viral antigen expression in CCD922-sk cells, and Figure 3B shows the effect of interferon on viral antigen expression in KB cells.
Figure 4 shows the survival curves for athymic mice bearing human ovarian ES-2 tumors and treated with either saline or NDV.
Figure 5 shows the interferon responsiveness of a number of human tumor and normal cell lines.
10/03 '05 17:07 FAX 613 9663 3099 F.B. RICE Co. o015 o
H
O Figure 6 shows the dose response of the SW620 tumor cell line to infection with PPMK107 and PPSINDBIS-Ar339.
Detailed Description of the Invention The present invention relates to the discovery of a novel mechanism by which viral replication selectively kills neoplastic cells deficient in an interferon (IFN)-mediated antiviral response. This invention also provides methods for selection, design, purification, and C use of viruses for the treatment of neoplastic diseases including cancer and large tumors. The O viruses of the invention selectively replicate in and kill neoplastic cells based on the selective ct 0 deficiency in these cells of an ]FN-mediated ani-viral response. Administration of the o appropriate dosage of virus results in neoplastic cell death, whereas normal cells, which possess an intact IFN-mediared anti-viral response, timit the replication of the virus and are not killed. Included in the subject of the invention is the use of paramyxoviruses such as NDV, and other viruses, for use in the treatment of diseases including neoplastic disease such as cancer, The invention also teaches screening and engineering of other viruses suitable for use as therapeutics of neoplastic diseases. Another embodiment of the invention involves a method of identifying tumor tissues that are candidates for viral therapy. Finally, the invention also describes the preparation of highly purified virus.
0 Rationale for the use of interferon-sesitive viruses including NDV to treat neoplastic disease NDV demonstrates selective killing of tumor cells.
Newcastle disease virus causes selective cytotoxic effects against many human tumor cells with markedly less effects en most normal human cells. In a differential cytoroxiciy assay, human cancer cells derived from renal carcinomas, pancreatic carcinoma, sarcomas, melanomas, breast carcinomas, ovarian carcinomas, bladder carcinomas, colon carcinoma, prostate carcinoma, small cell and non-small cell lung carcinomas, and glioblastomas were discovered to be approximately 3 to 4 orders of magnitude more sensitive to NDV than many normal human cells (renal epithelial cells, fibroblasts, keratinocytes, melanocytes, and 0 endothelial cells (see Example The differential cytotoxicity assay can also be applied to fresh isolates from the patient's cells or tumor tissue.
An in vitro assay is used to define the tumoricidal activity of NDV as described in Example 1, The assay measures the amount of virus required to kill 50% of the tested cell COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:08 FAX 613 9663 3099 F.B. RICE Co. Bao6 Sn 12 A Uw JWW*WA o culture in a five day time period. Examples 2 and 3 show the results of in vivo experiments in which virus was administered to athymic mice bearing human tumor xenografts by either t the intratumoral (Example 2) or intravenous (Example 3) mute. These results demonstrate Sthat NDV can cause regression of a variety of human tumor types in a standard animal model 0 for the testing of potential chemotherapeudc agents.
Evidence that NDV is specifically replicating within the tumor was demonstrated by immnuiohistochemical staining for virus antigen (Example Within 30 minutes of S intratumoral virus injection, the tumor tissue was negative for viral antigen. However, by day S 2 post treatment, intense immunostaining for viral antigen was seen within the tumor, C' indicating virus replication within the tumor. Importantly, virus replication was specific for o the tumor tssue since the neighboring connective tissue and skin were negative for viral Cl antigen.
Importantly, efficient replication of NDV is crucial for the ability of the virus to kill infected cells, as demonsrated in studies using UV-inacdvated non-clonal virus (Lorence, R., at al, 1994, J. Natl. Cancer Inst.. 86:1228-1233).
NDV can also cause regression of large tumors after intratumoral and intravenous administration (Examples 4 through Intratumoral NDV treatment of large intradermal A375 human melanoma xenografts (>10 mm in maximal dimension: tumor volume of >300 mm 3 in athymic mice lead to high rates of tumor regression (Examples 4 through 8).
Intravenous NDV treatment of large subcutaneous HTI080 human flbrosarcoma xenografts (>IO mm in maximal dimension) in athymic mice lead to complete or partial tumor regression in five out of six mice (Example 9).
The class I interferon family of cytokines are important negative modulators of viral S infection.
The class I interferons consist of the IFNa, found primarily in ells of hematopoietic origin, and IFND found primarily in fibroblasts and epithelial cells. (Joklik, W.K. 1990.
Interferons. pp 383-410. VYroloy, second edition, edited by BN. Fields, D.M. Knipe et al, Raven Press Ld., New York; and Sreevalsan, T. 1995. Biological Therapy with Interferon-a and f: Preclinical Studies. pp 347-364. Biologic Therapy of Cancer, second edition, edited by V.T. DeVita, Jr., S. Hellman, and S.A. Rosenberg, J.B. Uppincott Company, Philadelphia.] Both types of IFN function through an apparendy common mechanism of action that includes the degradation of double-stranded RNA intermediates of viral COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:08 FAX 813 9663 3099 F.B. RICE Co. o017 t 13 o replication, and the inhibition of cellular translation through the activity of a protein kins activatedby double-stranded RNA (Joklik, 1990. Interferons. pp 383410. Virology.
c Second Edition, edited by B.N. Fields, P.M. Knipe et al., Raven Press Ltd., New York; and Sreferences therein). Several viruses (influenza, EBV. SV40, adenovirus, vaccinia) have 0 5 evolved mechanisms by which one or more pathways of the IFN system are inactivated, thus allowing the efficient replication of the virus (Katze, M.G. 1995. Trends in Microbiol. 3:75- 78).
A wide variety of tumor cells are deficient In the ability to limit viral infection through C lo an IFN-dependent mechanism.
SHuman cervical carcinoma cells (HeLa) were over three-hundred-fold less sensitive to the C' inhibition of vesicular stomatitis virus replication following pre-reatmnent with IFN than a non-transformed fibroblast control cell line (Maheshwari 1983. Biochem, Biophys.
Res. Comm, 17:161-168). The subject Inventors have discovered that infection of a coculture of tumorigenic human head and neck carcinoma cells (KB) and normal human skin fibroblast cells (CCD922-sk) results in viral replication initially in both cell types, followed by a limiting of the infection in the normal cells versus continued replication and killing of the tumor cells (Example 10). Moreover, although IFN was being secreted by the normal cells into the culture medium, the tumor cells were unable to respond to the IFN at the !0 concentrations being produced to establish an antiviral state. Further evidence for the role of IFN in the differential sensitivity of tumor cells versus normal cells to killing by NDV was obtained in two separate experiments in which normal fibroblast cells (CCD922-sk) or normal epithelial keratinocyte cells (NHEK) were shown to become more sensitive to infection with NDV In the presence of neutralizing antibody to IFN (Examples 11 and 12).
Finally, parallel infection of normal fibroblasts (CCD922-sk) and human tumor cells (KB) in the presence of IFN revealed that the normal cells were at least 100-fold more sensitive to the antiviral effects of added IFN than were the tumor cells (Examples 13 and 14). Similar testing of variety tumor cell lines (total of 9) revealed a clear correlation in the relative sensitivity of a cell line to killing by NDV and an inability of the cell line to manifest an io interferon-mediated antiviral response (Example 26).
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:08 FAX 613 9863 3099 F.B. RICE Co. os18 14 o Inrferon and Cell Growth c There are several species of interferon (IFN) including natural and recombinant forms of Sat-IFN, P-IFN, r-IFN, and y-IFN as well as synthetic consensus forms as described in Zhang et al. (1996) Cancer Gene Therapy, 3:31-38). In addition to the anti-viral activities that lead to its discovery, IFN is now known to play an important role in the normal regulation of cell growth and differentiation. IFN is viewed as a negative growth regulator and several key proteins involved in the function and regulation of IFN activity have been o shown to act as tumor-suppresser proteins in normal cells (Tanaka et al, 1994 Cell 77:829- S839). Moreover, several other proteins known to antagonize the anti-viral activity of IFN 0 have been shown to have oncogenic potential when expressed inappropriately (see below, SBarber, ON, 1994, Proc.Natl. Acad. Sci. USA 91:4278-4282). Cells derived from a number C1 of human cancers have been shown to be deleted in the genes encoding IFN (amnes, CD, et al, 1991, Cancer Res.. 51:1684-1688), and partial or complete loss of IFN function has been observed in human cervical carcinoma (Petricoin, E, et al, 1994 Mol. Cell. Bio.. 14:1477- 1486), chronic lymphocytic leukemia (Xu, et al, 1994, Blod, 84:1942-1949), and malignant melanoma cells, (Linge, etal, 1995, Cancer Res, 55:4099-4104).
The IFN-inducible protein kinase (p68, PKR) has been shown to be an important regulator of cellular and viral protein synthesis. A correlation has emerged that links the expression or activity of the p68 kinase to the cellular state of differentiation. Thus, poorly 0 differentiated cells, such as those occurring in many cancers, are deficient in p68 function (Haines, ct al, 1993 Virchows Anh B Cell Pathol. 63:289-95). Cells that lack p68 activity are generally sensitive to viral mediated killing because the p 6 8 kinase is an important effector of the IFN-inducible antiviral state. The antiviral activity of p68 can be antagonized through a direct interaction with a cellular protein identified as p58. When cloned and overexpressed in N1H3T3 cells, p58 causes the cells to exhibit a transformed phenotype and anchorage-indepndent growth (Barber GN et al., 1994 Proc Nat] Acad Sci USA 91:4278-4282), and a number of human leukemia cell lines have been shown to overexpress the p58 protein (Korth MJ, at al., 1996 Gene 170:181-188). The activity of p68 kinase can also be antagonized by the Ras protein. Cells that express mutant, activated forms D of Ras have been shown to be defective in the activation of p68 kinase by double-stranded RNA (Mundshau, L.J, and Faller, 1992, J. Biol, Chem., 267:23092-23098).
Sensitivity to viral killing in undifferentiated cells can be reversed through the induction of a COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:09 FAX 613 0663 3099 F.B. RICE Co, S0oi o O more differentiated phenotype (Kalvakolanu, DYR and Sen. G.C. 1993 Proc Narl Acad Si USA 90:3167-3171).
Definitions Ceh l omoetent in an intrferon-medLated antiviral response As used herein, the term "cells competent in an interferon-mediated antiviral response" are cells which respond to low 0 levels 10 units per ml) of exogenous interferon by significantly reducing (at least o fold, more advantageously at least 100-fold, more advantageously at least 1000-fold, and o most advantageously at least,10,000-fold) the replication of an interferon-sensitivc virus as 0 compared to in the absence of interferon. The degree of virus replication is determined by Smeasuring the amount of virus infectious virus, viral antigen, viral nucleic acid).
C CCD922 normal fibroblasts are cells competent in an interferon-mediated antiviral response.
Cells degfcient in an interferon-mediated antiviral response, As used herein, the term "cels deficient in an interferon-mediated antiviral response" are cells which fail to meet the criteria listed above for a cell competent in an interferon-mediated antiviral response, that is, they fail to respond to low levels 10 units per ml) of exogenous interferon by significantly reducing the replication of an interferon-sensitive virus as compared to in the absence of interferon. KB oral carcinoma cells are cells deficient in an interferon-mediated 0 antiviral response, Clonal. Use of the term "clonal" virus is defined hereafter as virus derived from a single infectious virus particle and for which individual molecular clones have significant nucleic acid sequence homology. For example, the sequence homology is such that at least eight S individual molecular clones from the population of virions have sequence homology greater than 95%, more advantageously greater than 97%, more advantageously greater than 99%, and most advantageously 100% over 300 contiguous nucleotides.
Cstoedal. As used herein, the term "cytocidal" virus refers to a virus that infects cells ,o resulting in their death.
Desensitization. As used herein, the phrase desensitizarion refers to pretreatment with an agent that lessens the side effects caused by virus administration.
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:09 FAX 613 9663 3099 F.B. RICE Co. [020 16 0 Cl Desensitizing pose. As used herein, the phrase "desensitizing dose" refers to the amount of c virus required to lessen the side effects of subsequent doses of virus.
Differential Cvtotoxicitv Assay. As used herein, the phrase "differential cytocoxicity assay" for screening tumor cells or tissue using a virus refers to the virus infection of the tumor cells and one or more control cells or tissue; a determination of cell survivability or o death for each sample (for example, by the use of a dye indicator of cell viability as in detailed in Example 1) after one or more days of infection; and based on the results, an estimation of the sensitivity (for example, by IC50 determination as detailed in Example 1) of O the sample to the virus compared to the control(s).
InfLcting a Neoplasm. As used herein, the term "infecting a neoplasm" refers to the entry of viral nucleic acid into the neoplastic cells or tissues.
Interferon-sensitive. As used herein, the phrase "interferon-sensitive" virus NDV) means a virus that replicates significantly less (at least 10-fold less, advantageously at least 100-fold less, more advantageously at least 1000-fold less, and most advantageously at lease 10,000-fold less), in the presence of interferon compared to in the absence of interferon. This o0 is determined by measuring the amount of virus infectious virus, viral antigen, viral nucleic acid) obtained from cells competent in an interferon-mediated antiviral response in the presence or absence of low levels of exogenous interferon 10 units per ml).
Interferon-responsive. As used herein, the phrase "interferon-responsive" virus NDV) refers to a virus that following an infection at an moi (multiplicity of infection) of 1.0 at least less viral antigen is expressed in cells pretreated for 24 hours, and maintained with 500 Units/ml of the exogenous interferon-o, than in untreated cells. The measurements are made in cells competent in an interferon-mediated antiviral response at least 24 hours postinfection, and on the first day that would allow a determination of a 50% decrease in viral antigen expression.
Neoplasm and Neopuastic Disease. As used herein, "neoplasm" means new growth of tissue, including tumors, benign growths condylomas, papillomas) and malignant COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:09 FAX 613 9603 3099 F,B. RICE Co. l021L 17 O growths cancer). As used herein, "neoplastic disease" refers to disease manifested by the presence of a neoplasm.
Replication Competent. As used herein, the term "replicaton-competent" virs refers to a virus that produces infectious progeny in neoplastic cells.
Sabstantiallv Free of Contaminating E Proteins. The term "substantially free of Scon tminnating egg proteins" refers to a level of virus purity in which ovalburin is not 0 detectable in a Western blot as performed by one skilled in the ar by using 1.7 x 10 PFU i 0 of virus per well (3.3 cm in width) run on an SDS-PAGE (sodium dodecyl sulfate- Spolyacrylamidc gel electrophoresis) gel (1 mm thick); transferring the viral proteins from Cl the gel to a nitrocellulose membrane; and immunostaining for ovalbumin with the use of a rabbit anti-ovalbumin [Rabbit IgG fraction at a 1:200 dilution of a 4 mg/ml antibody concentration (from Cappel, Inc.) or equivalent polyclonal antibody] and. more advantageously, not detectable in an electrochemiluminescence assay with a sensitivity of 2.4 ng/ml.
Therapeuticallv effective amount As used herein, the term "therapeutically effective amount" when referring to the treatment of neoplastic disease refers to a quantity of virus 0 which produces the desired effect, cessation of neoplastic growth, tumor regression, improved clinical conditions, or increased survival.
Compounds of the Invention A diverse group of viruses are used to selectively kill neoplastic cells. Natural or engineered viruses can function as an antineoplastic agent. These viruses i) infect neoplastic cells resulting in their death; ii) are replication-competent in the neoplastic cells; and iii) are limited in killing of normal cells by the antiviral effects of interferon.
In an advantageous embodiment of the invention, the viruses possessing the above three characteristics they infect neoplastic cells resulting in their death; (ii) they are replication- 0 competent in the neoplastic cells; and (iii) they are limited in killing of normal cells by the antiviral effects of intrerfeon also induce interferon.
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:10 FAX 613 9663 3099 F,B. RICE Co.
R022 is O In another advantageous embodiment of the invention, the viruses possessing the above three characteristics also cause regression of human neoplasms; and/or are not neutralized in t the target human population because of the presence of pre-existing immunity.
In another advantageous embodiment, the viruses possessing the above three characteristics are cytocidal to tumor cells.
A Paramyxovirus (as used herein "Paramyxovirus" refers to a member of the Paramyxoviridae) can be used according to the present invention to treat a neoplasm Sincluding a large tumor or a host having a high tumor burden, The Paramyxoviridae family comprises three genera: paramyxoviruses; measles-like viruses (morbilli viruses); and 0 respiratory syncytial viruses (pneumoviruses). These viruses contain an RNA genome.
o Use of Paramyxoviridae viruses which are cytocidal, especially paramyxoviruses, e.g., C Newcastle disease virus and other avian paramyxoviruses such as avian paramyxovirus type 2, is an advantageous method of practicing the invention. Attenuated strains of these viruses are especially useful for treatment of neoplasms in accordance with the present invention.
NDV is an especially advantageous virus according to the present invention. NDV is categorized into three distinct classes according to its effects on chickens and chicken embryos. "Low virulence" strains are referred to as lentogenic and take 90 to 150 hours to kill chicken embryos at the minimum lethal dose (MLD): "moderate virulence" strains ar O referred to as mesogenic and take 60 to 90 hours to kill chicken embryos at the MLD; "high virulence" strains are referred to as velogenic and take 40 to 60 hours to kill chicken embryos at the MLD. See, Hanson and Brandly, 1955 (Science, 122:156-157), and Dardiri ct al., 1961 (Am. J. Vet. Res.. 918-920). All three classes ar useful, advantageously, mesogenic strains of NDV such as strain MKI07, strain NJ Roakin, and strain Connecticut-70726. (see Examples 21-23). See, Schloer and Hanson,1968. Virol., 2:40-47) for a listing of other mesogeic strains.
For certain purposes, it is desirable to obtain a clonal virus to ensure or increase the genetc homogeneity of a particular virus strain and to remove defective interfering particles.
Removal of defective interfering particles by cloning allows for increased purity in the final 0 product as assessed by the number of total virus panicles per infectious particle the number of particles per PFU).
Clonal virus can be produced according to any method available to the skilled worker.
For example, plaque purification is routinely utilized to obtain clonal virus. See, e.g., COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:10 FAX 613 9663 3099 F,B. RICE Co. [023 19 o Maassab et al., In: Plotkin and Mortimer, eds. Vaccines. Philadelphia: W.B. Saunders Co., 1994, pages 781-801. Triple plaque purification is especially desirable, where a plaque is t selected at each round of purification having the desired characteristics, such as a preferred Ssize, shape, appearance, or representative of the parental strain. Another means of generating 0 clonal virus is by recombinant DNA techniques applicable by one skilled in the art. Another means of obtaining a clonal virus applies the technique of limiting dilution by adding dilutions of the virus sample to give an average of one or less infectious virus particles per well containing a monolayer of a susceptible cell).
In an advantageous embodiment of the invention, purified virus is used to treat neoplastic diseases. An advantageous method for purification of egg derived viruses are as follows O (virus is not pelleted at any step in these methods): Ci Purification Method A a) generating a clonal virus plaque purification) b) inoculating eggs with the clonal virus c) incubating the eggs d) chilling the eggs e) harvesting the allantoic fluid from the eggs f) removing cell debris from the allantoic fluid h) ultracentrifugation of the allantoic fluid without pelleting using a discontinuous sucrose gradient) In another embodiment of the invention, additional steps, added after the removal of the cell debris (from the allantoic fluid) and before ultracentrifugarion, consist of: freezing then thawing the allantoic fluid removing contaminating material from the virus suspension by means of centrifugaion) In another embodiment of the invention, ultracentrifugation is accomplished by means of a continuous flow ultracentrifuge.
One embodiment of the invention relates to a method of purifying a replicationcompetent RNA virus comprising the steps of: a) generating a clonal virus, and b) purifying said clonal virus by ultracentrifugation without pellering, Another embodiment of the invention involves a method of purifying a pararnyxovirus NDV) comprising purifying the virus by ultracentrifugation COMS ID No: SBMI-01158839 Received by IP Australia: Time (H:rn) 17:19 Date 2005-03-10 10/03 '05 17:10 FAX 613 9883 3090 F.B. RICE Co.
1024 0 without pelleting. Optionally, the purifying step additionally comprises prior to the ultracentrifugation: a)plaque purifying to generate a clonal virus, b)inociilating eggs with the clonal virus, c)incubating the eggs, d)chilling the eggs, 0 c)harveing allantoic fluid from the eggs and, Sf)removing cell debris from the allantoic fluid.
SAnother embodiment of the invention involves a method of purifying a i replication-competent clonal virus from eggs or cell culture comprising the step of o ultracentrifugation without a step in which the virus is pelleted (see Example 31).
Cl Another embodiment of the invention involves a method of the purifying a paramyxovirus NDV) comprising purifying the virus by sequential tangential flow filtration Optionally, the virus can be additionally purified by gel permeation chromatography, where each of these steps occurs in the presence of a stabilizing buffer (Example a) plaque purifying to generate a clonal virus, b)inoculating eggs with the clonal virus, c)incubaing the eggs, d)chilling the eggs, e)harvesting allantoic fluid from the eggs and dilution of allantoic fluid with buffer, f)removing cell debris from the allantoic fluid by TFF g)purification of the virus by TFF b)purification of the virus by gel permeation chromatography Optionally, the virus obtained from the gel permeation step can be concentrated using
TFF.
Another embodiment of the invention involves a method of purifying a replicationcompetent clonal virus from eggs or cell culture comprising the step purifying the virus by sequential tangential flow filtration (TFF), and optionally followed by gel permeation chromatography, which can optionally be followed by TFP to concentrate the virus.
