WO2015017915A1 - Procédés de traitement de sujets naïfs pour les taxanes présentant des tumeurs primaires ou un cancer métastasique - Google Patents

Procédés de traitement de sujets naïfs pour les taxanes présentant des tumeurs primaires ou un cancer métastasique Download PDF

Info

Publication number
WO2015017915A1
WO2015017915A1 PCT/CA2014/000608 CA2014000608W WO2015017915A1 WO 2015017915 A1 WO2015017915 A1 WO 2015017915A1 CA 2014000608 W CA2014000608 W CA 2014000608W WO 2015017915 A1 WO2015017915 A1 WO 2015017915A1
Authority
WO
WIPO (PCT)
Prior art keywords
administered
subject
reovirus
amino acid
residue
Prior art date
Application number
PCT/CA2014/000608
Other languages
English (en)
Inventor
Matthew C. Coffey
Bradley G. Thompson
Jill Thompson
Thomas MULLIE
Original Assignee
Oncolytics Biotech Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oncolytics Biotech Inc. filed Critical Oncolytics Biotech Inc.
Publication of WO2015017915A1 publication Critical patent/WO2015017915A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/765Reovirus; Rotavirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • C12N2720/12211Orthoreovirus, e.g. mammalian orthoreovirus
    • C12N2720/12222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2720/12211Orthoreovirus, e.g. mammalian orthoreovirus
    • C12N2720/12232Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent

Definitions

  • Cancer is one of the leading causes of death. Although it has long been the focus of medical research, the main cancer therapies to date remain surgery, radiation therapy and chemotherapy. Each one of these therapies is subject to limitations that are not currently overcome including, for example, the differing effects of the same therapy to subjects with similar types of cancer.
  • a method of treating cancer in a subject comprising selecting a taxane naive subject with a primary tumor (e.g., head and neck cancer, lung cancer, liver cancer, lymphoma, kidney cancer or ovarian cancer) and administering to the subject a therapeutically effective amount of an oncolytic virus.
  • a primary tumor e.g., head and neck cancer, lung cancer, liver cancer, lymphoma, kidney cancer or ovarian cancer
  • kits for treating metastatic cancer in a subject comprising selecting a taxane naive subject with a metastatic cancer and administering to the subject a therapeutically effective amount of Carbo-Tax.
  • the methods further comprise administration of an oncolytic virus.
  • a method of treating cancer in a subject comprising selecting a taxane na ' ive subject with a primary tumor and administering to the subject a therapeutically effective amount of an oncolytic virus.
  • the method further comprises administration of Carbo-Tax to the subject.
  • the Carbo-Tax optionally comprises carboplatin and paclitaxel.
  • the provided methods include the selection of a certain class of subjects, specifically, subjects that are taxane naive with a primary tumor.
  • taxane naive subject refers to a subject that have never received taxane administration, e.g., paclitaxel, larotaxel, or docetaxel.
  • the selection of the taxane naive subject advantageously results in an increase in efficacy of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more if efficacy of the oncolytic virus as compared to the use of the oncolytic virus in the absence of selection of a taxane na ' fve subject.
  • the cancer is, for example, head and neck cancer, lung cancer, liver cancer, lymphoma, kidney cancer or ovarian cancer.
  • Cancer occurs at an originating site, e.g., breast, liver or ovary, which site is referred to as a primary tumor, e.g., primary liver cancer.
  • the provided methods comprise selecting subjects with a primary tumor.
  • the primary tumor or cancer is not metastatic.
  • the provided methods can include selection of taxane na ' ive subjects with non-metastatic cancer.
  • metastatic refers to the spread of a proliferative disease or disorder, e.g., cancer, from one organ or another non-adjacent organ or body part.
  • a second clinically detectable tumor formed from cancer cells of a primary tumor is referred to as a metastatic or secondary tumor.
  • subjects with cancer that is not metastatic or subjects with non-metastatic cancer refers to subjects having a primary tumor and not one or more secondary tumors.
  • the provided methods herein include surgical removal of the primary tumor prior to administration of the oncolytic virus.
  • the primary tumor is not surgically removed prior to administration of the oncolytic virus.
  • the cancer is ras-activated.
  • the cancer is characterized by interferon-resistance, p53 -deficiency, or Rb-deficiency.
  • the provided methods optionally, further comprise the step of selecting a subject with a primary tumor or cancer of a particular phenotype.
  • the provided methods further comprise the step of selecting a subject with ras-activated cancer.
  • the provided methods can include selecting a taxane naive subject with a cancer and further selecting subjects from this group with ras-activated cancer.
  • the provided methods comprise the step of diagnosing the phenotype of the cancer or primary tumor, for example, by determining whether the cancer is a ras-mediated cancer.
  • the provided methods comprise the step of determining whether the cancer is an interferon-resistant tumor, p53 deficient tumor or an Rb-deficient tumor.
  • Such methods for determining whether cancer has a certain phenotype are known. See, for example, U.S. Patent No. 7,306,902, which is incorporated herein by reference in its entirety.
  • the method includes selecting a taxane naive subject with a metastatic cancer and
  • the Carbo-Tax comprises carboplatin and paclitaxel.
  • the provided methods include the selection of a certain class of subjects, specifically, subjects that are taxane naive and who have metastatic cancer.
  • taxane naive subject refers to a subject that has never received taxane administration, e.g., paclitaxel, larotaxel, or docetaxel.
  • the selection of the taxane naive subject advantageously results in an increase of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in efficacy of the Carbo-Tax as compared to the use of Carbo-Tax in the absence of selection (e.g., in subjects previously exposed to taxane).
  • the provided methods include the selection of taxane naive subjects followed by the further selection of taxane naive subjects with metastatic cancer.
  • the selection of the taxane naive subjects with metastatic cancer results in an increase of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in the efficacy of the Carbo-Tax as compared to a control.
  • the efficacy of Carbo-Tax in taxane naive subjects with metastatic cancer is higher as compared to the efficacy of Carbo-Tax in taxane naive subjects with primary tumors (i.e., non-metastatic cancer).
  • an increase in efficacy can include a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% reduction or amelioration in the severity of an established disease or condition or symptom of the disease or condition.
  • the metastatic cancer is, for example, metastatic lung cancer, metastatic liver cancer, metastatic lymphoma, metastatic bone cancer or metastatic ovarian cancer.
  • the term metastatic refers to the spread of a proliferative disease or disorder, e.g., cancer, from one organ or another non-adjacent organ or body part. Cancer occurs at an originating site, e.g., breast, liver or ovary, which site is referred to as a primary tumor, e.g., primary liver cancer.
  • a second clinically detectable tumor formed from cancer cells of a primary tumor is referred to as a metastatic or secondary tumor.
  • the metastatic tumor and its cells are presumed to be similar to those of the original tumor.
  • the secondary tumor at the site of the lung consists of abnormal liver cells and not abnormal lung cells.
  • the secondary tumor in the lung is referred to a metastatic liver cancer.
  • metastatic cancer refers to a disease in which a subject has or had a primary tumor and has one or more secondary tumors.
  • non-metastatic cancer or subjects with cancer that is not metastatic refers to diseases in which subjects have a primary tumor but not one or more secondary tumors.
  • metastatic breast cancer refers to a disease in a subject with or with a history of a primary breast tumor and with one or more secondary tumors at a second location or multiple locations, e.g., in the lung.
  • the provided methods herein include surgical removal or other nontaxane treatment of the primary tumor prior to
  • the metastatic cancer is ras-activated.
  • the metastatic cancer is characterized by interferon-resistance, p53 -deficiency, or Rb-deficiency.
  • the provided methods optionally, further comprise the step of selecting a subject with metastatic cancer of a particular phenotype.
  • the provided methods further comprise the step of selecting a subject with ras-activated metastatic cancer.
  • the provided methods can include selecting a taxane naive subject with a metastatic cancer and further selecting subjects from this group with ras-activated metastatic cancer.
  • the provided methods comprise the step of diagnosing the phenotype of the metastatic cancer, for example, by determining whether the metastatic cancer is a ras-mediated metastatic cancer.
  • the provided methods comprise the step of determining whether the metastatic cancer is an interferon- resistant tumor, p53 deficient tumor or an Rb-deficient tumor.
  • Such methods for determining whether cancer has a certain phenotype are known. See, for example, U.S. Patent No. 7,306,902, which is incorporated herein by reference in its entirety.
  • the provided methods further comprise administering an oncolytic virus to the subject.
  • Oncolytic viruses that are used in the provided methods include, but are not limited to, oncolytic viruses that are members in the family of myoviridae, siphoviridae, podpviridae, teciviridae, corticoviridae, plasmaviridae, lipothrixviridae, fuselloviridae, poxyiridae, iridoviridae, phycodnaviridae, baculoviridae, herpesviridae, adnoviridae, papovaviridae, polydnaviridae, inoviridae, microviridae, geminiviridae, circoviridae, parvoviridae, hepadnaviridae, retroviridae, cyctoviridae, reoviridae, birnaviridae, paramyxoviridae, r
  • the oncolytic virus used in the provided methods is, for example, selected from the group consisting of a reovirus, a Newcastle disease virus (NDV), a vesicular stomatitis virus (VSV), an adenovirus, a vaccinia virus, a parapox orf virus and a herpes simplex virus.
  • NDV Newcastle disease virus
  • VSV vesicular stomatitis virus
  • adenovirus adenovirus
  • vaccinia virus a vaccinia virus
  • parapox orf virus a herpes simplex virus
  • a combination of at least two oncolytic viruses can also be employed to practice the provided methods.
  • a few oncolytic viruses are discussed below, and a person of ordinary skill in the art can practice the present methods using additional oncolytic viruses as well according to the disclosure herein and knowledge available in the art.
  • RNA Kinase RNA Kinase
  • VA1 RNA double-stranded RNA Kinase
  • VA1 RNA has extensive secondary structures and binds to PKR in competition with the double-stranded RNA (dsRNA) which normally activates PKR. Since it requires a minimum length of dsRNA to activate PKR, VA1 RNA does not activate PKR. Instead, it sequesters PKR by virtue of its large amount. Consequently, protein synthesis is not blocked, and adenovirus can replicate in the cell.
  • dsRNA double-stranded RNA
