AU767562B2 - Recombinant pox virus for immunization against tumor-associated antigens - Google Patents

Recombinant pox virus for immunization against tumor-associated antigens Download PDF

Info

Publication number
AU767562B2
AU767562B2 AU54194/01A AU5419401A AU767562B2 AU 767562 B2 AU767562 B2 AU 767562B2 AU 54194/01 A AU54194/01 A AU 54194/01A AU 5419401 A AU5419401 A AU 5419401A AU 767562 B2 AU767562 B2 AU 767562B2
Authority
AU
Australia
Prior art keywords
pox
virus
recombinant
vector
taa
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
AU54194/01A
Other versions
AU5419401A (en
Inventor
Linda R. Gritz
Gail P. Mazzara
Dennis L. Panicali
Jeffrey Schlom
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.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
Therion 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 AU37268/97A external-priority patent/AU731860B2/en
Application filed by US Department of Health and Human Services, Therion Biologics Corp filed Critical US Department of Health and Human Services
Priority to AU54194/01A priority Critical patent/AU767562B2/en
Publication of AU5419401A publication Critical patent/AU5419401A/en
Application granted granted Critical
Publication of AU767562B2 publication Critical patent/AU767562B2/en
Priority to AU2003266471A priority patent/AU2003266471A1/en
Assigned to THERION BIOLOGICS CORPORATION, GOVERNMENT OF THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES, THE reassignment THERION BIOLOGICS CORPORATION Request to Amend Deed and Register Assignors: THERION BIOLOGICS CORPORATION, United States Government as represented by the Department of Health and Human Services, The
Assigned to GOVERNMENT OF THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES, THE reassignment GOVERNMENT OF THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES, THE Alteration of Name(s) in Register under S187 Assignors: GOVERNMENT OF THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES, THE, THERION BIOLOGICS CORPORATION
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

.4
I.
P/00/011 Regulation 3.2
AUSTRALIA
Patents Act 1990 COMPLETE SPECIFICATION FOR A DIVISIONAL PATENT
ORIGINAL
TO BE COMPLETED BY APPLICANT Name of Applicant: Actual Inventor(s): THERION BIOLOGICS CORPORATION and UNITED STATES GOVERNMENT as represented by THE DEPARTMENT OF HEALTH AND HUMAN SERVICES Jeffrey Schlom; Dennis L. Panicali; Linda R. Gritz; Gail P. Mazzara CALLINAN LAWRIE, 711 High Street, Kew, Victoria 3101, Australia RECOMBINANT POX VIRUS FOR IMMUNIZATION AGAINST TUMOR-ASSOCIATED ANTIGENS Address for Service: Invention Title: The following statement is a full description of this invention, including the best method of performing it known to me:- 04/07/01 ,cf2158.fc, RECOMBINANT POX VIRUS FOR IMMUNIZATION
AGAINST
TUMOR-ASSOCIATED
ANTIGENS
Background of the Invention The immunotherapeutic approach to the treatment of cancer is based on the observation that human tumor cells express a variety of tumor-associated antigens (TAAs) that are not expressed in normal tissues. These antigens, which include viral tumor antigens, cellular oncogene proteins, and tumor-associated differentiation antigens, can serve as targets for the host immune system and elicit responses which result in tumor destruction. This immune response is mediated primarily by lymphocytes; T cells in general and class I MHC-restricted cytotoxic T lymphocytes in particular play a central role in tumor 10 rejection. Hellstrom, et al., (1969) Adv. Cancer Res. 12:167- 223; Greenberg, P.D. (1991) in Advances in Immunology, vol. 49 (Dixon, pp 281-355, Academic Press, Inc., Orlando, FL.
Unfortunately, as evidenced by the high incidence of cancer in the population, the immune response to neoplastic cells often fails to 15 eliminate tumors. The goal of active cancer immunotherapy is the augmentation of anti-tumor responses, particularly T cell responses, in order to effect complete tumor destruction.
Most attempts at active immunization against cancer antigens 20 have involved whole tumor cells or tumor cell fragments. However, the cloning of TAAs recognized by CD8 T cells has opened new possibilities for the immunotherapy of cancer based on the use of recombinant or synthetic anti-cancer vaccines. Boon, et al.,(1994) Annu. Rev. Immunol. 12:337-365; Brithcard, et al., (1993) J. Exp.
med. 178:489-495; Cox, et al., (1994) Science 264:716-719; 4, Houghton, A.N. (1994) J. Exp. Med. 180:1-4; Pardoll, D.M. (1994) Nature 369:357-358; Kawakami, et al., (1994) Proc. Natl. Acad.
Sci. U.S.A. 91:3515-3519; Kawakami, et al., (1994) Proc. Natl.
Acad. Sci. U.S.A. 91:6458-6462.
Two such antigens have been designated MART-1 (Melanoma Antigen Recognized by T cells 1) and gpl00. Proc. Natl. Acad. Sci.
U.S.A. 91:3515-3519. MART-1 and gp 100 appear to be expressed in virtually all fresh and cultured melanomas. With the exception of melanocyte and retina, no normal tissues express the antigens. The antigens may be responsible for mediating tumor regression in patients with advanced melanoma, since the tumor-infiltrating lymphocytes (TIL) used to identify MART-1 and gp100 were capable of effecting tumor regression in vivo. Thus, immunization of melanoma patients with 15 MART-1 or gp100 may boost their cellular immune responses against their cancers.
The use of recombinant vaccinia viruses for anti-tumor immunotherapy has been reviewed. (Hu, Hellstrom, and 20 Hellstrom K.E. (1992) in Vaccines: New Approaches to Immunological Problems Ellis, ed) pp 327-343, Butterworth-Heinemann, Boston.) Anti-tumor responses have been elicited using a recombinant vaccinia virus expressing a TAA designated carcinoembryonic antigen (CEA). CEA is a glycoprotein expressed at high level on the surface of nearly all tumors of the gastrointestinal tract, as well as on many mammary carcinomas and lung adenocarcinomas. (Muraro, et al., (1985) Cancer Res. 4S:5769-5780.) A recombinant vaccinia virus that expresses CEA (Kantor, et al. (1992) J. Natl. Cancer Inst.
84:1084-1091) was evaluated using a murine tumor model in which the human CEA gene was transduced into murine colon carcinoma cells. (Robbins, et al. (1991) Cancer Res. 51:3657-3662.) Mice immunized with the CEA/vaccinia recombinant were resistant to the growth of subsequently transplanted CEA-expressing tumors.
Moreover, when mice bearing established CEA-transduced murine carcinomas were treated with the recombinant virus, the tumors showed greatly reduced growth or complete regression. In rhesus monkeys, which carry an antigen on the surface of their granulocytes that cross-reacts with human CEA, immunization with the recombinant elicited anti-CEA antibodies, delayed type hypersensitivity, and lymphoproliferative responses. (Kantor, et al. (1992) Cancer Res.
52:6917-6925.) No toxicity was observed.
Prostate-specific antigen (PSA) is a 33,000-34,000 dalton glycoprotein that is produced in normal, benign, and cancerous prostate 15 epithelia, but not in other normal or malignant tissues. (Wang, et al. (1982) Meth Cancer Res. 19):179-197.) PSA is secreted into prostatic fluid and seminal plasma. (See, Wang, et al.) Elevation of PSA levels in serum is correlated with growth of the prostate, and prostate cancer patients show an exponential increase in PSA levels.
20 (Carter, et al. (1992) Cancer Research 52:3323-3328.) Due to its tissue specificity, PSA is a potential target antigen for immunotherapy against prostate cancer.
A number of laboratories have explored the use of recombinant poxviruses that express specific TAAs as immunotherapeutic vaccines.
The ability of recombinant poxviruses expressing a variety of antigens to serve as potential vaccines for the prevention of infectious disease has been well-documented. Immunization with live recombinant pox virus allows expression of foreign antigens that are presented to the immune system together with highly immunogenic, virus proteins, which may act as adjuvants to enhance immune responses to the foreign antigen. Austin, et al. (1979) Adv. Cancer Res. 50:301- 345. Finally, poxviruses are not oncogenic and do not integrate into the host cell genome, as replication and transcription of genetic material occurs in the cytoplasm of the infected cell.
Viruses of the family Poxviridae (pox viruses) are useful as vectors for the delivery of foreign genes and gene products in many clinical and research settings. Pox viruses of the genus Orthopoxvirus, particularly vaccinia, are used for several reasons. Among these are: its wide use in humans in the eradication of smallpox; its ability to infect a wide range of cells, including professional antigen presenting cells, and express the inserted gene product foreign gene product) in a manner that has the potential to be processed in the context of 15 class I and/or class II MHC molecules; and use as a recombinant vaccine in the treatment of certain tumors (Kantor, J. et al. (1992)).
Fowlpox virus (FPV) is a member of the avipox virus family.
Productive FPV infection is restricted in vivo to cells derived from avian 20 species; however, FPV-mediated gene expression does occur in infected non-avian cells. Taylor, J. et al., (1988) Vaccine 6:497-503.
Fowlpox virus based recombinant vaccines are described in: V Technological Advances in Vaccine Development (Alan R. Liss, Inc.) pp.
321-334. Furthermore, in vivo FPV-mediated gene expression in several mammalian species has been demonstrated. Six non-avian species immunized with live recombinant fowlpox virus expressing the rabies glycoprotein developed antibodies against this glycoprotein.
Immunization with this recombinant FPV elicited antibodies against this glycoprotein. Immunization with this recombinant FPV partially protected mice, cats, and dogs against a rabies virus challenge. There was no manifestation of proliferative infection or overt disease in any animals immunized with a variety of doses of live recombinant FPV. Taylor, et at.. (1988) Vaccine 6:497- 503. In another study, a recombinant FPV containing a measles fusion protein was shown to partially protect mice against lethal challenge the measles virus, although antibodies against the fusion protein were not detected. Wild, et at., (1990) Vaccine 8:441-442.
It was therefore postulated that protection was mediated by cellular immune responses.
These results suggest that recombinant FPV may have utility as a safe and effective alternative to vaccinia virus as a vaccine vector.
Summary of the Invention The present invention provides a method for treating a host having a tumor expressing a tumor associated antigen (TAA) selected from the group consisting of a carcinoembryonic antigen (CEA), prostate specific antigen (PSA), and mucin (MUC-1) comprising the step of: i) administering to a host a first recombinant pox virus vector system that encodes co-stimulatory molecule B7 and the TAA; and ii) administering, at least 1-3 months thereafter, a second recombinant pox virus vector system that is from a different genus than the first and that encodes co-stimulatory molecule B7 and TAA, thereby boosting said host.
20 The present invention also relates to recombinant pox viruses capable of expressing cell-encoded tumor associated antigens and/or immunomodulators.
Recombinant pox virus capable of expressing a cell-encoded tumor-associated antigen are produced by integrating into the pox virus genome sequences encoding the 25 antigen or immunogenic portions thereof. Tumor associated antigens include molecules expressed by tumor cells carcinoembryonic antigen, prostate-specific antigen (PSA).
MUCIN (MUC-1), melanoma associated antigens such as MART-1, etc.).
Immunomodulators include interleukin 2, B7.1 and B7.2.
Particularly preferred recombinant pox viruses include TBC-CEA (vaccinia, CEA), PROSTVAC (vaccinia, PSA), rV-B7.1 (vaccinia, B7), Rf-B7.1 (fowlpox, rF-CEA (fowlpox, CEA), rV-MUC-1 (vaccinia, MUC-1) and rF-PSA (fowlpox, PSA).
-6- 17/09/03,swl12158spa,6 -7- The present invention relates to a method for treating a host having a tumor expressing carcinoembryonic antigen comprising contacting the host with an effective amount of the recombinant pox virus TBC-CEA or rF-CEA.
The present invention also relates to a method for treating a host having a tumor expressing prostate specific antigen comprising contacting the host with an effective amount of the recombinant pox virus rF-PSA.
The present invention further relates to a method for treating a host having a tumor expressing muc-1 antigen comprising contacting the host with an effective amount of the recombinant pox virus rV-muc-1 or rF-muc-1.
The present invention even further relates to a method for treating a host having a tumor comprising contacting tumor cells of said host with an effective amount of the recombinant pox virus rV-B7.1 or rF-B7.1.
The present invention also relates to use of recombinant pox virus of the invention in the preparation of a medicament for the treatment of a tumor; a 15 tumor expressing carcinoembryonic antigen; a tumor expressing prostate specific antigen; or a tumor expressing muc-1 antigen.
l 04/07/01,cf12158.speci,7 Figure 1 is a plasmid map of pT1001, the donor plasmid used in the construction of PROSTVAC.
Figure 2 is a plasmid map of pT108, the donor plasmid used in the construction of TBC-CEA.
Figure 3 is a plasmid map of pT109, the donor plasmid used in the construction of rF-CEA.
Figure 4 is a plasmid map of pT1046, the donor plasmid used in the construction of rV-B7.1.
Figure 5 is a plasmid map of pT1058, the donor plasmid used in the construction of rF-B7.1.
igure 6 is a plasmid map of pT2078, the donor plasmid used in the construction of rF-PSA.
Figure 7 is a plasmid map of pT2068, the donor plasmid used in the construction of rV-muc-1.
Detailed Description of the Invention Pox viruses serve as effective vectors for inducing immunity against tumor-associated antigens.
Particularly preferred tumor-associated antigens are cell surface molecules. These are positioned for recognition by elements of the immune systems and thus are excellent targets for immunotherapy.
Tumor-associated antigens are expressed by certain tumor cells and provide effective targets for immunotherapy. Some examples are carcinoembryonic antigen (CEA) and prostate-specific antigen (PSA).
Immunomodulators can regulate immune responses, increasing the likelihood of a sufficient cellular immune response occurring, (e.g.
IL-2) or can provide cellular ligand necessary for stimulating a CTL response B7.1 or B7.2) Basic techniques for preparing pox viruses containing a heterologous DNA sequence encoding the tumor-associated antigen known to the skilled artisan and involve, for example, homologous recombination between the viral DNA sequences flanking the DNA sequence in a donor plasmid and homologous sequences present in the parental virus (Mackett, et al., Proc. Natl. Acad. Sci. USA 79:7415- 15 7419 (1982)). For example, recombinant viral vectors such as a pox viral vector can be used in delivering the gene. The vector can be o o constructed for example by steps known in the art, e.g. analogous to the methods for creating synthetic recombinants of the fowlpox virus described in U.S. Patent No. 5,093,258, the disclosure of which is 20 incorporated herein by reference. Other techniques include using a unique restriction endonuclease site that is naturally present or artificially inserted in the parental viral vector to insert the heterologous i DNA. See, U.S. Patent No. 5,445,953, incorporated herein by reference.
Pox viruses useful in practicing the present invention include orthopox, suipox, avipox and capripoxvirus.
Orthopox virus include vaccinia, ectromelia and raccoon pox.
The preferred orthopox is vaccinia. More preferred is a sub-clone of vaccinia having decreased virulence relative to a standard vaccine strain of vaccinia.
Avipox viruses include fowlpox, canary pox and pigeon pox. The preferred avipox is fowlpox.
A preferred suipox is swinepox.
For example, the DNA gene sequence to be inserted into the virus can be placed into a donor plasmid, an E. co/i plasmid construct. Separately the DNA gene sequence to be inserted is ligated to a promoter. The promoter-gene linkage is positioned in the plasmid construct so that the promoter-gene linkage is flanked on both ends by DNA homologous to a DNA sequence flanking a region of pox DNA S 15 which is the desired insertion region. With a parental pox viral vector, a pox promoter is used. The resulting plasmid construct is then amplified by growth within E. coil bacteria and isolated. Preferably, the plasmid also contains an origin of replication such as the E. coli origin of replication, and a marker such as an antibiotic resistance gene for 20 selection and propagation in E. coli.
Second, the isolated plasmid containing the DNA gene sequence to be inserted is transfected into a cell culture, chick embryo fibroblasts, along with the parental virus, poxvirus.
25 Recombination between homologous pox DNA in the plasmid and the viral genome respectively results in a recombinant poxvirus modified by the presence of the promoter-gene construct in its genome, at a site which does not affect virus viability.
As noted above, the gene is inserted into a region (insertion region), in the virus which does not affect virus viability of the resultant recombinant virus. The skilled artisan can readily identify such regions in a virus by, for example, randomly testing segments of virus DNA for regions that allow recombinant formation without seriously affecting virus viability of the recombinant. One region that can readily be used and is present in many viruses is the thymidine kinase (TK) gene. For example, the TK gene has been found in all pox virus genomes examined [leporipoxvirus: Upton, et al., J. Virology, 60:920 (1986) (shope fibroma virus); capripoxvirus: Gershon, et al., J. Gen. Virol., 70:525 (1989) (Kenya sheep-1); orthopoxvirus: Weir, et al., J. Virol., 46:530 (1983) (vaccinia); Esposito, et al., Virology, 135:561 (1984) (monkeypox and variola virus); Hruby, et al., PNAS, 80:3411 (1983) (vaccinia); Kilpatrick, et al., Virology, 143:399 (1985) (Yaba monkey tumor virus); avipoxvirus: Binns, et al., J. Gen. Virol. 69:1275 (1988) 15 (fowlpox); Boyle, et al., Virology, 156:355 (1987) (fowlpox); Schnitzlein, et al., J. Virological Methods, 20:341 (1988) (fowlpox, **ee quailpox); entomopox (Lytvyn, et al., J. Gen. Virol. 73:3235-3240 (1992)].
20 In vaccinia, in addition to the TK region, other insertion regions include, for example, Hindll M.
n fowlpox, in addition to the TK region, other insertion regions include, for example, BamHl J [Jenkins, et al., AIDS Research and Human Retroviruses 7:991-998 (1991)] the EcoRI-Hindlll fragment, BamHI fragment, EcoRV-Hindll fragment, BamHI fragment and the Hindlll fragment set forth in EPO Application No. 0 308 220 Al.
[Calvert, et al., J. of Virol. 67:3069-3076 (1993); Taylor, et al., Vaccine 6:497-503 (1988); Spehner, et al., (1990) and Boursnell, et al., J. of Gen. Virol. 71:621-628 (1990)].
In swinepox preferred insertion sites include the thymidine kinase gene region.
In addition to the requirement that the gene be inserted into an insertion region, successful expression of the inserted gene by the modified poxvirus requires the presence of a promoter operably linked to the desired gene, in the proper relationship to the inserted gene.
The promoter must be placed so that it is located upstream from the gene to be expressed. Promoters are well known in the art and can readily be selected depending on the host and the cell type you wish to target. For example in poxviruses, pox viral promoters should be used, such as the vaccinia 7.5K, or 40K or fowlpox C1. Artificial constructs containing appropriate pox sequences can also be used. Enhancer 15 elements can also be used in combination to increase the level of expression. Furthermore, the use of inducible promoters, which are also well known in the art, are preferred in some embodiments.
Vaccines 20 Live recombinant viruses expressing an immunogenic cell encoded tumor associated antigen can be used to induce an immune response against tumor cells which express the protein. These recombinant viruses may be administered by intradermal scarification, as was conventionally done for small pox vaccination, or by other 25 routes appropriate to the recombinant virus used. These may include ego. among others, intramuscular, subcutaneous, and intravenous routes.
Vaccination of a host organism with live recombinant vaccinia virus is followed by replication of the virus within the host.
For parenteral administration, the recombinant vectors will typically be injected in a sterile aqueous or non-aqueous solution, suspension or emulsion in association with a pharmaceuticallyacceptable carrier such as physiological saline.
A specific immune response to a tumor associated antigen can be generated by administering between about 10 5 -109 pfu of the recombinant pox virus, constructed as discussed above to a host, more preferably one uses >107 pfu. The preferred host is a human. At least one interval thereafter, which is preferably one to three months later, the immune response is boosted by administering additional antigen to the host. More preferably there is at least a second "boost" preferably at least one to three months after the first boost, more preferably 6-12 months after the first boost. The antigen for boosting may be administered using the same pox virus vector. The boosting antigen 15 may be administered as a whole protein, an immunogenic peptide fraction of the protein, or DNA encoding the protein or peptide. The boosting antigen may preferably be administered using a second pox virus vector from a different pox genus, or may be administered directly, for example, purified protein plus an adjuvant or in a liposome 20 formation. Cytokines, IL-2, IL-6, IL-12, IL-15, or co-stimulatory molecules, B7.1, 87.2, may be used as biologic adjuvants. The cytokines can be administered systemically to the host, either cytokines or costimulatory molecules can be co-administered via insertion of the genes encoding the molecules into the recombinant pox vector or a second recombinant poxvirus which is admixed with the recombinant poxvirus expressing the TAA.
Adjuvants include, for example, RIBI Detox (Ribi Immunochemical), QS21 (Cambridge Biotech), incomplete Freund's adjuvant or many others.
Generation of Cytotoxic T-Cells Cytotoxic T-cells specific for a tumor specific antigen can be established from peripheral blood mononuclear cells (PBMC) obtained from a host immunized as discussed above. For example, PBMC can be separated by using Lymphocyte Separation Medium gradient (Organon Teknika, Durham, NC, USA) as previously described fBoyum, et al., Scand J. Clin Lab Invest 21: 77-80 (1968)]. Washed PBMC are resuspended in a complete medium, for example, RPMI 1640 (GIBCO) supplemented with 10% pool human AB serum (Pel-Freeze Clinical System, Brown Dear, WI, USA), 2mM glutamine, 100 U/ml penicillin and 100 pg/ml of streptomycin (GIBCO). PBMC at a concentration of about 2 x 10 s cells in complete medium in a volume of, for example, 100 pl are added into each well of a 96-well flat-bottom assay plate 15 (Costar, Cambridge, MA, USA). The antigen or peptides are added into the cultures in a final concentration of about 50 pg/ml and incubated at 37 0 C in a humidified atmosphere containing 5% CO 2 for 5 days. After removal of peptide containing media, the cultures are provided with fresh human IL-2 (10U/mi) after 5 days and replenished with IL-2 20 containing medium every 3 days. Primary cultures are restimulated with the same peptide (50 pg/ml) on day 16. 5 x 10 s irradiated (4,000 rad) autologous PBMC are added in a volume of about 50 pl complete medium as antigen-presenting cells (APC). About five days later, the cultures are provided with human IL-2 containing medium as described 25 previously. Cells are restimulated for 5 days at intervals of 16 days.
Cell Therapy The cytotoxic T-cell can be cultured to amplify its number and then injected back into the host by a variety of means. Generally, between 1 x 10 s and 2 x 10" cytotoxic T-cells per infusion are administered in, for example, one to three infusions of 200 to 250 ml each over a period of 30 to 60 minutes. After the completion of the infusions, the patient may be treated with recombinant interleukin-2 with a dose of 720,000 IU per kilogram of body weight intravenously every eight hours; some doses can be omitted depending on the patient's tolerance for the drug. In addition, after infusion, additional antigen or fragments containing T-cell eliciting epitope(s) may be administered to the patient to further expand the T-cell number. The antigen or epitope may be formulated with an adjuvant and/or may be in a liposomal formulation.
The cytotoxic T-cells can also be modified by introduction of a viral vector containing a DNA encoding TNF and reintroduced into a host in an effort to enhance the anti-tumor activity of the cells. Other cytokines can also be used.
i 2 .20 REFERENCE EXAMPLE 1 CONSTRUCTION OF VECTORS Pox Viruses A number of pox viruses have been developed as live viral vectors for the expression of heterologous proteins (Cepko et al., Cell 37:1053-1062 (1984); Morin et al., Proc. Natl. Acad. Sci. USA 84:4626-4630 (1987); Lowe et al., Proc. Natl. Acad. Sci. USA, 84:3896-3900 (1987); Panicali Paoletti, Proc. Natl. Acad. Sci. USA, efl.
79:4927-4931 (1982); Machett et al., Proc. Natl. Acad. Sci. USA, 79:7415-7419 (1982)). Representative fowlpox and swinepox virus are available through the ATCC under accession numbers VR-229 and VR-363, respectively. The Wyeth strain of vaccinia is available through the ATCC under accession number VR-325.
The parental fowlpox virus is a plaque purified isolate of the POXVAC-TC vaccine strain of fowlpox virus (Schering Corp.).
The parental vaccinia virus (clone B-3-1) is a plaque-purified isolate of the Wyeth strain that was received from Flow Laboratories.
To obtain clone B-3-1, the Wyeth strain of vaccinia was passaged in the fetal rhesus lung line FRhL (ATCC Accession No.
CL160) as follows: Pass 1: Plaque was picked at 10s dilution Pass 2: Plaque was picked at 10-1.
6 dilution Pass 3: Plaque was picked at 10- 1 6 dilution Pass 4-7: Plaque B-3-1 was serially passaged using serum-free medium with sucrose phosphate glutamate (SPG) to prepare a small seed pool at the 7th passage level.
The virulence of this plaque isolate of the Wyeth vaccinia strain was assessed by determining the infections dose of the virus lethal to (LDso) of weaning mice infected intracranially. Two to three week old immunocompetent mice were inoculated with various doses of virus (7 mice/dose); the LD 5 o was determined on mice succubing between 2 and 12 days post-inoculation by calculating the 50% endpoint using the Reed-Muench method These values were compared to those obtained using a virus stock prepared by expanding virus directly from a vial of the CDC Smallpox Vaccine (Table 1).
Table 1 Neurovirulence of Wyeth Vaccinia Virus in Mice Experiment LDo (plaque forming units) Wyeth master virus stock Small pox vaccine 1 ND' 0 2 ND 105.6 3 1 0 6 .7 10 6 4 106.
8 10.4 5 107.3 N 7 .7
ND
7 10 10 5 7 not determined DNA Vectors For In Viv Recombination With A Parent Virus Genes that code for desired carcinoma associated antigens are inserted into the genome of a pox virus in such a manner as to allow them to be expressed.by that virus along with the expression of the normal complement of parent virus proteins. This can be accomplished by first constructing a DNA donor vector for in vivo recombination with a pox virus.
In general, the DNA donor vector contains the following elements: a prokaryotic origin of replication, so that the vector may 17 be amplified in a prokaryotic host; (ii) a gene encoding a marker which allows selection of prokaryotic host cells that contain the vector a gene encoding antibiotic resistance); (iii) at least one gene encoding a desired protein located adjacent to a transcriptional promoter capable of directing the expression of the gene; and (iv) DNA sequences homologous to the region of the parent virus genome where the foreign gene(s) will be inserted, flanking the construct of element (iii).
Methods for.constructing donor plasmids for the introduction of multiple foreign genes into pox virus are described in W091/19803, the techniques of which are incorporated herein by reference. In general, S: 15 all DNA fragments for construction of the donor vector, including fragments containing transcriptional promoters and fragments containing sequences homologous to the region of the parent virus genome into which foreign genes are to be inserted, can be obtained from genomic DNA or cloned DNA fragments. The donor plasmids can S 20 be mono-,di-, or multivalent can contain one or more inserted foreign gene sequences).
The donor vector preferably contains an additional gene which encodes a marker which will allow identification of recombinant viruses containing inserted foreign DNA. Several types of marker genes can be used to permit the identification and isolation of recombinant viruses.
These include genes that encode antibiotic or chemical resistance see Spyropoulos et al., J. Virol., 62:1046 (1988); Falkner and Moss., J. Viro/., 62:1849 (1988); Franke et al., Mol. Cell. Biol., 5:1918 (1985), as well as genes such as the E coli lacZ gene, that permit identification of recombinant viral plaques by colorimetric assay (Panicali et al., Gene, 47:193-199 (1986)).
Integration Of Foreign DNA Sequences Into The Viral Genome And Isolation Of Recombinants Homologous recombination between donor plasmid DNA and viral DNA in an infected cell results in the formation of recombinant viruses that incorporate the desired elements. Appropriate host cells for in vivo recombination are generally eukaryotic cells that can be infected by the virus and transfected by the plasmid vector. Examples of such cells suitable for use with a pox virus are chick embryo dermal (CED) cells, HuTK143 (human) cells, and CV-1 and BSC-40 (both monkey kidney) cells. Infection of cells with pox virus and transfection of these cells with plasmid vectors is accomplished by techniques 15 standard in the art (Panicali and Paoletti, U.S. Patent No. 4,603,112, W089/03429).
Following in vivo recombination, recombinant viral progeny can be identified by one of several techniques. For example, if the DNA donor vector is designed to insert foreign genes into the parent virus thymidine kinase (TK) gene, viruses containing integrated DNA will be TK' and can be selected on this basis (Mackett et al., Proc. Natl. Acad.
Sci. USA, 79:7415 (1982)). Alternatively, co-integration of a gene encoding a marker or indicator gene with the foreign gene(s) of 25 interest, as described above, can be used to identify recombinant progeny. One preferred indicator gene is the E. coli lacZ gene: recombinant viruses expressing P-galactosidase can be selected using a chromogenic substrate for the enzyme (Panicali et al., Gene, 47:193 (1986)).
Following in vivo recombination, recombinant viral progeny can be identified by one of several techniques. The presence of integrated foreign DNA can be detected by hybridization with a labeled DNA probe specific for the inserted DNA. Preferred techniques for selection, however, are based upon co-integration of a gene encoding a marker or indicator gene along with the gene of interest, as described above. A preferred indicator gene is the E. coil acZ gene which encodes the enzyme P-galactosidase. Selection of recombinant virus expressing pgalactosidase can be done by employing a chromogenic substrate for the enzyme. For example, recombinant viruses are detected as blue plaques in the presence of the substrate 5 -bromo-4-chloro-3-indolyl-p- D-galactoside or other halogenated-indolyl-p-D-galactoside BluoGal
T
Characterizing The Viral Antigens Expressed By Recombinant Viruses Once a recombinant virus has been identified, a variety of methods can be used to assay the expression of the polypeptide encoded by the inserted gene. These methods include black plaque assay (an in situ enzyme immunoassay performed on viral plaques), 20 Western blot analysis, radioimmunoprecipitation (RIPA), and enzyme immunoassay
(EIA).
EXAMPLE I Construction of Recombinant Vaccinia Virus (PROSTVAC) A 786 bp DNA fragment encoding the entire open reading frame of human prostate specific antigen was amplified by reverse transcriptase PCR (GeneAmp RNA PCR Kit, Perkin Elmer, Norwalk,
CT)
from total RNA extracted from the human metastatic prostate adenocarcinoma cell line, LNCaP.FGC (CRL 1740, American Type Culture Collection (ATCC), Rockville, MD). The predicted amino acid sequence derived from the PSA coding sequence was shown to be nearly identical to the published sequence (Lundwall, et al., FEBS Letters, 214:317-322, (1987), differing only in a change from asparagine to tyrosine at position 220. The PSA DNA fragment, containing the entire coding sequence for PSA, 41 nucleotides of the untranslated region, and 520 nucleotides of the 3' untranslated region, was inserted into the Xba I restriction endonuclease cleavage site of the vaccinia virus transfer vector pT116. The resulting plasmid, designated pT1001, contains the PSA gene under the control of the vaccinia virus 40K promoter (Gritz, et al., J. Virol. 64:5948-5957, (1990), and the E. coli lacZ gene under the control of the fowlpox virus C1 promoter (Jenkins, et al., AIDS Research and Human Retroviruses 7:991-998, (1991). The foreign genes are flanked by DNA sequences from the Hind III M region of the vaccinia genome. Figure 1 is a 15 plasmid map of PT1001.
A plaque-purified isolate from the Wyeth (New York City Board of Health) strain of vaccinia was used as the parental virus in the construction of the recombinant vaccinia virus, PROSTVAC. The generation of recombinant vaccinia virus was accomplished via homologous recombination between vaccinia sequences in the Wyeth :'""vaccinia genome and the corresponding sequences in pT1001 in vaccinia-infected RK,3 cells (CCL 37, ATCC) transfected with pT1001.
Recombinant virus was identified using a chromogenic assay, performed on viral plaques in situ, that detects expression of the lacZ gene product in the presence of halogenated indolyl-beta-D-galactoside (Bluo-gal), as described previously (Panacali, et al., 1986). Appropriate blue recombinant viruses were purified by four rounds of plaquepurification. Virus stocks were prepared by clarifying infected RK,, cell lysates followed by centrifugation through a 36% sucrose cushion.
EXAMPLE II Construction of Recombinant Fowlpox Virus (rF-CEA) The generation of recombinant fowlpox viruses is accomplished via homologous recombination in vivo between fowlpox DNA and a plasmid vector that carries the heterologous sequences to be inserted.
The plasmid vectors contain one or more chimeric genes, each comprising a poxvirus promoter linked to a protein coding sequence, flanked by viral sequences from a non-essential region of the fowlpox virus genome. The plasmid is transfected into cells infected with the parental fowlpox virus, and recombination between fowlpox sequences on the plasmid and the corresponding DNA in the viral genome results in the insertion into the viral genome of the chimeric genes on the plasmid.
S. The plasmid vector (pT109) used for insertion of the CEA gene into the parental fowlpox virus genome by in vivo recombination is illustrated in Figure 3. This vector contains the following elements: (1) a prokaryotic origin of replication to allow amplification of the vector in a bacterial host; the gene encoding resistance to the antibiotic ampicillin, to permit selection of prokaryotic host cells that contain the plasmid; DNA sequences homologous to the BamHIJ region of the fowlpox genome, which direct insertion of foreign sequences into this region via homologous recombination; a chimeric gene comprising 25 the vaccinia 40K transcriptional promotor linked to the CEA gene; a second chimeric gene comprising the fowlpox C1 transcriptional promoter linked to the E. co/i lacZ gene.
The gene encoding CEA was isolated at the National Cancer Institute from a cDNA library derived from RNA from a human colon carcinoma cell line as disclosed by Kaufman et al., (1991) Int. J.
Cancer 48, 900-907, the disclosure of which is herein incorporated by reference.
A plaque-purified isolate from the POXVAC-TC vaccine strain of fowlpox virus was used as the parental virus for this recombinant vaccine. In vivo recombination between the plasmid vector and the viral DNA resulted in the formation of a recombinant virus in which the CEA gene, under the transcriptional direction of the vaccinia promoter, and the lacZ gene, under the control of the C1 promoter, were inserted into the BamHIJ region of the fowlpox virus genome.
A chromogenic assay for P-galactosidase was used to identify recombinant viruses containing the lacZ and CEA sequences. This 15 method takes advantage of the ability of fowlpox virus to form distinct "i plaques when grown on monolayers of CED cells. After in vivo recombination, cells were infected with progeny virus until distinct o S* plaques were visible, at which time the plaques were overlaid with a chromogenic substrate for Beta-galactosidase (Bluo-gal). Viral plaques 20 expressing lacZ appeared blue against a clear background. Positive plaques were picked from the cell monolayer and their progeny were further propagated. Repeated rounds of plaque isolation and replating in a presence of Bluo-gal resulted in the purification of the desired recombinant. Positive recombinants were amplified to produce a seed stock on CED cells. The seed stock was then subjected to titration, genomic and protein expression analysis.
The structure of the plasmid transfer vector was verified by restriction endonuclease digestion using Xba I and BamHl. In addition, the products of digestion with these enzymes were subjected to Southern blot analysis using labeled probes corresponding to the CEA gene and to the fowlpox BamHl J sequences. The DNA fragments visualized by these methods were of the predicted sizes, and the presence of the CEA gene was unequivocally demonstrated, thus confirming the predicted structure of the plasmid.
The recombinant pox virus set forth below in Table 2 can be constructed using similar techniques.
.0 TABLE 2 RECOMBINANT DONOR TUMOR REFERENCE POX. PLASMID ASSOCIATED FOR GENE
GENE/IMMUNO-
MODULATOR
TBC-CEA pTlO8 CEA Beachmi t al.
rF-CEA pTl 09 7:3221-3230 W-137.1 pT1O046 B7.1 Frea ta. 1989.
rF-B7. 1 pT1 058 143:2714-272.2 rF-PSA pT2O78 PSA Hodge at al.. 1995.
Int. J1. Cancer.
3:231-237 rV-muc- 1 pT2O68 muc-1 Gendlerat al.. 1990.
.13. Ch Oem..
*rV indicates vaccinia recombinant rF indicates fowlpox recombinant Example III Patients enrolled in the Phase I study were previously diagnosed with rising PSA levels after undergoing radical prostatectomy, radiation therapy or both for treatment of prostate cancer. Patients were divided into three groups which received the following dose levels of PROSTVAC followed by two boosts in one month interval.
Group 1: 2.65 x 106 pfu of recombinant vaccine expressing PSA (rV-PSA) Group 2: 2.65 x 107 pfu rV-PSA Group 3: 2.65 x 108 pfu rV-PSA 10 out of 21 patients of Group 3 also received granulocyte-macrophage colony-stimulating factor (GM-CSF) as an immunostimulatory adjuvant (Group 3b).
S Cells from 7 of these patients were assayed for development of a PSA-specific cellular immune response.
::2o0 No vaccine-related toxicity was observed in any of the Groups beyond a lowgrade reaction at the site of inoculation. In 10 patients of Group 3b only one developed fever and rapid heartbeat after the first dose of GM-CSF but tolerated subsequent GM-CSF doses.
In 14 men out of 33 treated with PROSTVAC, with or without GM-CSF, PSA levels remained stable for at least 6 months. In addition, 9 patients remained stable for 11-25 months.
Of the 7 patients whose cells were assayed for development of PSA-specific cellular immune response, 5 demonstrated at least two-fold increase of precursor Tcells specific for PSA after vaccination. The stabilization of PSA levels in these patients correlated with the absence of any other indications of disease progression.
Equivalents Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
Where the terms "comprise", "comprises", "comprised" or "comprising" are used in this specification, they are to be interpreted as specifying the presence of the stated features, integers, steps or components referred to, but not to preclude the presence or addition of one or more other feature, integer, step, component or group thereof.
This is a divisional application of Australian Patent No. 731860 the disclosure of which is incorporated herein by way of reference.
*9*9
S

Claims (13)

1. A method for treating a host having a tumor expressing a tumor associated antigen (TAA) selected from the group consisting of a carcinoembryonic antigen (CEA), prostate specific antigen (PSA), and mucin (MUC-1) comprising the step of: i) administering to a host a first recombinant pox virus vector system that encodes co-stimulatory molecule B7 and the TAA; and ii) administering, at least 1-3 months thereafter, a second recombinant pox virus vector system that is from a different genus than the first and that encodes co-stimulatory molecule B7 and TAA, thereby boosting said host.
2. The method of claim 1, wherein said co-stimulatory molecule B7 is B7.1.
3. The method of claim 1, wherein said co-stimulatory molecule B7 is B7.2.
4. The method of claim 1, wherein the TAA is CEA. The method of claim 1, wherein the TAA is PSA.
6. The method of claim 1, wherein the TAA is MUC-1.
7. The method of claim 1, wherein said first recombinant pox virus vector system comprises a pox virus vector selected from the group consisting of an orthopox virus vector, an avipox virus vector, a suipox virus vector, and a capripox virus vector.
8. The method of claim 1, wherein said second recombinant pox virus vector system comprises a pox virus vector selected from the group consisting of an orthopox virus vector, an avipox virus vector, a suipox virus vector, and a capripox virus vector.
9. The method of claim 1, wherein the first recombinant pox vector system is an orthopox virus and the second recombinant pox vector system is an avipox.
10. The method of claim 9, wherein the orthopox virus is vaccinia.
11. The method of claim 1, wherein said first recombinant pox vector system comprises one pox vector containing DNA sequences encoding both B7 and the TAA. -27- 17/09/03,sw 2158spa,27
12. The method of claim 1 or 11, wherein the second recombinant pox vector system comprises one pox vector containing DNA sequences encoding both B7 and the TAA.
13. The method of claim 1, wherein the first recombinant vector system contains two pox vectors, one pox vector encoding B7 and the other pox vector encoding the TAA.
14. The method of treatment as claimed in claim 1, substantially as herein described with reference to Examples 1 or 2 and/or the accompanying Figures. Dated this 17 th day of September, 2003 THERION BIOLOGICS CORPORATION and UNITED STATES GOVERNMENT as represented by THE DEPARTMENT OF HEALTH AND HUMAN SERVICES By their Patent Attorneys: CALLINAN LAWRIE -28- 17/09/03,sw 2158spa,28
AU54194/01A 1996-07-25 2001-07-04 Recombinant pox virus for immunization against tumor-associated antigens Ceased AU767562B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU54194/01A AU767562B2 (en) 1996-07-25 2001-07-04 Recombinant pox virus for immunization against tumor-associated antigens
AU2003266471A AU2003266471A1 (en) 1996-07-25 2003-12-04 Recombinant pox virus for immunization against tumor-associated antigens

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/686280 1996-07-25
AU37268/97A AU731860B2 (en) 1996-07-25 1997-07-15 Recombinant pox virus for immunization against tumor-associated antigens
AU54194/01A AU767562B2 (en) 1996-07-25 2001-07-04 Recombinant pox virus for immunization against tumor-associated antigens

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU37268/97A Division AU731860B2 (en) 1996-07-25 1997-07-15 Recombinant pox virus for immunization against tumor-associated antigens

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2003266471A Division AU2003266471A1 (en) 1996-07-25 2003-12-04 Recombinant pox virus for immunization against tumor-associated antigens

Publications (2)

Publication Number Publication Date
AU5419401A AU5419401A (en) 2001-09-13
AU767562B2 true AU767562B2 (en) 2003-11-13

Family

ID=29425372

Family Applications (1)

Application Number Title Priority Date Filing Date
AU54194/01A Ceased AU767562B2 (en) 1996-07-25 2001-07-04 Recombinant pox virus for immunization against tumor-associated antigens

Country Status (1)

Country Link
AU (1) AU767562B2 (en)

Also Published As

Publication number Publication date
AU5419401A (en) 2001-09-13

Similar Documents

Publication Publication Date Title
AU731860B2 (en) Recombinant pox virus for immunization against tumor-associated antigens
US7118738B2 (en) Recombinant pox virus for immunization against MUC1 tumor-associated antigen
AU774076B2 (en) A recombinant vector expressing multiple costimulatory molecules and uses thereof
US6969609B1 (en) Recombinant vector expressing multiple costimulatory molecules and uses thereof
JP4078319B2 (en) A composition comprising a recombinant virus expressing an antigen and a recombinant virus expressing an immunostimulatory molecule
EP0789774A2 (en) Enhanced immune response by introduction of cytokine gene and/or costimulatory molecule b7 gene in a recombinant virus expressing system
AU2004289340B2 (en) System for treating and preventing breast cancer
AU718945B2 (en) Recombinant pox virus for immunization against tumor-associated antigens
US20050100558A1 (en) Heterologous boosting immunizations
CA2252406A1 (en) Heterologous boosting immunizations
AU767562B2 (en) Recombinant pox virus for immunization against tumor-associated antigens
AU2003266471A1 (en) Recombinant pox virus for immunization against tumor-associated antigens
EP1865065A1 (en) A recombinant vector expressing multiple costimulatory molecules and uses thereof
Chamberlain et al. THE USE OF RECOMBINANT POXVIRUSES AS A PARADIGM FOR THE DEVELOPMENT OF ANTI-CANCER VACCINES

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)