COMS ID No: SBMI-01158839 Received by IP Australia; Time 17:19 Date 2005-03-10 10/03 '05 17:10 F_.AP 613 9663 3099 F.B. RICE Co. 1025 0 21 o Clonal virus Use of these methods permits purification of a clonal virus [including Paramyxovirus NDV)] to at least 2 x 10' PFU/mg protein, advantageously to at least 3 x P FFU/mg protein, more advantageously to at least 5 x 10 9 PFU/mg protein, more advantageously to at least 1.0 x 10' PFU/mg protein, more advantageously to at least x 1010 PFU/mg protein, more advantageously to at least 3 x 1010 PFU/mg protein, more advantageously to at least 4 x 10' PFU/mg protein, more advantageously to at least 5 x PFU/mg protein, and most advantageously at least 6 x 10 PFU/mg.
C Use of these methods permits purification of a clonal virus [including Paramyxovirus S 10 NDV)] to level in which the number of virus particles per PFU is less than 10, more C advantageously less than 5, mor advantageously less than 3, more advantageously less than 2, and most advantageously less than 1.2. (Lower numbers of virus particles per PFU indicate a higher degree of purity.) RNA viruses In another embodiment, these methods permit purification (to the levels cited above for clonal viruses) of an RNA-virus [including a cyocidal RNA virus; a singlestranded RNA non-segmented, nonenveloped virus; a single-stranded RNA segmented, enveloped virus; a double-stranded RNA segmented, nonenveloped virus; ,o and a single-stranded RNA non-segmented, enveloped virus Paramyxovirus NDV) and Retoviruses].
DNA viruses In another embodiment, these methods permit purification (to the levels cited above for clonal viruses) of an interferon-sensi tive cytocidal virus selected from the group consisting of enveloped, double-stranded DNA viruses (including poxviruses); (b) nonenveloped, single-stranded DNA viruses; and nonenveloped, double-stranded DNA viruses.
Ega derived viruses In another embodiment, these methods permit purification of egg derived viruses to a level substantially free of contaminating egg proteins. It is prefared to limit the amount COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:11 FAX 613 963 3099 F.B. RICE .9.
O 22 o of egg proteins in virus preparations for human therapeutic use since major egg proteins like ovalbumin are allergens.
Viruses useful in the treatment of neoplastic diseases including cancer are shown in Table S 1. Additional examples of virus family members can be found in "Murphy A and Kingbury DW, 1990, In: Viroloy, 2" Edition (Ed. Fields, Raven Press, New York" hereby incorporated in its entirety These viruses are optionally screened for naturally occurring o variations (certain strains or isolates) that result in altered IFN production relative to the Sparental strain.
In .0 In another embodiment of this invention, candidate viruses, whether naturally occurring o or engineered, are tested for the ability to provide therapeutic utility in the treatment of neoplasms. In one embodiment, the amount of candidate vims required to kill 50% of cells deficient in an interferon-mediated antiviral response, KB head and neck carcinoma cells, is compared to the amount of virus required to kill 30% of a similar number of cells competent in an interferon-mediated antiviral response, for example normal skin fibroblasts.
The amount of killing is quantified by any number of means including trypan blue exclusion or MTT assay (see Example A significant reduction at least 5-fold) in the amount of virus required to kill cells deficient in an interferon-mediated antiviral response relative to the amount needed to kill cells competent in an interferon-mediated antiviral response indicates 0t that the virus being tested exhibits activity required for therapeutic utility in the treatment of neoplasms. Other NDV viruses and Sindbis virus are such natural occurring viruses that display tumor-selective killing (see Examples 21-23, and COMS ID No: SBMI-01158839 Received by IPAustralia: Time 17:19 Date 2005-03-10 10/03 '05 17:11 FAX 813 9063 3099 F.B RICE Co. [a 027 23 Table I Naturally Occurring Viruses for UBO in Cancer Therapy Virus Clan Virus Family Virus Examle RNA. negative stranded Paramyxoviridae Newcastle Disease Virus Avian Paramyxovirus Type 2 Measles Mumps Human Parainflucnza Orthomyxoviridae Influenza Virus Rhabdoviridac Vesicular Stomaritis Virus RNA. positive stranded Togaviridac Sindbis Virus Semliki Forest Virus Flaviviridae Yellow fever (attenuated) Piornaviridat Rhinovina Bovine enterovirus Coronaviridac Avian infectious bronchitis virus Human coronavinzses RNA, double stranded Reoviridac Reovius Ratavirus DNA Poxviridac Vacciua Virus Herpesviddae Herpes Simplex Virus, zype I COMS ID No: SBMI-01158839 Received by IF Ausralia: Time 17:19 Date 2005-03-10 10/03 '05 17:11 FAI 613 9663 3099 F.B. RICE Con.
o028 9F.B. RICE Co o 24 o An understanding of the factors involved in the establishment of an antiviral state allows for the creation of a screening assay for tumors that are likely to respond to viral therapy. In principle, patient derived tumor tissue obtained from biopsy is screened for the expression of Sp68 kinase, p58, or other factors involved in the regulation of an antiviral state or cellular differentiation. Other factors include, but arc not limited to, interferon response factor-1 (IRF-1), interferon stimulatory gene factor-3 (ISGF-3), c-Myc, e-Myb, and IFN receptors. In the case of c-Myc, c-Myb or p58, high level expression indicates that the tumor tissue or cells o are treatment candidates for virus therapy. In the case of p68, IRF-1, ISGF-3, and IFN 0 receptors, low level expression indicates that the tumor tissue or cells are treatment o. candidates for virus therapy.
0 At least 30% of human tumors are characterized by an activated Ras phenotype (Bos, 1989, Cancer res., 49:4682). An activated Ras phenotype can occur as a result of i) expression of Ras proteins with activating mutations, ii) overexpression of wild-type Ras protein or, iii) expression of unregulated cyrosine kinase receptors or other members of the Ras signaling pathway such as Grb2 or Sos. Cells with activated Ras phenotype have been shown to more sensitive to killing by NDV (Lorence, rt al., 1994, Cancer Res., 54:6017-6021) and by reovirus (Strong, JE.S., et al., 1998, EMBO, 17:3351-3362) than the same cells without an activated Ras phenotype. Activated Ras has been shown to inhibit the induction of responsive genes by interferon (Zullo, and Faller, 1988, Mol. Cell.
S Biol., 8:5080-5085) and the activation of PKR by dsRNA (Mundschau, LJ., and Faller, D.V., 1992, J. Biol. Chem., 267:23092-23098). Given the key role that PKR plays in the induction of the interferon-mediated antiviral response, the increased sensitivity of cells with an activated Ras phenotype to killing by NDV and reovirus provides even more evidence for the selective killing of cells deficient in an interferon-mediated antiviral response by viruses of the present invention.
Patient derived tumor tissue obtained from biopsy can be screened for the expression of i) activated Ras protein, ii) the GTP-bound fraction of Ras (active form), iii) activated form of MAPK ERKI or ERK2), or other indices of an activated Ras pathway. The presence of an activated Ras phenotype in the patient specimen indicates that tumor tissue is a 0 treatment candidate for virus therapy.
In another embodiment of this invention, primary tumor tissue or cells obtained from patient biopsies are expanded in culture and tested for sensitivity to killing by a suitable viral therapy. In one embodiment, the amount of virus required to kill 50% of the tumor tissue COMS ID No: SBMI-01158839 Received by IP Auslralia: Time 17:19 Date 2005-03-10 10/03 '05 17:12 FAX 613 9863 3099 F.B. RICE Co. 1h029 o O culture is compared to the amount required to kill 50% of a culture of normal cells as described above for the screening of candidate viruses. An increase of ten-fold or greater in Sthe sensitivity of the tumor cells relative to normal cells to killing by the viral agent indicates that the rumor cells are specifically sensitive to the cytocidal effects of the viral treatment In a further embodiment of the invention, the ability of the targeted tumor cells to respond to endogenously or exogenously supplied IPN is determined by conducting the above screen in the presence of IFN (alpha or beta form, using 10 units per ml, see Example 27).
SAn understanding of the cellular receptors required for virus attachment or entry will O allow additional screening for tumors that have high receptor expression and hence enhanced ci tfl 1t sensitivity to the interferon-sensiive virus. This is an additional level screening for patients othat are likely to respond to virus therapy. Advantageously for therapy with an interferon- C sensitive virus, the patient's tumor would be both resistant to interferon as well as having high expression of the cellular receptor for the virus. In principle, patient derived serum, tumor cells, tissues, or tissue sections are screened by immunoassay or immunostain for the amount of virus receptor present in the serum or on the tumor cells or tumor tissue. For example, Sindbis virus utilizes the high affinity laminin receptor to infect mammalian cells (Wang et al., 1992, J Virol., 66,4992-5001). This same receptor is known to be expressed in higher amounts in many diverse types of metastatic cancer. The Panc-1 pancreatic cancer cell line, and the colon adenocarcinoma cell line SW620 are known to express a high level of o high affinity laminin receptor mRNA (Campo at al, 1992, Am I Pathol 141:107301983; Yow at al., (1988) Proc. Natl Acad Sci, 85, 6394-6398) and are highly sensitive to Sindbis virus (Example 25). In contrast, the rectal adenocarcinoma cell line SW1463 Is known to express very low levels of high affinity laminin receptor mRNA (Yow et al,, (1988) Proc. Natl Acad Sci, 85, 6394-6398), and is more than 4 orders of magnitude more resistant to killing by PPSINDBIS-Ar339 than SW620 cells, Existing strains of NDV, or other viruses including RNA and DNA viruses, are screened or engineered for altered IFN responses advantageously increased IFN responses) in normal cells. In addition to the ability to elicit a strong IFN response, other viral characteristics are screened for or engineered into the virus. Viruses with altered receptor specificity Sindbis virus PPSINDBIS-Ar339, see Example 25), or low neurovirulence are included in the subject invention NDV virus PPNJROAKIN, see Example 24).
Advantageously, viruses of the invention have the capacity to spread through direct cell to cell contact COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:12 FAX 613 9663 3099 F.B. RICE Co. __030 0O 0 The invention described herein includes a broad group of viruses (see Tible 1) that are useful for treatment of neoplasms in a manner analogous to ihe indication for NDV. In addition, viruses that naturally would not be candidates for use, due to the presence of a mechanism(s) to inactivate the IFN response in normal cells, are optionally engineered to circumvent the above restrictions. If left unmodified, viruses with mechanisms to inactivate the interferon response would be more toxic to normal cells than viruses with such Smechanism removed. The subject invention provides the development of a vector that o can be easily manipulated; and the creation of a set of therapeutic viruses. Manipulations o include the addition of an IFN gene to permit the viral expression of a transgene expressing t) 1t IFN, or other activators of the IFN response pathway. Additional permutations include the engineered expression of pro-drug activating enzymes such as the Herpesvirus thymidine C' kinase or cytosine deaminase (B laese RM et al., 1994. Eur. J. Cancer 30A: 1190-1193) and the expression of suitable marker antigen to allow targeting of tumor cells by the immune system. An additional permutation include the engineered expression of receptor ligands to target cells with those receptors expression of receptors to other viruses to target cells infected with those viruses (see Mebastsion et al., 1997, Cell 90:841-847; and Schnell MJ et al., 1997, Cell 90:849-857.] Several Newcastle Disease virus strains in addition to the one cited above demonstrate selective killing of tumor cells. In a differential cytotoxicity assay using a second strain of mesogenic Newcastle Disease virus, tumor cells were found to be 3 orders of magnitude more sensitive than normal cells to killing by the virus (Example 21). Additionally, when a third mesogenic Newcastle Disease virus strain was used in a differential cytotoxicity assay, tumor cells were found to be 80 to 5000-fold more sensitive than normal cells to killing by the virus (Example 22). Both of these mesogenie Newcastle Disease virus strains also caused tumor growth regression following intratunoral administration to athymic mice bearing human tumor xenografts (Example 23).
In seprate experiments, the safety of three distinct Newcastle Disease virus strains were studied following intraceiebral inoculation in athymic and immune-competent mice. The results of this study showed that all three virus strains were well tolerated in mice with an intact immune system. Intracerebral inoculation into the brains of athymic mice revealed that one of the viruses was tolerated significantly better than the other two (Example 24). These results demonstrate that within a single virus family important differences in viral properties can occur and be can be exploited therapeutically for greater efficacy or increased safety.
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:12 FAX 613 9663 3099 F.B. RICE Co. 031 o 2 7 SAnother means by which increased efficacy and lower toxicity following treatment with roncolyic viruses can be achieved in through the use of interferon-sensitive viruses that require specific cell surface receptors that are preferentially expressed on tumor cells.
S Sindbis virus provides an example of this type of restriction, Sindbis virus infects mammalian cells using the high affinity laminin receptor (Wang et al., (1992) J, Virol. 66, 4992-5001). When normal and tumor cells were infected with Sindbis virus in a differential cytotoxicity assay, cells which both were tumorigenic and expressed the high affinity laminin O receptor were found to be more sensitive to killing by this virus than other cells (Example 0 25). Normal keratinocytes express the high affinity laminin receptor (Hand et al., (1985) l Cancer Res., 45, 2713-2719), but were resistant to killing by Sindbis in this assay. Moreover, Sanalysis of the interferon sensitivity and laminin receptor expression levels of normal keratinocytes and two different tumor cell lines demonstrates that PPSINDBIS-Ar339 selectively kills tumor cells that i) are deficient in an interferon-mediated antiviral response, and ii) express the high affinity laminin receptor.
PPSINDBIS-Ar339 also has potent tumor growth inhibitory properties when tested in vivo in athymic mice bearing subcutaneous SW620 adenocarcinoma tumors cells (Example 32).
Vesicular Stomatitis Virus (VSV) provides evidence for the generalized hypothesis of tumor-selective killing of by oncolytic viruses, an inherent deficiency in interferon responsiveness in tumor cells renders these cells sensitive to killing by interferon-sensitive replication-competent viruses. When VSV was used to infect non-tumorigenic human WISH cells and mmorigenic HT1080 or KB cells in the presence of exogenous interferon the tumorigenic cells were selectively killed (Example 26), Additional evidence is provided in Example 33. In this example, two unrelated viruses are shown to exhibit nearly identical behavior upon infection of a tumor cell line. The similar responsiveness of this cell line to each of these viruses demonstrates that the growth of the two unrelated viruses is controlled by similar mechanisms in this tumor cell line.
Below is a list of viruses that when modified to remove naturally-occurring antiinterferon activities, are useful for viral cancer therapy (see Table Modified viruses S (advantageously, but not necessarily, attenuated in addition to the anti-interferon modification, see Table 3) that have had endogenous anti-interferon activities destroyed or reduced, are useful for cancer therapy. This list includes, but is not be limited to, the viruses described below. Because of the similarity between viruses of a COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-3-10 10/03 '05 17:13 FAI 613 9663 3099 F.B. RICE Co. Q1032 8 28 common class, the identified mechanisms for each of the specific viruses listed below, are N also prsent in other members of that class of virus as identical or functionally analogous t mechanira. The broader group of viruses is added in parenthesis, Viruses, such as those Sbelow, that have a functional loss of andti-interferon activity, through any means, including 0 5 natural occuning mutations, as well as engineered deletions or point mutations, are useful in the methods of the subject invention.
0Viruses that exercise more than one mechanism are optionally modified to contain o mutations in one, some, or all of the activities. Mutations for some of the described activities are available in the general scientific conmmunity, S.0 Isolates of naturally occurring or engineered virus that are slower growing, compared to o the growth rate of wild-type virus, are particularly advantageous because a slower virus C'i growth rate will allow a cell or population of cells competent in an interferon response to establish an efficient antiviral state before viral replication can kill the cell or cell population.
The disabling of viral anti-interferon activities as a specific alteration of viral character that results in the augmentation of the interferon response in an infected cell, but still allows viral replication in neoplastic cells is included in the subject invention.
Table 2 shows existing viruses engineered tc remove anti-interferon activity.
Table 3 lists viruses engineered to be attenuated in virulence.
COMS ID No: SBMI-01156839 Received by IP Australia: Time 17:19 Date 2005-03-10 j 10/03 '05 17:13 FAX 13 9563 3099 7.8. RICE Co. j033 Table 2 Extant Viruses Engineered to Remove Anti-IFN Activity Virus Virus Funily Virus Auti4FN Reference Clan Activity RNA Reoiridae roovirus Omega-3 Iman and Jacobs (1988) Proc Nal Aced Sri USA 85:7887-7891.
DNA Pozviridac V inia K3L Beanie at al (1991) Virology 193- 419 EL Bense at A' (1996) Virus Genes 12:89-94 EIKR Symonsetal(1995) Cel 8: 551- 560 Adnoviridav Various VA-i Mahcmw ad Shen (1991) J Vim subtypes transcripts 64:5657-5662 Alphahrrpaa- FSV-1 Gamma 34.5 Cho et W (1996) Proc Nail Aed Sci viridae ne product USA 92:10516-10520 COMS ID No: SBMI-01158839 Received by IP Autralia: lime 17:19 Date 2005-03-10 10/03 '05 17:13 FAX 613 9863 3099 F.B. RICE Co, lih 03 Table 3 Known Attenuating Mutations in Selected Virueg Virus Virus Family Virus Atenuation Reference RNA Rhoviridn reovfu Omeg-I Spnng and Fialds (1982) Natur 297:68-70 rolavirus Bovine SaliDs Clark (1998)3 infect Di 15;570- (Wa3) 587 Togaviridae Sindbis Attenuating Polo and Johnsron (1990) J Viral mutations in the 64:4438-4444.
El ad B2 coding regions DNA Poxviridac Vwrcinia Vaccinia Bullet Ct a] (1985) Virology growh facor 164:192 Thymidiuc Buller er al (19&5) Narc 317:8 13kinas 815 Thymidylmte Hughes S eiii (1991)1 Bol Chem dnawe 26620103-20109 DNA igom err t al (1991) EMBOJ 10;4343- 4350 Ribonuievdde Child at a](1990) Virology reductasa 174:625-429 dUTPase Perkus at al (199 1) Virology 190,.406-410 Adenoviridae Various Ad-4. Ad-7 and Takafui ct al (1979)J1 Infect Ds subtypes Ad-ZI 140-4-53 Alphaherps. HSV-I Thynildine Fizld and Wildy (197) 'Hyg viridse kina 81:W-277 Ribonuccotide Goldstcin and Weller (1988) Roducma. Virology 166&41-i Garnma34,5 Chouuotal (1995) Proa Natl ad gene product Sd USA 92:0516-10520 ba'c' inverted Mgnwr otl (198)3 Inlfcs Dis repent 162;313-322 COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:13 FAX 613 9663 3099 F.B. RICE Co. 1035 it o Treatment of Neoplass The present invention relates to viral therapy of neoplasms, especially in animals having Scancer. In an advantageous embodiment, the invention relates to the treatment of tumors Swhich are 1 centimeter (cm) or more in size as measured in the greatest dimension. As used herein, "a 1 cm tumor" indicates that at least one dimension of the tumor is 1 cm in length.
Such tumors are more sensitive than expected to viral therapy, often at least as sensitive to O virus, if not more sensitive, than tumors which are smaller in size. In a more advantageous ro aspect of the invention, tumors greater than 1 cm. are treatd, tumors which are 2 cm or greater, from about 2 cm to about 5 cm, and greater than 5 cm.
The present invention can also be employed to treat hosts having a high tumor burden.
O As used herein, the phrase "tumor burden" refers to the total amount of tumor within the C body expressed as a percentage as body weight. Viral therapy of hosts having a tumor burden, from about 1% to about 2% of total body weight is surprisingly effective, e.g,, producing tumor regression and a reduction in the overall tumor load. This is especially unexpected since a tumor burden of approximately 2% of the total body weight a 1 kg tumor in a 60 kg human) is approximately the maximum cancer mass compatible with life.
See, Cotran et al., In Robbins Patholoaical Basis of Diseases. 4th Edition, WB Saunders, 1989, page 252. In the Examples, volumes up to 397 mm 3 for a melanoma cancer A375) in a mouse host showed complete regression in response to treatment with a .0 Newcastle disease virus a triple-plaque purified virus). Assuming that for tissue 1000 mm equals 1 gram, a tumor having a volume of 397 mm 3 comprises approximately 2% of the total body weight for a 20 gram mouse.
As shown in Examples 4 to 9 below, tumor regression was achieved with tumors at least 1 cm in size, while untreated, control animals began dying from tumor burden within several weeks. Thus, such diseased animals were successfully treated despite being within two weeks of death. Thus, in accordance with the present invention, an animal which is near terminal from its tumor burden can be created effectively with viral therapy. Consequently, the present invention can be used to treat patients who have not responded to conventional therapy, chemotherapy such as methotrexam. 5-fluorouracil, and radiation therapy.
1 0 The efficacy of NDV for the treatment of cancer following administration through the intraperitoncal route has also been examined. Using an ascites prevention model of ovarian cancer, inraperitoneal injection of NDV in mice harboring ES-2 human ovarian tumors resulted in increased survival compared to mice treated with saline (Example 16). When ES- COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:14 FAX 613 9663 3090 F.B. RICE Co. I030 32 o 2 cells wen used in an apparent ascites model, ascites fluid production was markedly decreased in virus-treated animals compared to saline controls (Example 17).
SIn another embodiment of the invention, the administration of virus results in I) the relief Sof tumor related symptoms, such as but not limited to deceased rate of ascites fluid s production, relief of pain, and relief of obstructive disease, and 2) the prolongation of life- Fifty-two patients have received the plaque purified NDV isolate by the intravenous route. Treatment responses include: regressions of individual tumors in 5 patients; Sstabilization of disease in 2 patients for 7 months, in 2 patients for 5 months and in 1 more Spatient that is ongoing at 3 months; and a reduction in pain medication (Example Administration and Formulation NC In one embodiment of the invention, tumor cells or tissue are screened in vitro to determine those patients with tumors sensitive to the virus. Tumor cells removed from the patient (by methods such as fine needle aspiration for solid tumors or by paracentesis for ovarian ascites tumors) are grown in vitro and incubated with virus. In this embodiment of the invention, patients are selected for therapy if the virus has a high activity against their tumor cells.
In an advantageous embodiment of the invention, the amount of virus administered results in regression of the tumor or tumors. As used herein, the term "regression" means that the tumor shrinks, in size, mass, or volume. Shrinkage in tumor size is demonstrated by various methods, including physical examination, chest film or other x-ray, sonography, CT scan, MRI, or a radionucieotide scanning procedure.
Various types of neoplasms Including cancers are treatable in accordance with the invention. The viruses of the present invention are useful to treat a variety of cancers, including but not limited to lung carcinoma, breast carcinoma, prostate cacinoma, colon adenocarcinoma, cervical carcinoma, endometrial carcinoma, ovarian carcinoma, bladder carcinoma, Wilm's tumor, fibrosarcoma, osteosarcoma, melanoma, synovial sarcoma, neuroblastoma, lymphoma, leukemia, brain cancer including glioblastoma. neuroendocrine carcinoma, renal carcinoma, head and neck carcinoma, stomach carcinoma, esophageal carcinoma, vulvular carcinoma, sarcoma, skin cancer, thyroid pancreatic cancer, and mesothelioma. The viruses of the present invention are also useful to treat a variety of benign tumors, including but not limited to condylomas, papillomas, meningiomas, and adenomas.