  • Ras-activated neoplastic cells are not subject to protein synthesis inhibition by PKR because ras inactivates PKR. These cells are therefore susceptible to viral infection even if the virus does not have a PKR-inhibitory system. Accordingly, if the PKR inhibitors in adenovirus, vaccinia virus, herpes simplex virus, or parapoxvirus orf virus are mutated so as not to block PKR function anymore, the resulting viruses do not infect normal cells due to protein synthesis inhibition by PKR, but they replicate in ras- activated neoplastic cells which lack PKR activities. By way of example, reoviruses selectively replicate and lyse ras-activated neoplastic cells.
  • the oncolytic virus is an adenovirus mutated in the VA1 region, a vaccinia virus mutated in the K3L and/or E3L region, a vaccinia virus mutated in the thymidine kinase (TK) gene, a vaccinia virus mutated in the vaccinia growth factor (VGF) gene, a herpes virus mutated in the ⁇ 134.5 gene, a parapoxvirus orf virus mutated in the OV20.0L gene, or an influenza virus mutated in the NS-1 gene.
  • TK thymidine kinase
  • VVF vaccinia virus mutated in the vaccinia growth factor
  • Vaccinia viruses mutated in the viral thymidine kinase (TK) gene are unable to make nucleotides needed for DNA replication.
  • the cellular TK levels are usually very low and the virus is unable to replicate.
  • loss of the tumor suppressor Rb or an increase in cyclin activity leads to E2F pathway activation and high levels of TK expression.
  • cancer cells have high TK levels and the mutated vaccinia virus can replicate and spread.
  • the vaccinia growth factor (VGF) gene is a homolog of mammalian epidermal growth factor (EGF) and can bind and activate the EGF Receptor (EGFR).
  • Vaccinia viruses mutated in the VGF gene are growth restricted to cells with activated EGF pathways, which is commonly mutated in cancers.
  • the viruses can be modified or mutated according to the known structure-function relationship of the viral PKR inhibitors. For example, since the amino terminal region of E3 protein interacts with the carboxy-terminal region domain of PKR, deletion or point mutation of this domain prevents anti-PKR function (Chang et al., PNAS 89:4825-4829 (1992); Chang, H. W. et al, Virology 194:537-547 (1993); Chang et al., J. Virol.
  • the K3L gene of vaccinia virus encodes pK3, a pseudosubstrate of PKR. Truncations or point mutations within the C-terminal portion of K3L protein that is homologous to residues 79 to 83 in eIF-2 abolish PKR inhibitory activity (Kawagishi-Kobayashi, M., et al., Mol. Cell. Biology 17:4146-4158 (1997)).
  • Delta24 virus which is a mutant adenovirus carrying a 24 base pair deletion in the El A region (Fueyo, J., et al., Oncogene 19(1):2-12 (2000)). This region is responsible for binding to the cellular tumor suppressor Rb and inhibiting Rb function, thereby allowing the cellular proliferative machinery, and hence virus replication, to proceed in an uncontrolled fashion.
  • Delta24 has a deletion in the Rb binding region and does not bind to Rb. Therefore, replication of the mutant virus is inhibited by Rb in a normal cell. However, if Rb is inactivated and the cell becomes neoplastic, Delta24 is no longer inhibited. Instead, the mutant virus replicates efficiently and lyses the Rb-deficient cell.
  • VSV vesicular stomatitis virus
  • VSV selectively kills neoplastic cells, optionally with the addition of interferon.
  • Newcastle disease virus replicates preferentially in malignant cells, and the most commonly used strain is 73-T (Reichard, K. W. et al., J. of Surgical Research 52:448-453 (1992); Zorn, U. et al., Cancer Biotherapy 9(3):22-235 (1994); Bar-Eli, N. et al., J.
  • Vaccinia virus propagates in several malignant tumor cell lines. Encephalitis virus has an oncolytic effect in a mouse sarcoma tumor, but attenuation may be required to reduce its infectivity in normal cells. Tumor regression has been described in tumor patients infected with herpes zoster, hepatitis virus, influenza, varicella, and measles virus (for a review, see Nemunaitis, J. Invest. New Drugs 17:375-386 (1999)).
  • the oncolytic virus is a modified, non-reovirus virus comprising a reovirus sigma 1 protein, wherein the reovirus sigma 1 protein replaces the native attachment protein of the non-reovirus virus, and wherein the modified virus does not comprise any portion of the native attachment protein of the non-reovirus virus.
  • the reovirus sigma 1 protein attaches to carrier cells that protect the virus from neutralizing antibodies during in vivo delivery to a tumor, for example, during systemic delivery.
  • the non-reovirus virus can be, but is not limited to, an adenovirus, a vaccinia virus, a herpes simplex virus or a parapox virus.
  • the modified, non-reovirus virus can be an oncolytic virus.
  • Such non-reovirus viruses can be made by replacing the native attachment protein of the non-reovirus virus with a reovirus sigma 1 protein, wherein the full-length sequence of the native attachment protein of the non- reovirus virus is replaced with a reovirus sigma 1 protein.
  • replacement of the native attachment protein of the virus with a reovirus sigma 1 protein allows the virus to attach to carrier cells which protect the virus from neutralizing antibodies during in vivo delivery.
  • the reovirus sigma- 1 protein is described in, for example, WO 2008/11004, which is incorporated by reference herein in its entirety.
  • the oncolytic virus is a reovirus.
  • Reovirus refers to any virus classified in the reovirus genus, whether naturally occurring, modified, or recombinant.
  • Reoviruses are viruses with a double-stranded, segmented RNA genome.
  • the virions measure 60-80 nm in diameter and possess two concentric capsid shells, each of which is icosahedral.
  • the genome consists of double-stranded RNA in 10-12 discrete segments with a total genome size of 16-27 kbp. The individual RNA segments vary in size.
  • the reovirus can be a mammalian reovirus or a human reovirus. All three types share a common complement-fixing antigen.
  • the human reovirus includes three serotypes: type 1 (strain Lang or TIL), type 2 (strain Jones, T2J), and type 3 (strain Dearing or strain Abney, T3D). The three serotypes are easily identifiable on the basis of neutralization and hemagglutinin- inhibition assays.
  • a reovirus according to this disclosure can be a type 3 mammalian orthoreovirus.
  • Type 3 mammalian orthoreoviruses include, without limitation, Dearing and Abney strains (T3D or T3 A, respectively). See, for example, ATCC Accession Nos. VR-232 and VR-824. As described previously, reoviruses use a host cell's ras pathway machinery to down-regulate double-stranded RNA-activated protein kinase (PKR) and thus replication in the cell. See, for example, U.S. Patent Nos. 6,1 10,461; 6,136,307; 6,261,555; 6,344,195; 6,576,234; and 6,811,775, which are incorporated by reference herein in their entireties.
  • PTR protein kinase
  • the reovirus may be naturally occurring or modified.
  • the reovirus is naturally- occurring when it can be isolated from a source in nature and has not been intentionally modified by humans in the laboratory.
  • the reovirus can be from a field source, that is, from a human who has been infected with the reovirus.
  • the reovirus may also be selected or mutagenized for enhanced oncolytic activity.
  • the reovirus may be modified but still capable of lytically infecting a mammalian cell having an active ras pathway.
  • the reovirus may be chemically or biochemically pretreated (e.g., by treatment with a protease, such as chymotrypsin or trypsin) prior to administration to the proliferating cells. Pretreatment with a protease removes the outer coat or capsid of the virus and may increase the infectivity of the virus.
  • the reovirus may be coated in a liposome or micelle (Chandran and Nibert, J. of Virology 72(l):467-75 1998).
  • the virion may be treated with chymotrypsin in the presence of micelle-forming concentrations of alkyl sulfate detergents to generate a new infectious subviral particle (IS VP).
  • the reovirus may be a recombinant reovirus.
  • the recombinant reovirus can be a reassortant reovirus, which includes genomic segments from two or more genetically distinct reoviruses. Recombination/reassortment of reovirus genomic segments may occur following infection of a host organism with at least two genetically distinct reoviruses. Recombinant/reassortant viruses can also be generated in cell culture, for example, by co-infection of permissive host cells with genetically distinct reoviruses.
  • the provided methods include the use of a recombinant reovirus resulting from reassortment of genome segments from two or more genetically distinct reoviruses, including but not limited to, human reovirus, such as type 1 (e.g., strain Lang), type 2 (e-g-, strain Jones), and type 3 (e.g., strain Dearing or strain Abney); non-human mammalian reoviruses; or avian reovirus.
  • human reovirus such as type 1 (e.g., strain Lang), type 2 (e-g-, strain Jones), and type 3 (e.g., strain Dearing or strain Abney); non-human mammalian reoviruses; or avian reovirus.
  • the provided methods include the use of recombinant reoviruses resulting from reassortment of genome segments from two or more genetically distinct reoviruses wherein at least one parental virus is genetically engineered, comprises one or more chemically synthesized genomic segment, has been treated with chemical or physical mutagens, or is itself the result of a recombination event.
  • the provided methods include the use of the recombinant reovirus that has undergone recombination in the presence of chemical mutagens, including but not limited to, dimethyl sulfate and ethidium bromide, or physical mutagens, including but not limited to, ultraviolet light and other forms of radiation.
  • the provided methods include the use of reoviruses with mutations
  • mutant reoviruses as described herein can contain a mutation that reduces or essentially eliminates expression of a sigma3 polypeptide or that results in the absence of a functional sigma3 polypeptide as described in U.S. Publication No. 2008/0292594, which is incorporated by reference herein in its entirety.
  • a mutation that eliminates expression of a sigma3 polypeptide or that results in the absence of a functional sigma3 polypeptide can be in the nucleic acid encoding the sigma3 polypeptide (i.e., the S4 gene) or in a nucleic acid that encodes a polypeptide that regulates the expression or function of the sigma3 polypeptide.
