WO2013116656A1 - Immunostimulatory compositions, particles, and uses related thereto - Google Patents

Immunostimulatory compositions, particles, and uses related thereto Download PDF

Info

Publication number
WO2013116656A1
WO2013116656A1 PCT/US2013/024355 US2013024355W WO2013116656A1 WO 2013116656 A1 WO2013116656 A1 WO 2013116656A1 US 2013024355 W US2013024355 W US 2013024355W WO 2013116656 A1 WO2013116656 A1 WO 2013116656A1
Authority
WO
WIPO (PCT)
Prior art keywords
particle
antigen
molecule
virus
adjuvant
Prior art date
Application number
PCT/US2013/024355
Other languages
French (fr)
Inventor
Periasamy Selvaraj
Original Assignee
Emory University
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 Emory University filed Critical Emory University
Priority to EP18196067.5A priority Critical patent/EP3520809A1/en
Priority to EP13743997.2A priority patent/EP2809345A4/en
Priority to CA2863658A priority patent/CA2863658C/en
Priority to US14/374,729 priority patent/US20150071987A1/en
Publication of WO2013116656A1 publication Critical patent/WO2013116656A1/en
Priority to US15/833,769 priority patent/US10987419B2/en
Priority to US17/132,320 priority patent/US20210154291A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001106Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001124CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001129Molecules with a "CD" designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/00117Mucins, e.g. MUC-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/001172Sialyl-Thomson-nouvelle antigen [sTn]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001169Tumor associated carbohydrates
    • A61K39/001173Globo-H
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00118Cancer antigens from embryonic or fetal origin
    • A61K39/001182Carcinoembryonic antigen [CEA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00119Melanoma antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/00119Melanoma antigens
    • A61K39/001191Melan-A/MART
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001193Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; PAP or PSGR
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001193Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; PAP or PSGR
    • A61K39/001194Prostate specific antigen [PSA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001193Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; PAP or PSGR
    • A61K39/001195Prostate specific membrane antigen [PSMA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5152Tumor cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • A61K2039/55527Interleukins
    • A61K2039/55533IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • A61K2039/55527Interleukins
    • A61K2039/55538IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6018Lipids, e.g. in lipopeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/62Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/62Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier
    • A61K2039/627Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier characterised by the linker