COMS ID No: SBMI-01158839 Received by IP Ausralia: Time 17:19 Date 2005-03-10 10/03 '05 17:14 FAX 813 9663 3099 F.B. RICE Co. 0037 33 o A therapeutically effective amount of virus is administered to a host having a neoplasm.
It is understood by those skilled in the art that the dose of virus administered will vary Sdepending on the virus selected, type of neoplasm, the extent of neoplastic cell growth or metastasis, the biological site or body compartment of the neoplasm(s), the strain of virus, the ute of administration, the schedule of administration, the mode of administration, and the identity of any other drugs or treatment being administered to the mammal, such as radiation, chemotherapy, or surgical tratment. These parameters are defined through maximum Stolerated dose (MTD) determination in animal models and scaling to human dosage as a function of relative body surface area or body mass. It is also understood that under certain .l ,0 circumstances, more than one dose of the virus is given. The optimal interval between such o multiple doses of the virus can be determined empirically and is within the skill of the art.
Cl NDV is generally administered from about 3 x 10 6 to about 5 x 10 1 t PFU of virus. For local administration directly into a tumor), total amounts of at least 3 x 10 6 PFU, more advantageously at least 3 x 10 7 PFU, more advantageously at least 3 x 10' PFU, more advantageously at least 3 x 10° PFU, more advantageously at least 3 x 1010 PFU, moe advantageously at least 3 x 1011 PFU, and most advantageously at least 5 x 1012 PFU are typically used. For systemic administration, amounts of at least I x 10a PFU of virus per square meter of body surface area, more advantageously at least 1 x 10' PFU of virus per square meter of body surface area more advantageously at least 5.9 x 10' PFU of virus per t0 square meter of body surface area, more advantageously at least 1.2 x 1010 PFU of virus per square meter of body surface area, more advantageously at least 4,8 x 10 10 PFU of virus per square meter of body surface area, more advantageously at least 7.2 x 10 1 PFU of virus per square meter of body surface area, and more advantageously at least 9.6 x 10Lo PFU of virus per square meter of body surface area, and most advantageously at least 3.0 x 1011 PFU of s virus per square meter of body surface area are used.
For intravenous administration, dosing schedules of once per week, two times per week and three times per week are used. A virus in accordance with the present invention, optonally with a chemotherapeutic agent, can be administered by various routes, enteral, parenteral, oral, nasal, metal, intrathecal, intravenous using a catheter), subcutaneous, ntratumor directly into its tissue or into vessels which perfuse it), perituioral, local, sublingual, buccal, topical, intramuscular, by inhalation, percutaneous, vaginal, int-arterial, intra-cranial, intrademal, epidural, systemically, topical, intraperitoneal, intrapleural, COMS ID No: SBMt-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:14 FAX 613 0663 3099 F.B. RICE Co. Q 038 in 34 0 o intravesicular (for bladder tumors), etc. For lung tumors, a bronchial route bronchial administration), a percutaneous route, or an endoscopic route can be used. Endoscopic c injections of gastrointestinal tumors, as well as suppository treatments of rectal tumors are -also used where appropriate.
Murine toxicity studies with NDV have indicated that the acute toxicity following intravenous virus administration is likely to be caused by cytoldkine mediated reactions.
0, Cytokine responses to repeated stimuli are known to be desensitized, or down-regulated, Sfollowing the initial induction event (Takahashi et al., (1991) Cancer Res. 51.,2366-2372).
SMice receiving a desensitizing dose of virus tolerate subsequent administration of higher S0 doses better than saline treated controls (Example 18). Mice intravenously injected with a o desensitizing dose of virus were able to tolerate approximately 10-fold more virus on an Cl second intravenous dose than mice receiving vehicle alone for the first injection.
The rate of virus administration by the intravenous route can significantly affect toxicity.
Two groups of athyinic mice were intravenously treated with identical doses of NDV which was administered either slowly (0.2 ml over 4 minutes) or rapidly (0.2 mil over 30 seconds).
Comparison of the maximal weight lose in each group revealed 50% less weight loss in the group receiving slow injection versus a rapid injection (Example 19).
In the clinical trial, patients received three injections of the plaque purified NDV isolate over the course of one week, Under these conditions,.a desensitizing effect of the initial dose !0 lessened the toxicity associated with the second and third doses, even when the second and third doses are two to eight times higher than the first dose (Example 20). These data parallel those obtained with the animal studies shown in Examples 18 and 28. Furthermore in the clinical trial, higher rates of tumor regression were noted when higher doses were achievable using a smaller desensitizing dose (see Table 19, Example 20). This again paralleled the data B obtained in animal model testing (Example 29).
One concern related to the use of oncolytic viruses in the treatment of cancer is the potential inhibitory effect the humoral immune response can exert on the therapy. In the clinical study, patients displaying stable disease after I month are eligible for a second course of treatment The second, and subsequent courses of treatment are therefore administered in the presence of neutralizing antibodies to NDV. Nevertheless, infectious virus could be found in patients' urine after dosing for the second course and tumor regression was observed after a second course, providing evidence that administration of high doses of virus can overcornme the effect of neutralizing antibodies and establish an infection within the patient COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:15 FAX 613 9663 3099 F.B. RICE Co. [0039 (Example 20). an advantageous embodiment of the invention, multiple courses of irus Stherapy are administered Examples of a course include; administering virus 3X per week for SI week, followed by a 3 week rest period; administering virus 3X per week for 4 weeks, i followed by a 2 week rest period; administering one dose of virus, followed a 4 week rest period; administering 3X per week for 6 weeks, followed by a 2 week rest period. In another embodiment, virus is the administered more than 2 weeks after administration of the initial dose of virus.
In an advantageous embodiment of the invention, a desensitizing dose is given before higher subsequent doses. The desensitizing dose level is determined from clinical indicators C t10 of toxicity such as hypotension, fatigue, liver transaminase elevation or other appropriate O indices, where the desensitizing dose level is equal to or below the maximum tolerated dose 0 (MTD) for a single adminiscration. Following desensitization, additional virus doses exceeding the desensitizing dose are given. In an advantageous embodiment, the subsequent virus doses ar equal to or greater than the single dose MTD. For example, desensitizing virus doses of at least 1 x 10 PFU/m 2 more advantageously at least 3 x 10' PFU/m a more advantageously at least 1 x 10' PFU/m 2 more advantageously at least 5.9 x 10' PFU/m 2 and most advantageously at least 1.2 x 1010 PFU/m 2 are used, After desensitization, additional virus doses at least 1 x 10 PFU/mn, more advantageously at least 3 x 10 s PFU/m', more advantageously at least 1 x 10 9 PFUm, more advantageously at least 5.9 x 10' PFU/m 2 o0 more advantageously at least 2.4 x 10' PFU/mr. more advantageously at least 4.8 x 10 10 PFUl/m, and more advantageously at least 9.6 x 10's PFU/m 2 and most advantageously at least 3.0 x 10" PFU/m are used. In another embodiment. TNFo. IL-2, or other cyrokines are administered, alone or in combination, for desensitization.
The time frame between doses including the time frame between desensitizing dose and the next dose is I to 14 days, advantageously I to 7 days. The desensitizing dose can be administered by various routes, intravenous, enteral, parenteral, oral, nasal, rectal, intrathecal, intravenous, subcutaneous, intratumor, peritumoral, local, sublingual, buccal, topical, intramuscular, by inhalation, percutaenous, vaginal, intra-arterial, intracranial, intradermal, epidural, systemically, topical, intraperitoneal, intrapleural, endoscopic, intrabronchial, etc. The subsequent doses can be administered by the same route as the desensitizing dose or by another route, intravenous, enteral, parenteral, oral, nasal, rectal, intrathecal, intravenous, subcutaneous, intratumor, peritumoral, local, sublingual, buocal, topical, intramuscular, by inhalation, percutaenous, vaginal, intra-arterial, intracranial, COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 200503-10 10/03 '05 17:15 FAX 013 9603 3099 F.B. RICE Co. Z040 36 0 intradrmal, epidural, systemically, topical, intrapeditoncal, intrapleural, endoscopic, intrabronchial, etc. The usefulness of IV desensitization for subsequent dosing by another t route is demonstrated in Example 28. Mice intravenously injected with a desensitizing dose of virs were able to tolerate approximately 5-fold more vims on an intraperitoneal second dose than mice receiving vehicle alone for the first injection, In preclinical testing, the increase in the maximum tolerated dose achievable using O desensitization allowed for increased antitumor efficacy as described in Example 29.
o Optionally, more than one route of administration can be used in either a sequential or Sconcurrent mode. Routes for either concurrent or sequential administration include but are not limited to intravenous, enteral, parenteral, oral, nasal, rectal, intrathecal, intravenous, osubcutaneous, intratumor, peritumoral, local, sublingual, buccal, topical, intramuscular, by l inhalation, percutacnous, vaginal, intra-arterial, intracranial, intradermal, epidural, systemically, topical, intraperitoneal, intrapleural, endoscopic, intrabronchial, etc. An example would be the administration of a intravenous desensitizing dose followed by an intraperitoneal dose.
In another advantageous embodiment of the invention, the virus is administered by slow infusion including using an intravenous pump. syringe pump, intravenous drip or slow injection over the course of 4 minutes to 24 hours, advantageously between 20 and minutes.
A virus, and optionally one or more chemotherapeutic agents, is administered by a single injection, by multiple injections, or continuously. The virus is administered before, at the same time, or after the administration of chemotherapeutic agents (such as but not limited to: busulfan, cyclophosphamide, methotrexate, cytarabine, bleomycin, platinum coordination complex such as carboplatin or cisplatin, doxorabicin, dacarbazinc, gemcitabine, melphalan, mercaptopurine, vinblasrine, 5-flucrouracil, taxol, and retinoic acid). Viral therapy in accordance with the present invention is optionally combined with other treatments, including, surgery, radiation, chemotherapy (see, Current Medical Diaenosis and Treatment. Ed. Tiamey et al., Appleton Lange, 1997. especially pages 78-94), and biological therapy. The virus is administered before, at the same time, or after the administration of biological agents such as other oncolytic agents [such as but not limited to: adenoviruses with one of its genes under transciptional control of a prostate cell specific response element (see Rodriques, R. et al, 1997, Cancer Res 57:2559-2563; adenoviruses which do not encode a Elb polypeptide capable of binding p53 (see Bischoff, et al, COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:15 FAX 613 9663 3099 F.B. RICE CO. 0041 o37 O 1996, Sciegn 274:373-376); a herpes simplex virus that is incapable of expressing a C functional gamma 34,5 gene product (see Mineta, T. et al, 1995, Nature Medicine, 1:938t 943)]; cycokines (such as but not limited to: colony stimulating factors such as GM-CSF; tumor necrosis factor, and interleuldns such as IL-1, IL-2, IL-6 and IL-10); viral vectors [such as but not limited to adenovirus encoding p53 (see Zhang, WW et al, 1994, Cance Gene Therapy 1:5-13)J; and cancer vaccines.
0 In one embodiment of the invention, therapy consists of the serial treatment with Santigenically distinct viruses which are cytotoxic and tumor selective via the IFN mechanism.
o This embodiment allows viral therapy over an extended period without immunological 0 interference.
Another embodiment involves the treatment of patients with IFN oIFN, pIFN or C yFN) prior to, concurent with, or following administration ofNDV (or other virus). The IFN is selected from the group class I (alpha, beta and omega) and class I (gamma), and recombinant version and analogs thereof as discussed in, for example, Sreevalsoun, 1995 (In: Biologic Theranv of Cancer, second edition, edited by V.T. DeVita, Jr., S. Hellman, and S.A. Rosenberg, J.B. Lippincott Company, Philadelphia. pp3 4 7-36 4 Normal cells respond to the IFN pre-treatment with an augmented IFN response to viral infection affording even greater safety to these cells. Tumor cells deficient in the IFN signaling pathway remain sensitive to killing by the virus. This allows even higher doses of viral therapy to be used.
o The IFN is administered in accordance with standard clinical guidelines for doses and regimens known to be effective for treating viral infections.
In another embodiment of the invention, other drugs, known to affect the IFN response pathway are also optionally used to increase the sensitivity of tumor cells, or increase the resistance of normal cells to the cytocidal effects of viral infection. This class of drugs includes, but is not limited to tyrosine kinase inhibitors, cimeidine, and mitochondrial inhibitors. One strategy for enhancing oncolytic activity of therapeutic viruses of the invention involves disruption of mitochondrial oxidative phosphorylation. Preferred agents are clinically acceptable drugs that inhibit respiratory chain function or mitochondrial protein synthesis. 4-quinolone antibiotics, menadione, chloramphenicol, chloroquine, and 0 tetacyclines are useful for potentiating oncolytic activity of anticancer viruses. Such agents are administered in clinically tolerated doses 0 to 24 hours prior to administadon of an oncolytic virus. Mitochondrial inhibitors are also optionally administered after the virus to further sensidze tumors supporting viral replication.
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:16 FAX 613 9663 3099 F.B. RICE Co, [042 38 o Hypoxia and hyperthermia are also known to modulate interferon responsiveness.
Therefore, in one embodiment of the invention, hypoxic regions of tumors are oxygenated c prior to or during exposure of tumors to a therapeutic oncolytic virus. Methods for accomplishing this include, but are not limited to, systemic administration of oxygenated fluorocarbon blood hemoglobin substitutes, erythropoietin, or vasodilators. Tumor oxygenation is also accomplished by delivering oxygen at supranormal concentrations in air via the lungs.
SIn another embodiment of the invention, immunosuppressants such as cyclosporin A, S1 azathiaprimc, leflunoide, anti-CD-40 ligand antibodies (Foy, et al., 1993, J. Exp.
Med. 178:1567-1575) and various cortcosteroid preparations such as, cortisol, predisone, o prednisolone, 6c-methylpredwsolone, fludrocortisone, corticosterone, triamcinolone, Cl paramethasone, betamethasoneand, and dexamethasone are administered before, during, or after administration of the virus. Alteratively, immunostimulatory compound, e.g., lipopeptides, can be administered with the virus.
In another embodiment of the invention, agents which inhibit TNF-alpha activity such as antibodies against TNF-alpha (see Example 30), soluble TNF-alpha receptors, corticosteroids, or other compounds, are administered before, during or after the virus.
An independent mechanism by which the amount of interferon produced in response to viral infection is increased through the use of nucleosides (Machida, 1979. Microbiol.
Immunol. 23:643-650), nucleoside precursors, or drugs that increase the cellular concentration of one or more nucleosides, are optionally used as an adjunct to viral therapy.
Certain purine nucleoside analogs, 2-chlorodeoxyadenosine and 2'-deoxycoformycin, reduce interferon production in vivo. Such compounds are used to further effect differences in interferon sensitivities of tumor cells versus normal cells and are optionally used as an 2 adjunct to viral therapy.
In one aspect, an effective amount of virus can be subdivided into smaller dose units and injected at the same time into different locations of the same tumor. For continuous administration, the desired agent(s) is administered via an implanted minipump or it is impregard into a desired polymer and then transplanted into a desired location directly into the tumor) for slow or delayed release.
A virus of the present invention is formulatd as a pharmaceutical preparaion by bringing it into a suitable dose form, together with at least one excipient or auxiliary, and, if desired, with one or more further active compounds. The preparations are utilized in both human and COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:16 FAI 613 9663 3099 F.B. RICE Co. i043 o 39 0 veterinary medicine. Suitable cxcipients include, organic and inorganic substances Cl which are appropriate for enteral or parntetrl ad ation, water, saline, tissue culture media. buffers. lysine, citrate, glycerol triacetate and other fatty acid glycerides, gelatin, soya lecithin, carbohydrates such as, mannitol, sucrose, lactose or starch, magnesium stearate, talc, cellulose or protein carriers, or a combination of the preceding compounds, such as mannitol/lysine, or mannitol/lysine/sucrose. The preparations are sterilized and/or contain additives, such as preservatives or stabilizers. For parenteral administration, e.g., o systemic or local injection, a virus preparation is formulated, as an aqueous suspension Sor emulsion.
Ct The invention also relates to a method of treating a disease in a mammal, in which the o diseased cells have defects in an interferon-mediated antiviral response, comprising C' administering to the mammal a therapeutically effective amount of an interferon-sensitive, replication-competent, clonal virus. For example, many viruses have evolved mechanisms to ablate the interferon-mediated antiviral response of the host cell. The hepatitis B and hepatitis C viruses are leading causes of liver dysfunction worldwide. These viruses are associated with progressive liver damage, cirrhosis, and hepatocollular carcinoma. Treatment with interferons is the current standard therapy for these diseases, but large segments of the population fail to respond to treatment, or auffer relapses following termination of therapy.
The terminal protein of the hepatitis B virus (HBV) has been shown to inhibit the cellular responses to interferon, and double-stranded RNA, a known activator of PKR (Foster, G.R., er al., 1991, Proc. Natl. Acad. Sci., USA, 88:2888-2892), Additionally, the core antigen of HBV has been shown to inhibit the expression of the beta interferon gene (Whiuen, et al., 1991, J Virol., 65:4699-4704), and the HBV-associated delta agent has been shown to block the activity of PKR in rabbit reticulolysates (Roberson, et al., 1996. J Virol., 70:5611-5617). The hepatitis C virus (HCV) also possesses an activity that represses the PKR protein kinase. The NS5A protein of HCV has been shown to directly inhibit PKR protein kinase activity (Ga le, et al., 1998, Clin. Diagn. Virol., 10:157-162). Patients failing initial interferon therapy for HBV or HCV infection are candidates for treatment with viruses of the current invention. The therapeutic virus, or viruses, are administered by any of the means described above, but would be advantageously administqred intravenously or through the intrahepaic artery There is evidence that human immunodeficiency virus (HIV) infected cells are also resistant to the effects of interferon, and that resistance is correlated with the presence of COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:16 FAX 613 9863 3099 F.B. RICE Co. 0044 o O AIDS (Kunzi, et 1995, J. Infect. Dis., 171:822-828; Edlin et al., 1992, Ann.
Intern. Med., 117:457-460). Mechanistically, the TAR RNA region of HIV has been shown t to interact with PKR and either activate or inhibit the activity of the kinase dependent on the Sconcentration of TAR RNA (Maitra, t al., 1994. Virology, 204:823-827).
S Additionally, the cellular TRBP and the viral Tat proteins are known to bind to the HIV TAR RNA region and inhibit the activity of PKR (Davies, at al., 1994. Proc. Natl. Acad. Sci., ON 91:47134717; Brand, et al., 1997, L Biol. Chem., 272:8388-8395). Cells infected with o HIV and resistant to the effects of interferon arc targets for killing by the viruses of this S invention.
ct A number of other human viral pathogens are known to inhibit one or more components S of the interferon-mediated antiviral state. Adenovirus and Epstein-Barr virus are all known Cl to express abundant, small RNA species that block the activation of PKR in infected cells (for a review see Clemens, M.L, et al., 1994, Biochimie, 76;770-778). Epstein-Barr virus is nearly 100% associated with endemic Burkitt's lymphoma, in addition to poorly and undifferentiated nasopharyngeal carcinoma, and is the causative agent of infectious mononucleosis. Vaccinia virus has been shown to encode proteins, E3L and K3L, that block the activation of PKR and serve as false substrate for activated PKR kinase, respectively (Davies, 1993, J. Virol., 67:1688-1692). The E3L protein has also been shown to inhibit the synthetase component of the cellular antiviral response (Rivas, et aL, 1998, Virology, 243:406414).
A homolog of the vaccinia virus E3L protein has also been described in a human orf parapoxvirus (Mclnnes, CJ., 1998, Virus Genes, 17:107-115). Vaccinia virus also encodes a soluble form of the type I interferon receptor that inhibits the induction of an antiviral state by interferon (Symons, J. 1995, Cell, 81:551-560). A cellular inhibitor of the PKR kinase is induced in cells infected with influenza virus (Lee, et. Al., 1990, Proc. Nal. Acad. Sci.
USA, 87:6208-6212) or poliovirus (Black, et al. 1989, J. Virol., 63:2244-2251).
Herpes simplex I virus also encodes a protein (gamma 34.5) that antagonizes the PER mediated shut-off of protein synthesis in infected cells (Chou, 1 et al,, 1995, Proc. Natl.
Acad. Sci. USA, 92:10516-10520). The NS1 protein of influeza vims and the sigma 3 protein of reovirus have been shown to inhibit the activation of PKR by double-stranded RNA (Lu, et al., 1995, Virology, 214:222-228; Imani, F. and Jacobs, 1988, Proc.
Natl. Acad. Sci. USA, 85:7887-7891). In each of the above examples of viral interference COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Dale 2005-03-10 10/03 '05 17:17 FAX 013 9663 3099 F.B, RICE Co. [045 41 Swith the cellular establishment of an antiviral state, treatment of infected cells with the Cl viruses of the present invention leads to the selective kldling of the infected cells.
SUnless indicated otherwise herein, details and conditions of viral therapy of this invention Sare in accordance with US Application Serial No. 08/260,536 whose disclosure is 0 5 incorporated herein by reference in its entirety. The entire disclosure of all applications, patents and publications, cited above and in the figures are hereby incorporated by reference.
o The following examples are illustrative, but not limiting of the methods and compositions Sof the present invention. Other suitable modifications and adaptations of a variety of .0 conditions and parameters normally encountered in clinical therapy which are obvious to othose skilled in the art are within the spirit and scope of this invention.
Example 1 PPMK107, (a triple plaque purified isolate of the NDV strain MK107) demonstrates a selective cytotaxic activity toward many human cancer cells compared to normal human cells.