  • a mutation that reduces the expression of a sigma3 polypeptide refers to a mutation that results in a decrease in the amount of sigma3 polypeptides, compared to a reovirus expressing wild type levels of sigma3 polypeptide, of at least 30% (e.g., at least 40%, 50%, 60%, 70%, 80%, 90%, or 95%).
  • a mutation that essentially eliminates expression of a sigma3 polypeptide refers to a mutation that results in a decrease in the amount of sigma3 polypeptides, relative to the amount of sigma3 polypeptides produced by a wild type reovirus, of at least 95% (e.g., 96%, 97%, 98%, 99%, or 100%).
  • a mutation that results in a decrease in or absence of a functional sigma3 polypeptide refers to a mutation that allows expression of the sigma3 polypeptide but that results in a sigma3 polypeptide that is not able to assemble or incorporate into the viral capsid. It would be understood that it may be desirable or necessary for sigma3 polypeptides to retain other functionalities (e.g., the ability to bind RNA) in order that the mutant reovirus retain the ability to propagate.
  • a mutation in a sigma3 polypeptide as described herein can result in a sigma3 polypeptide that is incorporated into the capsid at levels that are reduced relative to a sigma3 polypeptide that does not contain the mutation (e.g., a wild type sigma3 polypeptide).
  • a mutation in a sigma3 polypeptide as described herein also can result in a sigma3 polypeptide that cannot be incorporated into a viral capsid.
  • a sigma3 polypeptide may have reduced function or lack function due, for example, to an inability of the sigma3 polypeptide and the mul polypeptide to bind appropriately, or due to a conformational change that reduces or prohibits incorporation of the sigma3 polypeptide into the capsid.
  • a mutant reovirus as described herein also can contain one or more further mutations (e.g., a second, third, or fourth mutation) in one of the other reovirus capsid polypeptides (e.g., mul, lambda2, and/or sigmal).
  • Reoviruses containing a mutation affecting the sigma3 polypeptide and, optionally, a further mutation in any or all of the other outer capsid proteins can be screened for the ability of such mutant reoviruses to infect and cause lysis of cells.
  • neoplastic cells that are resistant to lysis by wild type reovirus can be used to screen for effective mutant reoviruses described herein.
  • a further mutation can reduce or essentially eliminate expression of a mul polypeptide or result in the absence of a functional mul polypeptide.
  • the mul polypeptide which is encoded by the M2 gene, is likely involved in cell penetration and may play a role in transcriptase activation.
  • Each virion contains about 600 copies of mul polypeptides, which are present in the form of 1 : 1 complexes with sigma3 polypeptides.
  • the mul polypeptide is myristolated on its N-terminus, and then the myristolated N- terminal 42 residues are cleaved off, resulting in a C-terminal fragment (mulC).
  • a further mutation can reduce or essentially eliminate expression of a lambda2 polypeptide or result in the absence of a functional lambda2 polypeptide, and/or a further mutation can reduce or essentially eliminate expression of a sigmal polypeptide or result in the absence of a functional sigmal polypeptide.
  • the lambda2 polypeptide is encoded by the L2 gene, is involved in particle assembly, and exhibits guanylyltransf erase and methyltransferase activity.
  • the sigmal polypeptide is encoded by the S 1 gene, is involved in cell-attachment and serves as the viral
  • the reovirus comprises a lambda-3 polypeptide having one or more amino acid modifications, a sigma-3 polypeptide having one or more amino acid modifications, a mu-1 polypeptide having one or more amino acid modifications, a mu-2 polypeptide having one or more amino acid modifications, or any combination thereof.
  • the reovirus has a lambda-3 polypeptide having one or more amino acid modifications; a sigma-3 polypeptide having one or more amino acid modifications; a mu-1 polypeptide having one or more amino acid modifications; and/or a mu-2 polypeptide having one or more amino acid modifications, as described in U.S. Serial No. 12/046,095, which is incorporated by reference herein in its entirety.
  • the one or more amino acid modifications in the lambda-3 polypeptide are a Val at residue 214, an Ala at residue 267, a Thr at residue 557, a Lys at residue 755, a Met at residue 756, a Pro at residue 926, a Pro at residue 963, a Leu at residue 979, an Arg at residue 1045, a Val at residue 1071, or any combination thereof, numbered relative to GenBank Accession No. M24734.1. It is noted that, when the amino acid sequence is a Val at residue 214 or a Val at residue 1071, the amino acid sequence further includes at least one additional change in the amino acid sequence.
  • the lambda-3 polypeptide includes the sequence shown in SEQ ID NO: 19.
  • the one or more amino acid modifications in the sigma-3 polypeptide are a Leu at residue 14, a Lys at residue 198, or any combination thereof, numbered relative to GenBank Accession No. K02739 (SEQ ID NO:25). It is noted that, when the amino acid sequence is a Leu at residue 14, the amino acid sequence further includes at least one additional change in the amino acid sequence.
  • the sigma-3 polypeptide includes the sequence shown in SEQ ID NO: 15.
  • the one or more amino acid modifications in the mu-1 polypeptide is an Asp at residue 73 numbered relative to GenBank Accession No. M20161.1 (SEQ ID NO:27).
  • the mu-1 polypeptide includes the sequence shown in SEQ ID NO: 17.
  • the amino acid modification mu-2 polypeptide is a Ser at residue 528 numbered relative to GenBank Accession No. AF461684.1 (SEQ ID NO:29).
  • the mu-1 polypeptide includes the sequence shown in SEQ ID NO: 17.
  • a reovirus as described herein having one or more modifications can further include a reovirus sigma-2 polypeptide.
  • Such a sigma-2 polypeptide has a Cys at one or more of position 70, 127, 195, 241, 255, 294, 296, or 340, numbered relative to GenBank Accession No. NP_694684.1 (SEQ ID NO:30).
  • the sigma-2 polypeptide includes the sequence shown in SEQ ID NO: 12.
  • the reovirus comprises an LI genome segment comprising one or more nucleic acid modifications, an S4 genome segment comprising one or more nucleic acid modifications, an Ml genome segment comprising one or more nucleic acid modifications, an M2 genome segment comprising one or more nucleic acid
  • the reovirus has an LI genome segment having one or more nucleic acid modifications; an S4 genome segment having one or more nucleic acid modifications; an Ml genome segment having one or more nucleic acid modifications; and/or an M2 genome segment having one or more nucleic acid modifications, as described in WO 2008/110004, which is incorporated by reference herein in its entirety.
  • the one or more nucleic acid modifications in the LI genome segment are a T at position 660, a G at position 817, an A at position 1687, a G at position 2283, an ATG at positions 2284-2286, a C at position 2794, a C at position 2905, a C at position 2953, an A at position 3153, or a G at position 3231, numbered relative to GenBank Accession No. M24734.1 (SEQ ID NO:22).
  • the LI genome segment includes the sequence shown in SEQ ID NO: 8.
  • the one or more nucleic acid modifications in the S4 genome segment is an A at position 74 and an A at position 624, numbered relative to GenBank Accession No.
  • the S4 genome segment includes the sequence shown in SEQ ID NO:4.
  • the nucleic acid modification in the M2 genome segment can be a C at position 248, numbered relative to GenBank
  • the M2 genome segment for example, includes the sequence shown in SEQ ID NO:6.
  • the nucleic acid modification in the Ml genome segment is a T at position 1595, numbered relative to GenBank Accession No. AF461684.1 (SEQ ID NO:28).
  • the Ml genome segment includes the sequence shown in SEQ ID NO:5.
  • a reo virus as described herein can include any modification or combination of modifications disclosed herein.
  • a reovirus as described herein includes genomic segments having the sequences shown in SEQ ID NOs: 1-10 or the polypeptides shown in SEQ ID NOs: 11 , 12, and 16-21, and either or both SEQ ID NO:13 or 14.
  • a reovirus as disclosed herein is identified as ID AC Accession No. 190907-01, which was deposited with the International Depositary of Canada (ID AC, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington St., Winnipeg, Manitoba Canada R3E 3R2 on September 19, 2007).
  • Sindbis virus can be used in the methods described herein.
  • Sindbis virus is a member of the alphavirus genus of the togaviridae family.
  • the Sindbis virus genome is a single-stranded RNA of 1 1703 nucleotides, capped at the 5' terminus and poly- adenylated at the 3' terminus.
  • the genome consists of a 49S untranslated region (UT), nonstructural proteins nsPl, nsP2, nsP3, and nsP4 followed by a promoter.
  • the promoter is followed by a 26S UT, structural proteins C, E3, E2, 6K, and El and finally a 3' UT and a poly-adenylated terminus.
  • the genomic 49S RNA is of plus sense, is infectious, and serves as mRNA in the infected cell.
  • Sindbis vectors systemically and specifically infect/detect and kill metastasized tumors in vivo, leading to significant suppression of tumor growth and enhanced survival (Hurtado et al., Rejuvenation Res. 9(l):36-44 (2006)).
  • Sindbis virus infects mammalian cells using the Mr 67,000 laminin receptor, which is elevated in tumor versus normal cells. Tumor overexpression of the laminin receptor may explain the specificity and efficacy that Sindbis vectors demonstrate for tumor cells in vivo.
  • Sindbis does not have to undergo genetic manipulation to target cancer cells or to be injected directly into tumors. Sindbis injected anywhere into a subject travels through the bloodstream to the target area (Tseng et al., Cancer Res.
  • Sindbis can also be genetically engineered to carry one or more genes that suppress the immune response to the virus and/or genes that stimulate an immune response against the tumor such as, for example, antitumor cytokine genes such as interleukin-12 and interleukin-15 genes.