Definitions

  • ProvengeTM is a recently FDA-approved autologous cellular immunotherapy treatment.
  • Peripheral blood leukocytes of a subject are harvested via leukapheresis. These enriched monocytes are incubated with prostatic acid phosphatase (PAP) conjugated to cytokine granulocyte macrophage colony stimulating factor (GM-CSF).
  • PAP prostatic acid phosphatase
  • GM-CSF cytokine granulocyte macrophage colony stimulating factor
  • GM-CSF cytokine granulocyte macrophage colony stimulating factor
  • GM-CSF cytokine granulocyte macrophage colony stimulating factor
  • GM-CSF cytokine granulocyte macrophage colony stimulating factor
  • GM-CSF cytokine granulocyte macrophage colony stimulating factor
  • GM-CSF cytokine granulocyte macrophage colony stimulating factor
  • GM-CSF cytokine granulocyte macrophag
  • B7-1 (also known as CD80) is a T cell costimulatory molecule that can be anchored in to autologous cancer cells to stimulate immune responses. McHugh et al., report the construction, purification and functional reconstitution of a glyco lipid anchored form of B7- 1 (CD80) on tumor cell membranes. Proc. Natl. Acad. Sci. USA 1995; 92:8059-8063. See also U.S. Patent 6,491,925. Glycosyl phosphatidylinositol anchored B7-1 (GPI-B7-1) molecules have been incorporated onto tumor cells and isolated tumor cell membranes to provide costimulation for allogenic T cell proliferation.
  • Glycosyl phosphatidylinositol anchored B7-1 (GPI-B7-1) molecules have been incorporated onto tumor cells and isolated tumor cell membranes to provide costimulation for allogenic T cell proliferation.
  • a method of tumor treatment or tumor vaccination generally comprises applying to a human being in need thereof a tumor therapeutic composition or tumor vaccine defined herein.
  • the tumor therapeutic composition or tumor vaccine can be produced by protein transfer of glycosyl- phosphatidylinositol (GPI)-anchored immunostimulatory or costimulatory molecules.
  • GPI glycosyl- phosphatidylinositol
  • the tumor therapeutic composition or tumor vaccine comprises a live tumor cell or tumor cell membranes that is or are modified by protein transfer to express one or more GPI-anchored immunostimulatory or costimulatory molecules.
  • the tumor therapeutic composition or tumor vaccine can be prepared by a method that comprises obtaining one or more GPI-anchored immunostimulatory or costimulatory molecules, and transferring the GPI-anchored immunostimulatory or costimulatory molecules onto a tumor cell or isolated tumor cell membranes by protein transfer.
  • the disclosure relates to non-naturally occurring particle comprising, a lipid membrane; a B7-1 and/or B7-2 molecule anchored to the lipid membrane on the exterior of the particle; and an antigen molecule such as a tumor specific antigen or cancer marker anchored to the lipid membrane on the exterior of the particle.
  • the particle further comprises an adjuvant molecule anchored to the lipid membrane on the exterior of the particle wherein the adjuvant molecule and antigen molecule are not the same molecule.
  • the adjuvant molecule is selected from IL-2, IL-12, ICAMl GM-CSF, flagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA).
  • the lipid membrane may be a phospholipid monolayer or phospholipid bilayer.
  • the particle is selected from a cell, allogeneic or autologous cancer cell or its membrane fragments or vesicles, liposome, virosome, micelle, polymer, and virus like particle.
  • the B7-1 molecule is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl-phosphatidylinositol, phospholipid, gly co lipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
  • the antigen molecule such as a tumor associated antigen or cancer marker is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl-phosphatidylinositol, phospholipid, glycolipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
  • the adjuvant molecule is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl-phosphatidylinositol,
  • phospholipid phospholipid, glycolipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
  • Particles comprising membranes such as tumor membranes carrying tumor antigens and immunostimulatory stimulatory molecules can be modified by incubating with lipophilic adjuvants such as lipopolysaccharides or an immunostimulatory unmethylated CpG oligonucleotides lipid conjugate.
  • lipophilic adjuvants such as lipopolysaccharides or an immunostimulatory unmethylated CpG oligonucleotides lipid conjugate.
  • antigen is a cancer marker molecule selected from HER-2, MUC-1, mucin antigens TF, Tn, STn, glycolipid globo H antigen, prostate-specific antigen, prostate-specific membrane antigen, early prostate cancer antigen-2 (EPCA-2), BCL-2, MAGE antigens such as CT7, MAGE- A3 and MAGE-A4, G-protein coupled estrogen receptor 1 , CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD1 17, melanoma-associated antigen (TA-90), peripheral myelin protein 22 (PMP22), epithelial membrane proteins (EMP-1, -2, and -3), HMB-45 antigen, MART-1 (Melan-A), S100A1, S100B and gp 100:209-217(210M).
  • MAGE antigens such as CT7, MAGE- A3 and MAGE-A4, G
  • the disclosure relates to virus like particles comprising B7-1 and/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule anchored to the lipid membrane on the exterior of the particle.
  • the antigen molecule is a cancer marker or tumor associated antigen or tumor-specific antigen selected from HER-2, MKI67, prostatic acid phosphatase (PAP), prostate-specific antigen (PSA), prostate-specific membrane antigen, early prostate cancer antigen-2 (EPCA- 2), BCL-2, MAGE antigens, antigens comprising a Mage Homology Domain (MHD), MAGE-1, CT7, MAGE-A3 and MAGE-A4, ERK5, G-protein coupled estrogen receptor 1 , CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD 117, melanoma-associated antigen (TA-90), peripheral
  • the virus like particle further comprising an adjuvant molecule anchored to a lipid membrane on the exterior of the particle wherein the adjuvant molecule and the antigen molecule are not the same molecule.
  • the adjuvant molecule is selected from is IL-2, IL- 12, ICAM1 GM-CSF, flagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA).
  • the disclosure relates to methods of treating cancer comprising administering an effective amount of a particle or a virus like particle as disclosed herein to a subject at risk of or diagnosed with cancer or a tumor optionally in combination with anti-CTLA-4 antibodies such as abatacept, belatacept, ipilimumab, tremelimumab, anti-PD-1 and PDL1 antibodies such as nivolumab, unmethylated CpG oligonucleotide, methyl jasmonate, cyclophosphamide, gemcitabine or other
  • the subject is a human subject and the virus like particle comprises a B7-land/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule wherein the antigen molecule is a viral protein.
  • anticancer agents contemplated include gefitinib, erlotinib, docetaxel, cis- platin, 5-fluorouracil, gemcitabine, tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin, vincristine, vinblastine, vindesine, vinorelbine taxol, taxotere, etoposide, teniposide, amsacrine, topotecan, camptothecin bortezomib anegrilide, tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene fulvestrant, bicalutamide, flutamide
  • the viral like particle has an hemagglutinin selected from influenza HI, H2, H3, H4, H5, H6, H7, H8, H9, H10, HI 1, H12, H13, H14, H15, and H16 optionally in combination with or individually influenza Nl, N2, N3, N4, N5, N6, N7, and N8.
  • the virus protein is an HIV envelope protein selected from gp 41, gp 120, and gp 160.
  • the disclosure relates to methods of treating or preventing a viral infection comprising administering an effective amount of a virus like particle disclosed herein to a subject at risk of, exhibiting symptoms of, or diagnosed with a viral infection.
  • the disclosure relates to particles comprising a cancer marker made by the process of mixing a cancer marker conjugated to a lipophilic moiety and a particle comprising a lipid membrane.
  • the cancer marker is HER-2 or PSA or PAP.
  • the disclosure relates to particles comprising a cancer marker and B7-1 and/or B7-2 made by the process of mixing a B7-1 and/or B7-2 conjugated to a lipophilic moiety and a particle comprising a lipid membrane and a cancer marker.
  • the disclosure relates to methods of treating or preventing breast cancer comprising administering an effective amount of a particle comprising B7-1 and/or B7-2, GM-CSF, and HER-2 to a subject in need thereof.
  • the method further comprises analyzing the subject for overexpression of HER-2, by measuring, detecting, sequencing, hybridizing with a probe, HER-2 polypeptide or a nucleic acid indicative of HER-2 expression, or sequencing a nucleic acid associated with HER-2, on a cancer cell or tumor cell isolated from the subject.
  • the disclosure relates to methods of treating or preventing prostate cancer comprising administering an effective amount of a particle comprising B7-1 and/or B7-2, GM-CSF, and PSA or PAP to a subject in need thereof.
  • the disclosure relates to methods of treating or preventing prostate cancer comprising administering an effective amount of a particle comprising B7-1 and/or B7-2, GM-CSF, IL-12, and PSA or PAP to a subject in need thereof.
  • compositions and method further comprises
  • the anticancer agent may be estradiol, tamoxifen, cetuximab and a HER-2 antibody, humanized antibody, or human chimera such as trastuzumab, pertuzumab.
  • the HER-2 antibodies may be administered before or after immune stimulation with particle.
  • the anticancer agent may be docetaxel, cabazitaxel, bevacizumab, alpharadin thalidomide, prednisone, abiraterone, finasteride and dutasteride, MDV3100, orteronel (TAK-700), omega-3 fatty acids such as ethyl esters of
  • EPA eicosapentaenoic acid
  • DHA docosahexaenoic acid
  • the tumor therapeutic composition or tumor vaccine comprises a microparticle with a lipid membrane encapsulating tumor antigens or peptides and one or more anchored immunostimulatory or costimulatory molecules expressed on the surface of the particle.
  • the tumor therapeutic composition or tumor vaccine can be prepared by a method that comprises obtaining one or more anchored immunostimulatory or costimulatory molecules, and transferring the anchored immunostimulatory or
  • microparticles can be formed of any biocompatible polymer capable of incorporating GPI-anchored immunostimulatory or costimulatory molecules.
  • biocompatible polymers include, but are not limited to, polyvinyl alcohols, polyvinyl ethers, polyamides, polyvinyl esters, polyvinylpyrrolidone,
  • polyglycolides polyurethanes, allyl celluloses, cellulose esters, hydroxypropyl derivatives of celluloses and cellulose esters, preformed polymers of poly alkyl acrylates, polyethylene, polystyrene, polyactic acid, polyglycolic acid, poly(lactide-co-glycolide),
  • polycaprolactones polybutyric acids, polyvaleric acid and copolymers thereof, alginates, chitosans, gelatin, albumin, zein and combinations thereof.
  • Anchored immunostimulatory or costimulatory molecules can be obtained by expressing the GPI-anchored immunostimulatory or costimulatory molecules in a cell, and isolating the GPI-anchored immunostimulatory or costimulatory molecules.
  • the anchored immunostimulatory or costimulatory molecules can be any substance that stimulates or costimulates immune reaction against a tumor cell that is capable of being expressed in a cell.
  • the immunostimulatory or costimulatory molecules useful here can be a cytokine molecule.
  • a useful cytokine can be, for example, one or more of cytokines IL-2, IL-4, IL-6, IL-12, IL-15, IL-18, IL-19, granulocyte -macrophage colony stimulating factor (GM-CSF), and combinations thereof.
  • the immunostimulatory or costimulatory molecules can be, for example, the
  • immunostimulatory or costimulatory molecules useful here can be a cytokine molecule.
  • the immunostimulatory or costimulatory molecules useful here can be, for example, B7-1, B7-2 and an intercellular adhesion molecule such as CD40L, ICAM-1, ICAM-2, and ICAM-3.
  • particle may be a wild type cell, cancer cell or immortalized cell.
  • the immunostimulatory or costimulatory molecules can be used alone or together and can be used in conjunction with antibody fusion proteins.
  • the tumor therapeutic composition or tumor vaccine described herein can be used therapeutically or prophylactically for the treatment or prevention of a tumor.
  • Representative tumors can be treated or prevented include, but are not limited to, breast cancer, prostate cancer, lung cancer, melanoma, liver cancer, leukemia, lymphoma, myeloma, colorectal cancer, gastric cancer, bladder carcinoma, esophageal carcinoma, head & neck squamous-cell carcinoma, sarcomas, kidney cancers, ovarian and uterus cancers, adenocarcinoma, glioma, and plasmacytoma, and combinations thereof.
  • the vaccine or therapeutic composition described herein can be GPI-anchored cytokine such as GPI-IL-2 and GPI-IL-12 alone or in combination with GPI- anchored costimulatory molecules such as GPI-B7-1, GPI-B7-2, GPI-ICAM-1, GPI-ICAM- 2 and GPI-ICAM-3.
  • GPI-anchored cytokine such as GPI-IL-2 and GPI-IL-12 alone or in combination with GPI- anchored costimulatory molecules such as GPI-B7-1, GPI-B7-2, GPI-ICAM-1, GPI-ICAM- 2 and GPI-ICAM-3.
  • GPI-anchored costimulatory molecules such as GPI-B7-1, GPI-B7-2, GPI-ICAM-1, GPI-ICAM- 2 and GPI-ICAM-3.
  • the vaccine and therapeutic composition can be any suitable pharmaceutical and therapeutic composition.
  • the vaccine and therapeutic composition can be any suitable pharmaceutical composition.
  • biocompatible microparticles such as biodegradable microparticles modified with GPI- anchored immunostimulatory molecules such as IL-2, IL-4, IL-6, IL-12, ICAM-1, ICAM-2, ICAM-3, B7-1, B7-2, CD40L, IL-15, IL-18, IL-19, granulocyte-macrophage colony stimulating factor (GM-CSF), and combinations thereof.
  • GPI-anchored immunostimulatory molecules such as IL-2, IL-4, IL-6, IL-12, ICAM-1, ICAM-2, ICAM-3, B7-1, B7-2, CD40L, IL-15, IL-18, IL-19, granulocyte-macrophage colony stimulating factor (GM-CSF), and combinations thereof.
  • GPI-anchored immunostimulatory molecules such as IL-2, IL-4, IL-6, IL-12, ICAM-1, ICAM-2, ICAM-3, B7-1, B7-2, CD40L, IL-15, IL
  • the vaccine or therapeutic compositions described herein can be tumor cells or membranes modified by protein transfer with GPI-anchored cytokines alone or/and in combination with other cytokines or/and other costimulatory molecules.
  • One such embodiment can be, for example, tumor membranes modified with purified GPI-IL-12.
  • particles like inactivated or partially attenuated virus, bacteria and virus-like particles can be modified to express immunostimulatory molecules by protein transfer with GPI-anchored cytokines and immunostimulatory molecules.
  • Vaccines and therapeutic compositions prepared in this manner can be used for preventing or treating viral, bacterial, or parasitic diseases or disorders.
  • the vaccine and therapeutic compositions described herein can be used for treating autoimmune disorders.
  • membrane anchored cytokines such as IL-10 and TGF-beta can also be used to induce tolerance or to suppress immunity which can be used in treating autoimmune diseases and transplant rejection.
  • Figure 1 illustrates the expression tumor associated antigens and immunostimulatory molecules onto particles containing a lipid membrane, e.g., CHO cells and envelope VLPs, using GPI anchoring for protein transfer.
  • a lipid membrane e.g., CHO cells and envelope VLPs
  • FIG. 2 shows data on protein transfer of (A) GPI-ICAM1 or (B) GPI-IL-12 onto sheep RBCs. Red: Background control; Black: Protein transfer of GPI-ISMs.
  • FIG. 3 shows data on Concentration dependent protein transfer of (A) GPI-ICAM-
  • Figure 4 shows data on the kinetics of protein transfer of GPI-ICAM-1 onto H5 influenza VLPs.
  • Figure 5 shows data on the specificity of protein transfer of GPI-ICAM1 onto VLPs.
  • Figure 6 shows data on the inhibition of protein transfer of GPI-ICAM1 via fatty acid binding proteins.
  • FIG. 7 shows data on the incorporation of two GPI-ISMs onto VLPs
  • Figure 8 shows a EM of VLPs (A) before and (B) after protein transfer with
  • Figure 9 shows data on the direct challenge with wild-type or GPI cytokine transfected 4T07 cells.
  • Figure 10 shows tumor size in individual mice post direct challenge with wild-type or
  • FIG 11 illustrates the production of extracellular portion of hHER-2 (hHER-
  • Figure 12 shows flow cytometry analysis of CHO cells expressing GPI-human HER-2 (hHER-2-CD59) using TA1 mAb.
  • Embodiments of the present disclosure will employ, unless otherwise indicated, techniques of medicine, organic chemistry, biochemistry, molecular biology, pharmacology, and the like, which are within the skill of the art. Such techniques are explained fully in the literature. It must be noted that, as used in the specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a support” includes a plurality of supports. In this specification and in the claims that follow, reference will be made to a number of terms that shall be defined to have the following meanings unless a contrary intention is apparent.
  • the term "combination with” when used to describe administration with an additional treatment means that the agent may be administered prior to, together with, or after the additional treatment, or a combination thereof.
  • the terms “prevent” and “preventing” include the prevention of the recurrence, spread or onset. It is not intended that the present disclosure be limited to complete prevention. In some embodiments, the onset is delayed, or the severity is reduced.
  • the terms “treat” and “treating” are not limited to the case where the subject (e.g. patient) is cured and the disease is eradicated. Rather, embodiments of the present disclosure also contemplate treatment that merely reduces symptoms, and/or delays disease progression.
  • Subject refers any animal, preferably a human patient, livestock, rodent, monkey or domestic pet.
  • protein and “polypeptide” refer to compounds comprising amino acids joined via peptide bonds and are used interchangeably.
  • amino acid sequence refers to an amino acid sequence of a protein molecule.
  • polypeptide or “protein” are not meant to limit the amino acid sequence to the deduced amino acid sequence, but such as amino acid deletions, additions, and modifications such as glycolsylations and addition of lipid moieties or other post-translational modifications.
  • the protein generally refers to the most frequent human isoform, variant, mutated form, or protein with substantially identity to the full-length or portion thereof. Typically, an appropriate fragment is of the extracellular domain.
  • portion when used in reference to a protein (as in “a portion of a given protein") refers to fragments of that protein.
  • the fragments may range in size from four amino acid residues or more than twenty or thirty or the entire amino sequence minus one amino acid.
  • reference sequence is a defined sequence used as a basis for a sequence comparison; a reference sequence may be a subset of a larger sequence, for example, as a segment of a full-length amino acid sequence of a protein. Generally, a reference sequence is at least 20 amino acids in length, frequently at least 25 amino acids in length, and often at least 50 amino acids in length.
  • two proteins may each (1) comprise a sequence (i.e., a portion of the complete amino acid sequence) that is similar between the two protein, and (2) may further comprise a sequence that is divergent between the two proteins, sequence comparisons between two (or more) proteins are typically performed by comparing sequences of the two proteins over a "comparison window" to identify and compare local regions of sequence similarity.
  • a “comparison window”, as used herein, refers to a conceptual segment of at least 20 contiguous nucleotide positions wherein a sequence may be compared to a reference sequence of at least 20 contiguous amino acids and wherein the portion of the sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • Optimal alignment of sequences for aligning a comparison window may be conducted by the local homology algorithm of Smith and Waterman (Smith and Waterman, Adv. Appl. Math. 2: 482 (1981)) by the homology alignment algorithm of Needleman and Wunsch (Needleman and Wunsch, J. Mol. Biol. 48:443 (1970)), by the search for similarity method of Pearson and Lipman (Pearson and Lipman, Proc. Natl. Acad. Sci. (U.S.) 85:2444
  • sequence identity means that two sequences are identical (i.e., on a nucleotide -by-nucleotide basis) over the window of comparison.
  • percentage of sequence identity is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical amino acids occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
  • substantially identical denotes a characteristic of a sequence, wherein the protein comprises a sequence that has at least 85 percent sequence identity, preferably at least 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison window of at least 20 amino acid positions, frequently over a window of at least 25-50 nucleotides, wherein the percentage of sequence identity is calculated by comparing the reference sequence to the sequence which may include deletions or additions which total 20 percent or less of the reference sequence over the window of comparison.
  • the disclosure relates to non-naturally occurring particle comprising, a B7-1 and/or B7-2 molecule anchored on the exterior of the particle; and an antigen molecule such as a tumor specific antigen or cancer marker anchored to the lipid membrane on the exterior of the particle.
  • the B7-1 and or B7-2 or antigen, or protein may be anchored onto the membrane of the particle through a variety of linkages, such as lipid palmatic acid, biotin-avidin interaction, or a GPI-anchor.
  • a contemplated sequence of B7-1 is MGHTR QGTS
  • a contemplated sequence is VIHVTKEVKE VATLSCGFINV SVEELAQTRI YWQKEKKMVL TMMSGDMNIW PEYKNRTIFD ITNNLSIVIL ALRPSDEGTY ECVVLKYEKD AFKREHLAEV TLSVKADFPT PSISDFEIPT SNIRRIICST SGGFPEPHLS WLENGEELNA INTTVSQDPE TELYAVSSKL DFNMTTNHSF MCLIKYGHLR VNQTFNWNTT KQEHFPDN (SEQ ID NO:2) or fragment thereof. See Stamper et al., Crystal structure of the b7-l/ctla-4 complex that inhibits human immune responses.
  • a contemplated fragment is KAMHVAQPAV VLASSRGIAS FVCEYASPGK ATEVRVTVLR QADSQVTEVC AATYMMGNELTFLDDSICTG TSSGNQVNLT IQGLRAMDTG LYICKVELMY PPPYYLGIGN GAQIYVIDPE PCPDSD (SEQ ID NO: 3) or fragment thereof.
  • the disclosure relates to non-naturally occurring particle comprising, a B7-1 and/or B7-2 molecule anchored on a lipid membrane; a B7-1 and/or B7- 2 molecule anchored to the lipid membrane on the exterior of the particle; and an antigen molecule such as a tumor specific antigen or cancer marker anchored to the lipid membrane on the exterior of the particle.
  • GPI-anchor A number of proteins commonly expressed by cells are attached to the cell membrane via a GPI-anchor. These proteins are post-translationally modified at their carboxy terminus to express this glycosylated moiety which is synthesized in the endoplasmic reticulum. These naturally expressing GPI-anchored molecules are widely distributed in mammalian cells and serve a host of different cellular functions, such as cell adhesion, enzymatic activity, and complement cascade regulation. Naturally occurring GPI- anchored proteins lack a transmembrane and cytoplasmic domain that otherwise anchor membrane proteins.
  • the GPI-anchor consists of a glycosylated moiety attached to phosphatidylinositol containing two fatty acids. The phosphatidylinositol portion, as well as an ethanolamine which is attached to the C-terminal of the extracellular domain of the membrane proteins, anchor the molecule to the cell membrane lipid bilayer.
  • transmembrane and cytoplasmic domains of a transmembrane surface protein need only be replaced by the signal sequence for GPI-anchor attachment that is found at the hydrophobic C-terminus of GPI-anchored protein precursors.
  • This method may be used to generate GPI- anchored proteins is not limited to membrane proteins; attaching a GPI-anchor signal sequence to secretory proteins would also convert them to a GPI-anchored form.
  • the method of incorporating the GPI-anchored proteins onto isolated cell surfaces or lipid particles is referred to here as protein transfer.
  • GPI-anchored molecules can be incorporated onto lipid membranes spontaneously. These GPI-anchored proteins can be purified from one cell type and incorporated onto different cell membranes. GPI-anchored proteins are used to customize of the lipid membranes disclosed herein for uses as a cancer vaccine. One may incorporate multiple molecules simultaneously onto the same cell membrane. One can control the level of protein expression by simply varying the concentration of the GPI-anchored molecules to be incorporated. The most significant outcome of this technology will be the reduction of time in preparing cancer vaccines from months to hours. These features make the protein transfer approach a more viable choice for the development of cancer vaccines for clinical settings. The molecules incorporated by means of protein transfer retain their functions associated with the extracellular domain. Cells and isolated membranes can be modified to express immunostimulatory molecules. In certain embodiments, the disclosure contemplates that the GPI-anchored molecules are incorporated onto the surface of albumin microparticles by this protein transfer method. GPI-anchored proteins attached to the surface of
  • microparticles are used to target and/or enhance the adjuvant activity of microparticles, thereby enhancing the capacity to function as a targeted antigen or drug delivery device for cancer treatment.
  • the GPI-B7-1 expression (by protein transfer) was stable up to 7 days on isolated membranes at 37°C and frozen membranes can be used up to 3 years of storage at - 80°C which makes the stability and storage a nonissue.
  • This approach for introducing proteins onto membranes provides advantages over other immunotherapies for cancer vaccine development.
  • This approach allows a protein to be added either singularly or in a combinatory manner to the tumor membrane surface.
  • This approach navigates around the necessity to establish tumor cells as is the case for gene transfer.
  • This GPI-mediated approach by protein transfer may be used for the co- stimulatory molecules, B7-land B7-2, GM-CSF, IL-2, and IL-12.
  • B7-land B7-2 GM-CSF
  • IL-2 IL-12
  • the disclosure relates to virus like particles comprising B7-1 and/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule anchored to the lipid membrane on the exterior of the particle for uses disclosed herein.
  • Influenza virus-like particles are particulate in nature and have shown to elicit robust immunity against antigens. Influenza VLPs have an outer lipid bilayer with properties similar to the cell membranes. Modification of influenza VLPs with a protein transfer method to incorporates tumor-associated antigens (TAAs) on the surface along with immunostimulatory molecules (ISMs) elicits enhanced immune responses directed against the TAAs.
  • TAAs tumor-associated antigens
  • ISMs immunostimulatory molecules
  • phosphatidylinositol to anchor the TAA, which can spontaneously incorporate onto the surface of the VLPs that contain a lipid bilayer upon incubation at 37°C (See Figure 1).
  • Incorporation of GPI-anchored forms of TAAs onto the surface of VLPs is used to direct the immune response against cancerous cells whereas the incorporation of immunostimulatory molecules (ISMs), such as GPI-anchored cytokines, costimulatory molecules, and adhesion molecules, onto the surface of VLPs is used to enhance the interaction between VLPs and antigen presenting cells (APCs) as well as lead to activation of these APCs and other immune effector cells.
  • ISMs immunostimulatory molecules
  • APCs antigen presenting cells
  • VLPs consist of a virus' capsid protein shell that presents viral antigens in an authentic conformation without the viral genome that is required for replication. Thus, they provide a safe approach for human use. VLPs contain a multivalent repetitive structure that is particulate in nature, allowing for recognition by many pattern recognition receptors and the induction of an enhanced innate and adaptive immune response. The particulate nature of VLPs allows for them to be readily taken up and presented by APCs, and thus could provide a means for breaking the immunosuppressive barrier initiated by the tumor microenvironment.
  • influenza virus-like particles may be produced using a variety of platform systems, including recombinant baculovirus vectors, transient plasmid expression systems, stable cell-line transformants, and plant expression systems.
  • VLPs are non-replicating particles that spontaneously self-assemble from expressed influenza virus proteins.
  • the viral hemagglutinin (HA) protein is sufficient for particle assembly and release from the cell.
  • the VLP comprises neuraminidase (NA).
  • NA neuraminidase
  • HA may present with a different type of
  • VLPs containing HA in mammalian cells, co-expression of NA or exogenous ly added NA was required for the effective release of influenza VLPs into culture media, implying an important role of the NA activity in cleaving sialic acids bound to HA of budding particles.
  • VLPs containing HA can be produced in insect cells in the absence of NA expression. Insect cells do not add sialic acids to the N-glycans during the posttranslational modification, which explains how VLPs containing HA but not NA are effectively released from insect cell surfaces. See Kang et al, Virus Res. 2009c, 143 (2), 140-6.
  • VLPs used herein are recombinant influenza VLPs that have been generated in insect cells infected with rBVs expressing influenza genes HA, NA, Ml, and M2.
  • VLPs used herein are recombinant influenza VLPs that have been generated in insect cells infected with rBVs expressing influenza genes HA, NA, and Ml .
  • VLPs used herein are recombinant influenza VLPs that have been generated in insect cells infected with rBVs expressing influenza genes of HA and Ml .
  • the VLP is obtained from influenza VLPs expressed from recombinant baculovirus (rBV) produced by replication in an insect cell system, e.g., Spodoptera frugiperda SF9 cells.
  • rBV recombinant baculovirus
  • the VLP is obtained from a modified vaccinia virus Ankara
  • MVA virus-like protein
  • H5N1 HA expressing expressing influenza H5N1 HA, NA, and M proteins to generate influenza VLPs produced by replication in mammalian cells. See Schmeisser et al., Vaccine, 2012, 30(23):3413-3422.
  • the disclosure relates to particles such as cells or virus like particles comprising B7-1 and/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule anchored to the lipid membrane on the exterior of the particle.
  • the antigen molecule is a cancer marker selected from HER-2, MKI67, prostatic acid phosphatase (PAP), prostate-specific antigen (PSA), prostate-specific membrane antigen, early prostate cancer antigen, early prostate cancer antigen-2 (EPCA-2), BCL-2, MAGE antigens such as CT7, MAGE- A3 and MAGE-A4, ER 5, G-protein coupled estrogen receptor 1, CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD 117, melanoma-associated antigen (TA-90), peripheral myelin protein 22 (PMP22), epithelial membrane proteins (EMP-1, -2, and -3), HMB-45 antigen, MART-1 (Melan-A), S100A1, S100B and gp 100:209-217(210M), MUC-1, mucin antigens TF, Tn, STn, glycolipid
  • the antigen is the human form.
  • HER-2 or Human Epidermal Growth Factor Receptor 2 refers to the human protein encoded by the ERBB2 gene that has been referred to as Neu, ErbB-2, CD340 (cluster of differentiation 340) or pl85. See Coussens et al, 1985, Science 230 (4730): 1132-9.
  • HER-2 is the extracellular domain or fragment thereof.
  • the protein comprises or consists essentially of the following sequence: TQVCTGTDMK LRLPASPETH LDMLRHLYQG CQVVQGNLEL
  • the protein comprises or consists essentially of the following sequence: DIQMTQSPSS LSASVGDRVT ITCRASQDVN TAVAWYQQKP GKAPKLLIYS ASFLYSGVPS RFSGSRSGTD FTLTISSLQP EDFATYYCQQ HYTTPPTFGQ GTKVEIKRTV AAPSVFIFPP SDEQLKSGTA SVVCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC (SEQ ID NO: 5) or fragment thereof.
  • the protein comprises or consists essentially of the following sequence: GTSHLVKCAE KEKTFCVNGG ECFMVKDLSN PSRYLCKCPN EFTGDRCQNY VMASF (SEQ ID NO: 6) or fragment thereof.
  • MKI67 or antigen identified by monoclonal antibody Ki-67, refers to the human protein that is encoded by the MKI67 gene. See Bullwinkel et al, 2006, J. Cell. Physiol. 206 (3): 624-35.
  • PAP Prostatic acid phosphatase or prostatic specific acid phosphatase
  • PSAP Prostatic acid phosphatase or prostatic specific acid phosphatase
  • PSA Prostate-specific antigen or gamma-seminoprotein or kallikrein-3
  • KLK3 Prostate-specific antigen or gamma-seminoprotein or kallikrein-3
  • PSMA Prostate-specific membrane antigen or Glutamate carboxypeptidase II
  • PSMA Prostate-specific membrane antigen or Glutamate carboxypeptidase II
  • Bcl-2 refers to an protein encoded by the BCL2 gene.
  • Bcl-2 has two isoforms that differ by two amino acids. Isoform 1 is known as 1G5M, and Isoform 2 is known as 1G50/1GJH. See Petros et al, 2001 , PNAS, 98: 3012-3017. Both isoforms are contemplated antigens.
  • the antigen is the entire protein, polypeptide, or a substantial fragment, or a fragment with conserved substitutions.
  • the fragment may contain 5, 10, 20, 50, 100, or halve of the amino acids in the full length antigen.
  • the fragment may be sufficient to mimic or replicate the folding of the full length antigen.
  • the conserved substitutions may be amino acids that are in the interior of the folded polypeptide.
  • a fragment is sufficient produce antibody production to the polypeptide.
  • the antigen may be a chimera containing the fragment.
  • the antigen may contain 1 , 2, or 3, or 5 to 10, or 10 to 20 or more conserved substitutions within the full length or polypeptide fragment which are typically outside of functional domains.
  • the antigen may have 80%, 90%, 95% or greater sequence identity to the full length or polypeptide fragment.
  • An antigen protein may or may not be glycosylated.
  • the virus like particles disclosed herein comprise an adjuvant molecule anchored to a lipid membrane on the exterior of the particle wherein the adjuvant molecule and the antigen molecule are not the same molecule.
  • the adjuvant molecule is selected from is IL-2, IL-12, ICAM1 , GM-CSF, flagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA).
  • co-stimulatory molecules, antigens, and adjuvant molecules may the individually conjugated to the lipophilic molecules or two or more or all of them may be conjugated together in a chimera and conjugated to a lipophilic molecule.
  • B7-1 may be conjugated to the adjuvant, HSA, in a chimera and the chimera is conjugated to a GPL
  • HSA heat stable antigen
  • a hybrid B7-1-HSA molecule on the cell surface membrane can function as a co-stimulatory molecule to induce T cell proliferation.
  • CHO cells and CHO transfectants expressing HSA, B7-1, and B7-1- HSA were used as stimulator cells in a T cell proliferation assay. See Wang et al,
  • TLR 9 ligands as adjuvants are contemplated such as
  • oligonucleotide sequence 5'-CG-3' is unmethylated and the oligonucleotide is greater than about 6 base pairs in length and is less than about 100 base pairs in length such as 5'- TGACTGTGAACGTTC GAGATGA-3 * (SEQ ID NO:8). It is contemplated that lipophilic molecules may be conjugated to the oligonucleotide for incorporation to the exterior of particles disclosed herein.
  • the antigen is also contained in the interior of the particle.
  • the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • HSA heat stable antigen
  • the antigen is HER-2 and the adjuvant is flagellin and/or GM-CSF.
  • the antigen is HER-2 and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • HSA heat stable antigen
  • the antigen is HER-2
  • the adjuvant is flagellin and/or GM- CSF
  • the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • the antigen is HER-2 and the adjuvant is IL-12.
  • the antigen is HER-2
  • the adjuvant is IL-12
  • the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • the antigen is PSA or PAP and the adjuvant is flagellin and/or or GM-CSF.
  • the antigen is PSA or PAP and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • HSA heat stable antigen
  • the antigen is PSA or PAP
  • the adjuvant is flagellin
  • the B7- 1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • the antigen is PSA or PAP and the adjuvant is IL-12.
  • the antigen is PSA or PAP, the adjuvant is IL-12, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • HSA heat stable antigen
  • the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • HSA heat stable antigen
  • the antigen is HER-2 and the adjuvant is fiagellin and/or
  • the antigen is HER-2 and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • HSA heat stable antigen
  • the antigen is HER-2
  • the adjuvant is fiagellin and/or GM- CSF
  • the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • the antigen is HER-2 and the adjuvant is IL-12.
  • the antigen is HER-2
  • the adjuvant is IL-12
  • the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • the antigen is PSA or PAP and the adjuvant is fiagellin and/or GM-CSF.
  • the antigen is PSA or PAP and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • HSA heat stable antigen
  • the antigen is PSA or PAP
  • the adjuvant is fiagellin or GM- CSF
  • the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
  • particle may be a wild type cell, cancer cell or immortalized cell.
  • the particle is a cell such as ZR-75-1, ZR-75-30, 184A1, UACC-812, UACC-893, HCC38, HCC70, HCC202, HCC1187, HCC1395, HCC 1428, HCC1500, HCC1569, HCC1599, HCC1806, HCC1937, HCC1954, HCC2157, HCC1419, HCC2218, AU-565, 184B5, MCF 10A, MCF 10F, MCF- 12 A, BT-20, MDA-kb2, BT-474, CAMA-1, MCF7, MDA-MB-134-VI, MDA-MB-157, MDA-MB-175-VII, MDA-MB-231, MDA-MB-361, SK-BR-3, BT-483, BT-549, DU4475, Hs 578T, MDA-MB-415, MDA- MB-436, MDA-MB-453, MDA-MB-468, T-
  • the particle is a cell such as Jurkat, Clone E6-1 (ATCC Number: TIB-152), RBL-2H3 (CRL-2256), MOLT-4 (CRL-1582), K-562 (CCL-243), CCRF-CEM (CCL-119), HL-60 (CCL-240), or KG-1 (CCL-246) for use in the treatment of cancer, leukemia, leukemia (AML), leukemia (CML), promyelocytic leukemia, basophilic leukemia, or acute T cell leukemia.
  • AML leukemia
  • CML promyelocytic leukemia
  • basophilic leukemia or acute T cell leukemia.
  • the particle is a cell such as NCI-H358 (CRL-5807), LL/2 (CRL-1642), Calu-3 (HTB-55), NCI-H441 (HTB-174), NCI-H1975 (CRL-5908), NCI-H23 (CRL-5800), NCI-H1299 (CRL-5803), NCI-H460 (HTB-177), NCI-H292 (CRL-1848), A- 549 (CCL-185), A-549 (CCL-185), A-549 (CCL-185), IMR-90 (CCL-186), MRC-5 (CCL- 171), or WI-38 (CCL-75) for use in the treatment of cancer, lung cancer, lung
  • adenocarcinoma adenocarcinoma, lung carcinoma, lewis lung carcinoma, or bronchioalveolar lung cancer.
  • the particle is a cell such as Ramos (CRL-1596), Daudi (CCL-213), Raji (CCL-86), EL4 (TIB-39), or U-937 (CRL-1593.2) for use in the treatment of cancer, lymphoma, B-cell lymphomas, histiocytic lymphoma, or Burkitt's lymphoma.
  • Ramos CRL-1596
  • Daudi CCL-213
  • Raji CL-86
  • EL4 TIB-39
  • U-937 CTL-1593.2
  • the particle is a cell such as HeLa (CCL-2) or HeLa S3
  • CCL-2.2 for use in the treatment of cancer, cervical cancer or cervical adenocarcinoma.
  • the particle is a cell such as COLO 205 (CCL-222), SW620 (CCL-227), SW480 (CCL-228), LoVo (CCL-229), LS 174T (CL-188), Caco-2 (HTB-37), HT-29 (HTB-38),DLD-1 (CCL-221), HCT 116 (CCL-247), T84 (CCL-248), CT26.WT (CRL-2638) for use in the treatment of cancer, colon cancer, colon carcinoma, or a colon adenocarcinoma.
  • COLO 205 CL-222
  • SW620 CL-227)
  • SW480 CL-228)
  • LoVo CL-229
  • LS 174T CL-188
  • Caco-2 HCT 116
  • T84 CTL-248
  • CT26.WT CT26.WT
  • the particle is a cell such as HCN-1A (CRL- 10442), U-87 MG (HTB-14), C6 (CCL-107), bEnd.3 (CRL-2299), or T98G (CRL- 1690) for use in the treatment of cancer, brain cancer, glioma, glioblastoma multiforme, glioblastoma- astrocytoma, or brain endothelioma cancer.
  • HCN-1A CL- 10442
  • U-87 MG HTB-14
  • C6 C6
  • CTL-2299 bEnd.3
  • T98G T98G
  • the particle is a cell such as 3197-3 (CRL- 1568), 3T3-Swiss albino (CCL-92), BALB/3T3 clone A31 (CCL-163), NTERA-2 cl.Dl (CRL-1973), 3T3-L1 (CL-173), NIH/3T3 (CRL-1658),SK-OV-3 (HTB-77), CHO-K1 (CCL-61), or F-12K (30- 2004) for use in the treatment of cancer, ovarian cancer, ovarian adenocarcinoma, or testicular cancer.
  • the particle is a cell such as 293T/17 (CRL-11268), 293 (CRL-1573), VERO C1008 (CRL-1568), Vera (CCL-81), MDCK (CCL-34), BHK-21 (CCL-10), Caki-1 (HTB-46), 786-0 (CRL- 1932), or COS-7 (CRL- 1651) for use in the treatment of cancer, renal cancer, or renal carcinoma.
  • the particle is a cell such as H9c2 (CRL-1446) for use in the treatment of cancer or cardiac tumors.
  • the particle is a cell such as A-431 (CRL-1555), Detroit 551 (CCL-110), BJ (CRL-2522), B16-F10 (CRL-6475), SK-MEL-28 (HTB-72), A375 (CRL- 1619), NCTC clone 929 (CCL-1), IRR-MRC-5 (55-X), or IRR-STO (56-X) for use in the treatment of cancer, skin cancer, squamous-cell carcinoma, melanoma, areolar lesions, or epidermoid carcinoma.
  • A-431 CL-1555
  • Detroit 551 CL-110
  • BJ CL-2522
  • B16-F10 CL-6475
  • SK-MEL-28 HTB-72
  • A375 CTL- 1619
  • NCTC clone 929 CCL-1
  • IRR-MRC-5 55-X
  • IRR-STO 56-X
  • the particle is a cell such as HT- 1080 (CCL-121) for use in the treatment of cancer or fibrosarcoma.
  • the particle is a cell such as AGS (CRL-1739) or NCI-N87
  • the particle is a cell such as HepG2/C3A (CRL-10741), Hep 3B2.1-7 (HB-8064), Hep G2 (HB-8065), or Hepa 1-6 (CRL-1830) for use in the treatment of cancer, liver cancer, heptoma, or hepatocellular carcinoma.
  • HepG2/C3A CRL-10741
  • Hep 3B2.1-7 HB-8064
  • Hep G2 HB-8065
  • Hepa 1-6 CRL-1830
  • the particle is a cell such as U266B1 (TIB- 196) for use in the treatment of cancer or multiple myeloma.
  • the particle is a cell such as IMR-32 (CCL-127), Neuro-2a (CCL-131), or SK-N-SH (HTB-11) for use in the treatment of cancer or neuroblastoma.
  • IMR-32 CCL-127
  • Neuro-2a CCL-131
  • SK-N-SH HTB-11
  • the particle is a cell such as Saos-2 (HTB-85), U-2 OS (HTB-96), or MG-63 (CRL-1427) for use in the treatment of cancer, bone cancer, or osteosarcoma.
  • Saos-2 HMB-85
  • U-2 OS HMB-96
  • MG-63 CTL-1427
  • the particle is a cell such as Beta-TC-6 (CRL-11506), AsPC-1 (CRL-1682), BxPC-3 (CRL-1687), MIA PaCa-2 (CRL-1420), PANC-1 (CRL- 1469), Capan-1 (HTB-79), or AR42J (CRL-1492) for use in the treatment of cancer, pancreatic cancer, or pancreatic carcinoma.
  • Beta-TC-6 CTL-11506
  • AsPC-1 CL-1682
  • BxPC-3 CL-1687
  • MIA PaCa-2 CL-1420
  • PANC-1 CL- 1469
  • Capan-1 Capan-1
  • AR42J AR42J
  • the particle is a cell such as PC- 12 (CRL-1721) for use in the treatment of cancer or pheochromocytoma.
  • the particle is a cell such as RPMI 8226 (CCL-155) for use in the treatment of cancer or plasmacytoma.
  • the particle is a cell such as PC-3 (CRL-1435), VCaP (CRL-2876), DU 145 (HTB-81), LNCaP clone FGC (CRL-1740), or 22Rvl (CRL-2505) for use in the treatment of cancer, prostate cancer, prostate adenocarcinoma.
  • the particle is a cell such as ARPE-19 (CRL-2302) for use in the treatment of cancer, eye cancer, or retinal cancer.
  • the particle is a cell such as RD (CCL-136) for use in the treatment of cancer, sarcoma, or rhabdomyosarcoma.
  • the particle is a cell such as a stem cells, mesenchymal stromal/stem, pluripotent stem cell, embryo, myoblast, hybridoma or macrophage, examples include RAW 264.7 (TIB-71), J774A.1 (TIB-67), C2C12 (CRL-1772), L6 (CRL-1458), Sp2/0-Agl4 (CRL-1581) for use in the treatment of cancer.
  • RAW 264.7 TIB-71
  • J774A.1 TIB-67
  • C2C12 C2C12
  • L6 CTL-1458
  • Sp2/0-Agl4 CTL-1581
  • DC dendritic cell
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with the administration of dendritic cell (DC)-based cancer vaccines.
  • DCs have the unique ability to take up and process antigens, move into secondary lymphoid tissues, and activate both helper and cytotoxic T cells.
  • Preparation of DC-based cancer vaccines involves loading DCs with known tumor-specific antigens, antigenic peptides, cDNA, or RNA isolated from tumor cells.
  • an object of this disclosure is to develop more effective methods to deliver tumor antigens to DCs.
  • One strategy is making hybrid cells by fusing tumor cells, tumor antigens, or conjugates with DCs and using the hybrid cells as vaccines.
  • Combination therapies with DC-based cancer vaccines may be used to treat melanoma, breast cancer, multiple myeloma, NHL, lymphatic leukemia, prostatic adenocarcinoma, lung cancer, and hepatocarcinoma
  • compositions disclosed herein and using any of the compositions in combination with antigen activated DCs for cancer treatments are used in combination with DCs fused with granulocyte macrophage colony-stimulating factor (GM-CSF) and prostatic acid
  • GM-CSF granulocyte macrophage colony-stimulating factor
  • PAP phosphatase
  • Provenge an autologous DC-based vaccine, was approved by the FDA for the treatment of men with advanced prostate cancer.
  • Provenge consists of patient-derived DCs pulsed ex vivo with a recombinant fusion protein (PA 2024) containing granulocyte macrophage colony-stimulating factor (GM-CSF) and prostatic acid phosphatase (PAP), an antigen found in 90-95% of prostate cancers.
  • PA 2024 recombinant fusion protein
  • GM-CSF granulocyte macrophage colony-stimulating factor
  • PAP prostatic acid phosphatase
  • compositions disclosed herein and using any of the compositions in combination with irradiated tumor cells for cancer treatments.
  • immunosuppressive cytokines such as TGF- ⁇ and IL-10
  • TGF- ⁇ and IL-10 the innate and adaptive immune responses are inhibited during tumor development.
  • the systemic administration of certain immunostimulatory cytokines has been used to alter the tumor microenvironment to mediate tumor recognition by immune cells.
  • certain immunostimulatory cytokines such as IL-2, IL-12, and IFN-a
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with cytokines such as IL-2, IL-12, and INF-a for cancer treatments.
  • Cytokines activate immune cells, such as NK and CD8+ T cells, and can also inhibit tumor angiogenesis.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with IL-2, IL-12, and INF-a for the treatment of metastatic melanoma and renal cell carcinoma (RCC).
  • T-cell growth cytokine IL-15
  • AICD activation-induced cell death
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with IL-15 as a potential cancer immunotherapeutic agent.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination intra-tumoral administration of cytokines, modification of tumor cells to secrete cytokines, and fusion of cytokines with antibodies for cancer treatments.
  • the cytokine is TNF-a.
  • the cancer is melanoma.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with administration of soluble GM-CSF and optionally a cytokine for cancer treatments. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with an antibody therapy for cancer treatment.
  • the contemplated anti-bodies are directed to epidermal growth factor receptor (EGFR), human EGFR-2 (HER-2), CD20 (an unglycosylated transmembrane phosphoprotein expressed on B and T cells), CD33 (a transmembrane protein expressed on cells of myeloid lineage and also on some lymphoid cells), CD52 (a highly glycosylated 12 amino acid membrane-anchored glycosylphosphatidylinositol (GPI) protein which is expressed on all circulating lymphocytes), and VEGF.
  • the antibody may be humanized, chimeric, a radiolabeled mouse antibody for targeted radiation.
  • compositions disclosed herein and using any of the compositions in combination with rituximab for the treatment of B-cell non-Hodgkin's lymphoma or chronic lymphocytic leukemia.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with ofatumumab for the treatment of B- cell non-Hodgkin's lymphoma or chronic lymphocytic leukemia.
  • compositions disclosed herein and using any of the compositions in combination with ibritumomab (tiuxetan) for the treatment of B-cell non-Hodgkin's lymphoma.
  • compositions disclosed herein and using any of the compositions in combination with tositumomab for the treatment of B- cell non-Hodgkin's lymphoma.
  • compositions disclosed herein and using any of the compositions in combination with gemtuzumab ozogamicin for the treatment of acute myeloid leukemia.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with alemtuzumab for the treatment of B- cell non-Hodgkin's lymphoma or chronic lymphocytic leukemia.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with trastuzumab for the treatment of breast cancer.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with bevacizumab for the treatment of breast, lung, or colon cancer. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with cetuximab for the treatment of brain and neck, or colon cancer.
  • the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with panitumomab for the treatment of colon cancer.
  • the disclosure relates to methods of treating cancer comprising administering an effective amount of a particle as disclosed herein to a subject at risk of or diagnosed with cancer or a tumor optionally in combination with another anticancer agent.
  • anticancer agents contemplated include gefitinib, erlotinib, docetaxel, cis-platin, 5-fluorouracil, gemcitabine, tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin, vincristine, vinblastine, vindesine, vinorelbine taxol, taxotere, etoposide, teniposide, amsacrine, topotecan, camptothecin bortezomib anegri
  • HER-2/neu a surface glycoprotein
  • HER-2/neu a surface glycoprotein
  • CHOKI cells were transfected to express GPI-ICAM1 or GPI-IL-12.
  • Expression of the GPI-ISMs was assessed by flow cytometry and verification of the GPI-anchor was confirmed by a phospholipase (PIPLC) treatment.
  • the transfectants were grown in large quantities, lysed, and the GPI-ISMs were purified by affinity chromatography. To determine if the purified GPI-ISMs still retained the GPI-anchor and could incorporate onto lipid bilayers by protein transfer, sheep red blood cells (RBCs) were used.
  • the GPI-ISMs were individually incubated with the RBCs at 37°C for 2 hours, washed and then analyzed by flow cytometry.
  • Figure 2 demonstrates that the purified GPI-ISMs were able to incorporate onto sheep RBCs.
  • VLPs are constructed by the rBV system through the expression of the hemagluttinin and matrix 1 protein in Sf9 insect cells. See Song et al., J Proteome Res. 2011, 10(8):3450-9.
  • PI-PLC treatment to cleave the GPI-anchor of GPI-ICAM1 either before incorporation ( Figure 5A) or after ( Figure 5B) incorporation was carried out.
  • GPI-ICAM1 incorporated with GPI-ICAM1 both led to decreased expression of ICAM1 on VLPs as detected by Western blotting to ICAM1, whereas when the PI-PLC inhibitors, ZnCl 2 , or 1,10-phenanthroline, were included, expression was retained.
  • GPI-ICAM1 was incubated with 1% fatty-acid- free bovine serum albumin (BSA) or 1% orosomucoid that bind to the GPI-anchor before protein transfer in order to competitively inhibit incorporation of GPI-ICAM1 onto VLP membranes.
  • BSA bovine serum albumin
  • Figure 6 shows that GPI-ICAM1 incubated first with 1% BSA or 1% orosomucoid showed decreased incorporation onto VLPs compared to those incorporated without prior incubation with BSA or orosomucoid.
  • VLPs were incubated with GPI-ICAM1 and GPI- IL-12 simultaneously at 37°C for 2 h.
  • Figure 7 shows that influenza VLPs can incorporate both GPI-ISMs on their surface and the expression of the first GPI-protein is not affected by the expression of the second GPI-protein.
  • GPIISMs such as GPI-IL-2, GPI-IL-12, GPI-B7-1, and GPI-ICAM-1
  • GPI-IL-2 GPI-IL-2
  • GPI-IL-12 GPI-B7-1
  • GPI-ICAM-1 GPI-ICAM-1
  • the incorporation of cytokines onto the surface of VLPs allows for a slow release depot of the cytokines into the administered microenvironment, leading to increased activation of immune effector cells at the vaccination site while decreasing chances of systemic toxicity.
  • the receptors of the ISMs, IL-2, IL-12, and ICAM-1 are found on APCs allowing for enhanced adhesion and activation of the APCs by the VLPs, thus leading to enhanced uptake and presentation.
  • the receptors for IL-12 and B7-1 are also found on
  • NK cells and mast cells allowing for the activation of a wide variety of immune effector cells to be elicited by the association of these ISMs onto the surface of VLPs. Since the immune response is directed against the antigens found on the VLPs, incorporating TAAs along with ISMs onto the surface of VLPs will direct the immune response towards the TAAs that are overexpressed on tumor cells as well.
  • GPI-anchored immune stimulatory molecules namely cytokines (IL-2, IL-12) and the costimulatory protein B7-1
  • IL-2, IL-12 cytokines
  • B7-1 costimulatory protein B7-1
  • mice were challenged (s.c.) with the following cells mixed in a 1 : 1 ratio with BD MatrigelTM (a solubilized basement membrane preparation derived from a mouse sarcoma): 4T07-WT, 4T07-B7/IL- 12 or PBS (control).
  • BD MatrigelTM a solubilized basement membrane preparation derived from a mouse sarcoma
  • the Matrigel/tumor, spleen and dLNs were harvested from the mice, digested and analyzed for cellular infiltrates by flow cytometry.
  • the expression of GPI-ISMs on the surface of tumor cells led to reduced angiogenesis as evidenced by a reduced level of blood vessels and decreased presence of
  • CD4+CD25+FOXP3+ regulatory T cells and CDl lb+Grl+ MDSCs locally at the tumor site and dLNs as well as systemically in the spleen. Additionally, there was a decrease in CD8+PD1+ exhausted T cells at the tumor site.
  • the GPI-ISMs increased the presence of CD4+ and CD8+ T cells as well as dendritic cells and B cells.
  • mice (five per group) were challenged subcutaneously (s.c.) with wild-type 4T07 or transfected 4T07-B7, GPI-IL-2, GPI-IL-12, B7/GPI-IL-2 or B7/GPI-IL- 12 cells (all 2xl0 5 cells in ⁇ PBS). Mice were injected s.c. in the rear flank and were monitored daily. Tumor size was measured using Vernier calipers every 2nd-3 rd day by taking 2x2 perpendicular measurements, and tumor size (mm 2 ) was calculated by multiplying the two diameters. Mice were euthanized when the tumor size reached close to 2 cm 2 .
  • tumor- free mice in the experimental groups were rechallenged on the opposite hind flank with wild-type 4T07 cells (2x105 in ⁇ PBS). Mice in each group were marked individually by ear punch and tumor growth was measured and recorded for each mouse separately.
  • the wild-type and transfected tumor cell lines all began to grow tumors in vivo, but while the wild-type tumors continued to increase in size, the tumors from the modified cell lines all regressed (See figure 9 and 10).
  • HER-2ECD is the extracellular portion of hHER-2.
  • the hHER-2 extracellular domain with CD59 GPI signal sequence were join and introduced by a EcoRI site, i.e., joining region: g/aattc introduced EcoRV site (gat/atc) before sequence and Apal (gggcc/c) site after sequence at the joining region as illustrated in figure 11.
  • g/aattc introduced EcoRV site (gat/atc) before sequence and Apal (gggcc/c) site after sequence at the joining region as illustrated in figure 11.
  • an optimized IL-2 Kozak sequence along with the restriction enzyme sites Hindlll and Kpnl were added. Following the hHER2ECD sequence an EcoRI site is added.
  • FIG. 12 shows flow cytometry analysis of CHO cells expressing GPI-human HER-2 (hHER-2-CD59) using TAl mAb. Testing shows that HER-2 expressed in CHO cells is GPI-anchored.
  • PIPLC is an enzyme which cleaves GPI anchor, reduces the level of expression.
  • PI-PLC treated CHOKl-hHER-2ECD-CD59 cells reduced hHER-2 cell surface expression by 98.4%. PIPLC will not have any effect on normal HER-2.
  • Nucleic acid encoding the hHER-2 extracellular domain E (Amino Acids 22-652) and GPI-anchor signal sequence (SEQ ID NO: 7) AAGGGGAGGT AACCCTGGCC CCTTTGGTCG GGGCCCCGGG CAGCCGCGCGCGCG CCCCTTCCCA CGGGGCCCTT TACTGCCG CGCGCCCGGC CCCCACCCCT CGCAGCACCC CGCGCCCCGC GCCCTCCCAG CCGGGTCCAG CCGGAGCCAT GGGGCCGGAGGATATC CCGCAGTGAG CACCATGGAG CTGGCGGCCT TGTGCCGCTG GGGGCTCCTC CTCGCCCTCTCT TGCCCCCCGG AGCCGCGAGC ACCCAAGTGT GCACCGGCAC AGACATGAAG CTGCGGCTCC CTGCCAGTCC CGAGACCCACCTGGACATGC TCCGCCACCT CTACCAGGGC TGCCAGGTGG TGCAGGGAAA CCTGGAACTC ACCTACCTGC CC

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Reproductive Health (AREA)
  • Pregnancy & Childbirth (AREA)
  • Gynecology & Obstetrics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)

Abstract

In some embodiments, described herein is a method of tumor treatment or tumor vaccination. The method generally comprises applying to a human being in need thereof a tumor therapeutic composition or tumor vaccine defined herein. The tumor therapeutic composition or tumor vaccine can be produced by protein transfer of glycosyl-phosphatidylinositol (GPI)-anchored immunostimulatory or costimulatory molecules.

Description

IMMUNOSTIMULATORY COMPOSITIONS, PARTICLES, AND
USES RELATED THERETO
ACKNOWLEDGEMENTS
This invention was made with government support under Grant R01CA138993 awarded by the National Institutes of Health. The government has certain rights in the invention.
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application Number 61/594,754 filed February 3, 2012 hereby incorporated by reference in its entirety.
BACKGROUND
Provenge™ is a recently FDA-approved autologous cellular immunotherapy treatment. Peripheral blood leukocytes of a subject are harvested via leukapheresis. These enriched monocytes are incubated with prostatic acid phosphatase (PAP) conjugated to cytokine granulocyte macrophage colony stimulating factor (GM-CSF). GM-CSF is thought to direct the target antigen to receptors on DC precursors, which then present PAP on their cell surface in a context sufficient to activate T cells for the cells that express PAP. Activated, PAP presenting DCs are administered to the subject to elicit an immune response retarding cancer growth. This strategy requires isolation and expansion of cells of the subject, and typically treatment does not entirely clear the subject of cancer or tumors. Thus, there is a need to identify improved methods.
B7-1 (also known as CD80) is a T cell costimulatory molecule that can be anchored in to autologous cancer cells to stimulate immune responses. McHugh et al., report the construction, purification and functional reconstitution of a glyco lipid anchored form of B7- 1 (CD80) on tumor cell membranes. Proc. Natl. Acad. Sci. USA 1995; 92:8059-8063. See also U.S. Patent 6,491,925. Glycosyl phosphatidylinositol anchored B7-1 (GPI-B7-1) molecules have been incorporated onto tumor cells and isolated tumor cell membranes to provide costimulation for allogenic T cell proliferation. See Nagarajan & Selvaraj, Vaccine, 2006 , 24(13):2264-74, U.S. Published Patent Application No. US 2007/0243159, Bozeman et al., Front Biosci. 2010;15:309-320. Bumgarner et al., report surface engineering of microparticles by novel protein transfer for targeted antigen/drug delivery. J Control Release. 2009;137:90-97. Cubas et al, report virus-like particle (VLP) lymphatic trafficking and immune response generation after immunization by different routes. J Immunotherapy, 2009, 32(2): 118-128. Kueng et al, report a general strategy for decoration of envelope viruses with functionally active lipid-modified cytokines, J Virology, 2007, 81, 8666-8676.
SUMMARY
In some embodiments, described herein is a method of tumor treatment or tumor vaccination. The method generally comprises applying to a human being in need thereof a tumor therapeutic composition or tumor vaccine defined herein. The tumor therapeutic composition or tumor vaccine can be produced by protein transfer of glycosyl- phosphatidylinositol (GPI)-anchored immunostimulatory or costimulatory molecules.
In one embodiment, the tumor therapeutic composition or tumor vaccine comprises a live tumor cell or tumor cell membranes that is or are modified by protein transfer to express one or more GPI-anchored immunostimulatory or costimulatory molecules. The tumor therapeutic composition or tumor vaccine can be prepared by a method that comprises obtaining one or more GPI-anchored immunostimulatory or costimulatory molecules, and transferring the GPI-anchored immunostimulatory or costimulatory molecules onto a tumor cell or isolated tumor cell membranes by protein transfer.
In certain embodiments, the disclosure relates to non-naturally occurring particle comprising, a lipid membrane; a B7-1 and/or B7-2 molecule anchored to the lipid membrane on the exterior of the particle; and an antigen molecule such as a tumor specific antigen or cancer marker anchored to the lipid membrane on the exterior of the particle. Typically, the particle further comprises an adjuvant molecule anchored to the lipid membrane on the exterior of the particle wherein the adjuvant molecule and antigen molecule are not the same molecule. In certain embodiments, the adjuvant molecule is selected from IL-2, IL-12, ICAMl GM-CSF, flagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA). The lipid membrane may be a phospholipid monolayer or phospholipid bilayer. Typically, the particle is selected from a cell, allogeneic or autologous cancer cell or its membrane fragments or vesicles, liposome, virosome, micelle, polymer, and virus like particle.
In certain embodiments, the B7-1 molecule is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl-phosphatidylinositol, phospholipid, gly co lipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule. In certain embodiments, the antigen molecule such as a tumor associated antigen or cancer marker is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl-phosphatidylinositol, phospholipid, glycolipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
In certain embodiments, the adjuvant molecule is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl-phosphatidylinositol,
phospholipid, glycolipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
Particles comprising membranes such as tumor membranes carrying tumor antigens and immunostimulatory stimulatory molecules can be modified by incubating with lipophilic adjuvants such as lipopolysaccharides or an immunostimulatory unmethylated CpG oligonucleotides lipid conjugate.
In certain embodiments, antigen is a cancer marker molecule selected from HER-2, MUC-1, mucin antigens TF, Tn, STn, glycolipid globo H antigen, prostate-specific antigen, prostate-specific membrane antigen, early prostate cancer antigen-2 (EPCA-2), BCL-2, MAGE antigens such as CT7, MAGE- A3 and MAGE-A4, G-protein coupled estrogen receptor 1 , CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD1 17, melanoma-associated antigen (TA-90), peripheral myelin protein 22 (PMP22), epithelial membrane proteins (EMP-1, -2, and -3), HMB-45 antigen, MART-1 (Melan-A), S100A1, S100B and gp 100:209-217(210M).
In certain embodiments, the disclosure relates to virus like particles comprising B7-1 and/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule anchored to the lipid membrane on the exterior of the particle. Typically, the antigen molecule is a cancer marker or tumor associated antigen or tumor-specific antigen selected from HER-2, MKI67, prostatic acid phosphatase (PAP), prostate-specific antigen (PSA), prostate-specific membrane antigen, early prostate cancer antigen-2 (EPCA- 2), BCL-2, MAGE antigens, antigens comprising a Mage Homology Domain (MHD), MAGE-1, CT7, MAGE-A3 and MAGE-A4, ERK5, G-protein coupled estrogen receptor 1 , CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD 117, melanoma-associated antigen (TA-90), peripheral myelin protein 22 (PMP22), epithelial membrane proteins (EMP-1, -2, and -3), HMB-45 antigen, MART-1 (Melan-A), S100A1, S100B and gp 100:209-217(210M). Typically, the virus like particle further comprising an adjuvant molecule anchored to a lipid membrane on the exterior of the particle wherein the adjuvant molecule and the antigen molecule are not the same molecule. In certain embodiments, the adjuvant molecule is selected from is IL-2, IL- 12, ICAM1 GM-CSF, flagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA).
In certain embodiments, the disclosure relates to methods of treating cancer comprising administering an effective amount of a particle or a virus like particle as disclosed herein to a subject at risk of or diagnosed with cancer or a tumor optionally in combination with anti-CTLA-4 antibodies such as abatacept, belatacept, ipilimumab, tremelimumab, anti-PD-1 and PDL1 antibodies such as nivolumab, unmethylated CpG oligonucleotide, methyl jasmonate, cyclophosphamide, gemcitabine or other
immunosuppression blocker or other anticancer agent. Typically, the subject is a human subject and the virus like particle comprises a B7-land/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule wherein the antigen molecule is a viral protein.
Other anticancer agents contemplated include gefitinib, erlotinib, docetaxel, cis- platin, 5-fluorouracil, gemcitabine, tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin, vincristine, vinblastine, vindesine, vinorelbine taxol, taxotere, etoposide, teniposide, amsacrine, topotecan, camptothecin bortezomib anegrilide, tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene fulvestrant, bicalutamide, flutamide, nilutamide, cyproterone, goserelin, leuprorelin, buserelin, megestrol anastrozole, letrozole, vorazole, exemestane, finasteride, marimastat, trastuzumab, cetuximab, dasatinib, imatinib, bevacizumab, combretastatin, thalidomide, and/or lenalidomide or combinations thereof.
In certain embodiments, the viral like particle has an hemagglutinin selected from influenza HI, H2, H3, H4, H5, H6, H7, H8, H9, H10, HI 1, H12, H13, H14, H15, and H16 optionally in combination with or individually influenza Nl, N2, N3, N4, N5, N6, N7, and N8.
In certain embodiments, the virus protein is an HIV envelope protein selected from gp 41, gp 120, and gp 160.
In certain embodiments, the disclosure relates to methods of treating or preventing a viral infection comprising administering an effective amount of a virus like particle disclosed herein to a subject at risk of, exhibiting symptoms of, or diagnosed with a viral infection. In certain embodiments, the disclosure relates to particles comprising a cancer marker made by the process of mixing a cancer marker conjugated to a lipophilic moiety and a particle comprising a lipid membrane. Typically, the cancer marker is HER-2 or PSA or PAP.
In certain embodiments, the disclosure relates to particles comprising a cancer marker and B7-1 and/or B7-2 made by the process of mixing a B7-1 and/or B7-2 conjugated to a lipophilic moiety and a particle comprising a lipid membrane and a cancer marker.
In certain embodiments, the disclosure relates to methods of treating or preventing breast cancer comprising administering an effective amount of a particle comprising B7-1 and/or B7-2, GM-CSF, and HER-2 to a subject in need thereof.
In certain embodiments, the method further comprises analyzing the subject for overexpression of HER-2, by measuring, detecting, sequencing, hybridizing with a probe, HER-2 polypeptide or a nucleic acid indicative of HER-2 expression, or sequencing a nucleic acid associated with HER-2, on a cancer cell or tumor cell isolated from the subject.
In certain embodiments, the disclosure relates to methods of treating or preventing prostate cancer comprising administering an effective amount of a particle comprising B7-1 and/or B7-2, GM-CSF, and PSA or PAP to a subject in need thereof.
In certain embodiments, the disclosure relates to methods of treating or preventing prostate cancer comprising administering an effective amount of a particle comprising B7-1 and/or B7-2, GM-CSF, IL-12, and PSA or PAP to a subject in need thereof.
In certain embodiments, the compositions and method further comprises
administering an immunostimulatory amount of particles disclosed herein in combination with an anticancer agent, individually as single agents and/or in a single pharmaceutical composition.
In the case of breast cancer the anticancer agent may be estradiol, tamoxifen, cetuximab and a HER-2 antibody, humanized antibody, or human chimera such as trastuzumab, pertuzumab. The HER-2 antibodies may be administered before or after immune stimulation with particle.
In the case of prostate cancer, the anticancer agent may be docetaxel, cabazitaxel, bevacizumab, alpharadin thalidomide, prednisone, abiraterone, finasteride and dutasteride, MDV3100, orteronel (TAK-700), omega-3 fatty acids such as ethyl esters of
eicosapentaenoic acid (EPA) and/or docosahexaenoic acid (DHA) or combinations thereof such as bevacizumab, docetaxel, thalidomide, and prednisone or abiraterone acetate in combination with prednisone.
In another embodiment, the tumor therapeutic composition or tumor vaccine comprises a microparticle with a lipid membrane encapsulating tumor antigens or peptides and one or more anchored immunostimulatory or costimulatory molecules expressed on the surface of the particle. The tumor therapeutic composition or tumor vaccine can be prepared by a method that comprises obtaining one or more anchored immunostimulatory or costimulatory molecules, and transferring the anchored immunostimulatory or
costimulatory molecules onto a particle encapsulating at least one tumor antigen or peptide, tumor lysate, tumor membranes, or combinations thereof by protein transfer.
The microparticles can be formed of any biocompatible polymer capable of incorporating GPI-anchored immunostimulatory or costimulatory molecules. For example, representative useful biocompatible polymers include, but are not limited to, polyvinyl alcohols, polyvinyl ethers, polyamides, polyvinyl esters, polyvinylpyrrolidone,
polyglycolides, polyurethanes, allyl celluloses, cellulose esters, hydroxypropyl derivatives of celluloses and cellulose esters, preformed polymers of poly alkyl acrylates, polyethylene, polystyrene, polyactic acid, polyglycolic acid, poly(lactide-co-glycolide),
polycaprolactones, polybutyric acids, polyvaleric acid and copolymers thereof, alginates, chitosans, gelatin, albumin, zein and combinations thereof.
Anchored immunostimulatory or costimulatory molecules can be obtained by expressing the GPI-anchored immunostimulatory or costimulatory molecules in a cell, and isolating the GPI-anchored immunostimulatory or costimulatory molecules. The anchored immunostimulatory or costimulatory molecules can be any substance that stimulates or costimulates immune reaction against a tumor cell that is capable of being expressed in a cell. For example, the immunostimulatory or costimulatory molecules useful here can be a cytokine molecule. In one embodiment, a useful cytokine can be, for example, one or more of cytokines IL-2, IL-4, IL-6, IL-12, IL-15, IL-18, IL-19, granulocyte -macrophage colony stimulating factor (GM-CSF), and combinations thereof. In another embodiment, the immunostimulatory or costimulatory molecules can be, for example, the
immunostimulatory or costimulatory molecules useful here can be a cytokine molecule. In another embodiment, the immunostimulatory or costimulatory molecules useful here can be, for example, B7-1, B7-2 and an intercellular adhesion molecule such as CD40L, ICAM-1, ICAM-2, and ICAM-3. In any of the embodiments, particle may be a wild type cell, cancer cell or immortalized cell.
The immunostimulatory or costimulatory molecules can be used alone or together and can be used in conjunction with antibody fusion proteins.
The tumor therapeutic composition or tumor vaccine described herein can be used therapeutically or prophylactically for the treatment or prevention of a tumor.
Representative tumors can be treated or prevented include, but are not limited to, breast cancer, prostate cancer, lung cancer, melanoma, liver cancer, leukemia, lymphoma, myeloma, colorectal cancer, gastric cancer, bladder carcinoma, esophageal carcinoma, head & neck squamous-cell carcinoma, sarcomas, kidney cancers, ovarian and uterus cancers, adenocarcinoma, glioma, and plasmacytoma, and combinations thereof.
In one embodiment, the vaccine or therapeutic composition described herein can be GPI-anchored cytokine such as GPI-IL-2 and GPI-IL-12 alone or in combination with GPI- anchored costimulatory molecules such as GPI-B7-1, GPI-B7-2, GPI-ICAM-1, GPI-ICAM- 2 and GPI-ICAM-3. Such a vaccine or therapeutic composition can be used for the treatment of tumor and other diseases such as viral, bacterial and parasitic diseases.
In another embodiment, the vaccine and therapeutic composition can be
biocompatible microparticles such as biodegradable microparticles modified with GPI- anchored immunostimulatory molecules such as IL-2, IL-4, IL-6, IL-12, ICAM-1, ICAM-2, ICAM-3, B7-1, B7-2, CD40L, IL-15, IL-18, IL-19, granulocyte-macrophage colony stimulating factor (GM-CSF), and combinations thereof.
In yet another embodiment, the vaccine or therapeutic compositions described herein can be tumor cells or membranes modified by protein transfer with GPI-anchored cytokines alone or/and in combination with other cytokines or/and other costimulatory molecules. One such embodiment can be, for example, tumor membranes modified with purified GPI-IL-12.
In a further embodiment, particles like inactivated or partially attenuated virus, bacteria and virus-like particles can be modified to express immunostimulatory molecules by protein transfer with GPI-anchored cytokines and immunostimulatory molecules.
Vaccines and therapeutic compositions prepared in this manner can be used for preventing or treating viral, bacterial, or parasitic diseases or disorders.
In some other embodiments, the vaccine and therapeutic compositions described herein can be used for treating autoimmune disorders. For example, membrane anchored cytokines such as IL-10 and TGF-beta can also be used to induce tolerance or to suppress immunity which can be used in treating autoimmune diseases and transplant rejection.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 illustrates the expression tumor associated antigens and immunostimulatory molecules onto particles containing a lipid membrane, e.g., CHO cells and envelope VLPs, using GPI anchoring for protein transfer.
Figure 2 shows data on protein transfer of (A) GPI-ICAM1 or (B) GPI-IL-12 onto sheep RBCs. Red: Background control; Black: Protein transfer of GPI-ISMs.
Figure 3 shows data on Concentration dependent protein transfer of (A) GPI-ICAM-
1 or (B) GPI-IL-12 onto H5-VLPs.
Figure 4 shows data on the kinetics of protein transfer of GPI-ICAM-1 onto H5 influenza VLPs.
Figure 5 shows data on the specificity of protein transfer of GPI-ICAM1 onto VLPs. Figure 6 shows data on the inhibition of protein transfer of GPI-ICAM1 via fatty acid binding proteins.
Figure 7 shows data on the incorporation of two GPI-ISMs onto VLPs
simultaneously.
Figure 8 shows a EM of VLPs (A) before and (B) after protein transfer with
GPI-ICAM1.
Figure 9 shows data on the direct challenge with wild-type or GPI cytokine transfected 4T07 cells. BALB/C mice (n=5/group) were challenged s.c. in the hind flank with 2xl05 cells in ΙΟΟμΙ PBS and were monitored every 2-3 days for tumor growth. Mean was calculated as the average of the tumor measurements from five mice per group. For the purpose of clarity, standard deviation was not included in the graph instead the values from individual mice in each group is given in figure 10.
Figure 10 shows tumor size in individual mice post direct challenge with wild-type or
transfected 4T07 murine mammary tumor cells. BALB/C mice (n=5/group) were challenged s.c. in the hind flank with 2xl05 cells in ΙΟΟμΙ PBS and were monitored every2- 3 days for tumor growth. Each data line represents an individual mouse per group.
Figure 11 illustrates the production of extracellular portion of hHER-2 (hHER-
2ECD). Before the sequence, an optimized IL-2 Kozak sequence along with the restriction enzyme sites Hindlll and Kpnl have been added. Following the hHER2ECD sequence an EcoRI site is added. At base pair position 1365 of hHER2, a change in base pair from T was made to C in order to remove an EcoRI restriction enzyme site at this position, however, the final amino acid still remains as an isoleucine.
Figure 12 shows flow cytometry analysis of CHO cells expressing GPI-human HER-2 (hHER-2-CD59) using TA1 mAb.
DETAILED DESCRIPTION
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure, the preferred methods and materials are now described.
All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference and are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the
publications are cited. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present disclosure is not entitled to antedate such publication by virtue of prior disclosure. Further, the dates of publication provided could be different from the actual publication dates that may need to be
independently confirmed.
As will be apparent to those of skill in the art upon reading this disclosure, each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present disclosure. Any recited method can be carried out in the order of events recited or in any other order that is logically possible.
Embodiments of the present disclosure will employ, unless otherwise indicated, techniques of medicine, organic chemistry, biochemistry, molecular biology, pharmacology, and the like, which are within the skill of the art. Such techniques are explained fully in the literature. It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a support" includes a plurality of supports. In this specification and in the claims that follow, reference will be made to a number of terms that shall be defined to have the following meanings unless a contrary intention is apparent.
Prior to describing the various embodiments, the following definitions are provided and should be used unless otherwise indicated.
As used herein, the term "combination with" when used to describe administration with an additional treatment means that the agent may be administered prior to, together with, or after the additional treatment, or a combination thereof.
As used herein, the terms "prevent" and "preventing" include the prevention of the recurrence, spread or onset. It is not intended that the present disclosure be limited to complete prevention. In some embodiments, the onset is delayed, or the severity is reduced.
As used herein, the terms "treat" and "treating" are not limited to the case where the subject (e.g. patient) is cured and the disease is eradicated. Rather, embodiments of the present disclosure also contemplate treatment that merely reduces symptoms, and/or delays disease progression.
"Subject" refers any animal, preferably a human patient, livestock, rodent, monkey or domestic pet.
The terms "protein" and "polypeptide" refer to compounds comprising amino acids joined via peptide bonds and are used interchangeably.
As used herein, an "amino acid sequence" refers to an amino acid sequence of a protein molecule. The terms such as "polypeptide" or "protein" are not meant to limit the amino acid sequence to the deduced amino acid sequence, but such as amino acid deletions, additions, and modifications such as glycolsylations and addition of lipid moieties or other post-translational modifications.
With regard to any of the antigens or adjuvants disclosed herein, the protein generally refers to the most frequent human isoform, variant, mutated form, or protein with substantially identity to the full-length or portion thereof. Typically, an appropriate fragment is of the extracellular domain.
The term "portion" when used in reference to a protein (as in "a portion of a given protein") refers to fragments of that protein. The fragments may range in size from four amino acid residues or more than twenty or thirty or the entire amino sequence minus one amino acid.
The following terms are used to describe the sequence relationships between two or more proteins: "reference sequence", "sequence identity", "percentage of sequence identity", and "substantial identity". A "reference sequence" is a defined sequence used as a basis for a sequence comparison; a reference sequence may be a subset of a larger sequence, for example, as a segment of a full-length amino acid sequence of a protein. Generally, a reference sequence is at least 20 amino acids in length, frequently at least 25 amino acids in length, and often at least 50 amino acids in length. Since two proteins may each (1) comprise a sequence (i.e., a portion of the complete amino acid sequence) that is similar between the two protein, and (2) may further comprise a sequence that is divergent between the two proteins, sequence comparisons between two (or more) proteins are typically performed by comparing sequences of the two proteins over a "comparison window" to identify and compare local regions of sequence similarity. A "comparison window", as used herein, refers to a conceptual segment of at least 20 contiguous nucleotide positions wherein a sequence may be compared to a reference sequence of at least 20 contiguous amino acids and wherein the portion of the sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
Optimal alignment of sequences for aligning a comparison window may be conducted by the local homology algorithm of Smith and Waterman (Smith and Waterman, Adv. Appl. Math. 2: 482 (1981)) by the homology alignment algorithm of Needleman and Wunsch (Needleman and Wunsch, J. Mol. Biol. 48:443 (1970)), by the search for similarity method of Pearson and Lipman (Pearson and Lipman, Proc. Natl. Acad. Sci. (U.S.) 85:2444
(1988)), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by inspection, and the best alignment (i.e., resulting in the highest percentage of homology over the comparison window) generated by the various methods is selected.
The term "sequence identity" means that two sequences are identical (i.e., on a nucleotide -by-nucleotide basis) over the window of comparison. The term "percentage of sequence identity" is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical amino acids occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. The terms "substantial identity" as used herein denotes a characteristic of a sequence, wherein the protein comprises a sequence that has at least 85 percent sequence identity, preferably at least 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison window of at least 20 amino acid positions, frequently over a window of at least 25-50 nucleotides, wherein the percentage of sequence identity is calculated by comparing the reference sequence to the sequence which may include deletions or additions which total 20 percent or less of the reference sequence over the window of comparison.
Particle anchored immunostimulatory or costimulatory molecules
In certain embodiments, the disclosure relates to non-naturally occurring particle comprising, a B7-1 and/or B7-2 molecule anchored on the exterior of the particle; and an antigen molecule such as a tumor specific antigen or cancer marker anchored to the lipid membrane on the exterior of the particle. In certain embodiments, the B7-1 and or B7-2 or antigen, or protein may be anchored onto the membrane of the particle through a variety of linkages, such as lipid palmatic acid, biotin-avidin interaction, or a GPI-anchor.
In one example, a contemplated sequence of B7-1 is MGHTR QGTS
PSKCPYLNFF QLLVLAGLSH FCSGVIHVTK EVKEVATLSC GHNVSVEELAQTRIYWQKEK KMVLTMMSGD MNIWPEYKNR TIFDITNNLS IVILALRPSD EGTYECVVLK
YEKDAFKREH LAEVTLSVKA DFPTPSISDF EIPTSNIRRI ICSTSGGFPE PHLSWLENGE ELNAINTTVS QDPETELYAV SSKLDFNMTT NHSFMCLIKY GHLRVNQTFN WNTTKQEHFP DNLLPSWAIT LISVNGIFVI CCLTYCFAPR CPvEPvRRNERL RRESVRPV (SEQ ID NO : 1 ) or fragment thereof.
In another example, a contemplated sequence is VIHVTKEVKE VATLSCGFINV SVEELAQTRI YWQKEKKMVL TMMSGDMNIW PEYKNRTIFD ITNNLSIVIL ALRPSDEGTY ECVVLKYEKD AFKREHLAEV TLSVKADFPT PSISDFEIPT SNIRRIICST SGGFPEPHLS WLENGEELNA INTTVSQDPE TELYAVSSKL DFNMTTNHSF MCLIKYGHLR VNQTFNWNTT KQEHFPDN (SEQ ID NO:2) or fragment thereof. See Stamper et al., Crystal structure of the b7-l/ctla-4 complex that inhibits human immune responses. Nature (2001) 410 :608. In another example, a contemplated fragment is KAMHVAQPAV VLASSRGIAS FVCEYASPGK ATEVRVTVLR QADSQVTEVC AATYMMGNELTFLDDSICTG TSSGNQVNLT IQGLRAMDTG LYICKVELMY PPPYYLGIGN GAQIYVIDPE PCPDSD (SEQ ID NO: 3) or fragment thereof.
In certain embodiments, the disclosure relates to non-naturally occurring particle comprising, a B7-1 and/or B7-2 molecule anchored on a lipid membrane; a B7-1 and/or B7- 2 molecule anchored to the lipid membrane on the exterior of the particle; and an antigen molecule such as a tumor specific antigen or cancer marker anchored to the lipid membrane on the exterior of the particle.
A number of proteins commonly expressed by cells are attached to the cell membrane via a GPI-anchor. These proteins are post-translationally modified at their carboxy terminus to express this glycosylated moiety which is synthesized in the endoplasmic reticulum. These naturally expressing GPI-anchored molecules are widely distributed in mammalian cells and serve a host of different cellular functions, such as cell adhesion, enzymatic activity, and complement cascade regulation. Naturally occurring GPI- anchored proteins lack a transmembrane and cytoplasmic domain that otherwise anchor membrane proteins. The GPI-anchor consists of a glycosylated moiety attached to phosphatidylinositol containing two fatty acids. The phosphatidylinositol portion, as well as an ethanolamine which is attached to the C-terminal of the extracellular domain of the membrane proteins, anchor the molecule to the cell membrane lipid bilayer.
In order to exploit this natural linkage using recombinant DNA techniques, the transmembrane and cytoplasmic domains of a transmembrane surface protein need only be replaced by the signal sequence for GPI-anchor attachment that is found at the hydrophobic C-terminus of GPI-anchored protein precursors. This method may be used to generate GPI- anchored proteins is not limited to membrane proteins; attaching a GPI-anchor signal sequence to secretory proteins would also convert them to a GPI-anchored form. The method of incorporating the GPI-anchored proteins onto isolated cell surfaces or lipid particles is referred to here as protein transfer.
GPI-anchored molecules can be incorporated onto lipid membranes spontaneously. These GPI-anchored proteins can be purified from one cell type and incorporated onto different cell membranes. GPI-anchored proteins are used to customize of the lipid membranes disclosed herein for uses as a cancer vaccine. One may incorporate multiple molecules simultaneously onto the same cell membrane. One can control the level of protein expression by simply varying the concentration of the GPI-anchored molecules to be incorporated. The most significant outcome of this technology will be the reduction of time in preparing cancer vaccines from months to hours. These features make the protein transfer approach a more viable choice for the development of cancer vaccines for clinical settings. The molecules incorporated by means of protein transfer retain their functions associated with the extracellular domain. Cells and isolated membranes can be modified to express immunostimulatory molecules. In certain embodiments, the disclosure contemplates that the GPI-anchored molecules are incorporated onto the surface of albumin microparticles by this protein transfer method. GPI-anchored proteins attached to the surface of
microparticles are used to target and/or enhance the adjuvant activity of microparticles, thereby enhancing the capacity to function as a targeted antigen or drug delivery device for cancer treatment.
The GPI-B7-1 expression (by protein transfer) was stable up to 7 days on isolated membranes at 37°C and frozen membranes can be used up to 3 years of storage at - 80°C which makes the stability and storage a nonissue. These studies suggest that the membrane vaccines are more suitable to stably express the GPI-anchored molecules than on intact cells, which lose the expression within 24 hr.
This approach for introducing proteins onto membranes provides advantages over other immunotherapies for cancer vaccine development. This approach allows a protein to be added either singularly or in a combinatory manner to the tumor membrane surface. This approach navigates around the necessity to establish tumor cells as is the case for gene transfer. This GPI-mediated approach by protein transfer may be used for the co- stimulatory molecules, B7-land B7-2, GM-CSF, IL-2, and IL-12. With these cytokines being attached to the tumor membrane via a GPI-anchor, it enables them to exert their effector functions locally at the vaccination site without the risk of systemic toxicity.
Virus like particles
In certain embodiments, the disclosure relates to virus like particles comprising B7-1 and/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule anchored to the lipid membrane on the exterior of the particle for uses disclosed herein.
Influenza virus-like particles (VLPs) are particulate in nature and have shown to elicit robust immunity against antigens. Influenza VLPs have an outer lipid bilayer with properties similar to the cell membranes. Modification of influenza VLPs with a protein transfer method to incorporates tumor-associated antigens (TAAs) on the surface along with immunostimulatory molecules (ISMs) elicits enhanced immune responses directed against the TAAs. One contemplated protein transfer approach utilizes glycosyl
phosphatidylinositol (GPI)- to anchor the TAA, which can spontaneously incorporate onto the surface of the VLPs that contain a lipid bilayer upon incubation at 37°C (See Figure 1).
Incorporation of GPI-anchored forms of TAAs onto the surface of VLPs is used to direct the immune response against cancerous cells whereas the incorporation of immunostimulatory molecules (ISMs), such as GPI-anchored cytokines, costimulatory molecules, and adhesion molecules, onto the surface of VLPs is used to enhance the interaction between VLPs and antigen presenting cells (APCs) as well as lead to activation of these APCs and other immune effector cells. The incorporation of GPI-TAAs and GPI- ISMs onto VLPs by protein transfer leads to an antitumor immune response and tumor regression.
VLPs consist of a virus' capsid protein shell that presents viral antigens in an authentic conformation without the viral genome that is required for replication. Thus, they provide a safe approach for human use. VLPs contain a multivalent repetitive structure that is particulate in nature, allowing for recognition by many pattern recognition receptors and the induction of an enhanced innate and adaptive immune response. The particulate nature of VLPs allows for them to be readily taken up and presented by APCs, and thus could provide a means for breaking the immunosuppressive barrier initiated by the tumor microenvironment.
In certain embodiments, influenza virus-like particles (VLPs) may be produced using a variety of platform systems, including recombinant baculovirus vectors, transient plasmid expression systems, stable cell-line transformants, and plant expression systems. Typically VLPs are non-replicating particles that spontaneously self-assemble from expressed influenza virus proteins. In some expression systems, the viral hemagglutinin (HA) protein is sufficient for particle assembly and release from the cell. Typically the VLP comprises neuraminidase (NA). HA may present with a different type of
glycosylation depending on whether they are obtained from. For the production of VLPs containing HA in mammalian cells, co-expression of NA or exogenous ly added NA was required for the effective release of influenza VLPs into culture media, implying an important role of the NA activity in cleaving sialic acids bound to HA of budding particles. In contrast, VLPs containing HA can be produced in insect cells in the absence of NA expression. Insect cells do not add sialic acids to the N-glycans during the posttranslational modification, which explains how VLPs containing HA but not NA are effectively released from insect cell surfaces. See Kang et al, Virus Res. 2009c, 143 (2), 140-6.
In certain embodiments, VLPs used herein are recombinant influenza VLPs that have been generated in insect cells infected with rBVs expressing influenza genes HA, NA, Ml, and M2.
In certain embodiments, VLPs used herein are recombinant influenza VLPs that have been generated in insect cells infected with rBVs expressing influenza genes HA, NA, and Ml .
In certain embodiments, VLPs used herein are recombinant influenza VLPs that have been generated in insect cells infected with rBVs expressing influenza genes of HA and Ml .
In some instances, the VLP is obtained from influenza VLPs expressed from recombinant baculovirus (rBV) produced by replication in an insect cell system, e.g., Spodoptera frugiperda SF9 cells.
In some instances, the VLP is obtained from a modified vaccinia virus Ankara
(MVA) system expressing expressing influenza H5N1 HA, NA, and M proteins to generate influenza VLPs produced by replication in mammalian cells. See Schmeisser et al., Vaccine, 2012, 30(23):3413-3422.
Tumor Associate Antigens and Cancer Markers
In certain embodiments, the disclosure relates to particles such as cells or virus like particles comprising B7-1 and/or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule anchored to the lipid membrane on the exterior of the particle. Typically, the antigen molecule is a cancer marker selected from HER-2, MKI67, prostatic acid phosphatase (PAP), prostate-specific antigen (PSA), prostate-specific membrane antigen, early prostate cancer antigen, early prostate cancer antigen-2 (EPCA-2), BCL-2, MAGE antigens such as CT7, MAGE- A3 and MAGE-A4, ER 5, G-protein coupled estrogen receptor 1, CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD 117, melanoma-associated antigen (TA-90), peripheral myelin protein 22 (PMP22), epithelial membrane proteins (EMP-1, -2, and -3), HMB-45 antigen, MART-1 (Melan-A), S100A1, S100B and gp 100:209-217(210M), MUC-1, mucin antigens TF, Tn, STn, glycolipid globo H antigen. Typically, the antigen is the human form. HER-2, or Human Epidermal Growth Factor Receptor 2, refers to the human protein encoded by the ERBB2 gene that has been referred to as Neu, ErbB-2, CD340 (cluster of differentiation 340) or pl85. See Coussens et al, 1985, Science 230 (4730): 1132-9.
In certain embodiments, HER-2 is the extracellular domain or fragment thereof. In one contemplated example the protein comprises or consists essentially of the following sequence: TQVCTGTDMK LRLPASPETH LDMLRHLYQG CQVVQGNLEL
TYLPTNASLS FLQDIQEVQG YVLIAHNQVR QVPLQRLRIV RGTQLFEDNY ALAVLDNGDP LNNTTPVTGA SPGGLRELQL RSLTEILKGG VLIQRNPQLC YQDTILWKDI FHKNNQLALT LIDTNRSRAC HPCSPMCKGS RCWGESSEDC QSLTRTVCAG GCARCKGPLP TDCCHEQCAA GCTGPKHSDC LACLHFNHSG ICELHCPALV TYNTDTFESM PNPEGRYTFG ASCVTACPYN YLSTDVGSCT LVCPLHN QEVTAEDGTQRCE KCSKPCARVC YGLGMEHLRE VRAVTSANIQ EFAGCKKIFG SLAFLPESFD GDPASNTAPL QPEQLQVFET LEEITGYLYI SAWPDSLPDL SVFQNLQVIR GRILHNGAYS LTLQGLGISW LGLRSLRELG SGLALIHHNT HLCFVHTVPW DQLFRNPHQA LLHTANRPED ECVGEGLACH QLCARGHCWG PGPTQCVNCS QFLRGQECVE ECRVLQGLPR EYVNARHCLP CHPECQPQNG SVTCFGPEAD QCVACAHYKD PPFCVARCPS GVKPDLSYMP IWKFPDEEGA CQPCPIN (SEQ ID NO: 4) or fragment thereof.
In one contemplated example, the protein comprises or consists essentially of the following sequence: DIQMTQSPSS LSASVGDRVT ITCRASQDVN TAVAWYQQKP GKAPKLLIYS ASFLYSGVPS RFSGSRSGTD FTLTISSLQP EDFATYYCQQ HYTTPPTFGQ GTKVEIKRTV AAPSVFIFPP SDEQLKSGTA SVVCLLNNFY PREAKVQWKV DNALQSGNSQ ESVTEQDSKD STYSLSSTLT LSKADYEKHK VYACEVTHQG LSSPVTKSFN RGEC (SEQ ID NO: 5) or fragment thereof.
In one contemplated example, the protein comprises or consists essentially of the following sequence: GTSHLVKCAE KEKTFCVNGG ECFMVKDLSN PSRYLCKCPN EFTGDRCQNY VMASF (SEQ ID NO: 6) or fragment thereof.
MKI67, or antigen identified by monoclonal antibody Ki-67, refers to the human protein that is encoded by the MKI67 gene. See Bullwinkel et al, 2006, J. Cell. Physiol. 206 (3): 624-35.
PAP, or Prostatic acid phosphatase or prostatic specific acid phosphatase (PSAP), refers to the human enzyme produced by the prostate in males. See Ostrowski & Kuciel, 1994, Clin. Chim. Acta 226 (2): 121-9. PSA, or Prostate-specific antigen or gamma-seminoprotein or kallikrein-3 (KLK3), refers to the human protein encoded by the KLK3 gene. See Menez et al., J Mol Biol. 2008, 376(4): 1021-33.
PSMA, or Prostate-specific membrane antigen or Glutamate carboxypeptidase II, refers to a human type 2 integral membrane glycoprotein found in prostate tissues. See William et al., Reviews on Recent Clinical Trials, 2007, 2, 182-190.
Bcl-2, or B-cell lymphoma 2 refers to an protein encoded by the BCL2 gene. Bcl-2 has two isoforms that differ by two amino acids. Isoform 1 is known as 1G5M, and Isoform 2 is known as 1G50/1GJH. See Petros et al, 2001 , PNAS, 98: 3012-3017. Both isoforms are contemplated antigens.
In certain embodiments, the antigen is the entire protein, polypeptide, or a substantial fragment, or a fragment with conserved substitutions. The fragment may contain 5, 10, 20, 50, 100, or halve of the amino acids in the full length antigen. The fragment may be sufficient to mimic or replicate the folding of the full length antigen. The conserved substitutions may be amino acids that are in the interior of the folded polypeptide. A fragment is sufficient produce antibody production to the polypeptide. The antigen may be a chimera containing the fragment. The antigen may contain 1 , 2, or 3, or 5 to 10, or 10 to 20 or more conserved substitutions within the full length or polypeptide fragment which are typically outside of functional domains. In certain embodiments, the antigen may have 80%, 90%, 95% or greater sequence identity to the full length or polypeptide fragment. An antigen protein may or may not be glycosylated.
Adjuvant Molecules
In certain embodiments, the virus like particles disclosed herein comprise an adjuvant molecule anchored to a lipid membrane on the exterior of the particle wherein the adjuvant molecule and the antigen molecule are not the same molecule. In certain embodiments, the adjuvant molecule is selected from is IL-2, IL-12, ICAM1 , GM-CSF, flagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA).
It is contemplated that the co-stimulatory molecules, antigens, and adjuvant molecules may the individually conjugated to the lipophilic molecules or two or more or all of them may be conjugated together in a chimera and conjugated to a lipophilic molecule. For example, B7-1 may be conjugated to the adjuvant, HSA, in a chimera and the chimera is conjugated to a GPL
One contemplated antigen is heat stable antigen (HSA). A hybrid B7-1-HSA molecule on the cell surface membrane can function as a co-stimulatory molecule to induce T cell proliferation. CHO cells and CHO transfectants expressing HSA, B7-1, and B7-1- HSA were used as stimulator cells in a T cell proliferation assay. See Wang et al,
Immunology Letters, 2006, 105(2): 185-192.
Contemplated TLR 9 ligands as adjuvants are contemplated such as
immunostimmulatory unmethylated CpG oligonucleotides, the cytosine of the
oligonucleotide sequence 5'-CG-3' is unmethylated and the oligonucleotide is greater than about 6 base pairs in length and is less than about 100 base pairs in length such as 5'- TGACTGTGAACGTTC GAGATGA-3* (SEQ ID NO:8). It is contemplated that lipophilic molecules may be conjugated to the oligonucleotide for incorporation to the exterior of particles disclosed herein.
In certain embodiments, the antigen is also contained in the interior of the particle.
In certain embodiments, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is HER-2 and the adjuvant is flagellin and/or GM-CSF.
In certain embodiments, the antigen is HER-2 and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is HER-2, the adjuvant is flagellin and/or GM- CSF, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is HER-2 and the adjuvant is IL-12.
In certain embodiments, the antigen is HER-2, the adjuvant is IL-12, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is PSA or PAP and the adjuvant is flagellin and/or or GM-CSF.
In certain embodiments, the antigen is PSA or PAP and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is PSA or PAP, the adjuvant is flagellin, the B7- 1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is PSA or PAP and the adjuvant is IL-12. In certain embodiments, the antigen is PSA or PAP, the adjuvant is IL-12, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is HER-2 and the adjuvant is fiagellin and/or
GM-CSF.
In certain embodiments, the antigen is HER-2 and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is HER-2, the adjuvant is fiagellin and/or GM- CSF, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is HER-2 and the adjuvant is IL-12.
In certain embodiments, the antigen is HER-2, the adjuvant is IL-12, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is PSA or PAP and the adjuvant is fiagellin and/or GM-CSF.
In certain embodiments, the antigen is PSA or PAP and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
In certain embodiments, the antigen is PSA or PAP, the adjuvant is fiagellin or GM- CSF, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
Cellular Particles
In any of the embodiments, particle may be a wild type cell, cancer cell or immortalized cell.
In certain embodiments, the particle is a cell such as ZR-75-1, ZR-75-30, 184A1, UACC-812, UACC-893, HCC38, HCC70, HCC202, HCC1187, HCC1395, HCC 1428, HCC1500, HCC1569, HCC1599, HCC1806, HCC1937, HCC1954, HCC2157, HCC1419, HCC2218, AU-565, 184B5, MCF 10A, MCF 10F, MCF- 12 A, BT-20, MDA-kb2, BT-474, CAMA-1, MCF7, MDA-MB-134-VI, MDA-MB-157, MDA-MB-175-VII, MDA-MB-231, MDA-MB-361, SK-BR-3, BT-483, BT-549, DU4475, Hs 578T, MDA-MB-415, MDA- MB-436, MDA-MB-453, MDA-MB-468, T-47D, EFM19, EFM192A, Hs 578Bst,
SUM44PE, SUM52PE, SUM102PT, SUM149PT, SUM190PT, 4T1 (CRL-2539), or CAL51 for use in the treatment of cancer, breast cancer, breast adenocarcinoma, or breast carcinoma. In certain embodiments, the particle is a cell such as Jurkat, Clone E6-1 (ATCC Number: TIB-152), RBL-2H3 (CRL-2256), MOLT-4 (CRL-1582), K-562 (CCL-243), CCRF-CEM (CCL-119), HL-60 (CCL-240), or KG-1 (CCL-246) for use in the treatment of cancer, leukemia, leukemia (AML), leukemia (CML), promyelocytic leukemia, basophilic leukemia, or acute T cell leukemia.
In certain embodiments, the particle is a cell such as NCI-H358 (CRL-5807), LL/2 (CRL-1642), Calu-3 (HTB-55), NCI-H441 (HTB-174), NCI-H1975 (CRL-5908), NCI-H23 (CRL-5800), NCI-H1299 (CRL-5803), NCI-H460 (HTB-177), NCI-H292 (CRL-1848), A- 549 (CCL-185), A-549 (CCL-185), A-549 (CCL-185), IMR-90 (CCL-186), MRC-5 (CCL- 171), or WI-38 (CCL-75) for use in the treatment of cancer, lung cancer, lung
adenocarcinoma, lung carcinoma, lewis lung carcinoma, or bronchioalveolar lung cancer.
In certain embodiments, the particle is a cell such as Ramos (CRL-1596), Daudi (CCL-213), Raji (CCL-86), EL4 (TIB-39), or U-937 (CRL-1593.2) for use in the treatment of cancer, lymphoma, B-cell lymphomas, histiocytic lymphoma, or Burkitt's lymphoma.
In certain embodiments, the particle is a cell such as HeLa (CCL-2) or HeLa S3
(CCL-2.2) for use in the treatment of cancer, cervical cancer or cervical adenocarcinoma.
In certain embodiments, the particle is a cell such as COLO 205 (CCL-222), SW620 (CCL-227), SW480 (CCL-228), LoVo (CCL-229), LS 174T (CL-188), Caco-2 (HTB-37), HT-29 (HTB-38),DLD-1 (CCL-221), HCT 116 (CCL-247), T84 (CCL-248), CT26.WT (CRL-2638) for use in the treatment of cancer, colon cancer, colon carcinoma, or a colon adenocarcinoma.
In certain embodiments, the particle is a cell such as HCN-1A (CRL- 10442), U-87 MG (HTB-14), C6 (CCL-107), bEnd.3 (CRL-2299), or T98G (CRL- 1690) for use in the treatment of cancer, brain cancer, glioma, glioblastoma multiforme, glioblastoma- astrocytoma, or brain endothelioma cancer.
In certain embodiments, the particle is a cell such as 3197-3 (CRL- 1568), 3T3-Swiss albino (CCL-92), BALB/3T3 clone A31 (CCL-163), NTERA-2 cl.Dl (CRL-1973), 3T3-L1 (CL-173), NIH/3T3 (CRL-1658),SK-OV-3 (HTB-77), CHO-K1 (CCL-61), or F-12K (30- 2004) for use in the treatment of cancer, ovarian cancer, ovarian adenocarcinoma, or testicular cancer.
In certain embodiments, the particle is a cell such as 293T/17 (CRL-11268), 293 (CRL-1573), VERO C1008 (CRL-1568), Vera (CCL-81), MDCK (CCL-34), BHK-21 (CCL-10), Caki-1 (HTB-46), 786-0 (CRL- 1932), or COS-7 (CRL- 1651) for use in the treatment of cancer, renal cancer, or renal carcinoma. In certain embodiments, the particle is a cell such as H9c2 (CRL-1446) for use in the treatment of cancer or cardiac tumors.
In certain embodiments, the particle is a cell such as A-431 (CRL-1555), Detroit 551 (CCL-110), BJ (CRL-2522), B16-F10 (CRL-6475), SK-MEL-28 (HTB-72), A375 (CRL- 1619), NCTC clone 929 (CCL-1), IRR-MRC-5 (55-X), or IRR-STO (56-X) for use in the treatment of cancer, skin cancer, squamous-cell carcinoma, melanoma, areolar lesions, or epidermoid carcinoma.
In certain embodiments, the particle is a cell such as HT- 1080 (CCL-121) for use in the treatment of cancer or fibrosarcoma.
In certain embodiments, the particle is a cell such as AGS (CRL-1739) or NCI-N87
(CRL-5822) for use in the treatment of cancer, stomach cancer, gastric carcinoma or gastric adenocarcinoma.
In certain embodiments, the particle is a cell such as HepG2/C3A (CRL-10741), Hep 3B2.1-7 (HB-8064), Hep G2 (HB-8065), or Hepa 1-6 (CRL-1830) for use in the treatment of cancer, liver cancer, heptoma, or hepatocellular carcinoma.
In certain embodiments, the particle is a cell such as U266B1 (TIB- 196) for use in the treatment of cancer or multiple myeloma.
In certain embodiments, the particle is a cell such as IMR-32 (CCL-127), Neuro-2a (CCL-131), or SK-N-SH (HTB-11) for use in the treatment of cancer or neuroblastoma.
In certain embodiments, the particle is a cell such as Saos-2 (HTB-85), U-2 OS (HTB-96), or MG-63 (CRL-1427) for use in the treatment of cancer, bone cancer, or osteosarcoma.
In certain embodiments, the particle is a cell such as Beta-TC-6 (CRL-11506), AsPC-1 (CRL-1682), BxPC-3 (CRL-1687), MIA PaCa-2 (CRL-1420), PANC-1 (CRL- 1469), Capan-1 (HTB-79), or AR42J (CRL-1492) for use in the treatment of cancer, pancreatic cancer, or pancreatic carcinoma.
In certain embodiments, the particle is a cell such as PC- 12 (CRL-1721) for use in the treatment of cancer or pheochromocytoma.
In certain embodiments, the particle is a cell such as RPMI 8226 (CCL-155) for use in the treatment of cancer or plasmacytoma.
In certain embodiments, the particle is a cell such as PC-3 (CRL-1435), VCaP (CRL-2876), DU 145 (HTB-81), LNCaP clone FGC (CRL-1740), or 22Rvl (CRL-2505) for use in the treatment of cancer, prostate cancer, prostate adenocarcinoma. In certain embodiments, the particle is a cell such as ARPE-19 (CRL-2302) for use in the treatment of cancer, eye cancer, or retinal cancer.
In certain embodiments, the particle is a cell such as RD (CCL-136) for use in the treatment of cancer, sarcoma, or rhabdomyosarcoma.
In certain embodiments, the particle is a cell such as a stem cells, mesenchymal stromal/stem, pluripotent stem cell, embryo, myoblast, hybridoma or macrophage, examples include RAW 264.7 (TIB-71), J774A.1 (TIB-67), C2C12 (CRL-1772), L6 (CRL-1458), Sp2/0-Agl4 (CRL-1581) for use in the treatment of cancer. Combination strategies for cancer treatment:
In some embodiments, In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with the administration of dendritic cell (DC)-based cancer vaccines, systemic administration of cytokines, targeted therapy using Abs or other anti-cancer agents.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with the administration of dendritic cell (DC)-based cancer vaccines. DCs have the unique ability to take up and process antigens, move into secondary lymphoid tissues, and activate both helper and cytotoxic T cells.
Preparation of DC-based cancer vaccines involves loading DCs with known tumor-specific antigens, antigenic peptides, cDNA, or RNA isolated from tumor cells. In certain embodiments, an object of this disclosure is to develop more effective methods to deliver tumor antigens to DCs. One strategy is making hybrid cells by fusing tumor cells, tumor antigens, or conjugates with DCs and using the hybrid cells as vaccines. Combination therapies with DC-based cancer vaccines may be used to treat melanoma, breast cancer, multiple myeloma, NHL, lymphatic leukemia, prostatic adenocarcinoma, lung cancer, and hepatocarcinoma
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with antigen activated DCs for cancer treatments. In one example, the compositions are used in combination with DCs fused with granulocyte macrophage colony-stimulating factor (GM-CSF) and prostatic acid
phosphatase (PAP) conjugate for cancer treatments.
Provenge, an autologous DC-based vaccine, was approved by the FDA for the treatment of men with advanced prostate cancer. Provenge consists of patient-derived DCs pulsed ex vivo with a recombinant fusion protein (PA 2024) containing granulocyte macrophage colony-stimulating factor (GM-CSF) and prostatic acid phosphatase (PAP), an antigen found in 90-95% of prostate cancers.
Another cell-based approach involves using irradiated whole tumor cells as potential cancer vaccines. This strategy allows the induction of a more polyclonal immune response through the presentation of a wide array of tumor antigens. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with irradiated tumor cells for cancer treatments.
The presence of immunosuppressive cytokines in the tumor microenvironment is an important factor in the establishment of tumors. Through the secretion of
immunosuppressive cytokines, such as TGF-β and IL-10, the innate and adaptive immune responses are inhibited during tumor development. In order to overcome this
immunosuppression, the systemic administration of certain immunostimulatory cytokines, such as IL-2, IL-12, and IFN-a, has been used to alter the tumor microenvironment to mediate tumor recognition by immune cells. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with cytokines such as IL-2, IL-12, and INF-a for cancer treatments.
Cytokines activate immune cells, such as NK and CD8+ T cells, and can also inhibit tumor angiogenesis. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with IL-2, IL-12, and INF-a for the treatment of metastatic melanoma and renal cell carcinoma (RCC).
T-cell growth cytokine, IL-15, promotes the activation of a variety of immune cells, namely NK, NKT, and memory CD8+ T cells, and can overcome activation-induced cell death (AICD) caused by IL-2. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with IL-15 as a potential cancer immunotherapeutic agent.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination intra-tumoral administration of cytokines, modification of tumor cells to secrete cytokines, and fusion of cytokines with antibodies for cancer treatments. In one embodiment, the cytokine is TNF-a. In one embodiment the cancer is melanoma.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with administration of soluble GM-CSF and optionally a cytokine for cancer treatments. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with an antibody therapy for cancer treatment. In certain embodiments, the contemplated anti-bodies are directed to epidermal growth factor receptor (EGFR), human EGFR-2 (HER-2), CD20 (an unglycosylated transmembrane phosphoprotein expressed on B and T cells), CD33 (a transmembrane protein expressed on cells of myeloid lineage and also on some lymphoid cells), CD52 (a highly glycosylated 12 amino acid membrane-anchored glycosylphosphatidylinositol (GPI) protein which is expressed on all circulating lymphocytes), and VEGF. In certain embodiments the antibody may be humanized, chimeric, a radiolabeled mouse antibody for targeted radiation.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with rituximab for the treatment of B-cell non-Hodgkin's lymphoma or chronic lymphocytic leukemia.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with ofatumumab for the treatment of B- cell non-Hodgkin's lymphoma or chronic lymphocytic leukemia.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with ibritumomab (tiuxetan) for the treatment of B-cell non-Hodgkin's lymphoma.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with tositumomab for the treatment of B- cell non-Hodgkin's lymphoma.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with gemtuzumab ozogamicin for the treatment of acute myeloid leukemia.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with alemtuzumab for the treatment of B- cell non-Hodgkin's lymphoma or chronic lymphocytic leukemia.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with trastuzumab for the treatment of breast cancer.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with bevacizumab for the treatment of breast, lung, or colon cancer. In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with cetuximab for the treatment of brain and neck, or colon cancer.
In certain embodiments, the disclosure contemplates compositions disclosed herein and using any of the compositions in combination with panitumomab for the treatment of colon cancer.
In certain embodiments, the disclosure relates to methods of treating cancer comprising administering an effective amount of a particle as disclosed herein to a subject at risk of or diagnosed with cancer or a tumor optionally in combination with another anticancer agent. Other anticancer agents contemplated include gefitinib, erlotinib, docetaxel, cis-platin, 5-fluorouracil, gemcitabine, tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea, adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin, vincristine, vinblastine, vindesine, vinorelbine taxol, taxotere, etoposide, teniposide, amsacrine, topotecan, camptothecin bortezomib anegrilide, tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene fulvestrant, bicalutamide, flutamide, nilutamide, cyproterone, goserelin, leuprorelin, buserelin, megestrol, anastrozole, letrozole, vorazole, exemestane, finasteride, marimastat, trastuzumab, cetuximab, dasatinib, imatinib, bevacizumab, combretastatin, thalidomide, and/or lenalidomide or combinations thereof.
EXAMPLES
Construct, express and purify GPI-TAAs
HER-2/neu, a surface glycoprotein, is overexpressed on many aggressive forms of breast cancer. One constructs a GPI-HER-2 by attaching extracellular domain of human HEPv-2 with a GPI-signal sequence and expresses it on CHOK1 cells by gene transfection. One grows CHO cells, lyses, and purifies GPI-HER-2 by affinity chromatography.
One constructs a pCDNA3.1 plasmid expression vector containing the DNA encoding the GPI-anchored form of human HER-2 attached with the GPI-anchor signal sequence from CD59 to the extracellular domain of HER-2 using PCR and ligation into the vector as described for making GPI-GM-CSF. See Poloso et al, Mol Immunol 38:803-816. One transfects CHOK1 cells with the plasmids encoding GPI-HER-2 and confirms the GPI- anchoring by PI-PLC treatment. One grows cells using roller bottles and lyses the collected cell pellets using the detergent octyl glucoside. Purification and incorporation of GPI-ICAM1, -IL-12, and -GM-CSF from CHOKI transfectants
CHOKI cells were transfected to express GPI-ICAM1 or GPI-IL-12. Expression of the GPI-ISMs was assessed by flow cytometry and verification of the GPI-anchor was confirmed by a phospholipase (PIPLC) treatment. The transfectants were grown in large quantities, lysed, and the GPI-ISMs were purified by affinity chromatography. To determine if the purified GPI-ISMs still retained the GPI-anchor and could incorporate onto lipid bilayers by protein transfer, sheep red blood cells (RBCs) were used. The GPI-ISMs were individually incubated with the RBCs at 37°C for 2 hours, washed and then analyzed by flow cytometry. Figure 2 demonstrates that the purified GPI-ISMs were able to incorporate onto sheep RBCs.
Optimization of incorporation of GPI-ISMs onto influenza H5 VLPs using protein transfer
These VLPs are constructed by the rBV system through the expression of the hemagluttinin and matrix 1 protein in Sf9 insect cells. See Song et al., J Proteome Res. 2011, 10(8):3450-9.
To determine the optimal conditions for incorporation of GPI-ISMs onto influenza H5 VLPs, protein transfer was conducted at different concentrations of GPI-ISMs and at different temperatures. As the concentration of GPI-ISMs was increased, the amount of incorporation, as detected by western blot, also increased (Figure 3). Optimal
incorporation occurred at 37°C. Blotting against the H5 VLPs by using serum from mice injected with H5 VLPs showed that the VLP protein expression was not altered by incorporation of the ISMs. The kinetics of GPI ICAMl incorporation was also determined to show that maximum incorporation occurs after only 2 hrs of incubation (Figure 4).
Incorporation of GPI-ISMs onto H5 VLPs by protein transfer is GPI-anchor dependent.
To determine if incorporation of GPI-ISMs onto VLPs occurred via the GPI-anchor or via non-specific binding, PI-PLC treatment to cleave the GPI-anchor of GPI-ICAM1 either before incorporation (Figure 5A) or after (Figure 5B) incorporation was carried out. PI-PLC treatment of GPI-ICAM1 before incorporation and PI-PLC treatment of VLPs that have been
incorporated with GPI-ICAM1 both led to decreased expression of ICAM1 on VLPs as detected by Western blotting to ICAM1, whereas when the PI-PLC inhibitors, ZnCl2, or 1,10-phenanthroline, were included, expression was retained. To further confirm that incorporation occurs via the GPI-anchor, GPI-ICAM1 was incubated with 1% fatty-acid- free bovine serum albumin (BSA) or 1% orosomucoid that bind to the GPI-anchor before protein transfer in order to competitively inhibit incorporation of GPI-ICAM1 onto VLP membranes. Figure 6 shows that GPI-ICAM1 incubated first with 1% BSA or 1% orosomucoid showed decreased incorporation onto VLPs compared to those incorporated without prior incubation with BSA or orosomucoid.
Incorporation of more than one GPI-protein simultaneously on the same VLPs by protein transfer
To determine if more than one GPI-protein could incorporate simultaneously onto the surface of VLPs by protein transfer, VLPs were incubated with GPI-ICAM1 and GPI- IL-12 simultaneously at 37°C for 2 h. Figure 7 shows that influenza VLPs can incorporate both GPI-ISMs on their surface and the expression of the first GPI-protein is not affected by the expression of the second GPI-protein.
Structural integrity of VLPs remains intact after protein transfer.
To determine if the structural integrity of the VLPs remains intact after
incorporation, electron microscopy of VLPs before and after incorporation was conducted. Figure 8 shows that even after incorporation, the VLP membranes remain intact suggesting that the protein transfer method is not detrimental to the VLP structural integrity. This data show that purified GPI-proteins are able to incorporate onto influenza VLPs within 2 h at 37°C via the GPI-anchor without disturbing the structural integrity of the VLPs. Study tumor regression and immune responses induced by vaccination with VLPs modified with GPI-HER-2 and GPI-ISMs by protein transfer in mice with established tumors
Protein transferred-VLPs that express the GPI-HER-2 in combination with
GPIISMs, such as GPI-IL-2, GPI-IL-12, GPI-B7-1, and GPI-ICAM-1, leads to tumor regression in mice with established tumors that express HER-2. Although it is not intended that the disclosure be limited by any particular mechanism, the incorporation of cytokines onto the surface of VLPs allows for a slow release depot of the cytokines into the administered microenvironment, leading to increased activation of immune effector cells at the vaccination site while decreasing chances of systemic toxicity. Furthermore, the receptors of the ISMs, IL-2, IL-12, and ICAM-1 are found on APCs allowing for enhanced adhesion and activation of the APCs by the VLPs, thus leading to enhanced uptake and presentation. The receptors for IL-12 and B7-1 are also found on
other immune cells such as NK cells and mast cells, allowing for the activation of a wide variety of immune effector cells to be elicited by the association of these ISMs onto the surface of VLPs. Since the immune response is directed against the antigens found on the VLPs, incorporating TAAs along with ISMs onto the surface of VLPs will direct the immune response towards the TAAs that are overexpressed on tumor cells as well.
To determine the efficacy of VLPs incorporated with GPI-TAAs and GPI-ISMs in regressing established tumors in vivo, one inoculates BALB/c mice with 4T07 tumor cells that expressing HER-2 and then start treatment a few days later (Table 1).
Figure imgf000030_0001
One injects live 4T07 tumor cells s.c. into the left flank of the mice and injects VLP in the right flank starting on days 4, 8, and 12 after tumor inoculation. If tumors do not regress, one uses a more vigorous 2-day interval immunization schedule. One monitors the mice daily and measures the size of the tumor. One screens for the production of antibodies to HER2/neu using flow cytometry or cell ELISA.
Expression of human breast cancer antigens in 4T07-WT cells
Using the 4T07 murine breast cancer model the effects of expressing GPI-anchored immune stimulatory molecules (GPI-ISMs), namely cytokines (IL-2, IL-12) and the costimulatory protein B7-1, were investigated on the surface of the tumor cells. BALB/c mice were challenged subcutaneously (s.c.) with either wild-type 4T07 cells (4T07-WT) or 4T07 cells expressing GPI-ISMs. Significant splenomegaly was observed in the mice challenged with 4T07-WT cells relative to the mice challenged with 4T07 cells expressing GPI-ISMs. This observed splenomegaly correlated with tumor size and a 4-5 fold increase in the percentage of splenic CDl lb+Grl+ MDSCs indicating the role of active immune suppression in the tumorigenicity of 4T07 breast cancer cells. Studies were conducted to analyze the effect of GPI-ISMs on infiltrating cells into the tumor microenvironment as well as in the spleen and draining lymph nodes (dLNs). Three groups of mice were challenged (s.c.) with the following cells mixed in a 1 : 1 ratio with BD Matrigel™ (a solubilized basement membrane preparation derived from a mouse sarcoma): 4T07-WT, 4T07-B7/IL- 12 or PBS (control). Seven days post challenge, the Matrigel/tumor, spleen and dLNs were harvested from the mice, digested and analyzed for cellular infiltrates by flow cytometry. The expression of GPI-ISMs on the surface of tumor cells led to reduced angiogenesis as evidenced by a reduced level of blood vessels and decreased presence of
CD4+CD25+FOXP3+ regulatory T cells and CDl lb+Grl+ MDSCs locally at the tumor site and dLNs as well as systemically in the spleen. Additionally, there was a decrease in CD8+PD1+ exhausted T cells at the tumor site. Along with the inhibition of immune suppressive cell populations, the GPI-ISMs increased the presence of CD4+ and CD8+ T cells as well as dendritic cells and B cells. These observations suggest that components of the active immune suppression evident in this model can be inhibited by expressing GPI- ISMs on the surface of the 4T07 tumor cells and could be effective in a therapeutic setting.
BALB/C female mice (five per group) were challenged subcutaneously (s.c.) with wild-type 4T07 or transfected 4T07-B7, GPI-IL-2, GPI-IL-12, B7/GPI-IL-2 or B7/GPI-IL- 12 cells (all 2xl05 cells in ΙΟΟμΙ PBS). Mice were injected s.c. in the rear flank and were monitored daily. Tumor size was measured using Vernier calipers every 2nd-3rd day by taking 2x2 perpendicular measurements, and tumor size (mm2) was calculated by multiplying the two diameters. Mice were euthanized when the tumor size reached close to 2 cm2. After 33 days of the initial challenge, tumor- free mice in the experimental groups were rechallenged on the opposite hind flank with wild-type 4T07 cells (2x105 in ΙΟΟμΙ PBS). Mice in each group were marked individually by ear punch and tumor growth was measured and recorded for each mouse separately. The wild-type and transfected tumor cell lines all began to grow tumors in vivo, but while the wild-type tumors continued to increase in size, the tumors from the modified cell lines all regressed (See figure 9 and 10).
Preparation and evaluation of hHER-2(ECD)-CD59 GPI
HER-2ECD is the extracellular portion of hHER-2. The hHER-2 extracellular domain with CD59 GPI signal sequence were join and introduced by a EcoRI site, i.e., joining region: g/aattc introduced EcoRV site (gat/atc) before sequence and Apal (gggcc/c) site after sequence at the joining region as illustrated in figure 11. Before the sequence, an optimized IL-2 Kozak sequence along with the restriction enzyme sites Hindlll and Kpnl were added. Following the hHER2ECD sequence an EcoRI site is added. At base pair position 1365 of hHER2, a change in base pair from T was made to C in order to remove an EcoRI restriction enzyme site at this position, however, the final amino acid still remains as an isoleucine. (2015bp). Figure 12 shows flow cytometry analysis of CHO cells expressing GPI-human HER-2 (hHER-2-CD59) using TAl mAb. Testing shows that HER-2 expressed in CHO cells is GPI-anchored. PIPLC is an enzyme which cleaves GPI anchor, reduces the level of expression. PI-PLC treated CHOKl-hHER-2ECD-CD59 cells reduced hHER-2 cell surface expression by 98.4%. PIPLC will not have any effect on normal HER-2.
Nucleic acid encoding the hHER-2 extracellular domain E (Amino Acids 22-652) and GPI-anchor signal sequence (SEQ ID NO: 7) AAGGGGAGGT AACCCTGGCC CCTTTGGTCG GGGCCCCGGG CAGCCGCGCG CCCCTTCCCA CGGGGCCCTT TACTGCGCCG CGCGCCCGGC CCCCACCCCT CGCAGCACCC CGCGCCCCGC GCCCTCCCAG CCGGGTCCAG CCGGAGCCAT GGGGCCGGAGGATATC CCGCAGTGAG CACCATGGAG CTGGCGGCCT TGTGCCGCTG GGGGCTCCTC CTCGCCCTCT TGCCCCCCGG AGCCGCGAGC ACCCAAGTGT GCACCGGCAC AGACATGAAG CTGCGGCTCC CTGCCAGTCC CGAGACCCACCTGGACATGC TCCGCCACCT CTACCAGGGC TGCCAGGTGG TGCAGGGAAA CCTGGAACTC ACCTACCTGC CCACCAATGC CAGCCTGTCC TTCCTGCAGG ATATCCAGGA GGTGCAGGGC TACGTGCTCA TCGCTCACAA CCAAGTGAGG CAGGTCCCAC TGCAGAGGCT GCGGATTGTG CGAGGCACCC AGCTCTTTGA GGACAACTAT GCCCTGGCCG TGCTAGACAA TGGAGACCCG CTGAACAATA CCACCCCTGT CACAGGGGCC TCCCCAGGAG GCCTGCGGGA GCTGCAGCTT CGAAGCCTCA CAGAGATCTT GAAAGGAGGG GTCTTGATCC AGCGGAACCC CCAGCTCTGC TACCAGGACA CGATTTTGTG GAAGGACATC TTCCACAAGA ACAACCAGCT GGCTCTCACACTGATAGACA CCAACCGCTC TCGGGCCTGC CACCCCTGTT CTCCGATGTG TAAGGGCTCC CGCTGCTGGG GAGAGAGTTC TGAGGATTGT CAGAGCCTGA CGCGCACTGT CTGTGCCGGT GGCTGTGCCC GCTGCAAGGG GCC ACTGCCC ACTGACTGCT GCCATGAGCA GTGTGCTGCC GGCTGCACGG GCCCCAAGCA CTCTGACTGC CTGGCCTGCC TCCACTTCAA CCACAGTGGC ATCTGTGAGC TGCACTGCCC AGCCCTGGTC ACCTACAACA CAGACACGTT TGAGTCCATG CCCAATCCCG AGGGCCGGTA TACATTCGGC GCCAGCTGTG TGACTGCCTG TCCCTACAAC TACCTTTCTA CGGACGTGGG ATCCTGCACC CTCGTCTGCC CCCTGCACAA CCAAGAGGTG ACAGCAGAGG ATGGAACACA GCGGTGTGAG AAGTGCAGCA AGCCCTGTGC CCGAGTGTGC TATGGTCTGG GCATGGAGCA CTTGCGAGAG GTGAGGGCAG TTACCAGTGC CAATATCCAG GAGTTTGCTG GCTGCAAGAA GATCTTTGGG AGCCTGGCAT TTCTGCCGGA GAGCTTTGAT GGGGACCCAG CCTCCAACAC TGCCCCGCTC CAGCCAGAGC AGCTCCAAGT GTTTGAGACT CTGGAAGAGA TCACAGGTTA CCTATACATC TCAGCATGGC CGGACAGCCT GCCTGACCTC AGCGTCTTCC AGAACCTGCA AGTAATCCGG GGACGAATTC TGCACAATGG CGCCTACTCG CTGACCCTGC AAGGGCTGGG CATCAGCTGG CTGGGGCTGC GCTCACTGAG GGAACTGGGC AGTGGACTGG CCCTCATCCA CCATAACACC CACCTCTGCT TCGTGCACAC GGTGCCCTGG GACCAGCTCT TTCGGAACCC GCACCAAGCT CTGCTCCACA CTGCCAACCG GCCAGAGGAC GAGTGTGTGG GCGAGGGCCT GGCCTGCCAC CAGCTGTGCG CCCGAGGGCA CTGCTGGGGT CCAGGGCCCA CCCAGTGTGT CAACTGCAGC CAGTTCCTTC GGGGCCAGGA GTGCGTGGAGGAATGCCGAG TACTGCAGGG GCTCCCCAGG GAGTATGTGA ATGCCAGGCA CTGTTTGCCGTGCCACCCTG AGTGTCAGCC CCAGAATGGC TCAGTGACCT GTTTTGGACC GGAGGCTGACCAGTGTGTGG CCTGTGCCCA CTATAAGGAC CCTCCCTTCT GCGTGGCCCG CTGCCCCAGC GGTGTGAAAC CTGACCTCTC CTACATGCCC ATCTGGAAGT TTCCAGATGA GGAGGGCGCA TGCCAGCCTT GCCCCATCAA CTGCACCCAC TCCTGTGTGG ACCTGGATGA CAAGGGCTGC CCCGCCGAGC AGAGAGCCAG CCCTCTGACGGAATTC
CTTGAAAATGGTGGGACATCCTTATCAGAGAAAACAGTTCTTCTGCTGGT GACTCCATTTCTGGCAGCAGCCTGGAGCCTTCATCCCTAACAGAAG GCCAAGGGGCCCTCCG

Claims

CLAIMS What we claim:
1. A non-naturally occurring particle comprising,
a lipid membrane;
a B7-1 or B7-2 molecule anchored to the lipid membrane on the exterior of the particle; and
an antigen molecule anchored to the lipid membrane on the exterior of the particle.
2. The particle of Claim 1 further comprising an adjuvant molecule anchored to the lipid membrane on the exterior of the particle wherein the adjuvant molecule and antigen molecule are not the same molecule.
3. The particle of Claim 2, wherein the adjuvant molecule is selected from molecules comprising IL-2, IL-12, ICAMl GM-CSF, flagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA).
4. The particle of Claims 1-3, wherein the lipid membrane is a phospholipid monolayer or phospholipid bilayer.
5. The particle of Claims 1-4 wherein the particle is a cell, allogeneic or autologous cancer cell or its membrane fragments or vesicles, liposome, virosome, micelle, polymer, or virus like particle.
6. The particle of Claims 1-5, wherein the B7-1 molecule is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl- phosphatidylinositol, phospholipid, glycolipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
7. The particle of Claims 1-6, wherein the antigen molecule is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl- phosphatidylinositol, phospholipid, glycolipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
8. The particle of Claims 1-7, wherein the adjuvant molecule is anchored to the lipid membrane on the exterior of the particle through a conjugated glycosyl- phosphatidylinositol, phospholipid, glycolipid, triglyceride, saturated or unsaturated fatty acid, or other lipophilic molecule.
9. The particle of Claims 1-8, wherein antigen is a cancer marker molecule selected from HER-2, MUC-1, mucin antigens TF, Tn, STn, glycolipid globo H antigen, prostatic acid phosphatase (PAP), prostate-specific antigen, prostate-specific membrane antigen, early prostate cancer antigen-2 (EPCA-2), bcl-2, G-protein coupled estrogen receptor 1 , CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD117, melanoma-associated antigen (TA-90), peripheral myelin protein 22 (PMP22), epithelial membrane proteins (EMP-1, -2, and -3), HMB-45 antigen, MART-1 (Melan-A), S100A1, and S100B.
10. The particle of Claims 1-9, wherein the antigen is contained in the interior of the particle.
11. The particle of Claims 1-10, wherein the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
12. The particle of Claims 1-10, wherein the antigen is HER-2 and the adjuvant is f agellin or GM-CSF.
13. The particle of Claims 1-10, wherein the antigen is HER-2 and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
14. The particle of Claims 1-10, wherein the antigen is HER-2, the adjuvant is flagellin or GM-CSF, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
15. The particle of Claims 1-10, wherein the antigen is HER-2 and the adjuvant is IL-12.
16. The particle of Claims 1-10, wherein the antigen is HER-2, the adjuvant is IL-12, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
17. The particle of Claims 1-10, wherein the antigen is PSA or PAP and the adjuvant is flagellin or GM-CSF.
18. The particle of Claims 1-10, wherein the antigen is PSA or PAP and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
19. The particle of Claims 1-10, wherein the antigen is PSA or PAP, the adjuvant is flagellin, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
20. The particle of Claims 1-10, wherein the antigen is PSA or PAP and the adjuvant is IL-12.
21. The particle of Claims 1-10, wherein the antigen is PSA or PAP, the adjuvant is IL- 12, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
22. A virus like particle comprising B7-1 or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule anchored to the lipid membrane on the exterior of the particle.
23. The virus like particle of claim 18, wherein the antigen molecule is a cancer marker selected from HER-2, prostatic acid phosphatase (PAP), prostate-specific antigen (PSA), prostate-specific membrane antigen, early prostate cancer antigen-2 (EPCA-2), BCL-2, MAGE antigens such as CT7, MAGE- A3 and MAGE-A4, G-protein coupled estrogen receptor 1 , CA15-3, CA19-9, CA 72-4, CA-125, carcinoembryonic antigen, CD20, CD31, CD34, PTPRC (CD45), CD99, CD 117, melanoma-associated antigen (TA-90), peripheral myelin protein 22 (PMP22), epithelial membrane proteins (EMP-1, -2, and -3), HMB-45 antigen, MART-1 (Melan-A), S100A1, and S100B.
24. The virus like particle of Claims 22-23 further comprising an adjuvant molecule anchored to a lipid membrane on the exterior of the particle wherein the adjuvant molecule and the antigen molecule are not the same molecule.
25. The virus like particle of Claim 24, wherein the adjuvant molecule is selected from is IL-2, IL-12, ICAM1, GM-CSF, flagellin, and heat stable antigen (HSA).
26. The virus like particle of Claims 22-25, wherein the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
27. The virus like particle of Claims 24-25, wherein the antigen is HER-2 and the adjuvant is flagellin or GM-CSF.
28. The virus like particle of Claims 22-25, wherein the antigen is HER-2 and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
29. The virus like particle of Claims 24-25, wherein the antigen is HER-2, the adjuvant is flagellin or GM-CSF, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
30. The virus like particle of Claims 24-25, wherein the antigen is HER-2 and the adjuvant is IL-12.
31. The virus like particle of Claims 24-25, wherein the antigen is HER-2, the adjuvant is IL-12, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
32. The virus like particle of Claims 24-25, wherein the antigen is PSA or PAP and the adjuvant is flagellin or GM-CSF.
33. The virus like particle of Claims 22-25, wherein the antigen is PSA or PAP and the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
34. The virus like particle of Claims 24-25, wherein the antigen is PSA or PAP, the adjuvant is flagellin or GM-CSF, the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
35. A method of treating cancer comprising administering an effective amount of a particle or a virus like particle of Claims 1-34 to a subject at risk of or diagnosed with cancer or a tumor optionally in combination with unmethylated CpG oligonucleotide, methyl jasmonate, cyclophosphamide, gencitabine or other immunosuppression blocker or other anticancer agent.
36. The method of Claim 35 wherein the subject is a human subject.
37. A virus like particle comprising B7-1 or B7-2 molecule anchored to a lipid membrane on the exterior of the particle and an antigen molecule wherein the antigen molecule is a viral protein.
38. The virus like particle of Claim 37, wherein the viral protein is an hemagglutinin selected from influenza HI, H2, H3, H4, H5, H6, H7, H8, H9, H10, HI 1, HI 2, HI 3, HI 4, H15, and H16.
39. The virus like particle of Claims 37-38, wherein the viral protein is a neuraminidase selected from influenza Nl, N2, N3, N4, N5, N6, N7, and N8.
40. The virus like particle of Claim 37, wherein the virus like particle is an HIV envelope protein selected from gp 41, gp 120, and gp 160.
41. The virus like particle of Claims 37-40 further comprising an adjuvant molecule selected from IL-2, IL-12, ICAMl GM-CSF, fiagellin, unmethylated, CpG oligonucleotide, lipopolysaccharides, lipid A, and heat stable antigen (HSA).
42. The virus like particle of Claim 37-41, wherein the B7-1 molecule is a B7-1 and heat stable antigen (HSA) hybrid chimera.
43. A method of treating or preventing a viral infection comprising administering an effective amount of a virus like particle of Claims 37-42 to a subject at risk of, exhibiting symptoms of, or diagnosed with a viral infection.
44. A particle comprising a cancer marker made by the process of mixing a cancer marker conjugated to a lipophilic moiety and a particle comprising a lipid membrane.
45. The particle of Claim 44, wherein the cancer marker is HER-2 or PSA or PAP.
46. The particle of Claims 44-45, wherein the lipophilic moiety is selected from glycosyl-phosphatidylinositol, phospholipid, glycolipid, triglyceride, saturated and unsaturated fatty acids.
47. A particle comprising a cancer marker and B7-1 or B7-2 made by the process of mixing a B7-1 or B7-2 conjugated to a lipophilic moiety and a particle comprising a lipid membrane and a cancer marker.
48. The particle of Claims 44-47, wherein the particle is a cancer cell, liposome, virosome, micelle, polymer, or virus like particle.
49. A method of treating or preventing breast cancer comprising administering an effective amount of a particle comprising B7-1 or B7-2, GM-CSF, and HER-2 to a subject in need thereof.
50. A method of treating or preventing prostate cancer comprising administering an effective amount of a particle comprising B7-1 or B7-2 , GM-CSF, and PSA or PAP to a subject in need thereof.
PCT/US2013/024355 2012-02-03 2013-02-01 Immunostimulatory compositions, particles, and uses related thereto WO2013116656A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP18196067.5A EP3520809A1 (en) 2012-02-03 2013-02-01 Immunostimulatory compositions.
EP13743997.2A EP2809345A4 (en) 2012-02-03 2013-02-01 Immunostimulatory compositions, particles, and uses related thereto
CA2863658A CA2863658C (en) 2012-02-03 2013-02-01 Immunostimulatory compositions, particles, and uses related thereto
US14/374,729 US20150071987A1 (en) 2012-02-03 2013-02-01 Immunostimulatory compositions, particles, and uses related thereto
US15/833,769 US10987419B2 (en) 2012-02-03 2017-12-06 Immunostimulatory compositions, particles, and uses related thereto
US17/132,320 US20210154291A1 (en) 2012-02-03 2020-12-23 Immunostimulatory compositions, particles, and uses related thereto

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261594754P 2012-02-03 2012-02-03
US61/594,754 2012-02-03

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/374,729 A-371-Of-International US20150071987A1 (en) 2012-02-03 2013-02-01 Immunostimulatory compositions, particles, and uses related thereto
US15/833,769 Continuation US10987419B2 (en) 2012-02-03 2017-12-06 Immunostimulatory compositions, particles, and uses related thereto

Publications (1)

Publication Number Publication Date
WO2013116656A1 true WO2013116656A1 (en) 2013-08-08

Family

ID=48905876

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/024355 WO2013116656A1 (en) 2012-02-03 2013-02-01 Immunostimulatory compositions, particles, and uses related thereto

Country Status (4)

Country Link
US (3) US20150071987A1 (en)
EP (2) EP3520809A1 (en)
CA (1) CA2863658C (en)
WO (1) WO2013116656A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015089114A1 (en) * 2013-12-09 2015-06-18 Bullet Biotechnology, Inc. Specific virus-like particle-cpg oligonucleotide vaccines and uses thereof
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
CN107614531A (en) * 2015-03-18 2018-01-19 纪念斯隆凯特琳癌症中心 CD99 composition and method is targetted in hematopoiesis and lymphoid malignancy
US9896483B2 (en) 2013-01-23 2018-02-20 The Board Of Trustees Of The Leland Stanford Junior University Stabilized hepatitis B core polypeptides
US20180128833A1 (en) * 2016-11-08 2018-05-10 Metaclipse Therapeutics Corporation Methods of treating with tumor membrane vesicle-based immunotherapy and predicting therapeutic response thereto
CN108778263A (en) * 2016-03-28 2018-11-09 维达克制药有限公司 Stabilizing pharmaceutical composition and application thereof for local application
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2019070955A1 (en) * 2017-10-04 2019-04-11 Georgia State University Research Foundation, Inc. Headless hemagglutin influenza vaccine

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150071987A1 (en) * 2012-02-03 2015-03-12 Emory University Immunostimulatory compositions, particles, and uses related thereto
AU2013359907B2 (en) 2012-12-11 2018-01-18 Albert Einstein College Of Medicine, Inc. Methods for high throughput receptor:ligand identification
KR20180069903A (en) 2015-11-02 2018-06-25 파이브 프라임 테라퓨틱스, 인크. CD80 extracellular domain polypeptides and their use in the treatment of cancer
CA3019005A1 (en) * 2016-05-18 2017-11-23 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
WO2017201131A1 (en) 2016-05-18 2017-11-23 Albert Einstein College Of Medicine, Inc. Variant pd-l1 polypeptides, t-cell modulatory multimeric polypeptides, and methods of use thereof
CA3043630A1 (en) 2016-12-22 2018-06-28 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
US11851471B2 (en) 2017-01-09 2023-12-26 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
KR102619015B1 (en) 2017-03-15 2023-12-28 큐 바이오파마, 인크. How to Regulate Your Immune Response
MX2019012849A (en) 2017-04-28 2019-11-28 Five Prime Therapeutics Inc Methods of treatment with cd80 extracellular domain polypeptides.
US20200199532A1 (en) * 2017-05-19 2020-06-25 Case Western Reserve University Compositions and methods for expanding ex vivo natural killer cells and therapeutic uses thereof
WO2019139896A1 (en) 2018-01-09 2019-07-18 Cue Biopharma, Inc. Multimeric t-cell modulatory polypeptides and methods of use thereof
GB201910651D0 (en) * 2019-07-25 2019-09-11 Autolus Ltd Virus-like particle
CN113521274A (en) * 2020-04-17 2021-10-22 北京科兴中维生物技术有限公司 COVID-19 inactivated vaccine composition and application
EP4149534A2 (en) 2020-05-12 2023-03-22 Cue Biopharma, Inc. Multimeric t-cell modulatory polypeptides and methods of use thereof
US20230190916A1 (en) * 2020-05-14 2023-06-22 Metaclipse Therapeutics Corporation Compositions and methods for detecting and treating a sars-cov-2 infection
US20230033099A1 (en) * 2021-07-28 2023-02-02 Emory University Dendritic Cells Pulsed With Tumor Membrane Vesicles And Uses In Treating Cancer

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020009468A1 (en) * 1996-08-15 2002-01-24 Periasamy Selvaraj Compositions and methods for cancer prophylaxis and/or treatment
WO2007039458A2 (en) * 2005-09-21 2007-04-12 Cytos Biotechnology Ag Hiv peptide conjugates and uses thereof
WO2007047831A2 (en) * 2005-10-18 2007-04-26 Novavax, Inc. Functional influenza virus like particles (vlps)
US20070243159A1 (en) * 2003-04-30 2007-10-18 Periasamy Selvaraj Therapeutic Compositions and Vaccines By Glycosyl-Phosphatidylinositol (Gpi)-Anchored Cytokines and Immunostimulatory Molecules

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6946133B1 (en) * 1996-03-20 2005-09-20 The United States Of America As Represented By The Department Of Health And Human Services Prostate specific antigen oligo-epitope peptide
US6730512B2 (en) * 1997-04-09 2004-05-04 Amdl, Inc. Combination immunogene therapy
US6054312A (en) * 1997-08-29 2000-04-25 Selective Genetics, Inc. Receptor-mediated gene delivery using bacteriophage vectors
US6749856B1 (en) * 1997-09-11 2004-06-15 The United States Of America, As Represented By The Department Of Health And Human Services Mucosal cytotoxic T lymphocyte responses
US20070269455A1 (en) * 1998-05-26 2007-11-22 Genitrix, Llc Cytokine-coated cells and methods of modulating an immune response to an antigen
US20020039573A1 (en) * 2000-01-21 2002-04-04 Cheever Martin A. Compounds and methods for prevention and treatment of HER-2/neu associated malignancies
US20020006413A1 (en) * 2000-01-27 2002-01-17 Sobol Robert E. Genetically engineered tumor cell vaccines
KR20020010206A (en) * 2000-07-27 2002-02-04 이시우 DNA vector comprising a single chain IL-12 and B7.1, and Anti-cancer cell vaccine transformed with the above vector
US20030232324A1 (en) * 2001-05-31 2003-12-18 Chiron Corporation Chimeric alphavirus replicon particles
US20040136963A1 (en) * 2001-06-22 2004-07-15 The Trustees Of The University Of Pennsylvania Simian adenovirus vectors and methods of use
US20030105054A1 (en) * 2001-08-27 2003-06-05 Greenville Hospital System GPI-anchored cytokines
US7745140B2 (en) * 2002-01-03 2010-06-29 The Trustees Of The University Of Pennsylvania Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool
US20040009146A1 (en) * 2002-02-26 2004-01-15 Osvaldo Podhajcer Anti-tumor vaccine and method
TWI259206B (en) * 2002-09-24 2006-08-01 Univ Nat Cheng Kung A DNA vaccine containing a tumor associated gene and a cytokine gene and the method producing thereof
US7429472B2 (en) * 2003-01-31 2008-09-30 Promega Corporation Method of immobilizing a protein or molecule via a mutant dehalogenase that is bound to an immobilized dehalogenase substrate and linked directly or indirectly to the protein or molecule
ES2653570T3 (en) * 2004-05-27 2018-02-07 The Trustees Of The University Of Pennsylvania Innovative artificial antigen presenting cells and uses thereof
EP2465538A3 (en) * 2004-10-21 2013-11-20 Ono Pharmaceutical Co., Ltd. Use of immunesuppressant receptor
EP2418278A3 (en) * 2005-05-09 2012-07-04 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
JP4824389B2 (en) * 2005-10-28 2011-11-30 株式会社医学生物学研究所 Cytotoxic T cell epitope peptide that specifically attacks Epstein-Barr virus infected cells and uses thereof
US7498142B2 (en) * 2006-01-31 2009-03-03 Yeda Research And Development Co., Ltd. Methods of identifying combinations of antibodies with an improved anti-tumor activity and compositions and methods using the antibodies
EP1844788B1 (en) * 2006-04-13 2010-07-14 Bio Life Science Forschungs- und Entwicklungsges.m.b.H. HER-2/neu multi-peptide vaccine
EP2111231A4 (en) * 2007-02-13 2010-12-15 Univ Northeastern Methods and compositions for improving immune responses
EP3222634A1 (en) * 2007-06-18 2017-09-27 Merck Sharp & Dohme B.V. Antibodies to human programmed death receptor pd-1
US8313896B2 (en) * 2008-04-04 2012-11-20 The General Hospital Corporation Oncolytic herpes simplex virus immunotherapy in the treatment of brain cancer
JP6055165B2 (en) * 2008-11-14 2016-12-27 株式会社Idファーマ Method for producing dendritic cells
US8779095B2 (en) * 2009-05-07 2014-07-15 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services LAT adapter molecule for enhanced T-cell signaling and method of use
CA3017298C (en) * 2009-05-15 2021-09-28 Irx Therapeutics, Inc. Compositions comprising primary cell-derived biologics for enhancing immune responses in patients
DK2770061T3 (en) * 2009-07-24 2019-01-07 Immune Design Corp NON-INTEGRATING LENTIVIRUS VECTORS
US20130022640A1 (en) * 2010-01-07 2013-01-24 Secretary, Department Of Health And Human Services Immune modulators relating to foxo3a
JP5894538B2 (en) * 2010-02-04 2016-03-30 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア ICOS critically regulates the proliferation and function of inflammatory human Th17 cells
US8557777B2 (en) * 2010-07-09 2013-10-15 The Board Of Trustees Of The University Of Illinois Methods for treating cancer using prostate specific antigen and tumor endothelial marker peptides
JP2015505326A (en) * 2012-01-18 2015-02-19 ニューメディシンズ,インコーポレーテッド IL-12 for radiation protection and radiation-induced toxicity alleviation
US20150071987A1 (en) * 2012-02-03 2015-03-12 Emory University Immunostimulatory compositions, particles, and uses related thereto
WO2013164754A2 (en) * 2012-05-04 2013-11-07 Pfizer Inc. Prostate-associated antigens and vaccine-based immunotherapy regimens
CN115093480A (en) * 2012-05-31 2022-09-23 索伦托药业有限公司 Antigen binding proteins that bind to PD-L1
US20180128833A1 (en) * 2016-11-08 2018-05-10 Metaclipse Therapeutics Corporation Methods of treating with tumor membrane vesicle-based immunotherapy and predicting therapeutic response thereto

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020009468A1 (en) * 1996-08-15 2002-01-24 Periasamy Selvaraj Compositions and methods for cancer prophylaxis and/or treatment
US20070243159A1 (en) * 2003-04-30 2007-10-18 Periasamy Selvaraj Therapeutic Compositions and Vaccines By Glycosyl-Phosphatidylinositol (Gpi)-Anchored Cytokines and Immunostimulatory Molecules
WO2007039458A2 (en) * 2005-09-21 2007-04-12 Cytos Biotechnology Ag Hiv peptide conjugates and uses thereof
WO2007047831A2 (en) * 2005-10-18 2007-04-26 Novavax, Inc. Functional influenza virus like particles (vlps)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
See also references of EP2809345A4 *
WANG Y.C. ET AL.: "B7-1-HSA (CD80-CD24), a recombinant hybrid costimulatory molecule retains ligand binding and costimulatory functions.", IMMUNOL. LETT., vol. 105, no. 2, 2006, pages 185 - 192, XP005480654 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9896483B2 (en) 2013-01-23 2018-02-20 The Board Of Trustees Of The Leland Stanford Junior University Stabilized hepatitis B core polypeptides
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015089114A1 (en) * 2013-12-09 2015-06-18 Bullet Biotechnology, Inc. Specific virus-like particle-cpg oligonucleotide vaccines and uses thereof
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
CN107614531A (en) * 2015-03-18 2018-01-19 纪念斯隆凯特琳癌症中心 CD99 composition and method is targetted in hematopoiesis and lymphoid malignancy
CN108778263A (en) * 2016-03-28 2018-11-09 维达克制药有限公司 Stabilizing pharmaceutical composition and application thereof for local application
US20180128833A1 (en) * 2016-11-08 2018-05-10 Metaclipse Therapeutics Corporation Methods of treating with tumor membrane vesicle-based immunotherapy and predicting therapeutic response thereto
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2019070955A1 (en) * 2017-10-04 2019-04-11 Georgia State University Research Foundation, Inc. Headless hemagglutin influenza vaccine
US11945841B2 (en) 2017-10-04 2024-04-02 Gerogia State University Research Foundation, Inc. Headless hemagglutin influenza vaccine

Also Published As

Publication number Publication date
EP2809345A1 (en) 2014-12-10
EP2809345A4 (en) 2015-11-25
US20210154291A1 (en) 2021-05-27
US10987419B2 (en) 2021-04-27
CA2863658C (en) 2023-03-14
US20180117145A1 (en) 2018-05-03
CA2863658A1 (en) 2013-08-08
US20150071987A1 (en) 2015-03-12
EP3520809A1 (en) 2019-08-07

Similar Documents

Publication Publication Date Title
US20210154291A1 (en) Immunostimulatory compositions, particles, and uses related thereto
WO2010030002A1 (en) Cell capable of expressing exogenous gitr ligand
Cai et al. Heterologous prime-boost enhances the antitumor immune response elicited by plant-virus-based cancer vaccine
JP2023506381A (en) Recombinant Polypeptides for Programming Extracellular Vesicles
CN115551537A (en) Tumor cell vaccine
US20230074462A1 (en) Methods and compositions for stimulating immune response
EP4021487A2 (en) Antigenic peptides for prevention and treatment of b-cell malignancy
WO2019101062A1 (en) Recombinant vaccine and application thereof
Yu et al. Current status and perspective of tumor immunotherapy for head and neck squamous cell carcinoma
AU2022200872B2 (en) Immunogenic compounds for cancer therapy
Ostrand‐Rosenberg et al. Expression of MHC class II and B7–1 and B7–2 costimulatory molecules accompanies tumor rejection and reduces the metastatic potential of tumor cells
Fournier et al. Targeting of IL-2 and GM-CSF immunocytokines to a tumor vaccine leads to increased anti-tumor activity
WO1995016775A1 (en) Tumor cell fusions and methods for use of such tumor cell fusions
AU2011252699B2 (en) The N-domain of carcinoembryonic antigen and compositions, methods and uses thereof
Danishmalik et al. Tumor regression is mediated via the induction of HER263-71-specific CD8+ CTL activity in a 4T1. 2/HER2 tumor model: no involvement of CD80 in tumor control
KR101040281B1 (en) Vaccine compositions comprising TAT-fusion Antigen protein and adjuvants
JP2023554154A (en) Cytokine protein treatment schedule
AU2022379949A1 (en) Methods and materials for treating cancer
CN115135335A (en) In vitro and in vivo gene delivery to immune effector cells using nanoparticles functionalized with Designed Ankyrin Repeat Proteins (DARPIN)
CN117715655A (en) Substances and methods for activating and targeting immune effector cells
JP2009108017A (en) Antitumor vaccine
Danishmalik et al. Tumor regression is mediated via the induction of HER2
Ianzano et al. Virus-like particle display of HER2 induces potent anti-cancer responses

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: 13743997

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 14374729

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2863658

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013743997

Country of ref document: EP