Human tumor cells and normal cells were grown to approximately 80% confluence in 24 well tissue culture dishes. Growth medium was removed and PPMKI07 was added in 10 fold dilutions ranging from 10'plaque forming units (PFU)/well to 10 PFU/well. Controls wells 0 with no virus added were included on each place. Virus was adsorbed for 90 minutes on a rocking platform at 370C. At the end of the incubation period, the viral dilutions were removed and replaced by 1 ml of growth medium. Plates were then incubated for 5 days at 37 0 C in 5% C02, then assessed qualitatively for the amount of cytopathic effect (CPE).
Cytotoxicity was quantified by using a colorimetric MTT (2-[4,5-dimethylthiazol-2-yl]-2,5diphenyl tetrazolium bromide) assay (Cell Tirer 96. catalog #G4000, Promega Corporation, Madison W1 53711) monitored at 570 nm, that detects mitochondrial enzyme activity (Mosman, 1983, J. Immunol. Methods 65:55). The viability in the virus treated wells was expressed as a percent of the activity in untreated control wells. The data was plotted graphically as PFUIwell vs. viability as a percent of controL The IC50 was calculated as the 0 amount of virus in PFU/well causing a 50% reduction in the amount of viable cells.
The results are given in Tables 4, 5 and 6. PPMMK107 demonstrated a high degree of cytotoxic activity against a diverse set of human cancer cells with 30 out of 39 malignant lines having an IC50 value less than 1000 compared to the relative insensitivity of normal COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:17 FAX 613 9063 3099 F.B. RICE Co. 12104 42 human cell types. The ma] ority of human cancer cells had IC50 values that war. 2 to 3 orden of magnitude lower than most normal human cell types.
Table 4 Summnary of Cytotoxicity faults TUMOR TYPE CELL LNE IC-58 (UM PIMROSARCOMA HTIOSO 2 PANCREATIC CARCINOMA PANC-I 3 RENAL CARCINOMA CAKI-1 9 CAIU-2 11 A498 LUNG CARCINOMA (Nosrrmall ccl) H-1299 (low 26 A427 2 A140 13 MELANOMA SKMEI2 SKMEL3 2 SKMfLS 4 A375 37 MALME3M 778 HrT144 28 BREAST CARCINOMA SKBR3 MDA-MB-468 44 ZR75-1 78 OVARIAN CARCINOMA SW626 4 PA-I 4 ES-2 13 SKOY-3 24 OVCARS 34 GUOBLASTOMA U07MG 373MG 765 U138 38 A172 BLADDER CARCNOMA HFF1197 3 UMUC-3 54 HT1376 422 NEUROELASTOMA DMR-32 41 COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 I 10/03 '05 17:17 FAX 613 9663 3099 F.B. RICE Co. 4047 CERVICAL CRCIOMA BeLa 4 PROSTATE CARCINOMA DU-145 31 PC3 3.1 EU03 COLON CARCINOMA SW620 1T29 >flt06 IEAD AND NE=S CARCIOMA KE 4 A253 2.7 E+03 FaDe 2-91403 H)-2 1.5 E04 NEUROEIPIW1oMA SK-N-MC SMALL CEL CARCINOMA (Lung) DM3- 114 48 DMS-153 1.1 NCI-H345 1.21+06 SMALL CELL CARCINOMA(Prosaz) NCI-H660 L.D LEUKEMIA (AML) C5624 SA04 LYMPHOMA (Eurkin's) Ramos 1.0 E+06 Daudi ,1.0 5E+06 Table 5. Summary of Cytotoxicity Assay Results Using Normal Human Coils.
Cell Type cani (PFU/well) Kerarinocyte NEEK 9.0 x lol Fibroblas= CCD-922 1.4 x I0= B. I x 10'1 Endothelial HPAEC 5.2 x 1 Renal RPTEC 2.7 x Melanocyte NHEM 5.1 x Ie Astrocye NHA 3.8 x 1 COMS ID Na: SBMI-01158839 Received by IP Australia: 'ime 17:19 Date 2005-03-10 10/03 '05 17:17 FAX 813 9883 3099 F.B. RICE Co.
0048 i.n 44 0 0 (N Table 6. Summary of Cytotoxlity Assay Results Using Rapidly Proliferating ct Normal Human Cells.
SRate of Proliferation 0 Cell Type In vitr In vive ICso (PFUIwell) Bone Marrow Cells, Moderate to High High' 6.2 x CD34 Enriched to o Breast Epithelial Cells Very low' Highs Human brast epitheial cells tested (HMEqC had a high rate of proliferation after imulation with bovine pituitary extract and human epidermal rowth factor. In marked conanat, nonnal breas o epithelial cells almost always have a very low deree of proliferaion in adult women with cancer.
Example 2 Use of PPMKIO7 for the Intratumoral Treatment of Human Tumor Xenografts :o mm and >5 mm) in Athymie Mice.
Athymic mice were injected intrademally with 10 million human tumor cells. After tumors reached a size range from between 5 and 10 mm, a single injection of PPMK107 (at a dose of 3 x 10 PFU) or saline was given. Almost all tumor types exhibited a rate of complete or partial regression of 50% to 100% (see Table 7) in mice treated with PPMKI07.
The one exception is the case of the U87MG experiment (experiment Although only one of 9 tumors treated with PPMK107 completely regressed, two more virus-treated tumors showed regression of 32% and 20% and two more virus-treated tumors had slower growth than all S tumors treated with saline control. Tumor regression was virtually absent in the saline control treated tumors: In all of these experiments (A through I listed in Table 7) only t0 one of 73 control tumors showed regression. These results indicate that diverse tumor types showed responses to intratumoral PPMK107 treatment.
To examine virus replication within the tumor, immunohistochemical staining for viral antigen (using a monoclonal antibody against the NDV P protein) was performed using the subcutaneous HT1080 fibrosarcoma model. Within 30 minutes of intratumoral injection of 3 x 10 PFU of PPMK107, the tumor tissue was negative for viral antigen. However, by day 2 post treatment, intense immunostaining for viral antigen was seen within the tumor, indicating virus replication within the tumor. Importantly, virus replication was specific for COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:18 FAX 613 9663 3099 F.B. RICE Co, [049 the turnor tissue since the neighboring connective tissue and skin Was negative for viral ntigen.
Table 7. PPMX1O7 Intratumoral Treatment of Subcutaneous Human Tumor Xenogrdts (<10 mm and >5 mm) in Athymi Mice.
Tumor Tumor Tvne EIxrtt I lnoa I 1 V rr -r P-u'IeCI Regression tumpitit Partial Regression A 3.OOE OS 12 11 11 HT1080 Pibrosaroma B 3.OOE 08 9 8 8 B 3.00E 08 8 8 8 PA-I Ovarian D 3.00OE 08 9 9 9 Carcinoma KB Oral E 3.00E 08 12 7 Carcinoma SKMELS Melanoma F 3.00E 08 8 5 7 A375 Melanoma G 3.OGE 08 8 5 7 H 3.OE 08 1 1 4 U87Mg Glioblastoma 1 3.0B 08 9 1 1 Examle 3 Use of PPMK107 for the Intravenous Tratment of Human Tumor Xenografas mm and >5.5 mm) In Athymic Mice.
Athymic mice were injected in'radennally with 10 million human HT1080 fibrosarcoma cells. After Eunors reached a size range from between 5 and 8 nun, a intravenous injection(s) of PPMK107 or saline wee mad. As shown in Table 8, at the highest virus dose level (1 x COMS ID No. SBMI1 158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:18 FAX 013 90663 3099 F.B. RICE Co.
46 PFU) complete tumor regression was seen in all seven mice. Single injections of 3 x and 6 x 10 resulted in regression rates of over 90%. While a single IV injection of 3 x gave only a 55% rate of tuwnor regression, three IV injections at this dose level yielded a 100% rate of response. Mice treated with IV saline exhibited no evidence of tumor regreslon. These results indicate that subcutaneous HT1O80 tumors are very responsive to IV treatment with PPMK107.
Table 8& PPMK1O7 Intravenous Treatment of Subcutaneous Human HT1080 Fibrosarcoma Xenografts and \5.5 mm) in Athymic Mice.
rn- nI 1 Dose Schedule Complete Regression Complete Partial Regression Regression 1.00 E 09 One 7 7 7 100% Injection 3.00E 08 One 10 9 10 100% Injection 6.00E 07 One II to 10 91% Injection 2.00E 07 One 11 5 6 Injecton 2.00E 07 Three 7 5 7 100% Injections every other day Saline One 10 0 0 0% Injection Saline Thret Injections Every Other Day 0%
I
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:18 FAX 613 9663 3099 F,B. RICE Co. a 051 47 A Example 4 t First Experiment Using PPMKI07 for Intratumoral Treatment of Large A375 SMelanoma Xenografts in Athymic Mice Athymic mice were injected intradenally with 10 million A375 human melanoma cells.
Ten days later, tumors of various sizes were treated with a single injection PPMK107 (doses of 3 x 10. 9 x 10, and 1.5 x 10 s PFU) or saline. For those tumor with a single largest o dimension of 10 to 11 mm, all nine completely regressed in response to intratumoral treatment with these doses of PPMK107, while of those tumors with a single largest dimension of 8 to 9.5 mm, twelve out of 24 completely regressed in response to virus therapy o (P<0.008; Table 9, section No tumor regression was seen in any mouse treated with 1 saline.
These same tumors when sorted by tumor volume also indicated a high percentage of complete regression in those of larger tumor volume, In response to these doses PPMK107, complete regression occurred in 14 out of 17 tumors with volumes >300 mm 3 (range of 304 to 397 mm 3 and in 7 out of 16 tumors with volumes <300 mm 3 (range of 144 to 295; P <0.023; Table 9, section B).
These results indicate chat tumors at least 1 cm in length or 300 mm in volume were at least as sensitive, if not more sensitive, to intratumoral PPMKI07 treatment than smaller S tumors.
COMS ID No: SBM1-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:18 FAX 513 9663 3099 F.B. RICE Co. li052 48 Table 9. Intrtumoral PPMK07 Treatment of ntraderma A375 Melanoma Xenografts.
A- Tunot Sorted Based on the Single Larget Dimension Tumor DiUrnuiou a to Tumor Diumension 10 to 11mm Treat Dosage Complete N Complete Wnt Regressionrs PPMYL107 1.53.10' 8 2 25% 3 3 100% PPMJUO7 9. xlO le 7 88% 3 3 100% PPMKI07 3.0 x 10 8 B 3 38% 3 3 100% Tal 24 12 50% 9 9 100%' Saline 6 0 0% 0 0% B. Tunar Srted Based on th luar Volume Tumor olume,: 300 mzn Tumor Volume: >300 rm' Treat Dosage Complete N Complete ant Regression Regression PPMK1O7 1-x i 6 7 33% 5 3 PFMK17 9.xlop 4 3 75% 7 7 100% PPMKI07 3.0 x0 1 6 2 33% 5 4 TOal 16 7 44% 17 14 82%' SUino .5 0 0% 1 0 a- O-OW for empletrepcssian in thePPMKI07 10 to II mmoupnrss thaPPMlr 8 to nun waled grup.
b--P c f23 for compiw reporssion in the PPM107-imeaed >3MO mm mup versus the PPMI1O7tneMld 00 m 3 PPMK107-reaed group COMS ID No: SBMI-01158839 Received by IP Australia: lime 17:19 Date 2005-03-10 10/03 '05 17:18 FAX 013 9683 3099 F.B. RICE Co. 1053 t 49 0 0 Cl Examle SSecond Experiment Using PPMK107 for Intratumoral Treatment of Large A375 SMelanoma Xenografts in Athymic Mice.
O 5 Tumors were established as in Example 4 ten days after tumor cell inoculation.
Treatment consisted of various doses of PPMK107 (3 x 10' PFU, 3 x 10 7 3 x 10", and 1.5 x 9 or saline. For tumors 10 to 11.5 mm in single largest dimension, complete or partial (at least 50%) regression occurred in all 28 tumors treated with PPMK107 using these doses in contrast to no regression in any of the saline-treated mice (Table 10, section A).
C o, When these same tumors were sorted by tumor volume, all 26 tumors greater than 300 Smm (range: 309 to 525 mm regressed completely or partially (at least 50%) in response to PPMK107 in contrast to none of the saline treated mice (Table 10. section B).
These results confirm that tumors at least 1 cm in length or 300 mm' in volume are sensitive to intraxumoral PPMK107 treatment.
COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 10/03 '05 17:10 FAX 013 9063 3099 F.B. RICE Co. 054 Table 10. Intratumonra PPMK1O7 Treatment of Intradermal A375 Melanoma ct Xmnogratts A. Tumors 10 to M s o mm 0srted Based on the Sige Larget Dimensio) TZ Do" Comlte Complte.4 PartIl 1 lI.xIO, 7 7 100% 7 x 10' 7 6 86% 7 100% 10 7 7 5 71% 7 x 100 7 5 71% 7 100% All PPMC 107 GOmups 28 23 82% 28 Saline 6 0 0% 0 0% B. Tunmrs 300 mn (sErted Based on the Tumor Volume) TX Dose N Complete Complete 4% Paltial x LO 7 7 100% 7 100% 3.a 10' 7 6 86% 7 100% I0 6 4 679b 6 3.Gx10W 6 4 67% 6 100% All PPMX107 Groups 26 21 81% 26 100% Saline 5 0 0 0% 5 Ecwnpe 6 Third Experiment Using FPMKI07 for [utratumoral Treatment of Large A375 Melanoma Xeuografi in Athynic Mice.
Tumors were established as h lxampc 4 nineteen days after tumor cell inoculation.
Inraturnorai treatment consisted of various doses of PPMIO07 (3 x 101a,3 x 106, 3 x 10'. 3 x 0 l, x IO0, 3 x i0 2 FU) or saline. For itors 125 to 14 mm in single largest dimension COMS ID No: SBMI-01158839 Received by IP Australia: Time 17:19 Date 2D05-03-I0 10/03 '05 17:19 FAX 613 9663 3099 F,B. RICE Co. 1055 S (volume range: 632 to 787 mm; avragvolume 69 mm), tumor regressins of at least c 50% occurred in two out of three mice treated with 3 x 10' PFU in contrast to no regrssion ct in both saline-treated mice (Table 11). Using the same dose of PPMK107 (3 x 10' PFU) to treat tumors with a single largest dimension of 10 to 12 mn (volume range: 320 to 600 rn3; average volume: 411 anm seven of 8 mice exhibited regression of at least 25% (P<0.001 for regression of at least 25% compared to the saline treated mice which exhibited no regressions, Table 11). Regressions of at least 25% for tumors of length 10 to 12 mm tumors Swere also seen in mice treated with 3 x 105 PFU, 3 x 10 PFU, 3 x 10 PFU, and 3 x 10 PFU, o but not for mice treated with 3 x 102 PFU or saline (Table 11).
tl t0 These results confirm that tumors at least 1 cm in length or 300 mm in volume are o sensitive to intratumoral PPMKI07 treatment.
COMS FD No: S8MI-01158839 Received by IP Australia: Time 17:19 Date 2005-03-10 2005201079 10 Mar 2005 Table 11 Third ZxPeriuient Using PPMIIDI far the Intratumorul Treatment of A375 Memnoma Xeuografth (at lenst 10 aIm 12.5en b w4 on- -vwIj Voum Mopg 1 Avg Volum Rwmlm a r47j&f" qy 5=m Rnmwear Ragrsiont 3.4)5403 3.09*08 1 a? fl 4 u 21 671 6321n727 1 LOut .1 4 632,078 0UTEU 1A0 4~i o 12 1010 12 mm Tumors II I 3.0B406 3.054 3.OE-F03 3J.E+02 slum 3210 Wa 411: 01 3 4 71 ci. d' S425to 6S2 5021~ 01 2 02S 833 6to 60D 01 1 01 281104 .34 2_ 2_ Al 379i.6661 I-*4 m-i mgddnrudon is defined uA ragrmim Ian 100% and cpud (o orearuis 50K1 4 Rhgmuimx "MZ5 end <M05 it defted u tumor rwuulua geate tn 25% and la tan 505k lnacbdus inftegtguidcis t @-4.O0bilbtr reulmater thpi25% In te 33443 gruwrai the n~fn. grop.
10/03 '05 17:12 FAX 61 3 9839 2951 [003 Example 7 ¢c Fourth Experiment Using PPMK107 for Intratumoral Treatment of Large A375 SMelanoma Xenografts in Athymic Mice.
o 5 Tumors of largest dimension 10 to 12 mm were established as in Example 4 thirteen days after tumor cell inoculation. Intratumoral treatment consisted of a single injection of 3 x 10 s PFU of PPMKI07 or saline. Volumes of those tumors treated with PPMK107 ranged from S295 to 600 mm 3 (average tumor volume of 437 mm 3 Groups of mice in each treatment Sgroup were euthanized on days 0, 2,3, 4,7, and 14 for tumor histology. For those mice 0 o observed for a minimum of 4 days, eleven out to 12 mice treated with PPMKI07 exhibited Sregression of at least 25% compared to none of 8 in the saline group (P<0.0001, Table 12).
Cl At 2 days after PPMK107 treament, two tumors already exhibited signs of regression but the degree of regression was less than Example 8 Fifth Experiment Using PPMK107 for Intratumoral Treatment of Large A375 Melanoma Xenografts in Athymic Mice.
Tumors of largest dimension 10 to 12 mm were established as in Example 4 twenty days after tumor cell inoculation. Intratumoral treatment consisted of a single injection of 3 x 10 8 0 PFU of PPMK107 or saline. Volumes of those tumors treated with PPMK107 ranged from 361 to 756 mm' (average tumor volume of 551 mm3). Nine out of 10 mice treated with PPMK107 exhibited a regression of at least 25% compared to none of 10 in the saline group (P<0.0001, Table 13).
Example 9 First Experiment Using PPMK107 for Intravenous Treatment of Large HT1080 Fibrosarcoma Xenografts.
Athymic mice were injected subcutaneously with 10 million HT1080 human fibrosarcoma cells. Six days later, tumors were treated with a single injection PPMKI07 (at a dose of 1.5 x 10' PFU) or saline. For those tumors 10 to 11 mm in single largest o dimension, five out of six tumors completely or partially regressed in response to a single intravenous injection of PPMK107 compared to none of the saline treated tumors (Table 14, P <0.025). These results indicate that tumors at least 1 em in length are sensitive to intravenous PPMK107 treatment COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 III i 2005201079 10 Mar 2005 Table12. Fouirth Experimnet UingPNsc7 far the Intrutumorni Treatment o(AS7S Melanoma Xemogrrts (eat Ieast to tar in Size).
Day Rutluiiod a Tetet Post Tthent N Complete Partial >25% and S#4A' Tota Numnber of *egresiont Regressm n Remalont 3.OE+D8 14 day. 3 0 2 1 3 1001 7days 3 0 21 3 100 3.ODE4+O 4 dys3 D 2 1 3 100 3.0*08 3 days 3 _0 0 2 3 67 All1IP2MIC107 Groups 12 0 6 5 11 92 d Saline 14 das2 01 0 0 00 saline 7 days 2 0 0 000 Sline 4 days 2 a 0 0 01 0 Saline 3Sdays2 0 DI 0 a 0 SL&Ine AllSullne Groups 8 0 001 01 01 P&A mrtia nin mn .V*la&A 4A. 1ne a a av n~ hviamu &uuzo wil qVu to or 9teC Msa 50756 C haunt orepeulon ptetrta2i% nd kur Own 7-4003 for CaVlfkcS rlWrtuRsjvshlnIn1PPMKIOI Supof 12 nile. vuulb.mdsgrow of9 mce, -J'C JOIftiibnreainte2%Imefl7gys12 micveuni te~ gr~e oup of mint= 2005201079 10 Mar 2005 Tales13 FMlt kpauent Using PEM0007 for the Tztrnsuuerl Tratment oIA375 Meolanhoma Noni ME Is 8hzel -llsgzusMal 4'UEd 50' Is debug ammo Wrveelo paturfin 24% ad tkn -P <MUlLbrf trdh uqrudate 1.2S% fa t PPMK1P7 pm untie ftine gp"q.
10/03 '05 17:13 FAI 61 3 9639 2951 1006 0 0 i try 0 0 0 cN r-.
O
O
i o~ o rs 56 Table 14. Intravenous Treatment of Subcutaneous HTI080 Human Fibrosarcoma Xenografts in Athymic Mice. Tumor Size 10-11 mm.
Treanent Dose N Compklete Complem Para Remresion Regression PPMK1O7 IE+09 6 4 67' Stline 4 0 0" 0 0 "P <0.025 (by Fisher's exact tes) for complete or partial regression (at east 50% reression) in the PPMK107 treatd group compared to aline group.
Example Specific Clearing of PPMK107 Infection from Normal but Not Tumor Cells.
In order to examine the mechanism of tumor-specific killing by NDV strain PPMK107.
representative tumor cells were chosen based on the following criteria: a) ability to form S tumors as xenografts in athymic mice; b) the tumor xenografts are specifically killed in vivo following administration of PPMK107; c) the tumors cells exhibit killing by PPMK107 in vitro at virus concentrations that are several logs below the concentration to kill resistant, normal cells; and d) tumor cells must be easily distinguished from the normal cells when present as a co-culture.
Xenograft tumors comprised of KB head and neck carcinoma cells exhibit 83% complete or partial regression in response to a single intratumoral injection of PPMK107, are more than four logs more sensitive to killing by PPMKI07 in vitro than ar normal primary skin fibroblasts (CCD922-sk), and are easily distinguished from CCD922-sk cells when present as a co-culture.
Accordingly, co-cultures of KB and CCD922-sk cells were infected at a multiplicity of infection the ratio of virus added per cell) of 0.0005 and the course of the infection followed for 5 days by immunohistochemical staining for a viral antigen (NDV P protein).
Infection of normal cells peaked at 2 days with little or no apparent cell death as determined by visual inspection of the cell monolayer, On the third day post-infection the amount of viral expression in the normal cells decreased significantly, while infection of the tumor cells was clearly apparent. The amount of viral antigen virtually disappeared in the normal cells on days 4 and 5, while the infection in the tumor cells progressed rapidly through the tumor cell population resulting in destruction of the majority of the tumor cells present in the coculture.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:13 FAX 61 3 9639 2951 1o007 ttn 57 Thus, nomal cells were infected and easily cleared the infection in a manner consistent owith the anti-viral effects of IFN. The tumor cells were unable to establish an anti-viral state in response and were killed by the unabated viral growth, despite the presence of Sphysiologically effective concentrations of IFN secreted into the media by the normal cells.
Example 1u Demonstration that Interferon is an Important Component of Viral Clearing in Normal CCD922-sk Cells SThe hypothesis that interferon was mediating the ability of CCD922-sk cells to clear the infection of PPMK107 was tested. Polyclonal neutralizing antibodies to human interferon-a n or human interferon-0, used alone or in combination, were added daily to cultures of SCCD922-sk cells infected with PPMK107 at an moi of 0.0005 and the progress of the infection followed for three days. The amount of viral antigen present in the cells increased in proportion to the concentration of neutralizing antibody, with the effect of the antiinterferon-P antibody being more marked than that of the anti-interferon-a antibody;.
consistent with reports that fibroblasts produce predominantly the beta form of interferon.
The ability to make the normally insensitive cells more susceptible to infection with PPMK107 through the addition of neutralizing antibody to interferon supports the hypothesis that a key difference between the sensitivity of normal and tumor cells to killing by PPMKI07 lies in the ability of normal cells, but not tumor cells, to establish an interferonmediated anti-viral response.
Example 12 Demonstration that Interferon-f is an Important Component of Viral Clearing in 2s Other Normal Cells.
In this experiment, it was determined that another normal cell (NHEK, normal human epithelial cells) known to be quite resistant to killing by PPMK107, was made more sensitive through the addition of polyclonal anti-interferon-p antibody to a culture of infected cells.
NHEK (normal human epithelial keratinocyte) cells were infected at an moi of either 0.0005 or 0.05 and had antibody added daily over five days.
In the cultures infected at the low moi (0.0005), antibody dependent augmentation of viral antigen expression was clear at five days post-infection, but was less clear earlier in the experiment. Antibody addition to cultures infected with PPMK107 at an moi of 0.05 resulted COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:14 FAX 61 3 9639 2951 @00oo8 o in a marked increase in viral antigen at 4 and 5 days post-infection. At 2 and 3 days postinfection the addition of neutralizing antibody resulted in less accumulation of viral antigen S(Fig. 1).
'The culture supernatants from the high moi samples were also titrated for the amount of 3 infectious virus present by plaque assay on human HT1080 fibrosarcoma umor cells; the standard assay system in our laboratory. Results from this analysis demonstrated that at five days post-infection ther was 19-fold increase in the amount of infectious virus in the O antibody-treated cultures relative to mock-treated controls (Fig. 1).
SThese results suggest a general mechanism by which normal cells are protected from c a killing by PPMKI07 through an interferon-related mechanism.
C
N Exampl 13 Comparison of the Effect of Interferon-) on PPMKI07 Infection in Tumor and Normal Cells.
A comparison of the effect of exogenously added interferon-0 on the infection of normal (CCD922-sk) and tumor cells of high (KB) or intermediate (HEp2) sensitivity PPMK107 was performed. Separate cultures of the three cells were treated with interferon-f at 20, 200, or 2000 units/ml I day pro- and 2 days post-infection at an moi of 0.0005, At 3 days post-infection the low level of viral antigen expression present in the normal cells :0 was eliminated at all doses of interferon used. Conversely, the addition of interferon to the highly sensitive KB tumor cells at concentrations of 2 or 200 units/ml decreased relative levels of viral antigen expression 2-fold, with complete suppression at 1000 units/mI interfearon. The intermediately sensitive HEp-2 cells responded to the exogenous interferon by clearing viral antigen expression at all of the interferon doses used (Fig. 2).
The pattern of sensitivity in the KB and CCD922-sk cells to the anti-viral effects of exogenously added interferon-P was inversely proportional to the sensitivity of these cells to killing by PPMKl07. The ability of the HEp-2 cells to respond to the effects of interferon indicates that these cells are able to efficiently utilize the concentrations of interferon used in this experiment. Similarly, the response of the KB cells to the high doses of interferon *O suggests that the inability to establish an interferon-mediated anti-viral response does not result from an absolute defect in the interferon pathway, but rather a relative insensitvity compared to normal cells.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:14 FAX 61 3 9639 2951 oo09 o 59 0 Exampule 14 k Effect of Low Concentrations of Interferon-P on the Infection of Normal and Tumor Cells by PPMK107.
o In this experiment normal (CCD922-sk) and tumor (KB) cells were treated with low concentrations of interferon-0 2, and 20 units/ml) 1 day before and 2 days post-infection with PPMKI07 at an moi of 0.05.
SUnder these conditions the normal cells experienced a dose-dependent decrease in the Samount of viral antigen, while the relative levels of viral antigen in the tumor cells was Sunaffected by the addition of exogenous interferon (Fig. 3).