  • the oncolytic virus may be naturally occurring or modified.
  • the virus may be chemically or biochemically pretreated (e.g., by treatment with a protease, such as chymotrypsin or trypsin) prior to administration to the neoplastic cells. Pretreatment with a protease removes the outer coat or capsid of the virus and may increase the infectivity of the virus.
  • the virus may be coated in a liposome or micelle (Chandran and Nibert, J. of Virology 72(l):467-75 (1998)) to reduce or prevent an immune response from a mammal that has developed immunity to the virus.
  • the virion may be treated with chymotrypsin in the presence of micelle forming concentrations of alkyl sulfate detergents to generate a new infectious subvirion particle.
  • the oncolytic virus may also be a reassortant virus or an ISVP.
  • the present methods include using any oncolytic virus according to the disclosure herein and knowledge available in the art.
  • the oncolytic virus may be naturally occurring or modified.
  • the oncolytic virus is naturally-occurring when it can be isolated from a source in nature and has not been intentionally modified by humans in the laboratory.
  • the oncolytic virus can be from a field source, that is, from a human who has been infected with the oncolytic virus.
  • the oncolytic virus may be a recombinant oncolytic virus.
  • the recombinant oncolytic virus results from the reassortment of genomic segments from two or more genetically distinct oncolytic viruses, also referred to herein as a reassortant.
  • Reassortment of oncolytic virus genomic segments may occur following infection of a host organism with at least two genetically distinct oncolytic viruses.
  • Recombinant viruses can also be generated in cell culture, for example, by co-infection of permissive host cells with genetically distinct oncolytic viruses.
  • the methods include the use of recombinant oncolytic virus resulting from reassortment of genome segments from two or more genetically distinct oncolytic viruses wherein at least one parental virus is genetically engineered, comprises one or more chemically synthesized genomic segment, has been treated with chemical or physical mutagens, or is itself the result of a
  • the methods include the use of the recombinant oncolytic virus that has undergone recombination in the presence of chemical mutagens, including but not limited to dimethyl sulfate and ethidium bromide, or physical mutagens, including but not limited to ultraviolet light and other forms of radiation.
  • chemical mutagens including but not limited to dimethyl sulfate and ethidium bromide
  • physical mutagens including but not limited to ultraviolet light and other forms of radiation.
  • the methods include the use of oncolytic viruses with mutations (including insertions, substitutions, deletions or duplications) in one or more genome segments.
  • mutations can comprise additional genetic information as a result of recombination with a host cell genome, or that comprise synthetic genes such as, for example, genes encoding agents that suppress anti-viral immune responses.
  • the oncolytic virus is a mutant oncolytic virus.
  • the oncolytic virus may be modified by incorporation of mutated coat proteins, such as for example, into the virion outer capsid.
  • the mutant oncolytic virus is, optionally, a mutant reovirus.
  • Mutant reoviruses as described herein can contain a mutation that reduces or essentially eliminates expression of a sigma3 polypeptide or that results in the absence of a functional sigma3 polypeptide as described in U.S. Serial No. 12/124,522, which is incorporated by reference herein in its entirety.
  • the mutant reoviruses used in the provided methods are mutated as described in U.S. Serial No. 12/046,095, which is incorporated by reference herein in its entirety.
  • a mutation as referred to herein can be a substitution, insertion or deletion of one or more nucleotides.
  • Point mutations include, for example, single nucleotide transitions (purine to purine or pyrimidine to pyrimidine) or transversions (purine to pyrimidine or vice versa) and single- or multiple-nucleotide deletions or insertions.
  • a mutation in a nucleic acid can result in one or more conservative or non-conservative amino acid substitutions in the encoded polypeptide, which may result in conformational changes or loss or partial loss of function, a shift in the reading frame of translation (frame-shift) resulting in an entirely different polypeptide encoded from that point on, a premature stop codon resulting in a truncated polypeptide (truncation), or a mutation in a virus nucleic acid may not change the encoded polypeptide at all (silent or nonsense). See, for example, Johnson and Overington, 1993, J. Mol. Biol. 233:716-38; Henikoff and
  • Mutations can be generated in the nucleic acid of an oncolytic virus using any number of methods known in the art.
  • site directed mutagenesis can be used to modify a reovirus nucleic acid sequence.
  • One of the most common methods of site- directed mutagenesis is oligonucleotide-directed mutagenesis.
  • oligonucleotide-directed mutagenesis an oligonucleotide encoding the desired change(s) in sequence is annealed to one strand of the DNA of interest and serves as a primer for initiation of DNA synthesis. In this manner, the oligonucleotide containing the sequence change is incorporated into the newly synthesized strand. See, for example, Kunkel, 1985, Proc.
  • nucleic acids containing a mutation can be generated using PCR or chemical synthesis, or polypeptides having the desired change in amino acid sequence can be chemically synthesized. See, for example, Bang and Kent, 2005, Proc. Natl. Acad. Sci. USA 102:5014-9 and references therein.
  • Viruses can be purified using standard methodology. See, for example, Schiff et al., "Orthoreoviruses and Their Replication," Ch 52, in Fields Virology, Knipe and Howley, eds., 2006, Lippincott Williams and Wilkins; Smith et al., 1969, Virology 39(4):791-810; and U.S. Patent Nos. 7,186,542; 7,049,127; 6,808,916; and 6,528,305, which are incorporated by reference herein in their entireties.
  • purified viruses refer to viruses that have been separated from cellular components that naturally accompany them. Typically, viruses are considered purified when they are at least 70% (e.g., at least 75%, 80%, 85%, 90%, 95%, or 99%) by dry weight, free from the proteins and other cellular components with which they are naturally associated.
  • compositions comprising Carbo-Tax and, optionally, one or more oncolytic viruses.
  • the pharmaceutical compositions comprise one or more additional therapeutic agents, for example, chemotherapeutic agents, anti-inflammatory agents or agents that decrease interstitial pressure.
  • the pharmaceutical compositions comprise Carbo-Tax.
  • compositions can comprise one agent or more than one agent.
  • each of the Carbo-Tax, oncolytic virus, and any other agent can be contained within separate pharmaceutical compositions or the same composition. If the oncolytic virus and agents are contained within separate pharmaceutical compositions, the compositions can be administered concomitantly or sequentially.
  • compositions are administered in vitro or in vivo in a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier can be a solid, semi-solid, or liquid material that can act as a vehicle, carrier or medium for the reovirus.
  • compositions containing an oncolytic virus and/or one or more of the provided agents can be in the form of tablets, pills, powders, lozenges, sachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • compositions containing an oncolytic virus are suitable for infusion.
  • crystalloids are aqueous solutions of mineral salts or other water-soluble molecules. Colloids contain larger insoluble molecules, such as gelatin; blood itself is a colloid.
  • the most commonly used crystalloid fluid is normal saline, a solution of sodium chloride at 0.9% concentration, which is close to the concentration in the blood (isotonic).
  • Ringer's lactate or Ringer's acetate is another isotonic solution often used for large-volume fluid replacement.
  • a solution of 5% dextrose in water, sometimes called D5W is often used instead if the patient is at risk for having low blood sugar or high sodium.
  • suitable carriers include phosphate-buffered saline or another physiologically acceptable buffer, lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose.
  • a pharmaceutical composition additionally can include, without limitation, lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • compositions can be formulated to provide quick, sustained or delayed release of a mutant reovirus after administration by employing procedures known in the art.
  • suitable formulations for use in a pharmaceutical composition can be found in Remington: The Science and Practice of Pharmacy, 22d Edition, Loyd et al. eds., Pharmaceutical Press and
  • a mutant reovirus can be mixed with a pharmaceutical carrier to form a solid composition.
  • tablets or pills can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • a tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • Liquid formulations that include a reovirus and/or agent for oral administration or for injection generally include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as corn oil, cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. These liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described herein. Such compositions can be administered by the oral or nasal respiratory route for local or systemic effect.
  • solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, orally or nasally, from devices which deliver the formulation in an appropriate manner.
  • transdermal delivery devices e.g., patches
  • transdermal patches may be used to provide continuous or discontinuous infusion of the viruses and agents as described herein.
  • the construction and use of transdermal patches for the delivery of pharmaceutical agents are well known in the art. See, for example, U.S. Patent No. 5,023,252.
  • patches can be constructed for continuous, pulsatile, or on- demand delivery of mutant reoviruses.
  • viruses and/or other agents can, if necessary, be coated in a liposome or micelle to reduce or prevent an immune response in a mammal that has developed immunity toward a virus or agent.
  • Such compositions are referred to as immunoprotected viruses or agents. See, for example, U.S. Patent Nos. 6,565,831 and 7,014,847.
  • the oncolytic virus is administered, for example, systemically, in a manner so that it can ultimately contact the target tumor or tumor cells.
  • the route by which the virus is administered, as well as the formulation, carrier or vehicle, depends on the location as well as the type of the target cells.
  • a wide variety of administration routes can be employed.
  • the virus can be administered by injection directly to the tumor.
  • the virus can be administered intravenously or intravascularly.
  • the virus is administered in a manner such that it can be transported systemically through the body of the mammal and thereby reach the tumor (e.g., intravenously or intramuscularly).
  • the virus can be administered directly to a single solid tumor, where it then is carried systemically through the body to metastases.
  • the virus can also be
  • intraperitoneally, intrathecally or intraventricularly e.g., for brain tumor
  • topically e.g., for melanoma
  • orally e.g., for oral or esophageal cancer
  • rectally e.g., for colorectal cancer
  • vaginally e.g., for cervical or vaginal cancer
  • nasally by inhalation spray or by aerosol formulation (e.g., for lung cancer).
  • the virus is administered continuously to a subject at least once per day or up to intermittently or continuously throughout the day on consecutive days, for a period of time.
  • the virus is administered, for example, to subjects by means of intravenous administration in any pharmacologically acceptable solution, or as an infusion over a period of time.
  • the substance may be administered systemically by injection (e.g., IM or subcutaneously) or taken orally daily at least once per day, or administered by infusion in a manner that results in the daily delivery into the tissue or blood stream of the subject.
  • the period of time is, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, or 24 hours, or any time between 1 and 24 hours, inclusive, or more.
  • the period of time is 5, 15, 30, 60, 90, 120, 150 or 180 minutes, or any time between 5 and 180 minutes, inclusive, or more.
  • the virus is administered by infusion for 60 minutes. Administrations can be repeated daily for 2, 3, 4, 5, 6, 7, 8, 9, 10, 14, 21, 28 days or any number of days between 2 and 28 days, inclusive, or longer.
  • the Carbo-Tax e.g., paclitaxel and carboplatin
  • the agents are administered via any of several routes of administration, including, topically, orally, parenterally, intravenously, intraperitoneally, intramuscularly, subcutaneously, intracavity, transdermally, intrahepatically, intracranially,
  • the therapeutic agents are administered continuously in the manner set forth in the description above with respect to oncolytic viruses.
  • the agent is administered to subjects by means of intravenous administration in any pharmacologically acceptable solution, or as an infusion over a period of time.
  • the agents are administered locally at or near the site of the tumor.
  • the agents are administered systemically.
  • effective amounts of taxanes, e.g., paclitaxel include from about 40-300
  • effective amounts of taxanes include 130-225 mg/m 2 .
  • effective amounts of platinum compounds, e.g., carboplatin include from about 5-1000
  • effective amounts of cisplatin include from about 175-200 mg/m and effective mounts for carboplatin include from about 200-600 mg/m .
  • Effective amounts of other agents range from 0.001-10,000 mg/kg body weight or any amount in between 0.001 and 10,000 mg/kg body weight, inclusive.
  • effective amounts of platinum compounds include approximately 2 to 7 mg/mL minute (AUC) as calculated by the Calvert formula.
  • effective amounts of platinum compounds include approximately 5 or 6 mg/mL minute (AUC) as calculated by the Calvert formula.
  • the platinum compounds are administered as an intravenous infusion over a period of 30 minutes.
  • the viruses as disclosed herein are administered in an amount that is sufficient (i.e., an effective amount) to treat the proliferative disorder.
  • a proliferative disorder is treated when administration of a virus to proliferating cells affects lysis (e.g., oncolysis) of the affected cells, resulting in a reduction in the number of abnormally, proliferating cells, a reduction in the size of a neoplasm, and/or a reduction in or elimination of symptoms (e.g., pain) associated with the proliferating disorder.
  • lysis e.g., oncolysis
  • the term oncolysis means at least 10% of the proliferating cells are lysed (e.g., at least about 20%, 30%, 40%, 50%, or 75% of the cells are lysed).
  • the percentage of lysis can be determined, for example, by measuring the reduction in the size of a neoplasm or in the number of proliferating cells in a mammal, or by measuring the amount of lysis of cells in vitro (e.g., from a biopsy of the proliferating cells).
  • An effective amount of a virus will be determined on an individual basis and may be based, at least in part, on the particular virus used; the individual's size, age, gender; and the size and other characteristics of the abnormally, proliferating cells. For example, for treatment of a human, approximately 10 to 10 plaque forming units (PFU) of a virus are used, depending on the type, size and number of proliferating cells or neoplasms present.
  • PFU plaque forming units
  • the effective amount can be, for example, from about 1.0 PFU/kg body weight to about 10 15 PFU/kg body weight (e.g., from about 10 2 PFU/kg body weight to about 10 13 PFU/kg body weight).
  • the effective amount is about 1x10 to about 1x10 PFU or TCID5 0 .
  • the effective amount is about 3xl0 10 to about lxlO 10 TCID 5 0.
  • 175 mg/m 2 of the paclitaxel of Carbo-Tax is administered to the subject and 3xl0 10 TCID 50 or lxlO 10 TCID 50 of a reovirus is administered to the subject.
  • 135 mg/m of the paclitaxel of Carbo-Tax is administered to the subject and 3xl0 10 TCID 50 or lxlO 10 TCID 50 of a reovirus is administered to the subject.
  • the paclitaxel of Carbo-TAx is administered as a three hour intravenous infusion.
  • the reovirus is administered as a one hour intravenous infusion.
  • 175 mg/m of the paclitaxel of the Carbo-Tax is administered to the subject; 5mg/ml minute (AUC as calculated by the Calvert formula) of the carboplatin of the Carbo-Tax, is administered to the subject; and 3xl0 10 TCID 0 or lxl0 10 TCID 5 o of a reovirus is administered to the subject.
  • 135 mg/m 2 of the paclitaxel of the Carbo-Tax is administered to the subject; 4mg/ml minute (AUC as calculated by the Calvert formula) of the carboplatin of the Carbo-Tax, is administered to the subject; and lxlO 10 TCID 50 of a reovirus is administered to the subject.
  • the paclitaxel is administered as a three hour intravenous infusion.
  • the carboplatin is administered as a thirty minute intravenous infusion.
  • the reovirus is administered as a one hour intravenous infusion.
  • the paclitaxel and carboplatin of Carbo-Tax are administered on the same day.
  • the paclitaxel and carboplatin are administered every 21 days.
  • the oncolytic virus is administered on the same day as the paclitaxel and carboplatin.
  • the oncolytic virus, paclitaxel and carboplatin are administered on a first day.
  • the oncolytic virus is also administered on a second, third, fourth and fifth day consecutively following the first day.
  • every 21 days, the oncolytic virus, paclitaxel and carboplatin are administered on a first day, and, the oncolytic virus is also administered on a second, third, fourth and fifth day consecutively following the first day.
  • Optimal dosages of viruses and therapeutic agents and compositions comprising viruses and agents depend on a variety of factors. The exact amount required will vary from subject to subject, depending on the species, age, weight and general condition of the subject, the severity of the disease being treated, the particular virus or vector used and its mode of administration. Thus, it is not possible to specify an exact amount for every composition. However, an appropriate amount can be determined by one of ordinary skill in the art using only routine experimentation given the guidance provided herein.
  • Effective dosages and schedules for administering the compositions may be determined empirically.
  • animal models for a variety of proliferative disorders can be obtained from the Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609 USA. Both direct (e.g., histology of tumors) and functional measurements (e.g., survival of a subject or size of a tumor) can be used to monitor response to therapies. These methods involve the sacrifice of representative animals to evaluate the population, increasing the animal numbers necessary for the experiments. Measurement of luciferase activity in the tumor provides an alternative method to evaluate tumor volume without animal sacrifice and allowing longitudinal population-based analysis of therapy.
  • the dosage ranges for the administration of compositions are those large enough to produce the desired effect in which the symptoms of the disease are affected.
  • the dosage should not be so large as to cause adverse side effects, such as unwanted cross- reactions and anaphylactic reactions.
  • the dosage can be adjusted by the individual physician in the event of any counterindications.
  • Dosages vary and are administered in one or more dose administrations daily, for one or several days.
  • the provided viruses and therapeutic agents are administered in a single dose or in multiple doses (e.g., two, three, four, six, or more doses).
  • the infusion can be a single sustained dose or can be delivered by multiple infusions. Treatment may last from several days to several months or until diminution of the disease is achieved.
  • Combinations of the provided viruses and therapeutic agents are administered either concomitantly (e.g., as an admixture), separately but simultaneously (e.g., via separate intravenous lines into the same subject), or sequentially (e.g., one of the compounds or agents is given first followed by the second).
  • the term combination is used to refer to either concomitant, simultaneous, or sequential administration of two or more agents.
  • the Carbo-Tax is administered prior to or at the same time as the oncolytic virus.
  • the first compound is administered minutes, hours, days, or weeks prior to administration of the second compound.
  • the first compound can be administered at 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 24, 36, 48, 60, or 72 hours, or any time between 1 and 72 hours, inclusive, prior to administration of a second compound.
  • the first compound is administered more than 72 hours prior to the second compound.
  • the first compound can be administered at 1, 5, 15, 30, 60, 90, 120, 150 or 180 minutes, or any time between 1 and 180 minutes, inclusive, prior to administration of a second compound.
  • the first compound is administered at 1, 2, 3, 4, 5, 6, 7, 14, 21, or 28 days, or any amount in between 1 and 28, inclusive, days prior to administration of the second compound.
  • the first compound is administered more than 28 days prior to the second compound.
  • the Carbo-Tax is administered from about 1 to 8 hours prior to administration of the oncolytic virus.
  • Oncolytic viruses or a pharmaceutical composition comprising such viruses are optionally packaged into a kit.
  • the kit also includes one or more agents or
  • kits comprising such agents including, but not limited to, Carbo-Tax.
  • the kit optionally, also includes one or more agents that inhibit a proinflammatory cytokine, one or more chemotherapeutic agents, one or more
  • a pharmaceutical composition can be formulated in a unit dosage form.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of a mutant reovirus calculated to produce the desired therapeutic effect in association with a suitable pharmaceutically acceptable carrier.
  • the provided methods may be combined with other tumor therapies such as chemotherapy, radiotherapy, surgery, hormone therapy and/or immunotherapy.
  • the oncolytic virus may be administered in conjunction with surgery or removal of the neoplasm. Therefore, provided herewith are methods for the treatment of a solid neoplasm comprising surgical removal of the neoplasm and administration of an oncolytic virus at or near to the site of the neoplasm.
  • the Carbo-Tax and/or oncolytic virus may be administered with an agent that decreases interstitial pressure and/or increases vascular permeability.
  • Agents that decrease interstitial pressure suitable for use in the provided methods include, but are not limited to, vasopressin; TNF; interleukin-1 (IL-1);
  • interferon-K interferon-K
  • substance P proteinase inhibitors such as N-alpha-tosyl-L-lysyl- chloromethyl-ketone (TLCK), tosyl phenylalanyl chloromethyl ketone (TPCK) and leupeptin; vascular endothelial growth factor (VEGF); nitroglycerine; serotonin; plasma kinins such as bradykinin; platelet-activating factor (PAF); prostaglandin Ej (PGEi); histamine; imatinib; zona occludens toxin (ZOT); interleukin-2; nitric oxide inhibitors such as L-N-monomethyl arginine (L-NMMA) and L-N-nitro-arginine methyl ester (L- NAME); and human growth factor receptor tyrosine kinase inhibitors such as gefitinib.
  • TLCK N-alpha-tosyl-L-lys
  • the interstitial pressure in the subject can be decreased by lowering extracellular calcium ion concentrations.
  • a low calcium ion concentration fluid can be perfused through the vasculature of the tissue to which the oncolytic virus is administered.
  • Suitable perfusate calcium ion concentrations may range from about 40 or 50 Tmol/L to about 500 Tmol/L, more preferably from about 50 Tmol/L to about 200 Tmol/L. A perfusate calcium concentration of about 50 Tmol/L is provided. Calcium ion (e.g., Ca 2+ ) concentration can also be lowered, for example, through use of a suitable buffer such as a chelating agent, for example,
  • EGTA ethylenebis(oxyethylenenitrilo)tet- racetic acid
  • EDTA ethylenediaminetetracetic acid
  • BAPTA l,2-bis-(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid
  • the interstitial pressure of a tumor can be reduced by removal of excess interstitial fluid.
  • Removal of excess interstitial fluid is accomplished by any known method, including, for example, by an artificial lymphatic system (ALS).
  • ALS artificial lymphatic system
  • permeabilizing photodynamic therapy can be used to enhance delivery of the oncolytic virus by enhancing vascular permeability.
  • P-PDT induced vascular leakiness allows the therapeutic agents to leave the vasculature and distribute into hyperproliferative tissue (e.g. the tumor bed) in higher concentrations than achievable without prior permeabilizing PDT. See U.S.
  • the provided methods further comprise administering to the subject an immunosuppressive agent.
  • the immunosuppressive agent is an agent that inhibits a pro-inflammatory cytokine.
  • a pro-inflammatory cytokine refers to a cytokine that directly or indirectly stimulates the immune system.
  • Pro-inflammatory cytokines include, but are not limited to, IL-1I, IL-3, IL-6, IL-12 p70, IL-17, MIP-1I, and RANTES.
  • the agent blocks T-cell responses while having little to no effect on B-cell activity.
  • the agent inhibits pro-inflammatory cytokines but does not inhibit or minimally inhibits production of NARA.
  • TNF-I antibodies such as infliximab, CDP571, CDP870, and adalimumab
  • recombinant, human soluble p55 TNF receptors such as onercept
  • soluble TNF receptor and Fc fragment fusion proteins such as etanercept
  • pegylated Fab fragments of humanized antibody to TNF such as certolizumab pegol
  • chimeric antibodies to anti-I chain of IL-2 receptor such as basiliximab or daclizumab
  • IL-12p40 antibodies such as ABT-874
  • IL-6 receptor antibodies such as MRA or tocilizumab
  • IFN-K antibodies such as fontolizumab
  • antibodies that inhibit IL-1 binding to the IL-1 receptor such as AMG108
  • caspase-1 inhibitors that inhibit cytokine-release such as diarylsulphonylurene
  • IL-15 antibodies such as mepolizumab
  • IL-8 antibodies such as
  • agents that inhibit pro-inflammatory cytokines include human recombinant lactoferrin, which inhibits cellular release of proinflammatory cytokines and
  • prometastatic cytokines including IL-6, IL-8, granulocyte macrophage colony- stimulating factor and TNF-a.
  • Inhibitors of dendritic cell derived IL-12 and IL-18, such as rapamycin and sanglifehrin, are also suitable for use in the provided methods.
  • Rapamycin is an immunosuppressant that inhibits T cell mTOR kinase activation
  • Sanglifehrin A is a cyclophilin-binding immunosuppressant that also inhibits IL-2 dependent T cell proliferation.
  • dietary rutin which suppresses the induction of pro-inflammatory cytokines such as IL- ⁇ , IL-6, and GM-CS.
  • compositions in the provided methods are, optionally, administered in conjunction with or in addition to known anticancer compounds or chemotherapeutic agents.
  • Chemotherapeutic agents are compounds which may inhibit the growth of tumors. Such agents, include, but are not limited to 5-fluorouracil; mitomycin C;
  • methotrexate methotrexate; hydroxyurea; cyclophosphamide; dacarbazine; mitoxantrone; anthracyclins
  • epirubicin and doxurubicin antibodies to receptors, such as herceptin; etoposide;
  • hormone therapies such as tamoxifen and anti-estrogens
  • interferons such as interferons
  • one or more of the proliferating cells associated with the disorder may have a mutation in which the ras gene (or an element of the ras signaling pathway) is activated, either directly (e.g., by an activating mutation in ras) or indirectly (e.g., by activation of an upstream or downstream element in the ras pathway).
  • Activation of an upstream element in the ras pathway includes, for example, transformation with epidermal growth factor receptor (EGFR) or Sos.
  • EGFR epidermal growth factor receptor
  • Activation of a downstream element in the ras pathway includes, for example, mutation within B-Raf. See, for example, Brose et al., 2002, Cancer Res. 62:6997-7000.
  • a proliferative disorder that results, at least in part, by the activation of ras, an upstream element of ras, or an element in the ras signaling pathway is referred to herein as a ras- mediated proliferative disorder.
  • the oncolytic virus is useful for treating proliferative disorders caused by mutations or dysregulation of PKR. See, for example, Strong et al., 1998, EMBO J. 17:3351-62.
  • treatment refers to a method of reducing the effects of a disease or condition or symptom of the disease or condition.
  • treatment can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% reduction or amelioration in the severity of an established disease or condition or symptom of the disease or condition.
  • the method for treating cancer is considered to be a treatment if there is a 10% reduction in one or more symptoms of the disease in a subject as compared to control.
  • the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100% or any percent reduction in between 10 and 100 as compared to native or control levels.
  • treatment does not necessarily refer to a cure or complete ablation of the disease, condition or symptoms of the disease or condition.
  • the treatment can be reflected in a reduced tumor size, for example.
  • the term subject can be a vertebrate, more specifically a mammal (e.g., a human, horse, pig, rabbit, dog, sheep, goat, non-human primate, cow, cat, guinea pig or rodent), a fish, a bird or a reptile or an amphibian.
  • a mammal e.g., a human, horse, pig, rabbit, dog, sheep, goat, non-human primate, cow, cat, guinea pig or rodent
  • a fish e.g., a fish
  • bird or a reptile or an amphibian e.g., a particular age or sex.
  • patient or subject may be used interchangeably and can refer to a subject with a disease or disorder.
  • patient or subject includes human and veterinary subjects.
  • Paclitaxel is administered as a 3 hour intravenous infusion at a dose of 135, 175 mg/m 2 or 200 mg/m 2 .
  • Carboplatin is then administered as a 30 minute intravenous infusion at a dose calculated by the Calvert formula (AUC 4 mg/mL minute, 5 mg/mL minute, or 6 mg/mL minute with GFR measured by 5 ICr EDTA).
  • reovirus is then administered as a 1 hour intravenous infusion at a dose of lxl 0 10 or 3xl0 10 TCID 50 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

La présente invention concerne un procédé de traitement d'un cancer chez un sujet comprenant la sélection d'un sujet naïf pour les taxanes présentant une tumeur primaire et l'administration au sujet d'une quantité thérapeutiquement efficace d'un virus oncolytique. L'invention concerne en outre des procédés de traitement d'un cancer métastasique chez un sujet comprenant la sélection d'un sujet naïf pour les taxanes présentant un cancer métastasique et l'administration au sujet d'une quantité thérapeutiquement efficace de Carbo-Tax. Les procédés comprennent facultativement en outre l'administration d'un virus oncolytique.
PCT/CA2014/000608 2013-08-08 2014-08-07 Procédés de traitement de sujets naïfs pour les taxanes présentant des tumeurs primaires ou un cancer métastasique WO2015017915A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361863631P 2013-08-08 2013-08-08
US201361863626P 2013-08-08 2013-08-08
US61/863,626 2013-08-08
US61/863,631 2013-08-08

Publications (1)

Publication Number Publication Date
WO2015017915A1 true WO2015017915A1 (fr) 2015-02-12

Family

ID=52460444

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2014/000608 WO2015017915A1 (fr) 2013-08-08 2014-08-07 Procédés de traitement de sujets naïfs pour les taxanes présentant des tumeurs primaires ou un cancer métastasique

Country Status (1)

Country Link
WO (1) WO2015017915A1 (fr)

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
FOUNTZILAS, G. ET AL.: "Paclitaxel and Carboplatin as First-Line Chemotherapy for Advanced Breast Cancer''.", ONCOLOGY., 1 January 1998 (1998-01-01), Retrieved from the Internet <URL:http://www.cancernetwork.com/review-article/paclitaxel-and-carboplatin- first-line-chemother apy-advanced-breast-cancer> [retrieved on 20141002] *
HEINEMANN, L. ET AL.: "Synergistic effects of oncolytic reovirus and docetaxel chemotherapy in prostate cancer''.", BMC CANCER., vol. 11, 2011, pages 221 *
KARAPANAGIOTOU, E. ET AL.: "Phase I/II Trail of Carboplatin and Paclitaxel Chemotherapy in Combination with Intravenous Oncolytic Reovirus in Patients with Advanced Malignancies''.", CLINICAL CANCER RESEARCH, vol. 18, 1 April 2012 (2012-04-01), pages 2080 *
ORR, G. ET AL.: "Mechanisms of taxol resistance related to microtubules", ONCOGENE, vol. 22, 2003, pages 7280 - 7295 *

Similar Documents

Publication Publication Date Title
US8658158B2 (en) Sensitization of chemotherapeutic agent resistant neoplastic cells with a virus
EP1984007B1 (fr) Utilisation d&#39;immunosuppression locale à faible dose pour améliorer une thérapie virale oncolytique
AU2002234453A1 (en) Sensitization of chemotherapeutic agent resistant neoplastic cells with reovirus
AU2009253682B2 (en) Modulating interstitial pressure and oncolytic viral delivery and distribution
CA2680661C (fr) Reovirus mutants et leurs procedes de fabrication et d&#39;utilisation
EP3068411B1 (fr) Virus oncolytiques et schémas posologiques de traitement du cancer renforcés
US20130243732A1 (en) Abrogating proinflammatory cytokine production during oncolytic reovirus therapy
WO2015017915A1 (fr) Procédés de traitement de sujets naïfs pour les taxanes présentant des tumeurs primaires ou un cancer métastasique
US20220105143A1 (en) T cell repertoire dynamics and oncolytic viral therapy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14835222

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14835222

Country of ref document: EP

Kind code of ref document: A1