In o 0 Eamule PPMK107 Purification Method A PPMK107 was derived from the mesogenic Newcastle disease virus strain Mass-MKI07 by triple plaque purification. Approximately 1000 PFUs (plaque forming units) of live PPMK107 were inoculated into the allantoic fluid cavity of each 10 day old embryonated chicken egg. After incubation at 36 0 C for 46 hours, the eggs were chilled and then the allantoic fluid was harvested. Cells and cell debris were removed from the allantoic fluid by to centrifugation at 1750 x g for 30 minutes. The clarified allantoic fluid (supernatant containing virus) was then layered over a 20%/55% discontinuous sucrose gradient) and centrifuged at approximately 100,000 x g for 30 minutes. The purified virus was harvested from the 20%/55% interface and dialyzed against saline to remove the sucrose.
Method B In another advantag ous embodiment, the clarified allantoic fluid was frozen at -70 0
C.
After thawing, the fluid was maintained at 1 to 40C overnight and then the contaminating material was removed f om the virus suspension by means of centrifugation (1750 x g for minutes). This material was further processed using the discontinuous sucrose gradient on 0t the ultracentrifuge as atove.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:14 FAX 61 3 9639 2851 010 In o wo 60 eMethod C In another advantageous embodiment, ultracentrifugation on the discontinuous sucrose O gradient was accomplished by means of a continuous flow ultracentrifuge.
Method D SIn another advantageous embodiment, harvested allantoic fluid is diluted with a buffer O containing 5% mannitol and 1.0% 1-lysine, pH 8.0 (ML buffer) and is clarified and 0 exchanged with ML buffer by tangential flow filtration (TF) through filters with a nominal ti 10 pore size of 0.45p- The permeate containing the clarified virus in ML buffer is collected and 0 virus is purified by TFF through filters with a nominal cut-off of 300,000 daltons in ML buffer. The concentrated, purified virus in ML buffer is collected as the retentate from this step and is again diluted with ML buffer before being applied to a Sephacryl 5500 (Pharmacia) gel permeation column equilibrated with ML buffer. Fractions containing purified virus are collected, pooled and can be recancentrated by TFF through filters with a nominal cut-off of 300,000 daltons with ML buffer.
Results Clonal Virus After generation of PPMK107 by plaque purification, eight individual molecular clones from the population of virions were found to have an identical sequence a homology of 100%) of over 300 contiguous nucleotides within the fusion protein gene of NDV.
PPMK107 is a clonal virus with a high degree of genetic homogeneity.
PFUnmg protein One quantitative means of measuring purity is by determination of a PFU/mg protein.
The activity of the virus preparations was determined by the plaque assay method using HT1080 and the protein content of the virus preparations was determined using the Modified 3o Lowrcy Assay (Bio-Rad, Hercules, CA) with bovine serum as the protein standards. Higher values indicate a greater level of purity. Using Method A, PPUAng values of at least 4.8 x 1010 were achieved (see Table 15). Using Method C, PFU/mg protein values of at least 2.0 x
I
were achieved. For a mesogenic strain of NDV, a literature value for this measurement COMS ID No: SBMI-01158845 Received by IPAustralia: Time 17:21 Date 2005-03-10 10/03 '05 17:14 FAX 61 3 9639 2951 0oil 0 0 i 0 0 0 ci 0 r-- 0 i o o rs 61 of purity has not been found. The best estimate for a mesogenic strain of NDV is the virus preparation (NDV MassMKl07, lot RU2, prepared as in Faaberg KS and Peoples, ME, 1988, J Virol 62:586; and Bratt, MA and Rubin, H. 1967, Virology 33:598-608). This RU2 lot was found to have a PFU/mg of 1.3 x 1' PFU/mg of protein. The purity values achieved by 5 Method A are approximately 40 times better than what the Peoples method achieved (see Table Particle per PFU Ratio Another quantitative means of measuring purity is by determination of a ratio of particles 0 per PFU, Lower values indicate a greater level of purity. Particle counts were done by electron microscopy using standard methods. Using either Method A or Method B, particles per PFU values near one were achieved (Table Table IS. Virus Purity PFU per Particle Virus Preparation Method Virus Lot me protein per PFU Preferred Method A PPMK1D7 L2 4.8 x 1010 0.80 L4A 6.9 x 10 o NT a 6.6 x 10 o 1
NT
0 L6 7.7 x 10 L 0.55 L7 6.1 x 10 t 1 NT Preferred Method C PPMK107 D004 2.0 x 1 0 'o 0.32 D005 4.5 x 10o 0.52 DO00 4.4 x 1010 NT Preferred Method D PPMK107 RD2 RD3 5.6 x 5.0 x 10"1 NT, Not Tested COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:15 FAX 61 3 9639 2951 0102 i 62 o Virus preparations using Methods A and C also permitted purification of NDV to a level Clq substantially free of contaminating egg proteins. For the PPMK107 lot 7 preparation using i Method A ovalbumin, was not detectable in a Wester blot using 1.7 x 109 PFU of purified virus per well (3.3 cm in width) run on an SDS-PAGE (sodium dodecyl sulfatepolyacrylamide gel electophoresis) gel (1 mm thick); a nitrocellulose membrane for transfer; and rabbit anti-ovalbumin (Cappel rabbit IgG fraction at a 1:200 dilution of a 4 mg/ml antibody concentration). For PPMK107 preparations using Method D and analyzed by o SDS-PAE followed by silver staining, no band corresponding to ovalbumin was observed.
So Example 16 tn o PPMK107 Treatment of Ascites-Forming ES-2 Ovarian Carcinoma in Athymic Mice.
C In this experiment, all of the athymic mice (female, NCR nu/nu. 8 weeks old) were given an intraperitoneal injection of 106 ES-2 cells. Seven days later before ascites had developed, they were treated intraperitoneally with saline or PPMK107 (at 1 x 10' PFU). As shown in Figure 4, there was a markedly improved survival in the animals treated with PPMK107 compared to saline. The majority of the mice in the saline treated group had developed ascites by seven days post-treatment and by day 38, all of these animals had died. In marked contrast, 92% of the mice treated with PPMK107 were still alive by day 38 and 25% of these animals were long term survivors (>120 day survival).
!0 Examnle 17 PPMKI07 Treatment of ES-2 Ovarian Carcinoma in Athymic Mice When Ascites is Present.
In this experiment, all of the athymic mice (female, NCR nu/nu, 8 weeks old) were given an intraperitoneal injection of 106 ES-2 cells. Fourteen days later when the majority of mice had developed ascites, the mice without ascites were excluded and the mice with ascites were randomized into 7 intrapmeitoneal treatment groups (PPMK107- one treatment on day 0; PPMKI07- two treatments for the first week; PPMK107- one treatment per week; PPMK107- two treatments per week; saline- one treatment on day 0, saline-two treatments 0 for the first week: saline-two treaments per week). A dose of 1 x 109 PFU/mouse was used for each virus treatment. All of the mice before the first treatment and any additional treatments were drained of the ascites fluid.
The degree of ascites Was quantified and noted as follows: COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:15 FAX 61 3 9639 2951 Q1013 Asites Degree of Ascites Score Animal appears normal- little or no ascites present Abdomen slightly distended; animal is capable of normal functions Abdomen distended; animal is slow-moving, hunched with a staggered gait.
Abdomen completely distended; animal moribund Death after ascites development As shown in Table 16, all of the saline-treated animals had more advanced ascites than the PPMK107-treated animals on both days 7 and 10. On day 7 post initial tratment, each S the saline group had ascites scores above 3.5 while all of the PPMK107-treated animals had ascites scores at 3.0 or below. Similarly on day 10 post initial teatment, each the saline group had ascites scores above 4.5 while all of the PPMKI07-treated animals had ascites scores at 4.1 or below. These results indicate that ascites fluid production was markedly decreased in virus-treated animals compared to saline conurols.
0 Table 16. PPMK107 Treatment of ES-2 Ovarian Carcinoma In Athymic Mice When Ascites is Present Treatment of Mice Ascites Score, Day 7 Ascites Score, Day Saline xl 12 4,3 4.7 Saline x2 12 3.7 4.6 Saline x2 per wk 12 4.3 4.8 PPMKI07 x 1 17 3.0 4.1 PPMKI07 x 2 17 2.3 3.6 PPMK107 xl per wk 17 2.6 2.6 PPMK107 x2 per wk 17 2.2 3.6 COMS ID No: SBMI-01158845 Received by IPAustralia: Time 17:21 Date 2005-03-10 10/03 '05 17:15 FAX 61 3 9639 2951 04 Examolc 18 Ct Use of a Desensitizing Dose of PPMK107 to Reduce the Letbality of a Subsequent Dose of PPMK107.
SC57BJU6 mice (seven weeks old) were injected intravenously on day 0 with either saline or a desensitizing dose of PPMK107 (3 x 10 PFU/mouse). Two days later each set of mice were further subdivided into groups for intravenous dosing with sallne or PPMKI17 (at doses o 9 9 10 of Ix 10, 2.5 x 10, 5 x 10, and Ii 100 PFUlmouse). As shown in Table 16 below, when saline was used to pretreat the mice, deaths were recorded in the mice subsequently dosed S with 2.5 x 10 5 x 10, and lx 1010 PFU. The doses of 5 x 11 and 1 x 10 PFU were 0 100% lethal to the mice prtreated with saline. In contrast, no deaths were seen in any group of mice given a desensitizing dose of PPMK107 on day 0 followed by PPMK107 injection two days later at dose levels up to 1 x 1010 PFU. These data indicate that PPMK107 can be used to desensitize the lethality of subsequent dosing with this sanme agent. Furthennore, the maximal tolerated dose of PPMKlO07 can be raised by an approximate order of magnitude when using this virus as a desensitizing agent Table 17. Use of a Desensitiing Dose of PPMK107 to Reduce the Lethality of a 3 Subsequent Dose of PPMKI07.
roup Injection on Day 0 Dose on Day 2 of of Mice Deaths Lethality I Saline Saline 8 0 0 2 Saline PPMK10I7, 1.OE4-09 8 0 0 3 Saline PPMK107, 2.5E--09 8 3 38 4 Saline PPMK107, 5.OE+09 8 8 100 Saline PPMK107, L.DE+10 8 8 100 6 PPMK107,3E+08 Saline 8 0 0 7 PPMKO7, 3E+08 PPMK107, LOE+09 8 0 0 8 PPMKl17, 3Ee08 PPMK107, 2,5E+09 8 0 0 9 PPMK107, 3Ei08 PPMX107, 5.DE+09 8 0 0 PPMK107, 3E.08 PPMK107, 1.OE+10 8 0 0 1 -s1 I 014 COMS ID No: SBMI-01158845 Received by IP Ausralia:Time 17:21 Date 2005-03-10 10/03 '05 17:15 FAX 81 3 9039 2951 O o Examle 19 Slower Intravenous Inection Rate Reduces the Toxicity of PPMK07.
STwenty two athymic mice (8 weeks old) were anesthetized with a combination of (O ketamine/xylazine and placed into a restrainer to help inhibit their movement during the injection process to allow for either a slow or rapid injection of PPMK107. For the slow injection group, 0.2 mL of 4 x 109 PFU of PPMK107 in saline was injected intravenously over a 4 minute period with 0.01 mL given every 10 to 15 seconds. The rapid injection group received the same dose and volume but over a 30 second period. As shown in Table 18, the o animals receiving their dose of PPMK107 over 4 minutes had half as much maximal weight 0 loss (recorded on day 2 after dosing) as the animals receiving the same IV dose over Sseconds. These results indicate that PPMK107 has less toxicity and is safer for intravenous administration when injected at such slower rates.
Table 18. Slower IV Injection of PPMK107 Results in Reduced Toxicity.
Group Length of Time of Mice Maximal Percent That Dose was Weight Loss Administered Rapid Injection of 30 seconds 11 12% 4E-09 Slow Injection of 4 minutes 11 6% 4E+09 Example Use of PPMKI07 in the Treatment of Patients with Advanced Cancer.
PPMK107 is currently being tested in a phase I clinical trial in the U.S.A. by the intravenous route. To date, a total of 52 patients with advanced solid tumors, no longer 0 amenable to established therapies, have been treated with PPMK107. Seventeen of these patients have received a single dose for the initial treatment course. Thirteen other patients have received three of the same doses per week for one week for the initial treatment course.
Twenty-two more patients have received three doses per week for one week for the initial treatment course with the first dose a desensitizing dose of 12 billion PFU/m 2 and the two subsequent higher doses of between 24 to 96 billion PFUl/m. The sizes of each patient's tumors were followed once per month. Patients with at least stable disease (less than io015 COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:16 FAX 61 3 9639 2951 Bo01 increase and less than 50% decrease in the sum of the products of all measurable tumors in the absence of any new lesions) were eligible for additional treatment courses each month.
Rearssions of Individual Tumors in Cancer Patients: Regressions of individual tumors were observed in 5 patients (one in the single dose regimen, one patient in the repeat same dose regimen and three patients in the lsensitizing dose regimen; Table 19). A higher rate of tumor regression (16% of patients) was noted in those receiving higher second and third doses as part of the desensitizing regimen than when patients received three of the same doses in the repeat same dose regimen tumor 0 regression).
Table 19. Regression of Individual Tumors in Patients with Advanced Cancer using PPMK107.
Regimen of Patients of Patients of Types of Cancer Treated with Tumor Patients with Tumor at this Dose Regressions with Regression Level Tumor Regression Single Doss 17 1 of 17 6% Colon Cncer Repeat Sar 13 1 of 13 8% Broas Cancer Dose Dewensitizing 22(19 3 of 19 16% Mesothelioma evaluated) Melanoma followed by Colon Cancer Two Higher Doses Total 52(49 5 of 49 10% At noted above evaluated) These cases are summarized below: Tumor Reression of a Palpable Colon Metastasis A 68 year old woman with colon carcinoma had a palpable abdominal tumor among her widespread metastases. After a single IV treatment with PPMKI07 at 12 billion PFU/m 2 this patient experienced a 91% regression of this single abdominal wall tumor over the course COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:16 FAX 61 3 9039 2951 017 67 0 of two weeks (Table 20 below). Mcasurements of the tumor one day after dosing (3.75 x 3 k cm) were similar to the baseline measurements of 4 x 3 cm. However, by day 7 post dosing, Sthe tumor had decreased in size to 2 x 2 cm and continued to decrease in size to 1.5 x 1.5 crm by day 14 after PPMK107 dosing. Previous to PPMK107 treeiment, this tumor mass had been rapidly growing with a 1065% increase in tumor volume in the two weeks before PPMK107 dosing. This patient was taken off study because of increased growth of the tumor ON elsewhere.
STable 20. Size of Palpable Abdominal Wall Tumor in Patient #123 (68 year old Female S 10 with Metastatic Colon Carcinoma) After a Single IV PPMK107 Dose of 12 Billion SPFU/n?.
cI Tumor Tumor Volume 8 Reduction Time After Dimensions x L W x W, in Tumor Date Dosing (L a W, em) c in Tum 7/23/98 Day 0 4x3 18. 7/24/98 Day I 3.75 x 3 16.9 6% 7/30/98 Day 7 2x2 4.0 78% 8/6/98 Day 14 1.5 x 1.5 1.7 91% B) Reeression of a Chest Wall Tumor in a Woman with Breast Cancer A 58 year old woman with breast carcinoma had a palpable chest wall mass apparent to visual inspection. During her second course of PPMKI07 treatment with three doses of 5.9 billion PFU/m 2 her chest wall tumor mass by visual and palpable inspection decreased This patient was taken off study after her third course of therapy because of increased growth of the cancer elsewhere, C) Regression of Abdominal Tumors in a Patient with Peritoneal Mesothetioma A 46 year old man with peritoneal mesothelioma had three large (8 to 10 cm) masses rgrers 50%, 42% and 10%, respectivly, after his first course of PPMK107 treatment consisting of a desensitizing dose of 12 billion PFU/mz followed by two doses at 48 billion PFU/m2. His other remaining large tumor mass (9.8 cm in size) remained stable after this COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:16 FAX 61 3 9839 2951 i__018 S68 0 first course of treanmnnt. This patient is currently still on study. His most recent CT sean still indicaed significant tumor regression of at least 30 to 36% from baseline for two of his Smctastass and overall disease stabilization.
D) Reeression of Metastatic Tumors in a Patient with Melanoma.
A 57 year old man with melanoma had two tumor masses completely regress after his Sfirst course of PPMK107 treatment consisting of a desensitizing dose of 12 billion FFU/m 2 Sfollowed by two doses st 48 billion PFU/m2. The two tumors which disappeared after o PPMK107 treatment were a palpable groin mass of 1 cm in size and a small lung metastasis.
t) lo This patient was taken off study because of increased growth of the tumor elsewhere.
0 0 E) Continued Remession of a Liver Metastasis in a Patient with Colon Cancer.
A liver metastasis in the caudate lobe of a 79 year old man with colon carcinoma regressed 59% after his first course of PPMK107 and regressed 97% after his second course.
Treatment consisting of a desensitizing dose of 12 billion PFUlm 2 followed by two doses at 72 billion PFU/m 2 At baseline before treatment, this tumor measured 3 x 3 cm (tumor volume of 13.5 cm 3 based on a 'A x Lx W 2 formula), After his first course of PPMKI07, it decreased 59% to 2.8 x 2 cm (tumor volume of 5.6 Three weeks after his second course of PPMK107. this same tumor mass in the caudate lobe was reported as measuring 3 x cm (tumor volume of 0.38 cm 3 a 97% decrease from baseline.
!o Stabilization of Cancer Twenty one other patients, all of whom previously had tumor progression with.
conventional cancer therapies, have experienced benefit in the form of stabilization of their advanced cancer after PPMKI07 dosing. These patients with stable disease represent those with diverse types of cancer including renal cancer, pancreatic cancer, breast cancer, bladder cancer, cholangiocarcinoma of the gallbladder, and lung cancer. Included among these cases are the following: seven month stable disease in a patient with renal cancer: seven month stable disease in a patient with lung cancer; five month stable disease in a patient with pancreatic cancer, five month stable disease in another patient with pancreatic }0 cancer, ongoing 3 month stable disease in a patient with renal cancer; ongoing 2 month stable disease in a patient with cholangiocarcinoma of the gallbladder.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:17 FAX 61 3 9638 2951 019 069 SReduction in Pain Medication One patient at the single dose 5,9 billion PFU/m2 dose level has benefited from 1 -PPMK107 treatment in the form of symptomatic relief of cancer pain as denoted by a reduction in narcotic pain medication.
Desensitization In the desensitizing regimen, a clear desensitizing effect from the first dose (at 12 billion o PFUAn 2 is seen on subsequent doses within that same week. In general, the reported side 0 effects from second and third doses have been of lower incidence and milder, even when 14r) these doses arc 2 to 8 times higher (at between 24 to 96 billion PFU/m 2 than the first dose.
o For example, while fever has been reported in 68% of patients (including 9% with a grade 3 c' fever spike) after the first dose, kit was only reported in 32% of patients (none with grade 3 fever) after the second dose and was reported in only 5% of patients after the third dose. As another example, chills were seen in 50% of The patients after the first dose, 18% of the patients after the second dose, and in only 14% of the patients after the third dose.
As another example, in the repeat same dose study, the first 4 patients in this multidose treatment regimen (three doses of 5.9 billion PFUJm2 per week for one week) had fever afier the first dose in spire of receiving prophylactic antipyretic treatment with acetanminophen and ibuprofen. The majority of these patients had no fever after receiving the second and third doses, even in cases in which they did not receive antipyretics, There is therefore strong evidence that administration of the first dose in the three times per week schedule reduces the toxicity for the second and third doses.
Dosing Through Neutralizing Antibodies in Serum Using the dose range in this phase I study (Q5.9 billion PFU/rnm2), there is also clear indication that one can effectively deliver virus to patients even if they have generated neutralizing antibodies. A 72 year old woman with pancreatic cancer at the 12 billion PFU/ml single dose level has had stable disease for 2 months since beginning PPMK107 treatment. A second course (consisting of a single IV dose of PPMK107) was administered one month after the first dose when the patient had produced neutralizing antibodies in her serum. Seven days after this second course, her urine was positive for PPMKX107 at a tjter of at least 40 PFU per mL.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:17 FAX 61 3 9639 2951 020 Additional evidence indicating that antitunor efficacy is achievable using the dose ranges (N in this trial billion PFU/m 2 in spite of the presence of neutralizing antibody is taken C from a 58 year old woman with breast cancer that had spread to her chest wall. As noted in the section discussing tumor regression, her chest wall tumor mass regressed -90% during her acecond course of therapy, This effect occurred between weeks 5 to 6 after her initial course, at a time in which her antibody had a titer of 1:256 (at the beginning of week 5) and S rose to >1:2560 (by the beginning of week Further evidence that virus can be delivered effectively to this patient in spite of the presence of neutralizing antibody is indicated by the positive urine sample (20 to 40 PFU/ml) seen at the end of week 5 (when her baseline urine C 0 at the beginning of week 5 had been negative).
o These results indicate that the neutralizing antibodies to PPMK107 in these patients' 0 serum was not able to completely inhibit the virus nor the virus's antitumor efficacy with a second treatment course.
Examele 21 Summary of Cytotoxlcity Assay Results with Newcastle Disease Virus PPNJROAKIN Human tumor cells and normal cells were grown to approximately 80% confluence in 24 well tissue culture dishes. Growth medium was removed and PPNIROAKIN, a plaque purified clone of the mesogenic Newcastle disease virus strain New Jersey Roakfn- 1946, was D added in 10 fold dilutions ranging from 10 plaque forming units (PFU)/well to 1 PFU/well.
Controls wells with no virus added were included on each plate. Virus was adsorbed for minutes on a rocking platform at 37C. At the end of the incubation period, the viral dilutions were removed and replaced by I ml of growth medium. Plates were then incubated for 5 days at 37 0 C in 5% C02. Cytotoxicity was quantified by using a colorimetric MTT (2- 4 .5-dimethylthiazol-2-yll-2,5-diphenyl tetrazolium bromide) assay (Cell Titer 96, catalog #G4000, Promega Corporation, Madison WI 53711) monitored at 570 nm, that detects mitochondrial enzyme activity (Mosman, 1983, J.ununoL Methods 65:55). The viability in the virus treated wells was expressed as a percent of the activity in untreated control wells, The data was plotted graphically as PFU/well vs. viability as a percent of 0 control. The IC50 was calculated as the amount of virus in PFUwell causing a reduction in the amount of viable cells- COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 .17:17 FAI 1 3 9639 2951 [a021 71
O
O
C Table 21. Summary of Cytotoxicity Assay Results with PPNJROAKIN.
c CelType Cell Line Icn (PFULTwnl o Fibrosarcoma HT1080 13.8 Head and Neck KB 2.4 Carcinoma 0 CN Normal Fiboblast CCD922sk 1.2 x 04 These results show that PPNJROAKIN demonstrates tumor-selective killing of at least two different human tumor cells (HT1080 and KB) relative to normal skin fibroblasts. The values for the two tumor cell lines are between 800 and 5000-fold lower than that for normal cells.
to Example 22 Summary of Cytotoxicity Assay Results with Newcastle Disease Virus PPCONN70726 Human tumor cells and normal cells were grown to approximately 80% confluence in 24 well tissue culture dishes. Growth medium was removed and PPCONN70726, a plaque purified clone of the mesogenic Newcastle disease virus strain Connecticut 70726-1946, was added in 10 fold dilutions ranging from 10 plaque forming units (PFU)/wll to 1 PFU/well.
Controls wells with no virus added were included on each plate. Virus was adsorbed for minutes on a rocking platform at 370C. At the end of the incubation period, the viral dilutions were removed and replaced by 1 ml of growth medium. Plates were then incubated for 5 days at 37°C in 5% C02. Cytotoxicity was quantified by using a colorimetric MfT (2- [4,5-dimhthylhiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assay (Cell Titer 96, catalog #04000, Promega Corporation, Madison WI 53711) monitored at 570 nm, that detects mitochondrial enzyme activity (Mosman, 1983, J.Immunol. Methods 65:55). The viability in the vims treated wells was expressed as a percent of the activity in untreated control wells. The data was plotted graphically as PFU/well vs. viability as a percent of control. The IC50 was calculated as the amount of virus in PFUlwell causing a reduction in the amount of viable cells.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:18 FAX 61 3 9639 2951 a022 n 72 0 0 Table 22. Summary of Cytotoxcity Assay Results with PPCONN70726.
Cell T Cell Line ICSn (PFU/well) JEd and Neck KB 18.1 Carcinoma Glioblastoma UB7MG 12.7 Glioblastoma U373MG 879 Normal Fibroblast CCD92sk 7.3 x These results show that PPCONN70726 demonstrates tumor-selective killing of at least o S three different human tumor cells (KB, U87MG, and U373MG) relative to nonual skin CN fibroblasts. The IC50 values for the two tumor cell lines are between 80 and 5000-fold lower than that for normal cells.
Examnple 23 .0 Intratumnbral Treatment of HT1080 Fibrosaroma Xenografts in Athymic Mice Using PPMK107, PPNJROAKIN, or PPCONN70726.
In this experiment athymic mice (female, NCR nu/nu, 5 to 6 weeks old) received a subcutaneous injection of 10 HTIO8 tumor cells. Four days later when tumors reached a size range of 6 to 85 mm, mice wer treated intratumorally with saline, PPMK107 (at 1 x 10' PFU), PPNJROAIN (at 1 x10' PFU), or PPCONNO70726 (at I x 0L PFU). As shown in Table 23 below, tumor regmassion was noted in mice treated with these three viruses (PPMK107, PPNJROAKIN, and PPCONN70726). After PPMKI07 treatment of 12 mice, four experienced complete tumor regression and six experienced partial regression. After PPNJROAKN treatment of 12 mice, one mouse experienced complete tumor regression and !o two experienced partial regression. After PPCONN70726 treatment of 12 mice, three experienced complete tumor regression and two experienced partial regression. No tumor regression was noted in any of the animals created with saline.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:18 FAI 61 3 9039 2951 [a023 iIs 0 Tble 23. Relgression of HTIOSO180 Fibrosarwma Tumors in Athymic Mice After Treatment with One of Three Viruses (PPMK107, PPNJROAEIN and PPCONN7726) C Each at a Dose of Ix 10' PFU.
o Regession Treatment ohf ice Partial (PR) Complete (CR) PR CR PPMK107 126 4 10 (83%) PPNJROAICN 12 213(2 2 1 3 PPCONN70726 .12 2 3 5 (42%) Saline 11 00 0 0( 0%) Example 24 Effects of PPMK107, PPNJROAKIN, PPCONN70726 After Intracerebral Injection in Immunoddcient Athymkl (nu/nu) and Immunocompetent Hetermygote (auI+) Mice.
Fifty-six athymnic mice (nu/nu) and 56 immunocompetent heteroxygote mice were 0 given stereotaxic intracerebral injections with either saline, PPMIC07, PPNJROAICIN, or PPCONN70726. Eight additional mica of each type were used as untreated controls. Viruses were used at one of two dose levels (2 x 104 or 3.5 x 10" PFU/mousc). As shown in Table 24 below, all of the heterozygote nu/+ mice treated with each of the three viruses at the two dose levels survived through day 39 with the exception of one mouse at the lower PPCONN70726 dose level that was euthanized for non-neurological symptoms, Athymrnic nu/u animals treated with either PPMKI07 or PPCONN70726 had significantly less survival than the heterozygotes. This was especially true for the highest PPMK1O07 or PPCONN70726 virus dose of 3.5 x 10' PFT/mouse where only 13% (1 of 8) of the athymic animals in each virus group survived through day 39. In contrast, there was 75% survival of the PPNIROAIN- 3 treated athymic mice at this same dose level (3.5 x 106 PEL/mouse). These data indicate that PPNJROAKIN is better tolerated in the brains of athymic mice than the other two virus strains.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:18 FAX 61 3 9639 2951 024 Table 24. Survival of Mice Following Intracerebral Injection of PPMK17, PPC0NN70726, and PPNJROAKIN Intracranial Injection of Mice e% Survival at Day 39 nzU Untreated 8 100 nu/+ Saline 100 nul/ PPMK107, ZE+04 8 100 nu+ PPMK107, 3.SE+06 8 100 nu/+ PPCONN70726, 2E4.0 8 88 nU/t PPCONN7O726, 3.5E+06 8 100 nl/ PPNJROAlN, 2E+04 8 100 nu+ I PPNflOAKIN, 3.5E-06 8 100 nu/nu -ntreted 8 100 nufnu Saline 8 100 nulnu PPMX1O7, ZE+04 8 ntlnu PPMK107, 3.5Ei.06 8 13 nulnu PPC0NN70726, 2Ei04 8 flu/u PPCONN70726, 3.5Bs06 8 13 hu/lnu PPNJROAKIN, 2Et04 8 100 uu/nu PPNJROAKIN, 3.SE06 8 I no one rln-survivang moun in this tmtauent group was eurbanized for non-neurojogicaI symnptoms.
Example Summary of Cytotoxicity Assay Results with Sindbis Virus PPSINDBIS-Ar339 Human tumor cells and normal cells were gown to approximateLy 80% conflIuencC in 24 well tissue culture dishes. Growth medium was removed and PPSINDBIS-Ar339, a plaque a purified clone of Sindbis Ar-339 was added in 10 fold dilutions ranging from 10' plaque forming units (PFU)/well to 1 PF1/well. Control wells with no virus added were included on each plate. Virus was adsorbed for 90 minutes on a rocking platform at 37 0 C. At the end of the incubation period, the viral dilutions were removed and replaced by 1 ri of growth medium. Plates were then incubated for 5 days at 37rC in 5% C02. Cytotaxicity was COMB ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:18 FAX 61 3 9639 2951 1025 o quantified by using a colorimetric MIfT (2-[4,5-dimethylhiazol-2-yl]-2,5-diphenyI ttrazolium bromide) assay (Cell Titer 96, catalog #G4000, Promega Corporation, Madison SWI 53711) monitored at 570 nm, that detects mitochondrial enzyme activity (Moman, T, 1983, 1. Immuunol. Methods 65:55). The viability in the virus treated wells was expressed as a percent of the activity in untreated control wells. The data was plotted graphically as PFUwell vs. viability as a percent of control. The IC50 was calculated as the amount of virus in PFUIwell causing a 50% reduction in the amount of viable cells.
0 CN 0 Table 25. Summary of Cytotoxicity Assay Results with PPSINDBJS-Ar339 o Cell Type Cell Line I1 (PFU/wll) 0 Pancreatic Carcinoma Panc-1* 69 Colorectal Carcinoma SW620* 13 Colorectal Carcinoma SW1463 1.8 x10 Non-small cell Lung carcinoma A427 >1 x 106 Non-small cell Lung A549 5.2 x carcinoma Renal carcinoma A498 2.4 x 104 Renal carcinoma Caki-I 3.4 x Fibrosarcoma HT1080 74 x Normal Keratinocyte NREX 2.0 x Normal Fibroblast CCD922sZk 1.6 x Cells known to overexpress the mRNA for the high affinity lamrninin receptor.
The cellular receptor for Sindbis virus on mammalian coils is the high affinity laminin receptor, that is expressed mainly on cells of epithelial lineage, but is often overexpressed in many metastatic cancer cells like the Panc-I pancreatic carcinoma line, and the SW620 colon adenonocarcinoma cell line (Campo et al., (1992) Am. J. Pathol. 141, 1073-1083; Yow et al., (1988) Proc. Natl Acad Sci. 85, 6394-6398). In contrast, the rectal adenocarcinoma cell line SW 1463 is known to express very low levels of high affinity laminin receptor mRNA (Yow et al.,.(1988) Proc. Natl Acad Sci. 85, 6394-6398), and is more than 4 order of 0 magiitude more irsistant to killing by PPSNDBIS-Ar339 than SW620 cells. These results demonstrate that cells that are tumnorigenic and express high levels of the high affinity COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:19 FAX 61 3 9039 2951 12 lih 026 tt~) 76 o la~Iminin receptor are more sensitive to illing by Siudhia Clone PP$ jNDBs.Ar339 than other tumor or normal cells.
Examople 2 o 5 VSV Killin Of Twnorlgenic and Non-Tumorigenie Cells in the presenc, of Interferon.
In 96 well plates. tumerigenic XB and RT1OSO cells (3 x 10' cells per well) and nontumorigenic WISH cels (2.5 x 10' cells per well) were seeded in the presence of serially diluted interferon..u ranging from 28001to22 Units/ml and allowed to incubate for 24 hours 0 at 370C.* The cells were then infected with vesicular stomatifis virus (VSV, Indiana strain) at 0 0 an moi of 10. Controls were included for cells without Interferon, and cells without Ininterfeman or virus. The cells wer= incubated at 3-1 for 24 hours. Cytotoxicirty was quantified o by using a colorimetric NrM 2 4 9 5 -dimethithiazol-2.y11-2,5-iphenyl retrazoliurn bromide) assay (Cell Titer 96, catalg #G4000, Prornega Corpomaion, Madison WI 53711) monitored at 570 ri, that detects mirochondrial enzyme activity (Moanian, 1983, J.
Irnmunol. Methods 65.,55), The viability in the vim~j treated wells was expressed as a Percent of die activity in control wells not receiving virus.
Table 26. Comparison of the Cell Killing Activity or VSV in Cells Treated with Exogenous [uterferom Percent Viable Calls WISH H'FIISG KE 0OU/mW ON 0 0 0 ITO-UiDt 50 6 0 1000 UWill)hIN 95 20 12 0 These results demonstrate thatWViS is able to selectively Idl tumor cells deficient in interferon responsiveness (see Example 27). WISH cells (hwnan aminion cells) are a well established call line for the use in interferon bioassays because of their ability to respond efficiently to interferons.
S Exime 27 In terferon Reapaivme in Celia Sensitive. or Resistant to Kiling by PPMK1O7.
IUidividual cell lines were grown to nea confluencein 96 well microffier plates and treated with between 5 and 5000 U/rd of IF~rxA for 24 hours. The cultures were then infected with PPMJC1O7 at an moi of 1.0 and cultured for an additional 24 hours. Following COMS ID No: SBMI-O1 158845 Received by IF Australia: lime 17.21 Date 2005-03-10 10/03 '05 17:19 FAX 61 3 9639 2951 21027 0 chemical fixation, the amount of viral expression was quantified by immunohistochemistry using a soluble indicator dye. The amount of virus growth is represented as the percent of P antigen expsmsed relative to control cells untreated with interferon (Figure In this assay, interferon responsive cells manifest at least a 50% decrease in the viral antigen in response to interferon.
The results of this experiment show a strong correlation between the resistance of the cell line to the antiviral effects of exogenous interferon and the relative sensitivity of the cell to o killing by PPMKI07 (indicated by the IC50 value shown in parentheses next to the cell line Sname in the graph legend, see Figure For example, following pretreatment with 5 U/ml of a interferon, 6 of 7 cell lines nonresponsive to interferon are sensitive to killing by SPPMK107; when pretreated with 500 U/ml of interferon, all (4 of 4) of the nonresponsive Cl cell lines are sensitive to killing by PPMK107.
The data above also present an example of a screening assay to identify candidate cells that are likely to be sensitive to killing by PPMKIO7 or other interferon-sensidve viruses.
For example, infected cells expressing significant more than 50% of controls) viral antigen following pretreatment with exogenous Interferon would be considered interferon deficient and thereby sensitive to viral oncolysis.
Use of a Desensitizing Intravenous Dose of PPMK107 to Reduce the Lethality of a Subsequent Intraperitoneal Dose of PPMKI07.
Mice were injected intravenously on day 0 with either saline or a desensitizing dose of PPMKI07 (3 x 108 PFU/mouse). Two days later each set of mice were further subdivided into groups for intraperitoneal dosing with saline or PPMK107 (at doses of 1 x 109, 2.5 x 109 is x 109, and 1 x 1010 PFU/mouse). As shown in Table 27 below, when saline was used to pretreat the mice, deaths were recorded in the mice subsequently dosed with 2.5 x 10 5 x 109, and 1 x 10 10 PFU. The doses of 2.5 x 10', 5 x 10 and l x 10 0 PFU were 25%, and 100% lethal, respectively, to the mice pretreated with saline. In contrast, using an IV desensitizing dose, no deaths were seen in the 2.5 x 109 and 5 x 109 PFU groups and only a 38% mortality was seen in the I x 1010 PFU group of mie. These data indicate that PPMK107 given by the intravenous route can be used to desensitize the toxicity of subsequent dosing of another route (namely in this example, the intraperitoneal route) with COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:19 FAX 81 3 9639 2951 ___il2 12)028 0 0 ci 0 0 0 ci 0 0 ci 73 this sams agefti. Furthennore, the maximal tolerated innrpcritoneal dose of PPIvtK1O7 can be raised by approximately 5-fold when using this virus as a desensitizing agentL Table 27. Usme of a Desensitizing Intravenous Dose of PPMK1O7 to Reduee the S Lethality or a Subsequent Intraperitoneal Doe of PPMIK1O1.
Goup IV Injection on [P Dose onDay 2 Of *tOf Day 0 Mece Deaths Letlity I Saline Saline a 0 0 2 Saline PPMJC1O7, LOE+09 8 0 0 3 Saline PPMKIO7, 2.SE-i-0 8 2 4 S aline PPMKIIJ7, 5.02409 8 4 so Saline PPMX1O7, 1.OE13-10 8 8 100 6 PFMK 107. 3E+08 Saline 8 0. 0 7 PPMX 107, 3E-i08 PPMv.K107, 1.OE±09 8 0 0 8 PPMXIO7, 3E-8 PIPMKO7, 2.5M)09 8 0 0 9 PPMKIO7. 3E-t-8 PPMKIOD7. 5.013+09 j_ S 0 PPWCIO7, 3E-08 PPMK1O7, 1.OE-i10 18 13 38 Use of a Desiensitizing Intravenous Daze of PPMK107 to Increas the Antitumrni~ Efficacy a Subsequent Inlraperitoneal Done of PPMKIO7.
In this example, athymic mice with visually apparent ascites tumors derived from the ES- 2 human ovarian carcinoma, cell line were tireated with incraperironeally with PFMKI07 either with our without prior intravenous desensitization two days previ ously with 3 x 108 PP/uiouac. As shown in Table 25, control mice treated innpeditoneally with saline rapidly died from ascites tumors with only 8% surviving by day 9 post treatment. Administering the maximum tolerated dome of PPMK107 inraperlconeally increased the siurvival Percentage to 46% at day 9. Using IV daeuicization, higher IP doses of PPMK1O7 such as 2.5 x 10p and x L69 PFU can be tolerated and lead to almnost a doubling of survival rate (83% and 79%, respetivYely) when compared to th-at achievable with the highest dose (1.0 xt 10'P PFU) without desensiuization.
COMS ID Na:SBMI-0115884 Received by IP Australia: Time (I-tm) 17:21 Date 2005-03-10 10/03 '05 17:19 FAX 61 3 9639 2951 1029 0 0 ci 0 0 0 ci 0 0 ci 79 Tahble 28. Survival of Mice with ES-2 Human Ovarian Carcinoma Aseites Tumors after Intraperitoneal PPMX07 Treatment P Treatment Survival (Day 9) Survival (Day 9) Group Without IV With IV Desensitization (PFUmoue) Desensitiation Saline 2 out of 24 3 out of 24 (13%) FA-E09 11 out of 24 11 out of 24 (46%) 2.5 E-09 a Dose Above MTD 20 out of 24 (83%) 5.0 E+09 a Dose Above MT 19 oat of 24 (79%) a- Dose above KTM without desensidzation but below MTD with desensiization.
Example Anti.Recnmbinant Murine TNF-alpha Antiserum Blocks IV Lethality of PPMK107 in Mice In this example, groups of 16 female C57BL/6 mice were dosed intravenously with 5.0 x PFU of PPMK107. Five hours previously different groups of 16 mice received either anti- 0 recombinant murine TNF-alpha antiserumn (100 ul from rabbits diluted 1:10 with saline), the equivalent amount of control rabbit serum or aline. As shown in Table 29 below, the lethality of PPMK107 at this dose is completed blocked using the rabbit antiserum against murine TNF-alpha.
S Table 29. Mortality of Mice Pretreated with Anti-Recombinant Murine TNFAlpha Antiserum Before IV Dosing with 5.OEFA-09 of PPMK107.
a- Monality siificanly Exact Test).
saline and rabbit serum contral neannet oups (P 0.05, Fisher's COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:20 FAX 61 3 9639 2951 @030 0 0 Cl Example 31 Purification fClonal Viruses.
A number of clonal RNA viruses were purified by either ultracentrifugation of the virus o 5 without pelleting or by sequential tangential flow filtration. For purification by one of the following methods: Method 1: As in Method A ofExaple Method 1: As in Method A of Example o io Method 2: As in Method D of Example o Method 3: Vero cells at approximately 70% confluence were infected at an moi of 0.01 and incubated at 37 C C for 18 hours. The flasks containing the infected cells were frozen at The cell were thawed at room temperature and then maintained on ice until harvest.
For harvest, the cells were scraped into the media present during the infection and clarified at 1750 x g for 30 minutes at 4°C. The clarified supernatams were pooled and layered over a 20%/55% discontinuous gradient and centrifuged at approximately 100.000 x g for minutes. The purified virus was harvested from the sucrose 20%/55% interface and dialyzed against calcium- and magnesium-free PBS to remove the sucrose, !0 The activity of the virus preparations was determined by the plaque assay method using cells for Newcastle disease virus and Vero cells for Vesicular stomatitis strains. The protein content of the virus preparations was determined using the NanoOrange Protein Quantification Kit (Molecular Probes, Inc., Eugene, OR) with bovine serum albumin as the protein standard.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:20 FAX 81 3 9039 2951.03 1603.1 81 Table 30. Specific Activity of a Number of Clonal RNA Viruse Purified by Ultraceatrlfdgaion without Pouting or Tangential Flow Fltratlon.
ViruS Virus Clone, Purl~cati Specific on Activity Method (PFUmg Newcastle diseasa-MI107 PPMK107 I 1.3E-i11 Newcastle diase-MKJO7 PPMX107 2 1-O11N 1 strain Newcastle disease.Roakin strain PPNJROAKIN I Newcastle discase-ConnH strain PPCONN7O726 1 2.IE+10 Vesicular Stomidis-Lniiana PPVSV 3 4.5TE+9 strain These meulus dernonzate the ability to purify different clonal RNA viruses to high specific- activity using the Methods. described in. this invention.
Sindibis VIMu PPSINDBJS.Ar339 Causes Tumor inhibition in Athyusic Mice with Human Tumor Cell Xenograft Atbymnic mice were injected subcutaneously with 10 million SW620 human Adenocarcinonia tumor cells. Five days later the wmiors (average size =78 mni') were Creatd with a single injection Of PPSINDBIS-M339 (10 mice, 5 x 104 PFU) or saline (9 mice).
Tumor size and mouse weighr were measured twice per week util study termination. By the twelfth day After treatmenr the average tumor size of the saline treated mice had increased by ant average of 896% (from 71.3 mi 3 to 639.1 mml COMS ID No: SBMI1-01158845 Received by IP Austrafla: Time 17:21 Date 2005-03-10 10/03 '05 17:20 FAX 61 3 9839 2951_ 032 Table 31. Intratumoral treatment of SW620 Colon Adenocarcinoma Human Xenografts PPSINDBIS-Ar339.
O
0 0 t-( c-i 0 0 i Time Post Tumor Growth Treatment Inhibition" 89% 9 days 12 days 87% tumor growth inhibition increase control group increase Tx eroup X 100 increase control group These data show that a single injection of PPSINDBIS-Ar339 results in significant tumor growth inhibition compared to treatment with saline. Treatment with PPSINDBIS-Ar339 was also well tolerated by the mice as evidenced by the absence of weight loss in either the saline 3 or virus treated groups.
Example 33 Viruses Belonging to Unrelated Families Have Similar Activity on Human Tumor Cell Lines.
Cytotoxicity assays were performed with the SW620 adenocarcinoma cell line and PPMK107 (Paramyxovirus family) or PPSINDBIS-Ar339 (Togavirus family) as described in Examples I and 25 (see Fig. These two unrelated viruses use distinct receptors to enter the host cell and replicate by mechanisms unique to each virus. Nevertheless, the response of the SW620 cell line to infection by these two viruses is remarkably similar indicating that the a mechanisms of tumor cell killing by these viruses share common elements.
Example 34 Use of PPMK107 in Combination with Chemotherapy for the Systemic Treatment of Human Tumor Xenografts in Athymic Mice Athymic mice were injected subcutaneously with 10 million human HT1080 fibrosarcoma cells. After tumors reached a size range from between 5 and 7 mm, mice were randomized into treatment groups. Mice received intraperitoneal Injections saline vehicle on treatment days 0, 2 and 4. PPMK107 at a dose of 2 x 101 PFU or saline vehicle was administered by COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:21 FAX 61 3 9639 2951 [a033 intravenous injection on treatment day 2 only, one hour after the intrperitoneal injection.
Caroboplatin (carbo) at a dose of 160 mg/kg was administeed by intraperitoneal injection on treatment day 0, 2, or4 as indicated. The percent of each group with complete regression (CR) and partial regression (PR) ii shown. Each tretmuent grup had 9 tumor-bearing mie.
Table 32. PPMK107 in Combination with Carboplatin for the Systemic TreatmeCit of Subcutaneous Human HT1iOBO Fibrosarcoma Xenografts In Athymic Mice.
Treatment CR+PR Saline, days 0,2 and 4 0
OO
(N PPMX1O7, day 2 44 Carbo, day 0 0 Carbo, day 2 11 Carbo, day 3 11 PPMK07, day 2 Carbo, day 0 67 o 0, PPMK107, day 2 Carbo, day 78 PPMK107, day 2 Carb, day 4 These results in Table 32 indicate that subcutaneous HT1OS80 tumors are responsive to IV .0 treatment with PPMK107 and that addition of carboplazin two days before, the same day, or two days after PPMR107 treatment resulted in a higher percentage of tumor regression (CR PR) than either PPMI107 alone or carboplatin alone.
Example Second Experiment on the Use of PPMK107 In Combination with Carboplatin for the Systemic Treatment of Buman HT1080 Tumor Xenografts in Athymic Mice Athymnic mice were injected subcutaneously with 10 million human 1T1080 fibrosarcoma cells as in Example 34. Mice received intraperitoneal injccdons caroboplatin (carbo) at a dose of 80 mg/kg or 120 mg/kg or saline vehicle on treatment day 0 followed by intravenous injection of PPMKI107 (at 6 x 10 or 2 x 10' PFULT) on creatment day 2. The percent of each COMS 0ID No: SBM 1-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:21 FAX 01 3 0030 2951 j04 [a 0.34 0 0 ci 0 0 0 ci 0 0 ci 34 grOUQ with complete regression (CR) and partial regression (PR) is shownl Each treatment group had 9 tumor-bearing mice.
Table 33. PPMKIO7 in Combination With Carboplatin for the Syistemiic Treatment Of Subcutaneous Human HTZO8Q Hlbrosarcma Xenografts in Athymic Mic.
Treament Doe a-+Pn PPNM07 6L.06 PFU 22 PPMK107 2E+07 PFU 56 Carbo 80mg/kg 33 Carbo l1flmg/kg 22 PPMK1076E+06 PFt) Carb 80 glkg78 Carbo 10 mg/kg 4 PPMIK10dEi.07 PEU 6 Carbo, 80 mg/kg PPMK107 2E+07 PFU Carbo 120 mg/kg 0 These results in Table 33 indicate that subcutaneows H 1080 tumors arm responsive to rV treatmientE wit PPMX1O7 at each dose level and that addition of carboplarin at either dose level two days before PFMKIO07 treaUnent reulted in a higher percentage of tumor regression 2(CR PR) than either PPMKI07 alone or carboplatin alon.
Example 3A Third Erpwerint on the Use of PPNM0X7 in Combination with Carbopiatin for the Systemic Tireatment of Human ff121080 Tumor Kenografa. in Atbymlc Me Athymic mice were injected subcutaneously with 10) million huan HT1OSO fibrosarcoina cells as in Example 34. M~ice received incraperitoneal injections caroboplacin (carbo) at a dose of 60 mg/kg or Waine vehicle on taumnn dayo0 followed bfy intravenous injection of COMS ID No: SBMI-01158845 Received by IP Australia: lime 17:21 Date 2005-03-10 10/03 '05 17:21 FAX 61 3 9639 2951 1035 as O PPMK107 (at 6 x 10 6 PFU) on treatment day 2. The percent of each group with complete regression (CR) and partial regression (PR) is shown. Each treatment group had 9 tumori bearing mice except for the carboplatin only group which had 8 mice.
Table 34. PPMK107 in Combination with Carboplatin for the Systemic Treatment of Subcutaneous Human HT1080 Fibrosarcoma Xenografts in Athymic Mice.
Treatment CR+PR 0 SSaline 0 Ci PPMK107 44 Carbo PPMK107 89 Carbo These results in Table 34 indicate that subcutaneous HT1080 tumors are responsive to IV treatment with PPMK107 and that addition of carboplatin two days before PPMK107 to treatment resulted in a higher percentage of tumor regression (CR PR) than either PPMK107 alone or carboplatin alone.
Example 37 Use of a Corticosteroid (Dexamethasone) to Reduce the Lethality of an Intravenous Dose of PPMK107.
Female athyrnic mice (six to seven weeks old) were injected intraperitoneally on days 0, 1, 2, 3 and 4 with either dexamethasone (one group at a dose of 25 mg/kg and another group at mg/kg) or saline. All animals were given an intravenous dose of PPMK107 (3 x 109 PFU/mouse) on day 2 (one hour following the IP dose of dexamathasone or saline). Mice were observed and the lethality tabulated in Table 33 below.
COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10 10/03 '05 17:21 FAX 61 3 9639 2951 86 0 Table 35. Use ofDexamethasone to Reduce the Lethality of an Intravenous Dose of PPNM17.
IP Treatment on Group Tat IV Treatment on Day 2 Lethality Days O,12,3,aud 4 Dexamethasone 1 PPMK107, 3.OE+09 7 mgfkg) Dnxamethasone 2 PPMK107, 3.OE09 0 mg/kg) o 3 Saline PPMKI07, 3.OEi09 67 0 SThe PPMK107 dose of 3 x 109 was lethal to 67% of the mice given saline control IP dosing. Dexamethasone markedly reduced lethality due to PPMK107 with only 7% mortality observed in the animals given 25 mgfkg and 0% mortality observed in the animals given mgfkg. These data indicate that a corticosteroid like dexamethsone can be used to reduce the toxicity of an intravenous dose of PPMI1O7.
0 The foregoing is intended as illustrative of the present invention but not limiting.
Numerous variations and modifications may be effected withom departing from the true spirit and scope of the invention.
G1030 COMS ID No: SBMI-01158845 Received by IP Australia: Time 17:21 Date 2005-03-10

Claims (24)

1. A method of infecting a neoplasm in a mammal comprising administering an 0interferon-sensitive, replication-competent clonal RNA virus, wherein said virus is to be administered over a period of at least 20 minutes.
2. The method according to claim 1, wherein said virus is to be administered at a dose of at least 5.9 X 10 9 PFU/m 2
3. The method according to either claim 1 or claim 2, wherein said virus is to be CN administered at a dose of at least 9.6 X 1010 PFU/m 2
4. The method according to any one of claims 1 3, wherein said virus is at a level of purity of at least 3 X 10 9 PFU/mg protein. The method according to any one of claims 1 4, wherein said infecting comprises administration of a first dose and at least one subsequent dose of said virus, and wherein the first dose is a desensitizing dose to be administered intravenously and the at least one subsequent dose is to be administered intravenously, and wherein said subsequent intravenous dose is at least 4.8 X 1010 PFU/m 2
6. The method according to claim 5, wherein said subsequent dose is at least 9.6 X 1010 PFU/m 2
7. The method according to any one of claims 1 6, wherein the virus is selected from the group of families consisting of Paramyxoviridae, Orthomyxoviridae, Rhabdoviridae, Togaviridae, Picomaviridae, Coronaviridae, and Reoviridae.
8. The method according to claim 7, wherein said Togavirus is a Sindbis virus.
9. The method according to claim 7, wherein said Rhabdovirus is a vesicular stomatitis virus (VSV).
10. The method according to claim 7, wherein said Paramyxovirus is a Newcastle disease virus (NDV). 00 88
11. The method according to any one of claims 1 10, wherein said virus is to be administered over a period of time between 20 to 60 minutes.
12. The method according to any one of claims 1 6 and 11, wherein said virus is a single stranded RNA virus. S13. The method according to any one of claims 1 12, wherein said interferon- sensitive, replication-competent clonal RNA virus is to be administered systemically. S14. The method according to any one of claims 1 13, wherein said neoplasm has been screened for protein, or mRNA encoding protein, selected from the group consisting of p68 protein kinase, C-Myc, C-Myb, ISGF-3, IRF-I, IFN receptor, and p58, Ras overexpression, Ras with activating mutations, overexpression of other mediators of the Ras signalling pathway and unregulated receptor tyrosine kinases. The method according to any one of claims 1 14, wherein said interferon- sensitive, replication-competent clonal RNA virus is to be administered over multiple courses.
16. The method according to any one of claims 1 15, wherein said interferon- sensitive, replication-competent clonal RNA virus is to be administered greater than two weeks after a first administration of the interferon-sensitive, replication-competent clonal RNA virus.
17. The method according to any one of claims 1 16, wherein an inhibitor of a TNF is to be administered before, during or after administration of said virus.
18. The method according to any one of claims 1 17, wherein an antibody to a TNF is to be administered before, during or after administration of said virus.
19. The method according to any one of claims 1 18, wherein said infecting comprises administration of a first dose and at least one subsequent dose of said virus, and wherein the first dose is a desensitizing dose to be administered 00 c intravenously, and wherein said subsequent intravenous dose is at least two-fold Sgreater than said first dose. The method according to any one of claims 1 19, wherein a desensitizing agent is to be administered before said virus.
21. The method according to claim 20, wherein said desensitizing agent is a TNF or an inducer of a TNF.
22. The method according to claim 20, wherein said desensitizing agent is an IL-2, C an endotoxin or another virus.
23. The method according to any one of claims 1 22, wherein a platinum coordination complex chemotherapeutic agent is to be administered before, during or after administration of said virus.
24. The method according to claim 23, wherein said platinum coordination complex is carboplatin.
25. The method according to any one of claims 1 24, wherein a corticosteroid immunosuppressant is to be administered before, during or after administration of said virus.
26. The method according to claim 25, wherein said corticosteroid is a dexamethasone.
27. Use of an interferon-sensitive, replication-competent clonal RNA virus in the manufacture of a medicament for infecting a neoplasm in a mammal, wherein said virus is to be administered over a period of at least 20 minutes.
28. A method of infecting a neoplasm in a mammal comprising administering an interferon-sensitive, replication-competent clonal RNA virus, substantially has herein described with reference to any one or more of the Examples and/or accompanying Figures. 00
29. Use of an interferon-sensitive, replication-competent clonal RNA virus in the Smanufacture of a medicament for infecting a neoplasm in a mammal, substantially has herein described with reference to any one or more of the Examples and/or accompanying Figures.
AU2005201079A 1999-04-15 2005-03-10 Treatment of neoplasms with viruses Ceased AU2005201079C1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2005201079A AU2005201079C1 (en) 1999-04-15 2005-03-10 Treatment of neoplasms with viruses

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09292376 1999-04-15
AU42469/00A AU4246900A (en) 1999-04-15 2000-04-17 Treatment of neoplasms with viruses
AU2005201079A AU2005201079C1 (en) 1999-04-15 2005-03-10 Treatment of neoplasms with viruses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU42469/00A Division AU4246900A (en) 1999-04-15 2000-04-17 Treatment of neoplasms with viruses

Publications (3)

Publication Number Publication Date
AU2005201079A1 AU2005201079A1 (en) 2005-04-07
AU2005201079B2 AU2005201079B2 (en) 2008-03-13
AU2005201079C1 true AU2005201079C1 (en) 2008-11-06

Family

ID=34397430

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005201079A Ceased AU2005201079C1 (en) 1999-04-15 2005-03-10 Treatment of neoplasms with viruses

Country Status (1)

Country Link
AU (1) AU2005201079C1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AUPQ425699A0 (en) 1999-11-25 1999-12-23 University Of Newcastle Research Associates Limited, The A method of treating a malignancy in a subject and a pharmaceutical composition for use in same
AU2002953436A0 (en) 2002-12-18 2003-01-09 The University Of Newcastle Research Associates Limited A method of treating a malignancy in a subject via direct picornaviral-mediated oncolysis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994025627A1 (en) * 1993-04-30 1994-11-10 Lorence Robert M Methods of treating and detecting cancer using viruses
WO1999008692A1 (en) * 1997-08-13 1999-02-25 Oncolytics Biotech, Inc. Reovirus for the treatment of neoplasia

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994025627A1 (en) * 1993-04-30 1994-11-10 Lorence Robert M Methods of treating and detecting cancer using viruses
WO1999008692A1 (en) * 1997-08-13 1999-02-25 Oncolytics Biotech, Inc. Reovirus for the treatment of neoplasia

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Cancer Detection and Prevention (1993) 17(6): 619-627 *
Cancer Research (1994) 54: 6017-6021 *
J Natl Cancer Inst (1988) 80(16): 1305-1312 *
J Natl Cancer Inst (1994) 86(16): 1228-1233 *

Also Published As

Publication number Publication date
AU2005201079A1 (en) 2005-04-07
AU2005201079B2 (en) 2008-03-13

Similar Documents

Publication Publication Date Title
CA2370187C (en) Treatment of neoplasms with viruses
US8105578B2 (en) Treatment of neoplasms with viruses
US7780962B2 (en) Treatment of neoplasms with RNA viruses
CA2305269C (en) Treatment of neoplasms with viruses
Lorence et al. Phase 1 clinical experience using intravenous administration of PV701, an oncolytic Newcastle disease virus
US9803216B2 (en) Genetically-engineered newcastle disease virus as an oncolytic agent, and methods of using same
JP5060694B2 (en) Oncolytic virus
CA2161671A1 (en) Methods of treating and detecting cancer using viruses
WO2008140621A2 (en) Transgenic oncolytic viruses and uses thereof
US7470426B1 (en) Treatment of neoplasms with viruses
CN109276580A (en) It is a kind of for treating the virus of tumour
AU2005201079C1 (en) Treatment of neoplasms with viruses
AU2005237176B2 (en) Treatment of neoplasms with viruses
AU2002325406B2 (en) Treatment of Neoplasms with Viruses
MXPA00003467A (en) Treatment of neoplasms with viruses
Nakhaei et al. Oncolytic virotherapy of cancer with vesicular stomatitis virus
Ohnesorge New approaches in the therapy of NUT carcinomas (NCs) involving immunovirotherapy and BET-protein inhibitors

Legal Events

Date Code Title Description
DA2 Applications for amendment section 104

Free format text: THE NATURE OF THE AMENDMENT IS: AMEND THE NAME OF THE COINVENTOR TO READ FROM BON BORSTEL, REID W. TO VON BORSTEL, REID W..

DA2 Applications for amendment section 104

Free format text: THE NATURE OF THE AMENDMENT IS: REMOVE CO- INVENTOR: BON BORSTEL, REID W..

DA2 Applications for amendment section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 19 JUN 2008.

DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: AMEND THE NAME OF THE INVENTOR TO READ: FROM: BON BORSTEL, REID W. TO: VON BORSTEL, REID W.

DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: REMOVE CO-INVENTOR: VON BORSTEL, REID W.

DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 19 JUN 2008

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired