WO2017177204A1 - Leveraging immune memory from common childhood vaccines to fight disease - Google Patents

Leveraging immune memory from common childhood vaccines to fight disease Download PDF

Info

Publication number
WO2017177204A1
WO2017177204A1 PCT/US2017/026719 US2017026719W WO2017177204A1 WO 2017177204 A1 WO2017177204 A1 WO 2017177204A1 US 2017026719 W US2017026719 W US 2017026719W WO 2017177204 A1 WO2017177204 A1 WO 2017177204A1
Authority
WO
WIPO (PCT)
Prior art keywords
varicella
orf22
zoster virus
strain oka
cancer
Prior art date
Application number
PCT/US2017/026719
Other languages
French (fr)
Inventor
Patrick Ho
Ravi KOLLA
Original Assignee
La Jolla Institute For Allergy And Immunology
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2016/068147 external-priority patent/WO2017112830A1/en
Application filed by La Jolla Institute For Allergy And Immunology filed Critical La Jolla Institute For Allergy And Immunology
Publication of WO2017177204A1 publication Critical patent/WO2017177204A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • 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
    • A61K39/001111Immunoglobulin superfamily
    • A61K39/001112CD19 or B4
    • 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
    • A61K39/001103Receptors for growth factors
    • A61K39/001104Epidermal growth factor receptors [EGFR]
    • 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
    • A61K39/001103Receptors for growth factors
    • A61K39/001109Vascular endothelial growth factor receptors [VEGFR]
    • 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
    • A61K39/001122Ephrin Receptors [Eph]
    • 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
    • A61K39/00113Growth factors
    • A61K39/001135Vascular endothelial growth factor [VEGF]
    • 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
    • A61K39/001136Cytokines
    • A61K39/00114Interleukins [IL]
    • 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
    • A61K39/001166Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • 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
    • A61K39/001171Gangliosides, e.g. GM2, GD2 or GD3
    • 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
    • A61K39/001192Glycoprotein 100 [Gp100]
    • 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
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • 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
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to the field of disease treatment.
  • the present invention relates to the implementation of treatments for disease, from the approach of targeting pre-existing human immunity as a vehicle to specifically target and destroy target cells.
  • Vaccines provide a mechanism to promote safe development of long-lived immune memory against a pathogen(s) by avoiding deleterious effects of actual pathogen infection.
  • a number of vaccines are routinely administered to children to prevent the contraction of debilitating infectious diseases and immune memory from these vaccines can be measured decades following administration.
  • T cells including CDS and CD4 cytotoxic T cells
  • CDS and CD4 cytotoxic T cells play a key role in the defense against intracellular pathogens and tumor cells.
  • T-cell immune responses are driven by the recognition of foreign peptides presented by major histocompatibility complex class I and class II (MHC I and MHC II) molecules at the cell surface.
  • MHC I and MHC II major histocompatibility complex class II
  • T-cell epitopes is therefore important for understanding disease pathogenesis and etiology as well as for vaccine design.
  • T-cell response has been implicated in providing vaccine-mediated protection. However few T- cell epitopes from vaccines have been identified.
  • the upregulated state translates into the body's own immune system recognizing its own cells as foreign and begins launching an attack on its own healthy cells.
  • a further problem with existing therapies is the risk associated with tissues beginning to build tolerance to the administered therapy whereby mutations enable cancer cells to bypass the therapeutic agent and proliferate, and they not only become unresponsive to the treatment but potentially further create an environment whereby no other alternative therapies are available.
  • a method of treating cancer comprising, a) providing a subject having or suspected of having a cancer and b) administering a vector (e.g., viral vector such as adenovirus, vaccinia or plasmid DNA) to the subject, wherein the vector directs expression of one or more peptide antigens on a target cell of the subject and wherein a memory or recall immune response is elicited against the one or more peptide antigens.
  • a vector e.g., viral vector such as adenovirus, vaccinia or plasmid DNA
  • a method of treating cancer comprising a) providing a subject having or suspected of having a cancer; and b) administering a composition to the subject, wherein the composition comprises one or more peptide antigen, wherein a memory or recall immune response is elicited against the one or more peptide antigens.
  • the subject prior to the administering of (b), the subject was vaccinated against a pathogen, non-limiting examples of which include a vaccination against chickenpox, measles, mumps, rubella, shingles/varicella zoster or polio.
  • a subject is vaccinated with another suitable live-attenuated or inactivated pathogen, or pathogen antigen(s).
  • the subject prior to the administering of (b), the subject was vaccinated with at least one of the one or more peptides selected from Table 1, or one or more other antigens derived from a pathogen.
  • the subject may have been vaccinated or re-vaccinated 1 day, 1 month, 6 months, or 1 year to 100 years prior to administering of (b).
  • the administering of (b) elicits a recall immune response in the subject, wherein the recall immune response is directed against the target cell and/or at least one of the one or more peptide antigens.
  • the one or more peptide antigens bind to MHC class I or MHC class II and can recall CD8 T-cell and/or CD4 T-cell responses.
  • the subject is further administered an anti-cancer therapy.
  • the anti-cancer therapy may be administered prior to, concurrently with and/or after the administering of vector or peptides of (b).
  • the anti -cancer therapy can be any suitable therapy.
  • the anti-cancer therapy comprises surgical resection.
  • the anti-cancer therapy comprises a radiation therapy, chemotherapy or an immunotherapy.
  • a radiation therapy comprises external beam radiation, proton therapy, or internal radiation therapy.
  • an immunotherapy comprises chimeric antigen receptor (CAR)-T immunotherapy.
  • CAR-T immunotherapy sometimes comprises administering CAR T-cells configured to specifically bind to and kill the target cell.
  • an immunotherapy comprises administering an antibody or antibody fragment that specifically binds to a cell-surface antigen, non-limiting examples of which include CD20, CD30, CD33, CD52, CD 138, CD274, CD279, PD-L1, CTLA-4, programmed cell death 1 (PD-1), CD33, EGFR, ERBB2 (CD340), ERBB3, VEGF, VEGFR, GM-2, gp-100, EPCAM, CD19, CD3, CEA, gpA33, mucins, TAG-72, CAIX, PSMA, folate-binding protein, GD2, GD3, Integrin aVp3, Integrin ⁇ 5 ⁇ 1, IGF1R, EPHA3, TRAILR1, TRAILR2, RA KL, FAP or Tenascin.
  • a cell-surface antigen non-limiting examples of which include CD20, CD30, CD33, CD52, CD 138, CD274, CD279, PD-L1, CTLA-4, programmed cell
  • the antibody may be bi-specific and may bind to two cell surface antigens.
  • the immunotherapy comprises administering an antibody or T-cell that specifically binds a checkpoint blocker.
  • an immunotherapy comprises administering an antibody or antibody fragment that specifically binds to a tumor marker.
  • a peptide antigen described herein is derived from a pathogen (e.g., a peptide or protein expressed by a pathogen).
  • the one or more peptide antigens are selected from a peptide listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • a target cell is a cell that is or has been infected with a pathogen.
  • a pathogen many types of cancers are induced or caused by a previous infection by a pathogen.
  • a target cell may be a cell that is or has been infected with a virus, bacteria, parasite or protozoa.
  • a target cell is not infected with a pathogen.
  • a pathogen can be a human pathogen and may be a virus, fungus, bacteria, parasite or protozoa.
  • a pathogen is a non-human pathogen, or the peptide antigen is conserved between human and another species, or wherein the peptide antigen cross-reacts between human and another species.
  • a pathogen is a virus, non-limiting examples of which include a virus of the order
  • a target cell is a cancer cell or neoplastic cell.
  • a target cell is a metastatic cancerous cell.
  • a cancer cell, metastatic cancerous cell or neoplastic cell include cells of a carcinoma, sarcoma, neuroblastoma, cervical cancer cell, hepatocellular cancer cell, bladder cancer cell, mesothelioma, glioblastoma, myeloma, lymphoma, neoplastic lymphocyte, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, lymphosarcoma, liposarcoma, osteosarcoma, chondrosarcoma, leiomyosarcoma, rhabdomyosarcoma or fibrosarcoma.
  • a subject is a mammal. In some aspects a subject is a non- human mammal. In some aspects, a subject is a human. In some embodiments, wherein prior to the administering of a vector or peptide of (b), a subject was infected with the pathogen, thereby inducing, eliciting or promoting a recall immune response in the subject to certain pathogen antigens.
  • a subject has, or is suspected of having a cancer.
  • a cancer include a hematological and solid tumors, a neoplasia derived from lung, thyroid, head or neck, nasopharynx, throat, nose, sinus, brain, spine, breast, adrenal gland, pituitary gland, thyroid, lymph, mouth, esophagus, stomach, duodenum, ileum, jejunum intestine, colon, rectum, uterus, urethra, ovary, cervix, endometrium, bladder, testicle, penis, prostate, kidney, pancreas, liver, bone, bone marrow, lymph, blood, muscle, or skin.
  • a cancer examples include a carcinoma, sarcoma, neuroblastoma, hepatocellular cancer, mesothelioma, glioblastoma, myeloma, lymphoma, myeloma, leukemia, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, cervical cancer, breast cancer, liver cancer, pancreatic cancer, brain cancer, lung cancer, skin cancer, ovarian cancer, and testicular cancer.
  • a cancer is an infection-induced cancer non-limiting examples of which include a hepatocellular carcinoma, cervical cancer, head and neck squamous cell carcinoma, adult T-cell leukemia/lymphoma, Burkitt lymphoma, Merkel cell carcinoma, kaposi sarcoma, bladder cancer and gastric cancer.
  • a cancer comprises a low mutational burden.
  • Non-limiting examples of a cancer comprising a low mutational burden include pilocytic astrocytoma, ALL, medulloblastoma, kidney chromophobe, thyroid, CLL, neuroblastoma, glioblastoma, and pancreatic cancer.
  • a method herein comprises administering a vector that
  • a vector directs expression of the one or more peptide antigens on the cell surface of the target cell.
  • the one or more peptide antigens are expressed or presented on the cell surface of the target cell. Presentation of a peptide antigen on the surface of a cell may include co-expression of a peptide in the presence of TAP (transporter associated with antigen processing).
  • a vector comprises a nucleic acid encoding one or more non-self-peptide antigens (e.g., one or more of the antigens of Table 1).
  • a vector comprises a promoter operatively linked to a nucleic acid encoding the one or more peptides.
  • a promoter of a vector is sometimes inducible by a factor, for example a transcription factor derived from a pathogen, a mutated human protein, and/or one or more viral proteins.
  • the promoter is selectively induced in tumor cells.
  • a factor is a viral transcription factor or a factor that is selectively expressed in tumor cells.
  • the administering of a vector or protein of (b) comprises administering nano-carriers, micro-carriers, microparticles, nanoparticles, liposomes, exosomes or recombinant virus to the subject, where the microparticles, nanoparticles, liposomes, exosomes or recombinant virus may comprise a vector or one or more peptides.
  • the administering of a vector or protein of (b) comprises selectively delivering nano-carriers, micro-carriers, microparticles, nanoparticles, liposomes, exosomes or recombinant virus to a target cell.
  • the administering of a vector of (b) comprises administering a nucleic acid to the subject, wherein the nucleic acid comprises the vector.
  • a vector include a plasmid, a linear nucleic acid molecule or a replicating or non-replicating viral vector.
  • a vector comprises a CRISPR element, and/or the vector is configured to express (CRISPR-associated protein-9 nuclease) CAS9.
  • a vector is integrated into the target cell by a method comprising CRISPR and (CRISPR-associated protein-9 nuclease) CAS9.
  • the administering comprises direct intra-tumoral injection. In some aspects, the administering comprises intravenous injection, intraperitoneal injection, subcutaneous injection, intramuscular injection, intravesical or intrathecal injection.
  • the method further comprises c) initiating, eliciting, promoting, or enhancing an immune response in the subject, wherein the immune response is directed against the target cell and/or target antigen.
  • the eliciting of (c) comprises initiating, eliciting, promoting or enhancing a recall response in the subject to the one or more peptide antigens.
  • the eliciting of (c) comprises administering a vaccine to the subject.
  • a vaccine is selected from a vaccine for chickenpox, measles, mumps, rubella, and polio.
  • the eliciting of (c) comprises administering a pharmaceutical composition to the subject, wherein the pharmaceutical composition comprises at least one of the one or more of the peptide antigens.
  • the pharmaceutical composition comprises an adjuvant.
  • the eliciting of (c) comprises administering an agent that modulates an immune response in the subject.
  • the agent increases, enhances, promotes or elicits an innate immune response.
  • the agent comprises an immune checkpoint blocker.
  • the agent increases, enhances, promotes or elicits a recall immune response.
  • the agent comprises an antibody.
  • the antibody specifically binds to the one or more peptide antigens.
  • the agent comprises a nucleic acid, synthetic chemical, or small molecule.
  • a method herein further comprising administering a
  • composition herein comprises an adjuvant, exosomes, microparticles, nanoparticles, and/or liposomes.
  • the target cell wherein TAP promotes peptide epitope:MHC expression.
  • Other peptide transporter can be provided such as the antennapedia homeodomain peptide to promote peptide:MHC binding or expression.
  • a composition is a pharmaceutical composition or pharmaceutical formulation comprising one or more peptides (e.g., a peptide listed in Table 1).
  • a composition (e.g., a pharmaceutical composition) comprises a vector configured to direct expression of one or more peptide antigens on a target cell of a subject, wherein the one or more peptide antigens is selected from a peptide listed in Table 1.
  • the present invention provides methods and compositions for eliciting an immune response against target cells.
  • known or predicted antigens from vaccines or other developed immunity are either engineered to or serve as tags for the targeted cells so as to provide a targeted identification method for the target cells that are recognized by the subjects existing immune system, inducing the subjects immune system to attack the tagged or target cells.
  • the present invention provides a targeted treatment for patients diagnosed with cancer.
  • the present invention is a preventative treatment for
  • At risk patients may be determined through various methods known in the art or in the future including but not limited to genetic predispositions and hereditary traits.
  • the treatment is administered as a primary treatment or in conjunction with other treatments well known in the art.
  • the disease target cells can be administered a factor, for example but not limited to a protein or variant of a protein, which is not present in healthy cells through restricted delivery to a specific tissue.
  • the target cells have a mutation not present in healthy cells that results in expression of factor, for example but not limited to a protein or variant of a protein, which is not present in healthy cells.
  • the expression of a factor that is unique to or predominantly specific to the disease cell enables expression of the antigen from a gene construct.
  • antigens are specifically expressed in target cells through administration of a gene construct encoding an antigen wherein expression or presentation of the antigen requires a factor (e.g., protein, gene, S P, transcription factor) selectively expressed in the target cells.
  • a factor e.g., protein, gene, S P, transcription factor
  • the gene construct contains the antigen with a promoter driven only by a factor selectively expressed in the target cells (e.g., a cancer cell-specific transcription factor).
  • the expression of transformed viral genes in target cells enables the selective expression of proteins or peptides in these cells.
  • a gene construct containing an antigen with a promoter driven by factors (e.g., proteins, peptides) expressed only in the virally -transformed cells provides for specific expression of the antigen only in the target cell and therefore provides labeling of the target cells (e.g., a cancer cell) with the antigen. This results in activation of pre-existing memory immune responses against the antigen providing a targeted immune response against the target cells to treat the cancer.
  • the methods of the present invention can be used to target pre-, active and metastatic cancerous cells.
  • cancerous cells enables the selective synthesis of proteins or peptides in these cells that are necessary for expression of the antigen from the gene construct.
  • the method involves administration of a gene construct containing the antigen driven by a viral transformed promoter that requires transformed viral genes found in the cancerous cell for expression of the antigen.
  • peptide antigens disclosed herein in cancers of low mutational burden or that are not sensitive to immune-based therapies such as, but not limited to, treatment with checkpoint inhibitors or CAR-T is contemplated.
  • Expression of peptide antigens disclosed herein in combination with other immune-based therapies in such cancers represents a novel utility to make these cancers immune sensitive.
  • delivery of peptide antigens disclosed herein to cancers with low potential for neoantigen/epitope development will promote the immunogenic potential of said cancer.
  • administration of peptide antigens herein in combination with any of the delivery platforms described herein will make said cancer reactive to other immune therapies such as checkpoint inhibitors and CAR-Ts.
  • the antigen to be selectively expressed are independently expressed and are independently expressed.
  • the presented by the target cells is an antigen from a vaccine including but not limited to childhood vaccines, e.g., chickenpox, measles, mumps, rubella, and polio.
  • the antigen to be selectively expressed and presented by the target cells is a subsequence, homologue, variant or derivative of an antigen from a vaccine.
  • the present inventors have identified peptides derived from vaccine strains of viruses used in live-attenuated vaccines and inactivated vaccine administered during child- and adult- hood including chickenpox, measles, mumps, rubella, and polio that are broadly bound by HLA class I and class II molecules represented in humans from diverse ethnic, racial, and geographic backgrounds that can be used to recall memory T-cell responses.
  • the herein described approach allows expression of peptides derived from chickenpox, measles, mumps, rubella and polio for which common childhood vaccines have been administered for the past five decades in target cells to allow targeting of memory immune responses from these common vaccines.
  • This approach is novel as it leverages existing, non-self memory immune responses to target T-cell responses against specific target cells including cancerous, transformed pre-cancerous cells, and metastasis as well as viral, parasitic and bacterial transformed cells.
  • compositions that comprise one or more peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof are directly administered to the cancer tissue of the patient.
  • the composition can be administered by for example but not limited to viral vectors, micro- or nano- particles, direct injection, lipid particles, exosomes, gene, and/or adjuvanted delivery methods.
  • a composition described herein is administered by direct tissue injection, intratumoral injection, intravenous injection, intraperitoneal injection, subcutaneous injection, intramuscular injection, intravesivular injection or intrathecal injection.
  • a subject is vaccinated prior to, concurrently with, or after the administering of (b).
  • a vaccination is a booster vaccination.
  • the booster vaccination comprises at least one peptide listed in Tables 1.
  • the booster vaccination is a booster vaccination against chickenpox, measles, mumps, rubella, or polio.
  • an anti-cancer therapy comprises administering
  • an anti-cancer therapy comprises administering, alone or in combination, one or more cancer vaccines optionally based on cancer-associated tumor antigens, or neo-antigens.
  • the one or more of the one or more peptide antigens are linked to a targeting antibody, antibody fragment or nanobody that is internalized by the target cell.
  • a pre-existing T-cell memory is measured prior to the administering of (b). In certain embodiments, such measurement of pre-existing T cell memory can be done to identify subjects for treatment or to determine appropriate treatment for a subject (e.g. companion diagnostic).
  • the method comprises boosting immune memory in the subject by administering a vaccines or antigen that stimulates a recall immune response against the one or more peptide antigens.
  • a diagnostic tool for measuring immune response to a vaccine or recall immunity to a target cell comprising measuring an immune response to one or more peptides listed Table 1.
  • the measurement of recall immunity is prior to administration of a peptide listed in any of Table 1.
  • the measurement of recall immunity is prior to administration of a vector configured to expresses at least one peptide listed in any of Table 1.
  • CD8/CD4 CD8/CD4
  • childhood vaccines leads to tumor cell killing and optionally a subsequent 'abscopal' effect with systemic, cancer-specific immune responses.
  • the one or more peptide antigens comprise full antigens or complete proteins derived from the pathogens.
  • a subject is vaccinated prior to,
  • the vaccination is a booster vaccination.
  • the booster vaccination comprises at least one peptide listed in Table 1.
  • the booster vaccination is a booster vaccination against chickenpox, measles, mumps, rubella, or polio.
  • An "associated symptom, disease or disorder” as used herein refers to a symptom, disease or disorder that develops in conjunction with, following or as a result of a disease state ⁇ e.g., cancer). This includes acute and chronic diseases or disorders, short and long term disease and disorders, as well as related symptoms thereof.
  • a subject is a mammal.
  • mammals include humans, non-human primates ⁇ e.g., apes, gibbons, chimpanzees, orangutans, monkeys, macaques, and the like), domestic animals (e.g., dogs and cats), farm animals (e.g., horses, cows, goats, sheep, pigs) and experimental animals (e.g., mouse, rat, rabbit, guinea pig).
  • a subject is a non-human mammal.
  • a subject is a rodent (e.g., a rat or mouse).
  • a subject is a human.
  • diagnosis refers to a relative probability that a disease or condition (e.g. cancer) is present in the subject. In some instances, as will be appreciated by any one of skill in the field of medicine, a diagnosis may not be absolute or definite.
  • virus bacteria
  • oncovirus bacteria
  • cancer virus bacteria
  • tumor virus bacteria
  • tumor virus bacteria
  • fung virus fungal virus
  • tumor virus a class of triggers that are associated with the development of or are the cause of a cancer, either currently known or yet to be confirmed or discovered.
  • the cause can be related to the insertion of viral or bacterial DNA or RNA into the genome of the target cell.
  • Vaccination regimes means vaccines, for example but not limited to MMR, oral polio vaccine, injectable polio vaccine, and chicken pox including those administered during childhood or adulthood.
  • recall immune response means an immune response to an antigen to which a subject has previously been exposed.
  • a recall or memory response is therefore an immune response subsequent to the initial antigen exposure and immune response.
  • a recall response may occur following exposure of the subject to the antigen for a second (secondary), third (tertiary), fourth, fifth, sixth, seventh, eighth, ninth, tenth, or any subsequent antigen exposure.
  • a recall or memory response is distinguished from a primary response to an antigen; a primary response is an immune response that occurs when a subject is exposed to an antigen for the first time. In a primary response, naive B and T cells become activated, develop higher specificity and affinity, expand, and develop memory cells. In contrast, recall immune responses are believed to be attributed to reactivation of long- lived, antigen-primed B and T lymphocytes that arise from differentiated B and T cells in a quiescent state. Thus, a "recall response” is an immune response in which antigen- primed cytotoxic T cell, Thl, Th2, and/or B cells participate.
  • target cell refers to cells, which are affected by a disease or disorder, contribute to a disease or disorder, predisposed to transform into a disease or disorder and/or are targets of treatment.
  • Target cells are distinguished from healthy or non-disease cells in that healthy or non-disease cells have normal function or are not desired targets of treatment.
  • Non-limiting examples of target cells comprise cells, tissue or derivatives thereof from cancer or any other desired targeted cells subset.
  • Target cells may include cancer cells that have been transformed by a virus or bacteria, including but not limited to a Burkitt's lymphoma cancer cell, nasopharyngeal cancer cell, Hodgkin lymphoma cancer cell and stomach cancer cell (Epstein Barr Virus), Kaposi sarcoma cancer cell (human herpes vims 8); cervical cancer cell, penis cancer cell, anus cancer cell, vagina cancer cell, vulva cancer cell, throat and mouth cancer cell (human papillomavirus), liver cancer ceil (hepatitis B and C viruses), breast cancer cell or gliobastoma multiform ceil (human cytomegalovirus), T cell leukemia cell and lymphoma cell (Human T-lymphotrophic virus 1), mesothelioma cell, brain cancer cell, bone cancer cell and lymphomas (Simian vims 40), stomach cancer cells (Helicobacter pylori and merkel cell cancer cell (Merkel cell poSyo
  • a target cell is a cancer cell or neoplastic cell.
  • a target cell is a metastatic cancerous cell.
  • Non-limiting examples of a cancer cell or neoplastic cell include a carcinoma, sarcoma, neuroblastoma, cervical cancer cell, hepatocellular cancer cell, mesothelioma, glioblastoma, myeloma, lymphoma, neoplastic lymphocyte, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, lymphosarcoma, liposarcoma, osteosarcoma, chondrosarcoma, leiomyosarcoma, rhabdomyosarcoma or fibrosarcoma.
  • a target cell is a cell that is or has been infected with a pathogen. In certain embodiments, a target cell is a cell that is or has been infected with a vims, bacteria, parasite or protozoa.
  • a pathogen can be a mammalian pathogen. In some embodiments a pathogen is a human pathogen. A pathogen can be a virus, bacteria, parasite or protozoa.
  • Non- limiting examples of a pathogen include a virus of the order Flaviviridae, Retroviridae, Orthomyxoviridae, Arenaviridae, or Mononegavirales, a virus of the family Togaviridae, Reoviridae, picornaviridae, herpesviridae or Bunyaviridae.
  • a virus is a human papillomavirus.
  • a virus is a hepatitis B or hepatitis C virus.
  • a virus is a Merkel cell polyomavirus.
  • compositions of the invention are administered to a subject or patient as part of a treatment described herein.
  • Delivery mechanisms refer to mechanisms (chemical, biological, physical and otherwise) that are utilized to introduce a package into the patient's system and cells.
  • the present invention is not limited to the delivery mechanisms disclosed herein and one skilled in the art would appreciate that other mechanisms known in the art, as a whole, in part and in combination, can be used to deliver the packages to the target and desired cells.
  • treatment may refer to any delay in onset, reduction in the frequency or severity of symptoms, amelioration of symptoms, improvement in patient comfort or function, decrease in the occurrence of a given condition or disease or condition or disease symptoms in a patient, decrease in severity of the condition or disease state etc.
  • the effect of treatment can be compared to an individual or pool of individuals not receiving a given treatment, or to the same patient prior to, or after cessation of, treatment.
  • treatment may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.
  • antigen refers to any protein, peptide, biological component, molecule or compound that induces an immune response.
  • epipe refers to the specific region of the antigen that elicits the immune response.
  • peptide and protein refers to polymers of amino acid residues. The terms include amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • a peptide or protein described herein includes an epitope.
  • Modified and variant proteins or peptides may therefore have greater or less activity or function than a reference protein or peptide but at least retain partial activity or function of the reference protein or peptide.
  • Constantly modified variants applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein, which encodes a polypeptide, also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
  • TGG which is ordinarily the only codon for tryptophan
  • substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid.
  • the following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
  • a "label” or a “detectable moiety” is a composition detectable by
  • useful labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and proteins or other entities, which can be made detectable, e.g., by incorporating a radiolabel into a peptide or antibody specifically reactive with a target peptide. Any method known in the art for conjugating an antibody to the label may be employed, e.g., using methods described in Hermanson, Bioconjugate Techniques 1996, Academic Press, Inc., San Diego.
  • recombinant when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
  • recombinant cells express genes that are not found within the native (non- recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
  • heterologous when used with reference to portions of a nucleic acid indicates that the nucleic acid comprises two or more subsequences that are not found in the same relationship to each other in nature.
  • the nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged to make a new functional nucleic acid, e.g., a promoter from one source and a coding region from another source.
  • a heterologous protein indicates that the protein comprises two or more subsequences that are not found in the same relationship to each other in nature (e.g., a fusion protein).
  • treatment may refer to any delay in onset, reduction in the frequency or severity of symptoms, amelioration of symptoms, improvement in patient comfort or function, decrease in the occurrence of a given condition or disease or condition or disease symptoms in a patient, decrease in severity of the condition or disease state etc.
  • the effect of treatment can be compared to an individual or pool of individuals not receiving a given treatment, or to the same patient prior to, or after cessation of, treatment.
  • treatment may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.
  • an "agent” as used herein refers to any molecule (e.g., antibody, nucleic acid, synthetic chemical, small chemical molecule, ligand mimetic) that can be administered to a subject.
  • An agent that modulates an immune response is a molecule (e.g., antibody, nucleic acid, synthetic chemical, small chemical molecule) that when administered to a subject modulates an immune response in the subject.
  • the agent modulates, inhibits, blocks, decreases, increases, enhances, promotes or elicits an acute or chronic immune response.
  • the agent modulates stimulation, activation proliferation, number or activity of immune proteins or cells (e.g., T cells, B cells, macrophages, monocytes, dendritic cells, cytokines).
  • the agent modulates an innate immune response.
  • the agent modulates a recall immune response.
  • the agent modulates an immune response to one or more of the peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • an agent is an agent that binds one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the agent binds one or more of the proteins listed in
  • Table 1 or a subsequence, homologue, variant or derivative thereof.
  • the agent binds the complex of an immune cell or immune protein bound to one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the agent modulates the activity or expression of one or more type of immune cell or protein (e.g., T cells, B cells, macrophages, monocytes, dendritic cells, cytokines) or a combination thereof.
  • the agent modulates the stimulation of T cells, activation of T cells, proliferation of T cells, number of T cells or activity of T cells, or a combination thereof.
  • the agent modulates the stimulation of B cells, activation of B cells, proliferation of B cells, number of B cells or activity of B cells, or a combination thereof.
  • the agent modulates stimulation, activation, proliferation, number or activity, or a combination thereof, of B cells generating antibodies (e.g., antibodies to one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof).
  • antibodies e.g., antibodies to one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the agent modulates production or activity of
  • cytokines cytokines, immune activators, chemokines, growth factors and T F family members
  • AREG Granzyme, Histamine, IFNct/ ⁇ , IFNy, IL12, IL15, IL16, IL17a-f, ⁇ .1 ⁇ , 112, IL21, IL22, IL23, IL25, IL27, GM-CSF, IL33, IL4, IL5, IL6, IL7, IL8, IL9, LFA-1, LIF, lymphotoxin, Mac-1, MCP-1, ⁇ , Perforin, TGF- ⁇ , TNF a, VLA-4, CCL1, CCL1 1, CLL17, CCL1, CCL20, CCL22, CCL3, CCL4, CCL5, CXCL10, CXCL13, CXCL8, RANTES, CXCL12, G-CSF, M-CSF, VEGF, PDGF, 4-1BB, APRIL, BAFF, CD27,
  • the agent modulates activity or expression of immune cells or proteins involved in an immune response to one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the immune response is an innate immune response. In other embodiments, the immune response is a recall immune response.
  • the agent modulates stimulation of T cells, activation of T cells, proliferation of T cells, number of T cells or activity of T cells, or a combination thereof, wherein the T cells are specific for one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the agent modulates stimulation of B cells, activation of B cells, proliferation of B cells, number of B cells or activity of B cells, or a combination thereof, wherein the B cells are specific for one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the method involves selectively expressing in the target cells (e.g., cancer cells, viral, bacterial, parasite or protozoa infected cells) an antigen that the subject has previously developed immunity against.
  • the method involves selectively expressing in target cells an antigen from a vaccine that the subject has previously received resulting in a recall immune response specific to the target cell.
  • the antigen is from a vaccine the subject received in childhood including but not limited to one or more of the vaccines for chickenpox, measles, mumps, rubella, and polio.
  • the expression of particular antigens that are non- self immunogenic peptide antigens results in a signature labeling of the target cells with non-self immunogenic peptide antigens.
  • the signature labeling is providing a means for the target cells to be singled out and differentiated from healthy ceils that are not expressing the particular antigens.
  • the expression of non-self immunogenic peptide antigens elicits a memory immune response against the target ceils that are expressing the particular antigens.
  • memory T cells e.g., CDS and/or CD4 T cells, specific to the non-self immunogenic peptides are reactivated.
  • the treatment comprises an antigen from a vaccine that is administered to a patient who has previously developed immunity against the antigen, resulting for example from childhood vaccination regimes, where the vaccine was effective in developing immunity against the disease for which the vaccine was designed.
  • the patient has been vaccinated against a disease and has developed immunity to these infections such that immunity can be detected, boosted and measured.
  • the patient received vaccination against the disease during childhood.
  • the patient has been previously infected with a virus, bacteria, parasite or protozoa expressing the antigen and has developed immunity to the antigen due to this prior infection.
  • a subject has been previously vaccinated against a pathogen.
  • the developed immunity is against chickenpox, measles, mumps, rubella and polio.
  • the subject' s immune system is able to recall an immune response developed from a previous vaccination regime or infections.
  • the subject may be administered a booster vaccination to prime immune memory (e.g., increase antibody titers, memory T cells) prior to administration of a method or composition of the present invention.
  • immune memory e.g., increase antibody titers, memory T cells
  • immunity may be induced using other vaccination methods other than childhood vaccines.
  • the antigen is selectively expressed in the target cells through administration of a gene construct encoding an antigen wherein expression or presentation of the antigen requires a factor (e.g., protein, gene, S P, transcription factor) selectively expressed in the target cells.
  • a factor e.g., protein, gene, S P, transcription factor
  • the gene construct contains the antigen with a promoter driven only by a factor selectively expressed in the target cells (e.g., a cancer cell-specific transcription factor).
  • this specific expression of the necessary factor in the target cells is a result of the target cells being infected by a virus, bacteria, parasite or protozoa such that there is expression of transformed viral genes or bacterial, parasitic or protozoa genes present in the target cells which encode for the necessary factors to express the antigen from the gene construct.
  • the gene construct comprises the antigen with a
  • a gene construct that contains a promoter driven only by a factor expressed by target cells that have been infected with a particular virus, bacteria, protozoa or parasite is
  • the subject administered to the subject, enters or is ingested by the subject cells and the viral, bacterial, parasitic or protozoa genes in the target cells enables the selective expression of proteins or peptides in these cells that are needed for expression of the antigen from the gene construct. Only the target cells that contain the viral, bacterial, parasitic or protozoa genes will express the antigen resulting in a recall immune response specific to the target cells.
  • the antigen is specifically expressed in target cells by- targeted delivery of the antigen to the target cells.
  • methods for targeted delivery of the antigen to the target cells including administration by direct intratumoral injection, micro- or nano-particles, liposome, PEGylation, lipofectamine, adenovirus, lentivirus, vaccinia virus or other viral gene vector, and/or CRISPR/CAS9.
  • the present invention comprise administering an optimized package comprising one or more antigens (e.g., peptide antigens).
  • antigens e.g., peptide antigens
  • peptide antigens include a peptide selected from Table 1, or a subsequence,
  • the optimized package included one or more epitopes from antigen against which the subject has immune memory, e.g., epitopes from childhood vaccines.
  • the optimized package compri ses one or more CD4 and/or CDS T- cell epitopes, for example, one or more of the epitopes listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the optimized package contains epitopes selected for their broad recognition by HLA class I molecules of subjects from diverse ethnic and geographical backgrounds.
  • the method includes administering the antigen and epitopes in combination with factors that induce an increase of epitope presentation by MHC molecules (e.g., TAP expression; antennapedia homeodomain peptide) and/or activation and recruitment of APCs (i.e. GM-CSF, anti-immune suppressive agents, or chemokines).
  • MHC molecules e.g., TAP expression; antennapedia homeodomain peptide
  • APCs i.e. GM-CSF, anti-immune suppressive agents, or chemokines
  • the present invention provides a gene construct encoding the antigen and genes for increased expression of cytokines, immune activators, chemokines, growth factors and TNF family members to amplify the inflammatory T cell response by the recalled immune system.
  • the gene constructs comprises genes for one or more of AREG, Granzyme, Histamine, IFNa/ ⁇ , ⁇ , IL12, IL15, IL16, IL17a-f, ILlb, 112, IL21, IL22, IL23, IL25, IL27, GM-CSF, IL33, IL4, IL5, IL6, IL7, IL8, IL9, LFA-1, LIF, lymphotoxin, Mac-1, MCP-1, ⁇ , Perforin, TGF- ⁇ , TNFa, VLA-4, CCL1, CCL1 1, CLL17, CCL1, CCL20, CCL22, CCL3, CCL4, CCL5, CXCL10, CXCL13, CXCL8, RANTES, CXCL12, G-CSF, M-CSF, VEGF, PDGF, 4-1BB, APRIL, BAFF, CD27, CD30, CD30L, CD70, DR3, DR4,
  • the treatment is directed towards hematological and solid tumors and cancer, including but not limited to tumors and cancers involving or caused by viral, parasitic or bacterial infection that can be targeted by local administration or tissue-targeting agents, or cancers of low mutational burden where reduced efficacy is observed following other cancer immunotherapies.
  • Non-limiting examples of cancers that may be treated by the present invention include a neoplasia derived from lung, thyroid, head or neck, nasopharynx, throat, nose, sinus, brain, spine, breast, adrenal gland, pituitary gland, thyroid, lymph, mouth, esophagus, stomach, duodenum, ileum, jejunum intestine, colon, rectum, uterus, urethra, ovary, cervix, endometrium, bladder, testicle, penis, prostate, kidney, pancreas, liver, bone, bone marrow, lymph, blood, muscle, and skin; a carcinoma, sarcoma, neuroblastoma, hepatocellular cancer, mesothelioma, glioblastoma, myeloma, lymphoma, myeloma, leukemia, adenoma, adenocarcinoma, glioma, glioblastoma
  • a cancer is an infection-induced cancer.
  • infection-induced cancers include a hepatocellular carcinoma, cervical cancer (human papilloniaviixis), head and neck squamous cell carcinoma (HNSCC), adult T-cell leukemia/lymphoma, liver cancer (hepatitis B and C viruses), Burkitt' s lymphoma and Kaposi sarcoma (Epstein-Barr virus), Merkel cell cancer (e.g., Merkel cell carcinoma) (Merkel cell polyomavirus), bladder cancer and gastric cancer.
  • a cancer comprises a low mutational burden, non- limiting examples of which include pilocytic astrocytoma, ALL, medulloblastoma, kidney chromophobe, thyroid, CLL, neuroblastoma, glioblastoma, and pancreatic cancer.
  • a method comprises administering a checkpoint blocker. In certain embodiments, a method comprises adoptive transfer of T-cells containing a chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • the present invention provides epitopes from childhood vaccines that can be administered in the methods described herein.
  • compositions comprising proteins and peptides comprising epitopes from childhood vaccines, including but not limited to chickenpox, measles, mumps, rubella, and polio.
  • the protein or peptide comprises, consists or consists essentially of one or more of the amino acid sequences listed in Table 1, or a
  • the present invention provides a composition
  • the present invention provides combinations of one or more proteins or peptides each comprising, consisting or consisting essentially of a different amino acid sequence listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • the present invention provides a composition
  • a composition comprising a gene construct encoding one or more epitopes from childhood vaccines.
  • the present invention provides a gene construct encoding one or more of the amino acid sequences listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • a composition comprises a viral construct configured to express one or more of the amino acid sequences listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
  • a composition comprises agents to improve peptide:HLA expression (e.g., TAP;
  • a composition comprises agents to improve immune infiltration. In certain embodiments a composition comprises agents to modulate checkpoint inhibition. In certain embodiments a composition comprises T cells that are adoptively transferred containing CAR specific to one or more of the peptides listed in Table 1.
  • epitopes are selected for their broad recognition by HLA class I or class II molecules of subjects from diverse ethnic, racial and geographical backgrounds.
  • the present invention provides a uniquely optimized package and appropriate delivery system for introducing the treatment into the subject.
  • the composition of the present invention comprises viral, bacterial, parasitic, protozoa or cancer specific transformed promoter, epitopes, proteins, factors that increase epitope expression in MHC pockets and genes that increase cytokine expression.
  • one or more epitopes are selected from the peptides in Table 1 to 10, or a subsequence, homologue, variant or derivative thereof.
  • the composition comprises CD4 and/or CD8 T-cell epitopes from the mumps vaccine.
  • the composition comprises T-cell (e.g., CD8 and/or CD4 T cells) epitopes from the measles vaccine.
  • the composition comprises CD4 and/or CD8 T-cell epitopes from the rubella vaccine.
  • the composition comprises CD4 and/or CD8 T-cell epitopes from the polio vaccine.
  • the composition comprises CD4 and/or CD8 T-cell epitopes from the chicken pox/varicella vaccine, comprising one or more or a combination of the peptides listed in Table 1.
  • the composition comprises proteins, peptides or epitopes from two or more different vaccines (e.g., measles, mumps, rubella, polio or chicken pox vaccine).
  • the composition comprises proteins, peptides or epitopes from two or more, three or more, four or more, or five or more different vaccines.
  • the composition may contain the same number of proteins, peptides or epitopes from each of the two or more vaccines or may contain a different number of proteins, peptides or epitopes from each of the two or more vaccines.
  • each T-cell epitope binds only a subset of naturally occurring HLA class II molecules in the human population and this repertoire of HLA class II molecules varies from one person to another and from one ethnic population to another
  • an epitope or combination of epitopes that have a HLA class II repertoire covering a high fraction of a worldwide population, and which also produce an immune response (e.g., T cell response) in a significant fraction of patients with the disease, disorder or condition to be treated.
  • an immune response e.g., T cell response
  • each T cell epitope binds only a subset of naturally occurring HLA class I molecules in the human population and this repertoire of HLA class I molecules varies from one person to another and from one ethnic population to another
  • an epitope or combination of epitopes that have a HLA class I repertoire covering a high fraction of a worldwide population, and which also produce an immune response (e.g., T-cell response) in a significant fraction of patients with the disease, disorder or condition to be treated.
  • an immune response e.g., T-cell response
  • the methods or compositions of the present invention it is desirable for the methods or compositions of the present invention to include as few peptides as possible, but at the same time to be able to treat the majority of patients in a worldwide population with the same method or composition.
  • Such a method or should contain a combination of peptides capable of binding to a HLA Class II and/or Class I repertoire covering a high fraction of a worldwide population, and the resulting peptide-HLA complexes should be recognized as epitopes (e.g., T cell epitopes) in the subject so as to induce an immune response.
  • composition of the present invention further comprises cytokines, immune activators, chemokines, growth factors or T F family members or a gene for increased expression of cytokines, immune activators,
  • the composition comprises one or more of or genes for one or more of AREG, Granzyme, Histamine, IFNa/ ⁇ , IFNy, IL12, IL15, IL16, IL17a-f, ILlb, 112, IL21, IL22, IL23, IL25, IL27, GM- CSF, IL33, IL4, IL5, IL6, IL7, IL8, IL9, LFA-1, LIF, lymphotoxin, Mac-1, MCP-1, ⁇ , Perforin, TGF- ⁇ , TNFa, VLA-4, CCL1, CCL1 1, CLL17, CCL1, CCL20, CCL22, CCL3, CCL4, CCL5, CXCL10, CXCL13, CXCL8, RANTES, CXCL12, G-CSF, M-CSF, VEGF
  • compositions are delivered to the target disease cells via one or more delivery systems depending on the indication, disease state, severity, clinical utility and other relevant parameters that may impact the desired efficacy of delivery and expression of the T-cell epitopes and/or antigens.
  • the delivery system is engineered to deliver the package to a specific cell subset of interest.
  • the composition is delivered to the target cell and other cells of interest using delivery methods comprising one or more but at least one of the methods from the group comprising: replicating or non-replicating viral vectors such as Adenovirus (AdV); Adeno-associated virus (AAV); Lentivirus; Vaccinia virus;
  • AdV Adenovirus
  • AAV Adeno-associated virus
  • Lentivirus Lentivirus
  • Vaccinia virus vaccinia virus
  • Oncolytic viruses Respective disease causing live, attenuated, or inactivated virus
  • Micro/nano particles Adjuvant emulsified; Electroporation; Bacteriophage; Lipid particles (e.g., Lipofectamine); Exosomes; Mineral oil / Freund's adjuvant; Hyperglycosylation; Pegylation; Poly(lactic/gly colic) acid; Direct injection; Conjugation via cell penetrating peptides/proteins; Lysosomal targeting agents; Gene delivery (e.g., Crispr-Cas9, flox/LoxP).
  • the one or more delivery methods can be used in combination, alone, and in combinations in part thereof. Parts of particular delivery methods may be combined to other delivery methods to form new delivery methods. Other delivery methods well known in the art can be utilized to delivery the composition to the target cells.
  • one or more epitopes are selected from the peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof, are delivered to the target cell using the delivery methods disclosed herein.
  • a peptide's immunogenicity more particularly one or more epitopes from the peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof, may be modified.
  • the modifications to the peptides and/or antigens administered comprise one or more of the modifications from the group comprising: Oxidization; Reduction; Citrullination; Glycatation (including but not limited to: prenylation, myristylation, palmitolation); Nitration; Di-sulfide bonding (Cysteine); Transglutamination; Deamidation; Misfolding; Single amino acid substitutions; Genetic frameshift mutations.
  • the modifications are not intended to be limiting by scope or category but rather an illustration of how the peptides may be constructed in the delivery mechanisms.
  • compositions for use according to the methods of the invention described herein can be chosen by the individual physician in view of the patient's condition. See e.g., Fingl et al. 1975, in "The Pharmacological Basis of Therapeutics," Ch. 1 p. 1; which is incorporated herein by reference in its entirety. Any suitable route of administration can be used for
  • routes of administration include topical or local (e.g., transdermally or cutaneously, (e.g., on the skin or epidermis), in or on the eye, intranasally, transmucosally, in the ear, inside the ear (e.g., behind the ear drum)), enteral (e.g., delivered through the
  • parenteral administration e.g., parenterally, e.g., intravenously, intra-arterially, intramuscularly, intraperitoneally, intradermally, subcutaneously, intracavity, intracranially, intraarticular, into a joint space, intracardiac (into the heart), intracavernous injection, intralesional (into a skin lesion), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intrauterine, intravaginal, intratumoral, intravesical , intravitreal), the like or combinations thereof.
  • parenteral administration e.g., parenterally, e.g., intravenously, intra-arterially, intramuscularly, intraperitoneally, intradermally, subcutaneously, intracavity, intracranially, intraarticular, into a joint space, intracardiac (into the heart), intracavernous injection, intralesional (into a skin lesion), intraosse
  • a composition herein is provided to a subject.
  • a composition that is provided to a subject is often provided to a subject for self- administration or for administration to a subject by another (e.g., a non-medical professional).
  • a composition described herein can be provided as an instruction written by a medical practitioner that authorizes a patient to be provided a composition or treatment described herein (e.g., a prescription).
  • a composition can be provided to a subject wherein the subject self-administers a composition orally, intravenously or by way of an inhaler, for example.
  • compositions herein can be formulated to be compatible with a particular route of administration or use.
  • Compositions for parenteral, intradermal, or subcutaneous administration can include a sterile diluent, such as water, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents.
  • the preparation may contain one or more preservatives to prevent microorganism growth (e.g., antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as EDTA; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose).
  • a composition herein is substantially free of a chelator (e.g., a zinc chelator, e.g., EDTA or EGTA).
  • compositions for injection include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ), hydroxypropyl-b- cyclodextrin or other cyclodextrin derivatives, phosphate buffered saline (PBS).
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and polyethylene glycol), and suitable mixtures thereof.
  • Fluidity can be maintained, for example, by the use of a coating such as lecithin, or by the use of surfactants.
  • Antibacterial and antifungal agents include, for example, parabens, chlorobutanol, phenol, ascorbic acid and thimerosal. Including an agent that delays absorption, for example, aluminum monostearate and gelatin can prolong absorption of injectable compositions.
  • Polysorbate 20 and polysorbate 80 can be added into the formulation mixture, for example, up to 1%.
  • Other non-limiting additives include histidine HC1, ⁇ , ⁇ -trehalose dehydrate, sucrose, lactose or mannitol.
  • compositions for use according to the methods of the invention in a local rather than systemic manner, for example, via direct application to the skin, mucous membrane or region of interest for treating, including using a depot or sustained release formulation.
  • active ingredients can be administered alone.
  • active ingredients can be administered in combination with one or more additional materials, for example, as two separate compositions or as a single composition where the additional material(s) is (are) mixed or formulated together with an active ingredient.
  • an active ingredient can be formulated with additional excipients, or additional active ingredients.
  • the active ingredients when administered in the forms described herein can attain concentrations at a target tissue such as the nose, mucous membranes, the bronchi, the skin, etc. that cannot be attained by the usual intravascular administration of the active ingredients.
  • compositions can be manufactured by any suitable manner, including, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes.
  • compositions for use in accordance with the invention thus can be formulated in any suitable manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • Proper formulation can depend upon the route of administration chosen.
  • any suitable formulation, ingredient, excipient, the like or combinations thereof as listed in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 18th edition, 1990 can be used with a composition described herein.
  • the various materials listed herein, alone or in combination, can be incorporated into or used with the materials described in Remington's.
  • the composition may be formulated, for example, as a topical formulation.
  • the topical formulation may include, for example, a formulation such as a gel formulation, a cream formulation, a lotion formulation, a paste formulation, an ointment formulation, an oil formulation, and a foam formulation.
  • the composition further may include, for example, an absorption emollient.
  • At least part of the affected area of the mammal is contacted with the composition on a daily basis, on an as-needed basis, or on a regular interval such as twice daily, three times daily, every other day, etc.
  • the composition can be administered for a period of time ranging from a single as needed administration to administration for 1 day to multiple years, or any value there between, (e.g., 1 - 90 days, 1 - 60 days, 1 - 30 days).
  • the dosages described herein can be daily dosages or the dosage of an individual administration, for example, even if multiple administrations occur (e.g., 2 sprays into a nostril).
  • compositions described herein relate to methods of treating or preventing cancer through administration of compositions described herein to the upper respiratory track/bronchi in a mammal in need thereof, for example, by contacting at least part of the upper respiratory tract/bronchi of a mammal with a therapeutically effective amount of a composition as described above or elsewhere herein.
  • the composition can be, for example, formulated as an aerosol formulation, including formulated for use in a nebulizer or an inhaler.
  • the composition further may include other pharmaceutically acceptable components such as a preservative.
  • compositions that include, for example, a composition as described herein and an aerosolized pharmaceutically acceptable carrier solution or dry powder.
  • the compositions may be formulated, for example, to be substantially absorbed by a bronchus.
  • the compositions also may include, for example, one or more of dichlorodifluoromethane, trichlorofluoromethane,
  • compositions can be formulated for use in a nebulizer or an inhaler, for example.
  • carrier defines a chemical compound that facilitates the incorporation of a compound into cells or tissues.
  • DMSO dimethyl sulfoxide
  • a pharmaceutical carrier for a composition described herein can be selected from castor oil, ethylene glycol, monobutyl ether, diethylene glycol monoethyl ether, corn oil, dimethyl sulfoxide, ethylene glycol, isopropanol, soybean oil, glycerin, zinc oxide, titanium dioxide, glycerin, butylene glycol, cetyl alcohol, and sodium hyaluronate.
  • a pharmaceutical carrier for certain of such hydrophobic compounds can be a co-solvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • a common co-solvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant POLYSORBATE 80TM, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • co-solvent system can be varied considerably without destroying its solubility and toxicity characteristics.
  • identity of the co-solvent components can be varied: for example, other low-toxicity nonpolar surfactants can be used instead of POLYSORBATE 80TM; the fraction size of polyethylene glycol can be varied; other biocompatible polymers can replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides can substitute for dextrose.
  • other delivery systems for hydrophobic pharmaceutical compounds, excipients, or additives can be employed, if required. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs and drug compositions.
  • Certain organic solvents such as dimethyl sulfoxide also can be employed, although usually at the cost of greater toxicity.
  • the compounds can be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • the pharmaceutical compositions described herein can be administered to a patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s).
  • the compounds and compositions can be formulated with salts or excipients, such as for example, sodium or meglumine. Techniques for formulation and administration of the compounds of the instant application can be found in "Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, 18th edition, 1990.
  • compositions described herein can preferably be stable over an extended period of time, for example on the order of months or years.
  • Compositions described herein in some embodiments, comprise a preservative.
  • the preservative can comprise a quaternary ammonium compound, such as benzalkonium chloride, benzoxonium chloride, benzethonium chloride, cetrimide, sepazonium chloride, cetylpyridinium chloride, or domiphen bromide (BRADOSOL®).
  • the preservative can comprise an alkyl-mercury salt of thiosalicylic acid, such as thiomersal, phenylmercuric nitrate, phenylmercuric acetate or phenylmercuric borate.
  • the preservative can comprise a parabens, such as methylparaben or propylparaben.
  • the preservative can comprise an alcohol, such as chlorobutanol, benzyl alcohol or phenyl ethyl alcohol.
  • the preservative can comprise a biguanide derivative, such as chlorohexidine or polyhexamethylene biguanide.
  • the preservative can comprise sodium perborate, imidazolidinyl urea, and/or sorbic acid.
  • the preservative can comprise stabilized oxychloro complexes, such as known and commercially available under the trade name PURITE®).
  • the preservative can comprise polyglycol-polyamine condensation resins, such as known and commercially available under the trade name POLYQUART® from Henkel KGaA.
  • the preservative can comprise stabilized hydrogen peroxide generated from a source of hydrogen peroxide for providing an effective trace amount of resultant hydrogen peroxide, such as sodium perborate tetrahydrate.
  • the preservative can be benzalkonium chloride.
  • the preservative can enable a composition to be used on multiple occasions.
  • the preservative can reduce the effects of one or more of acid exposure, base exposure, air exposure, heat, and light on the active ingredient.
  • the compounds and compositions used herein can include any suitable buffers, such as for example, sodium citrate buffer and/or sequestering agents, such as edetate disodium sequestering agent.
  • Ingredients, such as meglumine, may be added to adjust the pH of a composition or compound described herein.
  • Compounds and compositions described herein may comprise sodium and/or iodine, such as organically bound iodine.
  • Compositions and compounds used herein may be provided in a container in which the air is replaced by another substance, such as nitrogen.
  • compositions suitable for use in the technology which include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose.
  • a "therapeutically effective amount” means an amount to prevent, treat, reduce the severity of, delay the onset of or inhibit a symptom of a cancer.
  • the symptom can be a symptom already occurring or expected to occur. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • a therapeutically effective amount can describe the amount necessary for a significant quantity of the composition to contact the desired region or tissue where prevention or treatment of a cancer is desired.
  • compositions described herein can be administered at a suitable dose, e.g., at a suitable volume and concentration depending on the route of administration.
  • dosages of an administered composition, vector, factor, agent or peptide can be from 0.001 - 200 grams, or 0.001 - 50 grams, 0.001 - 10 grams, etc.
  • the amount can be, for example, from 0.001 - 0.1 grams, 0.1 - 5 grams, 5 - 10 grams, 10 - 15 grams, 15 - 20 grams, 20 - 25 grams, 25 - 30 grams, 30 - 35 grams, 35 - 40 grains, 40 - 45 grams, 45 - 50 grams and 50 - 200 grams.
  • a composition, vector, factor, agent or peptide can be provided in a concentration, for example, of 1 mg/kg to 5000 mg/kg, 50 mg/kg to 1000 mg/kg, 50 mg/kg to 500 mg/kg, 150 mg/kg to 350 mg/kg or 350 mg/kg to 1000 mg/kg.
  • composition, vector, factor, agent or peptide described herein can be administered at a concentration of at least 50 mg/kg, at least 100 mg/kg, at least 150 mg/kg, at least 200 mg/kg, at least 150 mg/kg, at least 200 mg/kg, at least 250 mg/kg, at least 300 mg/kg, at least 350 mg/kg, at least 400 mg/kg, at least 450 mg/kg, or at least 500 mg/kg.
  • a composition, vector, factor, agent or peptide described herein can be administered at a concentration of about 100 mg/kg, about 150 mg/kg, about 200 mg/kg, about 150 mg/kg, about 200 mg/kg, about 250 mg/kg, about 300 mg/kg, about 350 mg/kg, about 400 mg/kg, about 450 mg/kg, or about 500 mg/kg.
  • Volumes suitable for intravenous administration are well known. For example, 50 ml - 100 ml of a composition, vector, factor, agent or peptide at a working concentration of between about 200 mg/kg to about 350 mg/kg can be safely administered intravenously to an adult human subject.
  • the amount delivered can be any suitable amount, for example, in order to contact the desired tissue in a therapeutically effective manner.
  • the compositions can be delivered to the nose, and the amount delivered to each nostril can be from about 20 microliters to about 1500 microliters, 50 microliters to about 1000 microliters, or 50 microliters to about 500 microliters.
  • composition described herein can be delivered to the nose in a volume of about 50, 100, 200, 300, 400 or 500 microliters for example.
  • compositions can, if desired, be presented in a pack or dispenser device, which can contain one or more unit dosage forms containing the active ingredient.
  • the pack can for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device can be accompanied by instructions for administration.
  • the pack or dispenser can also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, can be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier can also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • compositions, formulations, combination products and materials described herein can be included as part of kits, which kits can include one or more of the compositions, vectors, factors, agents or peptides described herein, formulations of the same, combination drugs and products and other materials described herein.
  • the products, compositions, kits, formulations, etc. can come in an amount, package, product format with enough medication to treat a patient for 1 day to 1 year, 1 day to 180 days, 1 day to 120 days, 1 day to 90 days, 1 day to 60 days, 1 day to 30 days, or any day or number of days there between.
  • kits including a composition, vector, factor, agent or peptide of the invention, combination compositions and pharmaceutical formulations thereof, packaged into suitable packaging material.
  • a kit optionally includes a label or packaging insert including a description of the components or instructions for use in vitro, in vivo, or ex vivo, of the components therein.
  • Exemplary instructions include instructions for a method, treatment protocol or therapeutic regimen.
  • a kit can contain a collection of such components, e.g., two or more conjugates alone, or in combination with another therapeutically useful composition (e.g., an anti-proliferative or immune-enhancing drug).
  • the term "packaging material” refers to a physical structure housing the components of the kit.
  • the packaging material can maintain the components sterilely, and can be made of material commonly used for such purposes (e.g., paper, corrugated fiber, glass, plastic, foil, ampules, vials, tubes, etc.).
  • Kits can include labels or inserts.
  • Labels or inserts include "printed matter," e.g., paper or cardboard, or separate or affixed to a component, a kit or packing material (e.g., a box), or attached to an ampule, tube or vial containing a kit component.
  • Labels or inserts can additionally include a computer readable medium, optical disk such as CD- or DVD-ROM/RAM, DVD, MP3, magnetic tape, or an electrical storage media such as RAM and ROM or hybrids of these such as magnetic/optical storage media, FLASH media or memory type cards.
  • Labels or inserts can include identifying information of one or more components therein, dose amounts, clinical pharmacology of the active ingredient(s) including mechanism of action, pharmacokinetics (PK) and pharmacodynamics (PD). Labels or inserts can include information identifying manufacturer information, lot numbers, manufacturer location and date.
  • Labels or inserts can include information on a condition, disorder, disease or symptom for which a kit component may be used.
  • Labels or inserts can include instructions for the clinician or for a subject for using one or more of the kit components in a method, treatment protocol or therapeutic regimen. Instructions can include dosage amounts, frequency or duration, and instructions for practicing any of the methods, treatment protocols or therapeutic regimes set forth herein. Kits of the invention therefore can additionally include labels or instructions for practicing any of the methods and uses of the invention described herein.
  • Labels or inserts can include information on any benefit that a component may provide, such as a prophylactic or therapeutic benefit. Labels or inserts can include information on potential adverse side effects, such as warnings to the subject or clinician regarding situations where it would not be appropriate to use a particular composition. Adverse side effects could also occur when the subject has, will be or is currently taking one or more other medications that may be incompatible with the composition, or the subject has, will be or is currently undergoing another treatment protocol or therapeutic regimen which would be incompatible with the composition and, therefore, instructions could include information regarding such incompatibilities.
  • Kits can additionally include other components. Each component of the kit can be enclosed within an individual container and all of the various containers can be within a single package. Invention kits can be designed for cold storage. [00131] The invention has been described herein using specific embodiments for the purposes of illustration only. It will be readily apparent to one of ordinary skill in the art, however, that the principles of the invention can be embodied in other ways. Therefore, the invention should not be regarded as being limited in scope to the specific embodiments disclosed.
  • Effective immunity against a pathogen entails (i) the development of highly specific, neutralizing, and non-self reactive antibodies and T cells and (ii) results in the development of immune memory cells that patrol the body and provide lifelong protection.
  • Immune memory cells are uniquely positioned to rapidly and robustly expand and neutralize a specific pathogen if repeat exposure occurs.
  • Vaccines provide a mechanism to promote safe development of long-lived immune memory against a pathogen(s) by avoiding the deleterious effects of actual pathogen infection. The inventor approach activates and redirects the long-lived immune memory derived from vaccines to fight cancer.
  • Peptide antigens are used in combination with other cancer and cancer- immune-based therapies to increase efficacy of patient treatment.
  • Peptide antigens disclosed herein are delivered in combination with viral vectors including oncolytic viruses like T-Vec (Imlygic).
  • T-Vec is the first oncolytic virus approved in the US and has been shown of utility in the treatment of patients with recurrent melanoma.
  • Peptide antigens disclosed herein are cloned into different viruses, including the herpes simplex virus type 1 (HSV1) virus, adenovirus (AdV), adeno-associated virus (AAV) and vaccinia virus, for expression in tumor tissue to combine oncolytic potential with increased tumor immunogenicity.
  • HSV1 herpes simplex virus type 1
  • AdV adenovirus
  • AAV adeno-associated virus
  • vaccinia virus are in advanced clinical development. These viruses have potential for selective tumor-mediated killing through direct tumor injection or engineered tumor specificity after systemic delivery.
  • CTLA-4 (Ipilumimab), PDL-1 and PD1 (Nivolumab / Pembrolizumab) are two presently approved targets of antibody-based immune checkpoint inhibitors. These antibodies non-specifically block inhibitory receptors on T cells that maintain T cells in an activated state. Recent publications highlight the efficacy of these therapies may be tied to the mutational burden of the specific tumor and in tumors with low mutational burden, checkpoint inhibitors may be of limited utility. Peptide antigens disclosed herein are delivered to the tumor in combination with checkpoint inhibitors to bring immune memory derived from childhood vaccines to more robustly kill cancerous and precancerous tissue.
  • Chimeric Antigen receptor T cell therapies are presently in advanced clinical trials for various cancers. These therapies entail the isolation of patient T cells and re- engineering of T-cell specificity to the target of choosing. While these therapies have shown therapeutic promise in single-cell type tumors such as B cell lymphomas, solid tumors that are under immune-suppression and lack a single marker for targeting are presently intractable. In particular instances, the introduction of peptide antigens disclosed herein and simultaneous engineering of T cells with specificity for the delivered epitopes enable robust and targeted killing of epitope expressing cells.
  • cytokine and TNF-super family based therapies are in advanced clinical development for the treatment of cancer. Specifically, tumor specific expression of cytokines like IL12 and TNF superfamily members such as OX40 reduce tumor immune suppression and promote inflammation.
  • oncolytic virus therapies are in development in which IL12 and OX40 are expressed by the therapeutic virus.
  • combination with peptide antigens disclosed herein increase T cell infiltration and activation in the tumor tissue.
  • Rituximab represents a particularly successful candidate in the treatment of B cell-based tumors such as non-Hodgkin's lymphoma.
  • Therapeutic antibodies recognize surface antigens of the tumor tissue and induce immune responses through phagocytosis, complement deposition, and cell cytotoxicity.
  • combined delivery of peptide antigens described herein with mAbs increase the immune potential of the tumor leading to increased killing of tumor tissue by the immune system.
  • Popular developing therapies in clinical development utilize cancer-specific mutations to enable generation or expansion of tumor-specific immunity within a patient.
  • patient cancer-specific mutations are identified by a variety of sequencing methods and mutation gene products are identified and assessed for the ability to be bound by HLA molecules or the existence of mutation-specific T cells within the donor. Vaccination of these peptides/antigens in combination with adjuvant and checkpoint blockers are then used to expand mutation specific T cells.
  • Various tumors have lower mutational burden and therefore cancer vaccine approaches are of limited practical utility.
  • combination of administration of patient mutation specific peptides with childhood vaccine peptide antigens improve the therapeutic potential against the tumor.
  • T cells are brought into closer proximity to malignant tissue by fusion of two antibodies or generation of bi-specific antibodies.
  • Such an approach has shown early clinical potential by combined target of CD 19 and T-cell markers such as CD3 to bring T cells in proximity to the cellular target and promote T- cell-mediated killing.
  • delivery of peptide antigens described herein to target tissue and co-administration of BiTE to promote T-cell infiltration into the target tissue improve the frequency or concentration of T cells with potential to specifically kill the tumor tissue.
  • a panel of 16 HLA A (A*01 :01, A*26:01, A*32:01, A*02:01, A*02:03, A*02:06, A*68:02, A*2301, A*24:02, A*03 :01, A* l l :01, A*30:01, A*31 :01, A*33 :01, and A*68:01) and 11 HLA B alleles (B*40:01, B*44:02, B*44:03, B*57:01, B*58:01, B* 15:01 B*07:02, B*35:01, B*51 :01, B*53 :01, and B*08:01) were selected,
  • Binding predictions were performed using the consensus prediction tool available on the IEDB web site (Accessed online at ⁇ URL:http:/www.iedb.org/>). For each allele and length combination, peptides from each included vaccine strain were selected if they were in the top 1% of binders. Redundant sequences across various vaccine strains were removed and non-redundant sequences are listed in Table 1.
  • the binding affinity of 15-mer peptides derived from childhood vaccines were predicted using the panel of 27 HLA class II DRB1, 3/4/5, DQA1/DQB1, and DPB 1 molecules that provide broad ethnic and geographic population coverage using a consensus prediction approach. Peptides with predicted binding scores in the top 20% for a given allele were considered potential binders. Peptides predicted to bind 13 or more HLA molecules at this threshold were considered promiscuous binders.
  • viral genes can become incorporated into the host genome. This viral transformation event can lead to uncontrolled proliferation and the development of various diseases and disorders including cancer. Viral, parasitic and bacterial transformation or infection of healthy cells has been identified as a common underlying cause of a great number of human cancers.
  • specific viral genes are incorporated into the genome of cancers cells and maintain expression within the cancerous cells throughout the lifecycle of the cancer cell. For example, in cervical cancer, which has been linked to infection by human papillomaviruses, transformation of viral genes E6 and E7 has been widely established.
  • the measurement of pre-existing immune responses derived from childhood vaccines are performed to assess and quantify the presence of circulating antibodies and/or frequency of vaccine-specific immune cells in the individual.
  • the proteins and/or peptides listed in Table 1 are used as part of a kit to specifically measure B cell response by antibody ELISA and/or the CD4 and CD8 T-cell response by cytokine ELISA or ELISPOT to inform the frequency and functionality of vaccine immune memory of the patient.
  • the results obtained from use of the diagnostic kit inform whether a patient has sufficient immune memory for therapeutic application or would benefit from receipt of a booster vaccination prior to administration of the therapeutic to increase the frequency of vaccine-specific immune cells that recognize and promote an anti-tumor immune response.
  • Varicella-zoster virus (strain Oka) ORF22 233 242 LIYGSCDTYF
  • Varicella-zoster virus (strain Oka) ORF22 252 260 VTAQHPLLL
  • Varicella-zoster virus (strain Oka) ORF22 256 264 HPLLLSPPN
  • Varicella-zoster virus (strain Oka) ORF22 262 270 PPNSTTIVL
  • Varicella-zoster virus (strain Oka) ORF22 272 280 KSSIVPLHQ
  • Varicella-zoster virus (strain Oka) ORF22 291 300 TLHSTLTNTV
  • Varicella-zoster virus (strain Oka) ORF22 296 305 LTNTVALDPR
  • Varicella-zoster virus (strain Oka) ORF22 319 328 ETTSTGSGVL
  • Varicella-zoster virus (strain Oka) ORF22 326 335 GVLDCRRRRR
  • Varicella-zoster virus (strain Oka) ORF22 395 403 EVANTPDVV
  • Varicella-zoster virus (strain Oka) ORF22 410 418 DTPESSPTF
  • Varicella-zoster virus (strain Oka) ORF22 415 424 SPTFSRN NV
  • Varicella-zoster virus (strain Oka) ORF22 420 429 RNMNVGSSLK
  • Varicella-zoster virus (strain Oka) ORF22 470 478 TVITGHTNV
  • Varicella-zoster virus (strain Oka) ORF22 478 486 VPIWVTRPL 98 Varicella-zoster virus (strain Oka) ORF22 478 487 VPIWVTRPLV
  • Varicella-zoster virus (strain Oka) ORF22 1 116 1 125 PALSFVLSLY 251 Varicella-zoster virus (strain Oka) ORF22 1 117 1 125 ALSFVLSLY
  • Varicella-zoster virus (strain Oka) ORF22 1779 788 LPIWIKTQYM 404 Varicella-zoster virus (strain Oka) ORF22 781 790 IWIKTQYMTF

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)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Developmental Biology & Embryology (AREA)
  • Vascular Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Dermatology (AREA)
  • Molecular Biology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Reproductive Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Presented herein are novel methods of treating cancer, which methods result in the expression of peptide antigens on a cancer. The peptide antigens are selected to initiate a recall immune response in a subject (e.g., a pre-existing immune response (memory response) that may have been induced by a prior vaccination or infection), which immune response is directed toward the cancer. In certain embodiments, the methods herein are used in combination with existing cancer therapies.

Description

LEVERAGING IMMUNE MEMORY
FROM COMMON CHILDHOOD VACCINES TO FIGHT DISEASE
RELATED PATENT APPLICATIONS
[0001] This patent application claims the benefit of U.S. Provisional Patent Application No. 62/320,488 filed on April 9, 2016, entitled LEVERAGING IMMUNE MEMORY FROM COMMON CHILDHOOD VACCINES TO FIGHT DISEASE, naming Patrick Ho and Ravi Kolla as inventors, and International Patent Application No. PCT/US2016/068147 filed on December 21, 2016, entitled LEVERAGING IMMUNE MEMORY FROM COMMON CHILDHOOD VACCINES TO FIGHT DISEASE, naming Patrick H. Ho and Ravi Kolla as inventors. The entire content of the foregoing patent applications is incorporated herein by reference, including all text, tables and drawings.
REFERENCE TO A "SEQUENCE LISTING," A TABLE, OR A COMPUTER PROGRAM LISTING APPENDIX
Sequence Listing file Name: 2015-121-04_ST25.txt
Sequence Listing file Size: 122 KB
The entire contents of the sequence listing are hereby expressly incorporated by reference.
FIELD OF INVENTION
[0002] The present invention relates to the field of disease treatment. In particular the present invention relates to the implementation of treatments for disease, from the approach of targeting pre-existing human immunity as a vehicle to specifically target and destroy target cells. BACKGROUND
[0003] Effective immunity against a pathogen entails (i) the development of highly specific, neutralizing and non-self reactive antibodies and T cells and (ii) results in the development of immune memory cells that patrol the body and provide life long protection. Immune memory cells are uniquely positioned to rapidly and robustly expand and neutralize a specific pathogen if repeated exposure occurs.
[0004] Vaccines provide a mechanism to promote safe development of long-lived immune memory against a pathogen(s) by avoiding deleterious effects of actual pathogen infection. A number of vaccines are routinely administered to children to prevent the contraction of debilitating infectious diseases and immune memory from these vaccines can be measured decades following administration.
[0005] It is known in the art that T cells, including CDS and CD4 cytotoxic T cells, play a key role in the defense against intracellular pathogens and tumor cells. T-cell immune responses are driven by the recognition of foreign peptides presented by major histocompatibility complex class I and class II (MHC I and MHC II) molecules at the cell surface. The identification of these peptides (T-cell epitopes) is therefore important for understanding disease pathogenesis and etiology as well as for vaccine design. T-cell response has been implicated in providing vaccine-mediated protection. However few T- cell epitopes from vaccines have been identified.
[0006] Current popular approaches in immunotherapy utilize cellular targets that illicit immune responses against cancer and non-cancers alike (CAR-T, Rituximab, etc.). Developing therapies to target cancer-specific antigens requires the identification of neo- antigens, self-proteins which have undergone mutation to allow recognition by the immune system. These approaches are promising, however they present possible issues with tolerance and risk of autoimmunity.
[0007] In addition, there exists in the art current immunotherapies that utilize cellular targets that are relatively widely expressed throughout the body and are not pointedly specific to a particular cancer. In this relatively broader approach, the idea is to eradicate as many of the cells that present these wide markers, which naturally include healthy and cancerous cells. This blanket approach results in the immune response attacking cells that are both cancerous and non cancerous, resulting in deleterious side effects and in many instances the patient may no longer be able to continue therapy due to negative side effects outweighing the potential benefits. The upregulation of the immune system in an attempt to equip the body's own immune system with fighter cells to attack cancer cells, has significant risk associated with inducing autoimmunity. In particular instances, whereby the treatment induces upregulation of the immune response, the upregulated state translates into the body's own immune system recognizing its own cells as foreign and begins launching an attack on its own healthy cells. A further problem with existing therapies is the risk associated with tissues beginning to build tolerance to the administered therapy whereby mutations enable cancer cells to bypass the therapeutic agent and proliferate, and they not only become unresponsive to the treatment but potentially further create an environment whereby no other alternative therapies are available. 08] Based on the foregoing, there is a need in the art for methods to target immunotherapies to specific disease cells without targeting healthy cells or significantly minimizing the effect on healthy cells and is not reliant upon antigens or epitopes of considerable homology to self, for example, a cancer immunotherapy method that provides an immune response specifically targeted to cancer cells that does not target healthy cells. Increasing evidence regarding checkpoint inhibitor targeting therapies (e.g., anti-CTLA4, anti-PDl or anti-PDL-1) shows reduced utility in cancers of low mutational burden. Unlike current treatments that target a broad range of immune response cells, which may or may not translate into the desired effect on the specific disease cells, the present invention discloses a treatment that has a targeted approach towards known epitopes recognized by the immune system from previous immunity. This approach has significant implications for patient care and treatment success by promoting more rapid, non-self, and targeted cytolytic activity. SUMMARY
[0009] In certain aspects, presented herein is a method of treating cancer comprising, a) providing a subject having or suspected of having a cancer and b) administering a vector (e.g., viral vector such as adenovirus, vaccinia or plasmid DNA) to the subject, wherein the vector directs expression of one or more peptide antigens on a target cell of the subject and wherein a memory or recall immune response is elicited against the one or more peptide antigens. In certain aspects, presented herein is a method of treating cancer comprising a) providing a subject having or suspected of having a cancer; and b) administering a composition to the subject, wherein the composition comprises one or more peptide antigen, wherein a memory or recall immune response is elicited against the one or more peptide antigens. In some embodiments, prior to the administering of (b), the subject was vaccinated against a pathogen, non-limiting examples of which include a vaccination against chickenpox, measles, mumps, rubella, shingles/varicella zoster or polio. In certain embodiments a subject is vaccinated with another suitable live-attenuated or inactivated pathogen, or pathogen antigen(s). In some embodiments, prior to the administering of (b), the subject was vaccinated with at least one of the one or more peptides selected from Table 1, or one or more other antigens derived from a pathogen. The subject may have been vaccinated or re-vaccinated 1 day, 1 month, 6 months, or 1 year to 100 years prior to administering of (b). Accordingly, in certain aspects, the administering of (b) elicits a recall immune response in the subject, wherein the recall immune response is directed against the target cell and/or at least one of the one or more peptide antigens.
[0010] In certain aspects the one or more peptide antigens bind to MHC class I or MHC class II and can recall CD8 T-cell and/or CD4 T-cell responses.
[0011] In some aspects, the subject is further administered an anti-cancer therapy.
The anti-cancer therapy may be administered prior to, concurrently with and/or after the administering of vector or peptides of (b). The anti -cancer therapy can be any suitable therapy. In some embodiments, the anti-cancer therapy comprises surgical resection. In some embodiments, the anti-cancer therapy comprises a radiation therapy, chemotherapy or an immunotherapy. In some embodiments, a radiation therapy comprises external beam radiation, proton therapy, or internal radiation therapy. In some embodiments, an immunotherapy comprises chimeric antigen receptor (CAR)-T immunotherapy. CAR-T immunotherapy sometimes comprises administering CAR T-cells configured to specifically bind to and kill the target cell. In some embodiments, an immunotherapy comprises administering an antibody or antibody fragment that specifically binds to a cell-surface antigen, non-limiting examples of which include CD20, CD30, CD33, CD52, CD 138, CD274, CD279, PD-L1, CTLA-4, programmed cell death 1 (PD-1), CD33, EGFR, ERBB2 (CD340), ERBB3, VEGF, VEGFR, GM-2, gp-100, EPCAM, CD19, CD3, CEA, gpA33, mucins, TAG-72, CAIX, PSMA, folate-binding protein, GD2, GD3, Integrin aVp3, Integrin α5β 1, IGF1R, EPHA3, TRAILR1, TRAILR2, RA KL, FAP or Tenascin. In some embodiments, the antibody may be bi-specific and may bind to two cell surface antigens. In some embodiments, the immunotherapy comprises administering an antibody or T-cell that specifically binds a checkpoint blocker. In some embodiments, an immunotherapy comprises administering an antibody or antibody fragment that specifically binds to a tumor marker.
[0012] In some aspects a peptide antigen described herein is derived from a pathogen (e.g., a peptide or protein expressed by a pathogen). In some aspects, the one or more peptide antigens are selected from a peptide listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
[0013] In some embodiments, a target cell is a cell that is or has been infected with a pathogen. For example, many types of cancers are induced or caused by a previous infection by a pathogen. For example, a target cell may be a cell that is or has been infected with a virus, bacteria, parasite or protozoa. In some embodiments, a target cell is not infected with a pathogen.
[0014] A pathogen can be a human pathogen and may be a virus, fungus, bacteria, parasite or protozoa. In some embodiments, a pathogen is a non-human pathogen, or the peptide antigen is conserved between human and another species, or wherein the peptide antigen cross-reacts between human and another species. In some embodiments a pathogen is a virus, non-limiting examples of which include a virus of the order
Flaviviridae, Retroviridae, Orthomyxoviridae, Arenaviridae, or Mononegavirales, or a virus of the family Togaviridae, Reoviridae, picornaviridae, herpesviridae or Bunyaviridae.
[0015] In certain aspects, a target cell is a cancer cell or neoplastic cell. In some
embodiments, a target cell is a metastatic cancerous cell. Non-limiting examples of a cancer cell, metastatic cancerous cell or neoplastic cell include cells of a carcinoma, sarcoma, neuroblastoma, cervical cancer cell, hepatocellular cancer cell, bladder cancer cell, mesothelioma, glioblastoma, myeloma, lymphoma, neoplastic lymphocyte, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, lymphosarcoma, liposarcoma, osteosarcoma, chondrosarcoma, leiomyosarcoma, rhabdomyosarcoma or fibrosarcoma.
[0016] In some aspects, a subject is a mammal. In some aspects a subject is a non- human mammal. In some aspects, a subject is a human. In some embodiments, wherein prior to the administering of a vector or peptide of (b), a subject was infected with the pathogen, thereby inducing, eliciting or promoting a recall immune response in the subject to certain pathogen antigens.
[0017] In some aspects, a subject has, or is suspected of having a cancer. Non- limiting examples of a cancer include a hematological and solid tumors, a neoplasia derived from lung, thyroid, head or neck, nasopharynx, throat, nose, sinus, brain, spine, breast, adrenal gland, pituitary gland, thyroid, lymph, mouth, esophagus, stomach, duodenum, ileum, jejunum intestine, colon, rectum, uterus, urethra, ovary, cervix, endometrium, bladder, testicle, penis, prostate, kidney, pancreas, liver, bone, bone marrow, lymph, blood, muscle, or skin. Other non-liming examples of a cancer include a carcinoma, sarcoma, neuroblastoma, hepatocellular cancer, mesothelioma, glioblastoma, myeloma, lymphoma, myeloma, leukemia, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, cervical cancer, breast cancer, liver cancer, pancreatic cancer, brain cancer, lung cancer, skin cancer, ovarian cancer, and testicular cancer. In certain embodiments, a cancer is an infection-induced cancer non-limiting examples of which include a hepatocellular carcinoma, cervical cancer, head and neck squamous cell carcinoma, adult T-cell leukemia/lymphoma, Burkitt lymphoma, Merkel cell carcinoma, kaposi sarcoma, bladder cancer and gastric cancer. In some embodiments, a cancer comprises a low mutational burden. Non-limiting examples of a cancer comprising a low mutational burden include pilocytic astrocytoma, ALL, medulloblastoma, kidney chromophobe, thyroid, CLL, neuroblastoma, glioblastoma, and pancreatic cancer.
[0018] In some aspects, a method herein comprises administering a vector that
selectively directs expression of one or more peptide antigens on a target cell. In some embodiments, a vector directs expression of the one or more peptide antigens on the cell surface of the target cell. In some embodiments, the one or more peptide antigens are expressed or presented on the cell surface of the target cell. Presentation of a peptide antigen on the surface of a cell may include co-expression of a peptide in the presence of TAP (transporter associated with antigen processing). In some embodiments, a vector comprises a nucleic acid encoding one or more non-self-peptide antigens (e.g., one or more of the antigens of Table 1). In some embodiments, a vector comprises a promoter operatively linked to a nucleic acid encoding the one or more peptides. A promoter of a vector is sometimes inducible by a factor, for example a transcription factor derived from a pathogen, a mutated human protein, and/or one or more viral proteins. In certain embodiments, the promoter is selectively induced in tumor cells. In certain embodiments, a factor is a viral transcription factor or a factor that is selectively expressed in tumor cells.
[0019] In certain aspects, the administering of a vector or protein of (b) comprises administering nano-carriers, micro-carriers, microparticles, nanoparticles, liposomes, exosomes or recombinant virus to the subject, where the microparticles, nanoparticles, liposomes, exosomes or recombinant virus may comprise a vector or one or more peptides. In certain aspects, the administering of a vector or protein of (b) comprises selectively delivering nano-carriers, micro-carriers, microparticles, nanoparticles, liposomes, exosomes or recombinant virus to a target cell. In some embodiments, the administering of a vector of (b) comprises administering a nucleic acid to the subject, wherein the nucleic acid comprises the vector. Non-limiting examples of a vector include a plasmid, a linear nucleic acid molecule or a replicating or non-replicating viral vector. In some embodiments, a vector comprises a CRISPR element, and/or the vector is configured to express (CRISPR-associated protein-9 nuclease) CAS9. In some embodiment, a vector is integrated into the target cell by a method comprising CRISPR and (CRISPR-associated protein-9 nuclease) CAS9.
[0020] In some aspects, the administering comprises direct intra-tumoral injection. In some aspects, the administering comprises intravenous injection, intraperitoneal injection, subcutaneous injection, intramuscular injection, intravesical or intrathecal injection.
[0021] In certain aspects of a method herein, after the administering of b), the method further comprises c) initiating, eliciting, promoting, or enhancing an immune response in the subject, wherein the immune response is directed against the target cell and/or target antigen. In some embodiments, the eliciting of (c) comprises initiating, eliciting, promoting or enhancing a recall response in the subject to the one or more peptide antigens. In certain embodiments, the eliciting of (c) comprises administering a vaccine to the subject. In certain embodiments, a vaccine is selected from a vaccine for chickenpox, measles, mumps, rubella, and polio. In certain embodiments, the eliciting of (c) comprises administering a pharmaceutical composition to the subject, wherein the pharmaceutical composition comprises at least one of the one or more of the peptide antigens. In some embodiments, the pharmaceutical composition comprises an adjuvant. In certain embodiments, the eliciting of (c) comprises administering an agent that modulates an immune response in the subject. In some embodiments, the agent increases, enhances, promotes or elicits an innate immune response. In certain embodiments, the agent comprises an immune checkpoint blocker. In certain embodiments, the agent increases, enhances, promotes or elicits a recall immune response. In certain
embodiments, the agent comprises an antibody. In certain embodiments, the antibody specifically binds to the one or more peptide antigens. In some embodiments, the agent comprises a nucleic acid, synthetic chemical, or small molecule.
[0022] In certain aspects, a method herein further comprising administering a
checkpoint blocker, or providing TAP (transporter associated with antigen processing) to the target cell, wherein TAP promotes peptide epitope:MHC expression. Other peptide transporter can be provided such as the antennapedia homeodomain peptide to promote peptide:MHC binding or expression. [0023] In some aspects a composition herein comprises an adjuvant, exosomes, microparticles, nanoparticles, and/or liposomes. In some embodiments, the
microparticles, nanoparticles, or liposomes are configured to selectively deliver one or more peptide antigens to a target cell. In some embodiments, a composition is a pharmaceutical composition or pharmaceutical formulation comprising one or more peptides (e.g., a peptide listed in Table 1). In some embodiments, a composition (e.g., a pharmaceutical composition) comprises a vector configured to direct expression of one or more peptide antigens on a target cell of a subject, wherein the one or more peptide antigens is selected from a peptide listed in Table 1.
[0024] In an embodiment, the present invention provides methods and compositions for eliciting an immune response against target cells. In an embodiment, known or predicted antigens from vaccines or other developed immunity are either engineered to or serve as tags for the targeted cells so as to provide a targeted identification method for the target cells that are recognized by the subjects existing immune system, inducing the subjects immune system to attack the tagged or target cells. In certain embodiments, there is provided the presentation of antigens from vaccines specifically on target cells of interest to elicit a recall immune response to these antigens that is available as a result of childhood or other vaccination. In certain embodiments, the present invention provides a targeted treatment for patients diagnosed with cancer.
[0025] In an embodiment, the present invention is a preventative treatment for
subjects who have been determined to be at risk of developing particular cancer. At risk patients may be determined through various methods known in the art or in the future including but not limited to genetic predispositions and hereditary traits. In an
embodiment the treatment is administered as a primary treatment or in conjunction with other treatments well known in the art.
[0026] In an embodiment, the disease target cells can be administered a factor, for example but not limited to a protein or variant of a protein, which is not present in healthy cells through restricted delivery to a specific tissue. [0027] In an embodiment, the target cells have a mutation not present in healthy cells that results in expression of factor, for example but not limited to a protein or variant of a protein, which is not present in healthy cells. In an embodiment, the expression of a factor that is unique to or predominantly specific to the disease cell enables expression of the antigen from a gene construct. In specific embodiments of the present invention, antigens are specifically expressed in target cells through administration of a gene construct encoding an antigen wherein expression or presentation of the antigen requires a factor (e.g., protein, gene, S P, transcription factor) selectively expressed in the target cells. For example, the gene construct contains the antigen with a promoter driven only by a factor selectively expressed in the target cells (e.g., a cancer cell-specific transcription factor).
[0028] In other embodiments, the expression of transformed viral genes in target cells enables the selective expression of proteins or peptides in these cells. A gene construct containing an antigen with a promoter driven by factors (e.g., proteins, peptides) expressed only in the virally -transformed cells, provides for specific expression of the antigen only in the target cell and therefore provides labeling of the target cells (e.g., a cancer cell) with the antigen. This results in activation of pre-existing memory immune responses against the antigen providing a targeted immune response against the target cells to treat the cancer. In certain embodiments, the methods of the present invention can be used to target pre-, active and metastatic cancerous cells.
[0029] In certain embodiments, the expression of transformed viral genes in
cancerous cells enables the selective synthesis of proteins or peptides in these cells that are necessary for expression of the antigen from the gene construct. In such embodiments, the method involves administration of a gene construct containing the antigen driven by a viral transformed promoter that requires transformed viral genes found in the cancerous cell for expression of the antigen.
[0030] In certain embodiments, specific utility of peptide antigens disclosed herein in cancers of low mutational burden or that are not sensitive to immune-based therapies such as, but not limited to, treatment with checkpoint inhibitors or CAR-T is contemplated. Expression of peptide antigens disclosed herein in combination with other immune-based therapies in such cancers represents a novel utility to make these cancers immune sensitive. Specifically, delivery of peptide antigens disclosed herein to cancers with low potential for neoantigen/epitope development will promote the immunogenic potential of said cancer. Thereby, administration of peptide antigens herein in combination with any of the delivery platforms described herein will make said cancer reactive to other immune therapies such as checkpoint inhibitors and CAR-Ts.
[0031] In different embodiments, the antigen to be selectively expressed and
presented by the target cells is an antigen from a vaccine including but not limited to childhood vaccines, e.g., chickenpox, measles, mumps, rubella, and polio. In certain embodiments, the antigen to be selectively expressed and presented by the target cells is a subsequence, homologue, variant or derivative of an antigen from a vaccine.
[0032] Towards this end, the present inventors have identified peptides derived from vaccine strains of viruses used in live-attenuated vaccines and inactivated vaccine administered during child- and adult- hood including chickenpox, measles, mumps, rubella, and polio that are broadly bound by HLA class I and class II molecules represented in humans from diverse ethnic, racial, and geographic backgrounds that can be used to recall memory T-cell responses.
[0033] The herein described approach allows expression of peptides derived from chickenpox, measles, mumps, rubella and polio for which common childhood vaccines have been administered for the past five decades in target cells to allow targeting of memory immune responses from these common vaccines. This approach is novel as it leverages existing, non-self memory immune responses to target T-cell responses against specific target cells including cancerous, transformed pre-cancerous cells, and metastasis as well as viral, parasitic and bacterial transformed cells.
[0034] In an embodiment, the compositions that comprise one or more peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof, are directly administered to the cancer tissue of the patient. In an embodiment, the composition can be administered by for example but not limited to viral vectors, micro- or nano- particles, direct injection, lipid particles, exosomes, gene, and/or adjuvanted delivery methods. In some embodiments, a composition described herein is administered by direct tissue injection, intratumoral injection, intravenous injection, intraperitoneal injection, subcutaneous injection, intramuscular injection, intravesivular injection or intrathecal injection.
[0035] In some embodiments, a subject is vaccinated prior to, concurrently with, or after the administering of (b). In certain embodiments, a vaccination is a booster vaccination. In some embodiments, the booster vaccination comprises at least one peptide listed in Tables 1. In some embodiments, the booster vaccination is a booster vaccination against chickenpox, measles, mumps, rubella, or polio.
[0036] In some embodiments, an anti-cancer therapy comprises administering
combinations of one or more small molecules, immune checkpoint inhibitors optionally selected from IDO/TDO inhibitors, and epigenetic regulators such as HDAC inhibitors. In some embodiments, an anti-cancer therapy comprises administering, alone or in combination, one or more cancer vaccines optionally based on cancer-associated tumor antigens, or neo-antigens.
[0037] In some embodiments, the one or more of the one or more peptide antigens are linked to a targeting antibody, antibody fragment or nanobody that is internalized by the target cell.
[0038] In some embodiments, a pre-existing T-cell memory is measured prior to the administering of (b). In certain embodiments, such measurement of pre-existing T cell memory can be done to identify subjects for treatment or to determine appropriate treatment for a subject (e.g. companion diagnostic).
[0039] In some embodiments, a few days to weeks prior to the administering of (b), the method comprises boosting immune memory in the subject by administering a vaccines or antigen that stimulates a recall immune response against the one or more peptide antigens.
[0040] In some aspects presented herein is a diagnostic tool for measuring immune response to a vaccine or recall immunity to a target cell comprising measuring an immune response to one or more peptides listed Table 1. In some embodiments, the measurement of recall immunity is prior to administration of a peptide listed in any of Table 1. In some embodiments, the measurement of recall immunity is prior to administration of a vector configured to expresses at least one peptide listed in any of Table 1.
[0041] In some embodiments, a recall and targeting of a pre-existing immune
responses (CD8/CD4) from childhood vaccines leads to tumor cell killing and optionally a subsequent 'abscopal' effect with systemic, cancer-specific immune responses.
[0042] In some embodiments, the one or more peptide antigens comprise full antigens or complete proteins derived from the pathogens.
[0043] In some aspects of a method herein, a subject is vaccinated prior to,
concurrently with, or after the administering of a vector or peptide of (b). In some embodiments, the vaccination is a booster vaccination. In certain embodiments, the booster vaccination comprises at least one peptide listed in Table 1. In certain
embodiments, the booster vaccination is a booster vaccination against chickenpox, measles, mumps, rubella, or polio.
DETAILED DESCRIPTION
Definitions
[0044] An "associated symptom, disease or disorder" as used herein refers to a symptom, disease or disorder that develops in conjunction with, following or as a result of a disease state {e.g., cancer). This includes acute and chronic diseases or disorders, short and long term disease and disorders, as well as related symptoms thereof.
[0045] The terms "subject," "patient," "individual," etc. are not intended to be
limiting and can be generally interchanged. That is, an individual described as a "patient" does not necessarily have a given disease, but may be merely seeking medical advice. In some embodiments, a subject is a mammal. Non-limiting examples of mammals include humans, non-human primates {e.g., apes, gibbons, chimpanzees, orangutans, monkeys, macaques, and the like), domestic animals (e.g., dogs and cats), farm animals (e.g., horses, cows, goats, sheep, pigs) and experimental animals (e.g., mouse, rat, rabbit, guinea pig). In certain embodiments a subject is a non-human mammal. In certain embodiments, a subject is a rodent (e.g., a rat or mouse). In some embodiments, a subject is a human.
[0046] The term "diagnosis" refers to a relative probability that a disease or condition (e.g. cancer) is present in the subject. In some instances, as will be appreciated by any one of skill in the field of medicine, a diagnosis may not be absolute or definite.
[0047] The terms "virus", "bacteria", "oncovirus", "cancer virus", "tumor virus" are intended to encompass a class of triggers that are associated with the development of or are the cause of a cancer, either currently known or yet to be confirmed or discovered. In some embodiments, the cause can be related to the insertion of viral or bacterial DNA or RNA into the genome of the target cell.
[0048] "Vaccination regimes" means vaccines, for example but not limited to MMR, oral polio vaccine, injectable polio vaccine, and chicken pox including those administered during childhood or adulthood.
[0049] As used herein, "recall response," "adaptive immune response",
"recall immune response," "memory response," "memory immune response" and grammatical variations thereof means an immune response to an antigen to which a subject has previously been exposed. A recall or memory response is therefore an immune response subsequent to the initial antigen exposure and immune response. For example, a recall response may occur following exposure of the subject to the antigen for a second (secondary), third (tertiary), fourth, fifth, sixth, seventh, eighth, ninth, tenth, or any subsequent antigen exposure.
[0050] A recall or memory response is distinguished from a primary response to an antigen; a primary response is an immune response that occurs when a subject is exposed to an antigen for the first time. In a primary response, naive B and T cells become activated, develop higher specificity and affinity, expand, and develop memory cells. In contrast, recall immune responses are believed to be attributed to reactivation of long- lived, antigen-primed B and T lymphocytes that arise from differentiated B and T cells in a quiescent state. Thus, a "recall response" is an immune response in which antigen- primed cytotoxic T cell, Thl, Th2, and/or B cells participate.
[0051] As used herein a "target cell", "target cells" in singular or plural, or grammatical variations thereof, refer to cells, which are affected by a disease or disorder, contribute to a disease or disorder, predisposed to transform into a disease or disorder and/or are targets of treatment. Target cells are distinguished from healthy or non-disease cells in that healthy or non-disease cells have normal function or are not desired targets of treatment. Non-limiting examples of target cells comprise cells, tissue or derivatives thereof from cancer or any other desired targeted cells subset.
[0052] Target cells may include cancer cells that have been transformed by a virus or bacteria, including but not limited to a Burkitt's lymphoma cancer cell, nasopharyngeal cancer cell, Hodgkin lymphoma cancer cell and stomach cancer cell (Epstein Barr Virus), Kaposi sarcoma cancer cell (human herpes vims 8); cervical cancer cell, penis cancer cell, anus cancer cell, vagina cancer cell, vulva cancer cell, throat and mouth cancer cell (human papillomavirus), liver cancer ceil (hepatitis B and C viruses), breast cancer cell or gliobastoma multiform ceil (human cytomegalovirus), T cell leukemia cell and lymphoma cell (Human T-lymphotrophic virus 1), mesothelioma cell, brain cancer cell, bone cancer cell and lymphomas (Simian vims 40), stomach cancer cells (Helicobacter pylori and merkel cell cancer cell (Merkel cell poSyomavirus)
[0053] In some embodiments, a target cell is a cancer cell or neoplastic cell. In certain embodiments, a target cell is a metastatic cancerous cell. Non-limiting examples of a cancer cell or neoplastic cell include a carcinoma, sarcoma, neuroblastoma, cervical cancer cell, hepatocellular cancer cell, mesothelioma, glioblastoma, myeloma, lymphoma, neoplastic lymphocyte, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, lymphosarcoma, liposarcoma, osteosarcoma, chondrosarcoma, leiomyosarcoma, rhabdomyosarcoma or fibrosarcoma.
[0054] In some embodiments, a target cell is a cell that is or has been infected with a pathogen. In certain embodiments, a target cell is a cell that is or has been infected with a vims, bacteria, parasite or protozoa. [0055] A pathogen can be a mammalian pathogen. In some embodiments a pathogen is a human pathogen. A pathogen can be a virus, bacteria, parasite or protozoa. Non- limiting examples of a pathogen include a virus of the order Flaviviridae, Retroviridae, Orthomyxoviridae, Arenaviridae, or Mononegavirales, a virus of the family Togaviridae, Reoviridae, picornaviridae, herpesviridae or Bunyaviridae. In certain embodiments a virus is a human papillomavirus. In certain embodiments, a virus is a hepatitis B or hepatitis C virus. In some embodiments, a virus is a Merkel cell polyomavirus.
[0056] As used herein "package" and "agent" are used synonymously and refer to the compositions of the invention that are administered to a subject or patient as part of a treatment described herein.
[0057] "Delivery mechanisms" refer to mechanisms (chemical, biological, physical and otherwise) that are utilized to introduce a package into the patient's system and cells. The present invention is not limited to the delivery mechanisms disclosed herein and one skilled in the art would appreciate that other mechanisms known in the art, as a whole, in part and in combination, can be used to deliver the packages to the target and desired cells.
[0058] As used herein, the terms "treatment", "treat" and "prevent" may refer to any delay in onset, reduction in the frequency or severity of symptoms, amelioration of symptoms, improvement in patient comfort or function, decrease in the occurrence of a given condition or disease or condition or disease symptoms in a patient, decrease in severity of the condition or disease state etc. The effect of treatment can be compared to an individual or pool of individuals not receiving a given treatment, or to the same patient prior to, or after cessation of, treatment. As indicated above, treatment may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.
[0059] As used herein "antigen" refers to any protein, peptide, biological component, molecule or compound that induces an immune response. The term "epitope" refers to the specific region of the antigen that elicits the immune response. [0060] The terms "peptide" and "protein" refers to polymers of amino acid residues. The terms include amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer. In certain embodiments, a peptide or protein described herein includes an epitope.
[0061] As used herein "variant" and grammatical variations thereof, refers to a
protein or peptide that deviates from a reference protein or peptide sequence. Modified and variant proteins or peptides may therefore have greater or less activity or function than a reference protein or peptide but at least retain partial activity or function of the reference protein or peptide.
[0062] "Conservatively modified variants" applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, conservatively modified variants refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein, which encodes a polypeptide, also describes every possible silent variation of the nucleic acid. One of skill will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid, which encodes a polypeptide, is implicit in each described sequence with respect to the expression product, but not with respect to actual probe sequences.
[0063] As to amino acid sequences, one of skill will recognize that individual
substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid.
Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
[0064] The following eight groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
[0065] A "label" or a "detectable moiety" is a composition detectable by
spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means. For example, useful labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and proteins or other entities, which can be made detectable, e.g., by incorporating a radiolabel into a peptide or antibody specifically reactive with a target peptide. Any method known in the art for conjugating an antibody to the label may be employed, e.g., using methods described in Hermanson, Bioconjugate Techniques 1996, Academic Press, Inc., San Diego.
[0066] The term "recombinant" when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (non- recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
[0067] The term "heterologous" when used with reference to portions of a nucleic acid indicates that the nucleic acid comprises two or more subsequences that are not found in the same relationship to each other in nature. For instance, the nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged to make a new functional nucleic acid, e.g., a promoter from one source and a coding region from another source. Similarly, a heterologous protein indicates that the protein comprises two or more subsequences that are not found in the same relationship to each other in nature (e.g., a fusion protein).
[0068] As used herein, the terms "treatment", "treat" and "prevent" may refer to any delay in onset, reduction in the frequency or severity of symptoms, amelioration of symptoms, improvement in patient comfort or function, decrease in the occurrence of a given condition or disease or condition or disease symptoms in a patient, decrease in severity of the condition or disease state etc. The effect of treatment can be compared to an individual or pool of individuals not receiving a given treatment, or to the same patient prior to, or after cessation of, treatment. As indicated above, treatment may be complete (no detectable symptoms) or partial, such that fewer symptoms are observed than would likely occur absent treatment.
[0069] An "agent" as used herein refers to any molecule (e.g., antibody, nucleic acid, synthetic chemical, small chemical molecule, ligand mimetic) that can be administered to a subject. An agent that modulates an immune response is a molecule (e.g., antibody, nucleic acid, synthetic chemical, small chemical molecule) that when administered to a subject modulates an immune response in the subject. In certain embodiments, the agent modulates, inhibits, blocks, decreases, increases, enhances, promotes or elicits an acute or chronic immune response. In some embodiments, the agent modulates stimulation, activation proliferation, number or activity of immune proteins or cells (e.g., T cells, B cells, macrophages, monocytes, dendritic cells, cytokines). In certain embodiments, the agent modulates an innate immune response. In certain embodiments, the agent modulates a recall immune response. In particular embodiments, the agent modulates an immune response to one or more of the peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof. In particular aspects of the methods described herein an agent administered to a subject to treat cancer when administered to a subject in a therapeutically effective dose or amount. In some embodiments, an agent is an agent that binds one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
[0070] In some embodiments, the agent binds one or more of the proteins listed in
Table 1, or a subsequence, homologue, variant or derivative thereof. In some
embodiments, the agent binds the complex of an immune cell or immune protein bound to one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
[0071] Thus in particular embodiments, the agent modulates the activity or expression of one or more type of immune cell or protein (e.g., T cells, B cells, macrophages, monocytes, dendritic cells, cytokines) or a combination thereof. In some embodiments, the agent modulates the stimulation of T cells, activation of T cells, proliferation of T cells, number of T cells or activity of T cells, or a combination thereof. In some embodiments, the agent modulates the stimulation of B cells, activation of B cells, proliferation of B cells, number of B cells or activity of B cells, or a combination thereof. In some embodiments, the agent modulates stimulation, activation, proliferation, number or activity, or a combination thereof, of B cells generating antibodies (e.g., antibodies to one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof).
[0072] In some embodiments, the agent modulates production or activity of
cytokines, immune activators, chemokines, growth factors and T F family members (e.g., AREG, Granzyme, Histamine, IFNct/β, IFNy, IL12, IL15, IL16, IL17a-f, Π.1 β, 112, IL21, IL22, IL23, IL25, IL27, GM-CSF, IL33, IL4, IL5, IL6, IL7, IL8, IL9, LFA-1, LIF, lymphotoxin, Mac-1, MCP-1, ΜΙΡΙα, Perforin, TGF-β, TNF a, VLA-4, CCL1, CCL1 1, CLL17, CCL1, CCL20, CCL22, CCL3, CCL4, CCL5, CXCL10, CXCL13, CXCL8, RANTES, CXCL12, G-CSF, M-CSF, VEGF, PDGF, 4-1BB, APRIL, BAFF, CD27, CD30, CD30L, CD70, DR3, DR4, DR5, FAS, FASL, GITR, HVEM, LIGHT, LTB, OX40, Ox40L, TACI, TRAIL, TWEAK, IL1 1, IL14 or IL10).
[0073] In some embodiments, the agent modulates activity or expression of immune cells or proteins involved in an immune response to one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof. In certain
embodiments, the immune response is an innate immune response. In other embodiments, the immune response is a recall immune response. In particular embodiments, the agent modulates stimulation of T cells, activation of T cells, proliferation of T cells, number of T cells or activity of T cells, or a combination thereof, wherein the T cells are specific for one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof. In particular embodiments, the agent modulates stimulation of B cells, activation of B cells, proliferation of B cells, number of B cells or activity of B cells, or a combination thereof, wherein the B cells are specific for one or more of the proteins listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
[0074] Methods of treatment
[0075] Provided herein are methods of treating cancer in patients. The method involves selectively expressing in the target cells (e.g., cancer cells, viral, bacterial, parasite or protozoa infected cells) an antigen that the subject has previously developed immunity against. In particular embodiments, the method involves selectively expressing in target cells an antigen from a vaccine that the subject has previously received resulting in a recall immune response specific to the target cell. In particular embodiment, the antigen is from a vaccine the subject received in childhood including but not limited to one or more of the vaccines for chickenpox, measles, mumps, rubella, and polio.
[0076] In a preferred embodiment, the expression of particular antigens that are non- self immunogenic peptide antigens (e.g., proteins or epitopes) results in a signature labeling of the target cells with non-self immunogenic peptide antigens. The signature labeling is providing a means for the target cells to be singled out and differentiated from healthy ceils that are not expressing the particular antigens. In an embodiment, the expression of non-self immunogenic peptide antigens elicits a memory immune response against the target ceils that are expressing the particular antigens. In a preferred embodiment, memory T cells, e.g., CDS and/or CD4 T cells, specific to the non-self immunogenic peptides are reactivated.
[0077] In particular embodiments, the treatment comprises an antigen from a vaccine that is administered to a patient who has previously developed immunity against the antigen, resulting for example from childhood vaccination regimes, where the vaccine was effective in developing immunity against the disease for which the vaccine was designed. In particular embodiments, the patient has been vaccinated against a disease and has developed immunity to these infections such that immunity can be detected, boosted and measured. In certain, embodiment, the patient received vaccination against the disease during childhood. In other embodiments, the patient has been previously infected with a virus, bacteria, parasite or protozoa expressing the antigen and has developed immunity to the antigen due to this prior infection. In some embodiments, a subject has been previously vaccinated against a pathogen. In a preferred embodiment the developed immunity is against chickenpox, measles, mumps, rubella and polio.
[0078] In a preferred embodiment, the subject' s immune system is able to recall an immune response developed from a previous vaccination regime or infections. In some embodiments, the subject may be administered a booster vaccination to prime immune memory (e.g., increase antibody titers, memory T cells) prior to administration of a method or composition of the present invention. In an embodiment, immunity may be induced using other vaccination methods other than childhood vaccines.
[0079] In particular embodiments, the antigen is selectively expressed in the target cells through administration of a gene construct encoding an antigen wherein expression or presentation of the antigen requires a factor (e.g., protein, gene, S P, transcription factor) selectively expressed in the target cells. For example, the gene construct contains the antigen with a promoter driven only by a factor selectively expressed in the target cells (e.g., a cancer cell-specific transcription factor). In certain embodiments, this specific expression of the necessary factor in the target cells is a result of the target cells being infected by a virus, bacteria, parasite or protozoa such that there is expression of transformed viral genes or bacterial, parasitic or protozoa genes present in the target cells which encode for the necessary factors to express the antigen from the gene construct.
[0080] In certain embodiments, the gene construct comprises the antigen with a
promoter driven only by a factor selectively expressed in the target cells (e.g., a cancer cell-specific or infected cell-specific transcription factor). Thus, in certain embodiments, a gene construct that contains a promoter driven only by a factor expressed by target cells that have been infected with a particular virus, bacteria, protozoa or parasite is
administered to the subject, enters or is ingested by the subject cells and the viral, bacterial, parasitic or protozoa genes in the target cells enables the selective expression of proteins or peptides in these cells that are needed for expression of the antigen from the gene construct. Only the target cells that contain the viral, bacterial, parasitic or protozoa genes will express the antigen resulting in a recall immune response specific to the target cells.
[0081] In other embodiments, the antigen is specifically expressed in target cells by- targeted delivery of the antigen to the target cells. Non-limiting examples of methods for targeted delivery of the antigen to the target cells including administration by direct intratumoral injection, micro- or nano-particles, liposome, PEGylation, lipofectamine, adenovirus, lentivirus, vaccinia virus or other viral gene vector, and/or CRISPR/CAS9.
[0082] In an embodiment, the present invention comprise administering an optimized package comprising one or more antigens (e.g., peptide antigens). Non-limiting examples of peptide antigens include a peptide selected from Table 1, or a subsequence,
homologue, variant or derivative of a peptide listed in Table 1. In particular
embodiments, the optimized package included one or more epitopes from antigen against which the subject has immune memory, e.g., epitopes from childhood vaccines. In specific embodiments, the optimized package compri ses one or more CD4 and/or CDS T- cell epitopes, for example, one or more of the epitopes listed in Table 1, or a subsequence, homologue, variant or derivative thereof. In certain embodiments, the optimized package contains epitopes selected for their broad recognition by HLA class I molecules of subjects from diverse ethnic and geographical backgrounds.
[0083] In an embodiment, the method includes administering the antigen and epitopes in combination with factors that induce an increase of epitope presentation by MHC molecules (e.g., TAP expression; antennapedia homeodomain peptide) and/or activation and recruitment of APCs (i.e. GM-CSF, anti-immune suppressive agents, or chemokines). The increased expression accelerates the T-cell response and ability to clear the target cells. In an embodiment, the present invention provides a gene construct encoding the antigen and genes for increased expression of cytokines, immune activators, chemokines, growth factors and TNF family members to amplify the inflammatory T cell response by the recalled immune system. In different embodiments, the gene constructs comprises genes for one or more of AREG, Granzyme, Histamine, IFNa/β, ΠΤΝΓγ, IL12, IL15, IL16, IL17a-f, ILlb, 112, IL21, IL22, IL23, IL25, IL27, GM-CSF, IL33, IL4, IL5, IL6, IL7, IL8, IL9, LFA-1, LIF, lymphotoxin, Mac-1, MCP-1, ΜΙΡΙα, Perforin, TGF-β, TNFa, VLA-4, CCL1, CCL1 1, CLL17, CCL1, CCL20, CCL22, CCL3, CCL4, CCL5, CXCL10, CXCL13, CXCL8, RANTES, CXCL12, G-CSF, M-CSF, VEGF, PDGF, 4-1BB, APRIL, BAFF, CD27, CD30, CD30L, CD70, DR3, DR4, DR5, FAS, FASL, GITR, HVEM, LIGHT, LTB, OX40, OX40L, TACI, TRAIL, TWEAK, IL1 1, IL14 and IL10. 84] In particular embodiments, the treatment is directed towards hematological and solid tumors and cancer, including but not limited to tumors and cancers involving or caused by viral, parasitic or bacterial infection that can be targeted by local administration or tissue-targeting agents, or cancers of low mutational burden where reduced efficacy is observed following other cancer immunotherapies. Non-limiting examples of cancers that may be treated by the present invention include a neoplasia derived from lung, thyroid, head or neck, nasopharynx, throat, nose, sinus, brain, spine, breast, adrenal gland, pituitary gland, thyroid, lymph, mouth, esophagus, stomach, duodenum, ileum, jejunum intestine, colon, rectum, uterus, urethra, ovary, cervix, endometrium, bladder, testicle, penis, prostate, kidney, pancreas, liver, bone, bone marrow, lymph, blood, muscle, and skin; a carcinoma, sarcoma, neuroblastoma, hepatocellular cancer, mesothelioma, glioblastoma, myeloma, lymphoma, myeloma, leukemia, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, and a melanoma; a cervical cancer, breast cancer, liver cancer, pancreatic cancer, brain cancer, lung cancer, skin cancer, ovarian cancer, and a testicular cancer. In some embodiments, a cancer is an infection-induced cancer. Non-limiting examples of infection-induced cancers include a hepatocellular carcinoma, cervical cancer (human papilloniaviixis), head and neck squamous cell carcinoma (HNSCC), adult T-cell leukemia/lymphoma, liver cancer (hepatitis B and C viruses), Burkitt' s lymphoma and Kaposi sarcoma (Epstein-Barr virus), Merkel cell cancer (e.g., Merkel cell carcinoma) (Merkel cell polyomavirus), bladder cancer and gastric cancer. [0085] In some embodiments, a cancer comprises a low mutational burden, non- limiting examples of which include pilocytic astrocytoma, ALL, medulloblastoma, kidney chromophobe, thyroid, CLL, neuroblastoma, glioblastoma, and pancreatic cancer.
[0086] In some embodiments a method comprises administering a checkpoint blocker. In certain embodiments, a method comprises adoptive transfer of T-cells containing a chimeric antigen receptor (CAR).
Pharmaceutical compositions
[0087] In an embodiment, the present invention provides epitopes from childhood vaccines that can be administered in the methods described herein. Thus there is presently provided compositions comprising proteins and peptides comprising epitopes from childhood vaccines, including but not limited to chickenpox, measles, mumps, rubella, and polio. In certain embodiments, the protein or peptide comprises, consists or consists essentially of one or more of the amino acid sequences listed in Table 1, or a
subsequence, homologue, variant or derivative thereof.
[0088] In particular embodiments, the present invention provides a composition
comprising combinations of epitopes from childhood vaccines. Thus, in particular embodiments, the present invention provides combinations of one or more proteins or peptides each comprising, consisting or consisting essentially of a different amino acid sequence listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
[0089] In other embodiments, the present invention provides a composition
comprising a gene construct encoding one or more epitopes from childhood vaccines. Thus in certain embodiments, the present invention provides a gene construct encoding one or more of the amino acid sequences listed in Table 1, or a subsequence, homologue, variant or derivative thereof. In some embodiments a composition comprises a viral construct configured to express one or more of the amino acid sequences listed in Table 1, or a subsequence, homologue, variant or derivative thereof. In certain embodiments a composition comprises agents to improve peptide:HLA expression (e.g., TAP;
antennapedia homeodomain peptide). In certain embodiments a composition comprises agents to improve immune infiltration. In certain embodiments a composition comprises agents to modulate checkpoint inhibition. In certain embodiments a composition comprises T cells that are adoptively transferred containing CAR specific to one or more of the peptides listed in Table 1.
[0090] In different embodiments of the present invention, epitopes are selected for their broad recognition by HLA class I or class II molecules of subjects from diverse ethnic, racial and geographical backgrounds.
[0091] In a further embodiment, the present invention provides a uniquely optimized package and appropriate delivery system for introducing the treatment into the subject. Thus, in a preferred embodiment the composition of the present invention comprises viral, bacterial, parasitic, protozoa or cancer specific transformed promoter, epitopes, proteins, factors that increase epitope expression in MHC pockets and genes that increase cytokine expression. In particular embodiments, one or more epitopes are selected from the peptides in Table 1 to 10, or a subsequence, homologue, variant or derivative thereof.
[0092] In particular embodiments, the composition comprises CD4 and/or CD8 T-cell epitopes from the mumps vaccine. In particular embodiments, the composition comprises T-cell (e.g., CD8 and/or CD4 T cells) epitopes from the measles vaccine. In particular embodiments, the composition comprises CD4 and/or CD8 T-cell epitopes from the rubella vaccine. In particular embodiments, the composition comprises CD4 and/or CD8 T-cell epitopes from the polio vaccine. In particular embodiments, the composition comprises CD4 and/or CD8 T-cell epitopes from the chicken pox/varicella vaccine, comprising one or more or a combination of the peptides listed in Table 1.
[0093] In particular embodiments, the composition comprises proteins, peptides or epitopes from two or more different vaccines (e.g., measles, mumps, rubella, polio or chicken pox vaccine). In different embodiments, the composition comprises proteins, peptides or epitopes from two or more, three or more, four or more, or five or more different vaccines. In different embodiments, the composition may contain the same number of proteins, peptides or epitopes from each of the two or more vaccines or may contain a different number of proteins, peptides or epitopes from each of the two or more vaccines. [0094] As each T-cell epitope binds only a subset of naturally occurring HLA class II molecules in the human population and this repertoire of HLA class II molecules varies from one person to another and from one ethnic population to another, in certain embodiments of the presentation invention, it is desirable to select a mix of several peptides covering different HLA class II molecules to generate an immune response in a broad segment of the population. Further, in other embodiments of the present invention, it is desirable to select peptides that elicit an immune response (e.g., T-cell response) in a significant fraction of patients with the disease, disorder or condition to be treated. Thus, in particular embodiments, it is desirable to select an epitope or combination of epitopes that have a HLA class II repertoire covering a high fraction of a worldwide population, and which also produce an immune response (e.g., T cell response) in a significant fraction of patients with the disease, disorder or condition to be treated.
[0095] As each T cell epitope binds only a subset of naturally occurring HLA class I molecules in the human population and this repertoire of HLA class I molecules varies from one person to another and from one ethnic population to another, in certain embodiments of the presentation invention, it is desirable to select a mix of several peptides covering different HLA class I molecules to generate an immune response in a broad segment of the population. Further, in other embodiments of the present invention, it is desirable to select peptides that elicit an immune response (e.g., T-cell response) in a significant fraction of patients with the disease, disorder or condition to be treated. Thus, in particular embodiments, it is desirable to select an epitope or combination of epitopes that have a HLA class I repertoire covering a high fraction of a worldwide population, and which also produce an immune response (e.g., T-cell response) in a significant fraction of patients with the disease, disorder or condition to be treated.
[0096] In particular embodiments, it is desirable for the methods or compositions of the present invention to include as few peptides as possible, but at the same time to be able to treat the majority of patients in a worldwide population with the same method or composition. Such a method or should contain a combination of peptides capable of binding to a HLA Class II and/or Class I repertoire covering a high fraction of a worldwide population, and the resulting peptide-HLA complexes should be recognized as epitopes (e.g., T cell epitopes) in the subject so as to induce an immune response.
[0097] In particular embodiments, the composition of the present invention further comprises cytokines, immune activators, chemokines, growth factors or T F family members or a gene for increased expression of cytokines, immune activators,
chemokines, growth factors or TNF family members to amplify the inflammatory T-cell response by the recalled immune system. In different embodiments, the composition comprises one or more of or genes for one or more of AREG, Granzyme, Histamine, IFNa/β, IFNy, IL12, IL15, IL16, IL17a-f, ILlb, 112, IL21, IL22, IL23, IL25, IL27, GM- CSF, IL33, IL4, IL5, IL6, IL7, IL8, IL9, LFA-1, LIF, lymphotoxin, Mac-1, MCP-1, ΜΙΡΙα, Perforin, TGF-β, TNFa, VLA-4, CCL1, CCL1 1, CLL17, CCL1, CCL20, CCL22, CCL3, CCL4, CCL5, CXCL10, CXCL13, CXCL8, RANTES, CXCL12, G-CSF, M-CSF, VEGF, PDGF, 4-lBB, APRIL, BAFF, CD27, CD30, CD30L, CD70, DR3, DR4, DR5, FAS, FASL, GITR, HVEM, LIGHT, LTB, OX40, Ox40L, TACI, TRAIL,
TWEAK, IL1 1, IL14 and IL10.
Delivery Methods
[0098] In an embodiment, particular compositions are delivered to the target disease cells via one or more delivery systems depending on the indication, disease state, severity, clinical utility and other relevant parameters that may impact the desired efficacy of delivery and expression of the T-cell epitopes and/or antigens. In a preferred embodiment, the delivery system is engineered to deliver the package to a specific cell subset of interest.
[0099] In particular embodiments, the composition is delivered to the target cell and other cells of interest using delivery methods comprising one or more but at least one of the methods from the group comprising: replicating or non-replicating viral vectors such as Adenovirus (AdV); Adeno-associated virus (AAV); Lentivirus; Vaccinia virus;
Oncolytic viruses; Respective disease causing live, attenuated, or inactivated virus;
Micro/nano particles; Adjuvant emulsified; Electroporation; Bacteriophage; Lipid particles (e.g., Lipofectamine); Exosomes; Mineral oil / Freund's adjuvant; Hyperglycosylation; Pegylation; Poly(lactic/gly colic) acid; Direct injection; Conjugation via cell penetrating peptides/proteins; Lysosomal targeting agents; Gene delivery (e.g., Crispr-Cas9, flox/LoxP). The one or more delivery methods can be used in combination, alone, and in combinations in part thereof. Parts of particular delivery methods may be combined to other delivery methods to form new delivery methods. Other delivery methods well known in the art can be utilized to delivery the composition to the target cells. In particular embodiments, one or more epitopes are selected from the peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof, are delivered to the target cell using the delivery methods disclosed herein.
[00100] A peptide's immunogenicity, more particularly one or more epitopes from the peptides listed in Table 1, or a subsequence, homologue, variant or derivative thereof, may be modified. In particular embodiments, the modifications to the peptides and/or antigens administered comprise one or more of the modifications from the group comprising: Oxidization; Reduction; Citrullination; Glycatation (including but not limited to: prenylation, myristylation, palmitolation); Nitration; Di-sulfide bonding (Cysteine); Transglutamination; Deamidation; Misfolding; Single amino acid substitutions; Genetic frameshift mutations. The modifications are not intended to be limiting by scope or category but rather an illustration of how the peptides may be constructed in the delivery mechanisms.
[00101] The exact formulation and route of administration for a composition for use according to the methods of the invention described herein can be chosen by the individual physician in view of the patient's condition. See e.g., Fingl et al. 1975, in "The Pharmacological Basis of Therapeutics," Ch. 1 p. 1; which is incorporated herein by reference in its entirety. Any suitable route of administration can be used for
administration of a composition or compound described herein. Non-limiting examples of routes of administration include topical or local (e.g., transdermally or cutaneously, (e.g., on the skin or epidermis), in or on the eye, intranasally, transmucosally, in the ear, inside the ear (e.g., behind the ear drum)), enteral (e.g., delivered through the
gastrointestinal tract, e.g., orally (e.g., as a tablet, capsule, granule, liquid, emulsification, lozenge, or combination thereof), sublingual, by gastric feeding tube, rectally, and the like), by parenteral administration (e.g., parenterally, e.g., intravenously, intra-arterially, intramuscularly, intraperitoneally, intradermally, subcutaneously, intracavity, intracranially, intraarticular, into a joint space, intracardiac (into the heart), intracavernous injection, intralesional (into a skin lesion), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intrauterine, intravaginal, intratumoral, intravesical , intravitreal), the like or combinations thereof.
[00102] In some embodiments, a composition herein is provided to a subject. A composition that is provided to a subject is often provided to a subject for self- administration or for administration to a subject by another (e.g., a non-medical professional). For example, a composition described herein can be provided as an instruction written by a medical practitioner that authorizes a patient to be provided a composition or treatment described herein (e.g., a prescription). In another example, a composition can be provided to a subject wherein the subject self-administers a composition orally, intravenously or by way of an inhaler, for example.
[00103] Compositions herein can be formulated to be compatible with a particular route of administration or use. Compositions for parenteral, intradermal, or subcutaneous administration can include a sterile diluent, such as water, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents. The preparation may contain one or more preservatives to prevent microorganism growth (e.g., antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as EDTA; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose). In certain embodiments, a composition herein is substantially free of a chelator (e.g., a zinc chelator, e.g., EDTA or EGTA).
[00104] Compositions for injection include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, NJ), hydroxypropyl-b- cyclodextrin or other cyclodextrin derivatives, phosphate buffered saline (PBS). The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and polyethylene glycol), and suitable mixtures thereof. Fluidity can be maintained, for example, by the use of a coating such as lecithin, or by the use of surfactants. Antibacterial and antifungal agents include, for example, parabens, chlorobutanol, phenol, ascorbic acid and thimerosal. Including an agent that delays absorption, for example, aluminum monostearate and gelatin can prolong absorption of injectable compositions. Polysorbate 20 and polysorbate 80 can be added into the formulation mixture, for example, up to 1%. Other non-limiting additives include histidine HC1, α,α-trehalose dehydrate, sucrose, lactose or mannitol.
[00105] Alternatively, one can administer compositions for use according to the methods of the invention in a local rather than systemic manner, for example, via direct application to the skin, mucous membrane or region of interest for treating, including using a depot or sustained release formulation.
[00106] In some embodiments, active ingredients (e.g., peptides or vectors) can be administered alone. In other embodiments, active ingredients can be administered in combination with one or more additional materials, for example, as two separate compositions or as a single composition where the additional material(s) is (are) mixed or formulated together with an active ingredient. For example, without being limited thereto, an active ingredient can be formulated with additional excipients, or additional active ingredients. In some aspects, when administered in the forms described herein the active ingredients can attain concentrations at a target tissue such as the nose, mucous membranes, the bronchi, the skin, etc. that cannot be attained by the usual intravascular administration of the active ingredients.
[00107] The pharmaceutical compositions can be manufactured by any suitable manner, including, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes.
[00108] Pharmaceutical compositions for use in accordance with the invention thus can be formulated in any suitable manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation can depend upon the route of administration chosen. In particular, any suitable formulation, ingredient, excipient, the like or combinations thereof as listed in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 18th edition, 1990, can be used with a composition described herein. The various materials listed herein, alone or in combination, can be incorporated into or used with the materials described in Remington's. Any suitable techniques, carriers, and excipients can be used, including those understood in the art; e.g., in Remington's Pharmaceutical Sciences, above. The pages in the attached Appendix from Remington's Pharmaceutical Sciences are incorporated herein by reference in their entirety, including without limitation for all of the types of formulations, methods of making, etc.
Formulations
[00109] In some embodiments, the composition may be formulated, for example, as a topical formulation. The topical formulation may include, for example, a formulation such as a gel formulation, a cream formulation, a lotion formulation, a paste formulation, an ointment formulation, an oil formulation, and a foam formulation. The composition further may include, for example, an absorption emollient.
[00110] In some embodiments, at least part of the affected area of the mammal is contacted with the composition on a daily basis, on an as-needed basis, or on a regular interval such as twice daily, three times daily, every other day, etc. The composition can be administered for a period of time ranging from a single as needed administration to administration for 1 day to multiple years, or any value there between, (e.g., 1 - 90 days, 1 - 60 days, 1 - 30 days). The dosages described herein can be daily dosages or the dosage of an individual administration, for example, even if multiple administrations occur (e.g., 2 sprays into a nostril).
[00111] Some embodiments relate to methods of treating or preventing cancer through administration of compositions described herein to the upper respiratory track/bronchi in a mammal in need thereof, for example, by contacting at least part of the upper respiratory tract/bronchi of a mammal with a therapeutically effective amount of a composition as described above or elsewhere herein. The composition can be, for example, formulated as an aerosol formulation, including formulated for use in a nebulizer or an inhaler. The composition further may include other pharmaceutically acceptable components such as a preservative.
[00112] Other embodiments relate to aerosol compositions that include, for example, a composition as described herein and an aerosolized pharmaceutically acceptable carrier solution or dry powder. The compositions may be formulated, for example, to be substantially absorbed by a bronchus. The compositions also may include, for example, one or more of dichlorodifluoromethane, trichlorofluoromethane,
dichlorotetrafluoroethane, carbon dioxide, and the like. The compositions can be formulated for use in a nebulizer or an inhaler, for example.
Pharmaceutical Carriers
[00113] The term "carrier" defines a chemical compound that facilitates the incorporation of a compound into cells or tissues. For example dimethyl sulfoxide (DMSO) is a commonly utilized carrier as it facilitates the uptake of many organic compounds into the cells or tissues of an organism. In some embodiments, a pharmaceutical carrier for a composition described herein can be selected from castor oil, ethylene glycol, monobutyl ether, diethylene glycol monoethyl ether, corn oil, dimethyl sulfoxide, ethylene glycol, isopropanol, soybean oil, glycerin, zinc oxide, titanium dioxide, glycerin, butylene glycol, cetyl alcohol, and sodium hyaluronate.
[00114] In certain embodiments comprising hydrophobic excipients, additives, or other hydrophobic components, a pharmaceutical carrier for certain of such hydrophobic compounds can be a co-solvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. A common co-solvent system used is the VPD co-solvent system, which is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant POLYSORBATE 80™, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. Naturally, the proportions of a co-solvent system can be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the co-solvent components can be varied: for example, other low-toxicity nonpolar surfactants can be used instead of POLYSORBATE 80™; the fraction size of polyethylene glycol can be varied; other biocompatible polymers can replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides can substitute for dextrose. [00115] Alternatively, other delivery systems for hydrophobic pharmaceutical compounds, excipients, or additives can be employed, if required. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs and drug compositions. Certain organic solvents such as dimethyl sulfoxide also can be employed, although usually at the cost of greater toxicity. Additionally, the compounds can be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. The pharmaceutical compositions described herein can be administered to a patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carriers or excipient(s). The compounds and compositions can be formulated with salts or excipients, such as for example, sodium or meglumine. Techniques for formulation and administration of the compounds of the instant application can be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, 18th edition, 1990.
[00116] Furthermore, the compounds and compositions used herein can preferably be stable over an extended period of time, for example on the order of months or years. Compositions described herein, in some embodiments, comprise a preservative. The preservative can comprise a quaternary ammonium compound, such as benzalkonium chloride, benzoxonium chloride, benzethonium chloride, cetrimide, sepazonium chloride, cetylpyridinium chloride, or domiphen bromide (BRADOSOL®). The preservative can comprise an alkyl-mercury salt of thiosalicylic acid, such as thiomersal, phenylmercuric nitrate, phenylmercuric acetate or phenylmercuric borate. The preservative can comprise a parabens, such as methylparaben or propylparaben. The preservative can comprise an alcohol, such as chlorobutanol, benzyl alcohol or phenyl ethyl alcohol. The preservative can comprise a biguanide derivative, such as chlorohexidine or polyhexamethylene biguanide. The preservative can comprise sodium perborate, imidazolidinyl urea, and/or sorbic acid. The preservative can comprise stabilized oxychloro complexes, such as known and commercially available under the trade name PURITE®). The preservative can comprise polyglycol-polyamine condensation resins, such as known and commercially available under the trade name POLYQUART® from Henkel KGaA. The preservative can comprise stabilized hydrogen peroxide generated from a source of hydrogen peroxide for providing an effective trace amount of resultant hydrogen peroxide, such as sodium perborate tetrahydrate. The preservative can be benzalkonium chloride.
[00117] The preservative can enable a composition to be used on multiple occasions.
The preservative can reduce the effects of one or more of acid exposure, base exposure, air exposure, heat, and light on the active ingredient. The compounds and compositions used herein can include any suitable buffers, such as for example, sodium citrate buffer and/or sequestering agents, such as edetate disodium sequestering agent. Ingredients, such as meglumine, may be added to adjust the pH of a composition or compound described herein. Compounds and compositions described herein may comprise sodium and/or iodine, such as organically bound iodine. Compositions and compounds used herein may be provided in a container in which the air is replaced by another substance, such as nitrogen.
Dosages and Products
[00118] Certain embodiments provide pharmaceutical compositions suitable for use in the technology, which include compositions where the active ingredients are contained in an amount effective to achieve its intended purpose. A "therapeutically effective amount" means an amount to prevent, treat, reduce the severity of, delay the onset of or inhibit a symptom of a cancer. The symptom can be a symptom already occurring or expected to occur. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
[00119] In other embodiments, a therapeutically effective amount can describe the amount necessary for a significant quantity of the composition to contact the desired region or tissue where prevention or treatment of a cancer is desired.
[00120] Compositions described herein can be administered at a suitable dose, e.g., at a suitable volume and concentration depending on the route of administration. Within certain embodiments of the invention, dosages of an administered composition, vector, factor, agent or peptide can be from 0.001 - 200 grams, or 0.001 - 50 grams, 0.001 - 10 grams, etc. In some aspects the amount can be, for example, from 0.001 - 0.1 grams, 0.1 - 5 grams, 5 - 10 grams, 10 - 15 grams, 15 - 20 grams, 20 - 25 grams, 25 - 30 grams, 30 - 35 grams, 35 - 40 grains, 40 - 45 grams, 45 - 50 grams and 50 - 200 grams. In some non- limiting aspects, a composition, vector, factor, agent or peptide can be provided in a concentration, for example, of 1 mg/kg to 5000 mg/kg, 50 mg/kg to 1000 mg/kg, 50 mg/kg to 500 mg/kg, 150 mg/kg to 350 mg/kg or 350 mg/kg to 1000 mg/kg. In certain embodiments a composition, vector, factor, agent or peptide described herein can be administered at a concentration of at least 50 mg/kg, at least 100 mg/kg, at least 150 mg/kg, at least 200 mg/kg, at least 150 mg/kg, at least 200 mg/kg, at least 250 mg/kg, at least 300 mg/kg, at least 350 mg/kg, at least 400 mg/kg, at least 450 mg/kg, or at least 500 mg/kg. In certain embodiments a composition, vector, factor, agent or peptide described herein can be administered at a concentration of about 100 mg/kg, about 150 mg/kg, about 200 mg/kg, about 150 mg/kg, about 200 mg/kg, about 250 mg/kg, about 300 mg/kg, about 350 mg/kg, about 400 mg/kg, about 450 mg/kg, or about 500 mg/kg. Volumes suitable for intravenous administration are well known. For example, 50 ml - 100 ml of a composition, vector, factor, agent or peptide at a working concentration of between about 200 mg/kg to about 350 mg/kg can be safely administered intravenously to an adult human subject.
[00121] In some embodiments, the amount delivered can be any suitable amount, for example, in order to contact the desired tissue in a therapeutically effective manner. In certain embodiments, the compositions can be delivered to the nose, and the amount delivered to each nostril can be from about 20 microliters to about 1500 microliters, 50 microliters to about 1000 microliters, or 50 microliters to about 500 microliters. In some embodiments, composition described herein can be delivered to the nose in a volume of about 50, 100, 200, 300, 400 or 500 microliters for example.
[00122] The compositions can, if desired, be presented in a pack or dispenser device, which can contain one or more unit dosage forms containing the active ingredient. The pack can for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device can be accompanied by instructions for administration. The pack or dispenser can also be accompanied with a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the drug for human or veterinary administration. Such notice, for example, can be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier can also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
Kits
[00123] In some embodiments the compositions, formulations, combination products and materials described herein can be included as part of kits, which kits can include one or more of the compositions, vectors, factors, agents or peptides described herein, formulations of the same, combination drugs and products and other materials described herein. In some embodiments the products, compositions, kits, formulations, etc. can come in an amount, package, product format with enough medication to treat a patient for 1 day to 1 year, 1 day to 180 days, 1 day to 120 days, 1 day to 90 days, 1 day to 60 days, 1 day to 30 days, or any day or number of days there between.
[00124] The invention provides kits including a composition, vector, factor, agent or peptide of the invention, combination compositions and pharmaceutical formulations thereof, packaged into suitable packaging material. A kit optionally includes a label or packaging insert including a description of the components or instructions for use in vitro, in vivo, or ex vivo, of the components therein. Exemplary instructions include instructions for a method, treatment protocol or therapeutic regimen.
[00125] A kit can contain a collection of such components, e.g., two or more conjugates alone, or in combination with another therapeutically useful composition (e.g., an anti-proliferative or immune-enhancing drug). The term "packaging material" refers to a physical structure housing the components of the kit. The packaging material can maintain the components sterilely, and can be made of material commonly used for such purposes (e.g., paper, corrugated fiber, glass, plastic, foil, ampules, vials, tubes, etc.).
[00126] Kits can include labels or inserts. Labels or inserts include "printed matter," e.g., paper or cardboard, or separate or affixed to a component, a kit or packing material (e.g., a box), or attached to an ampule, tube or vial containing a kit component. Labels or inserts can additionally include a computer readable medium, optical disk such as CD- or DVD-ROM/RAM, DVD, MP3, magnetic tape, or an electrical storage media such as RAM and ROM or hybrids of these such as magnetic/optical storage media, FLASH media or memory type cards.
[00127] Labels or inserts can include identifying information of one or more components therein, dose amounts, clinical pharmacology of the active ingredient(s) including mechanism of action, pharmacokinetics (PK) and pharmacodynamics (PD). Labels or inserts can include information identifying manufacturer information, lot numbers, manufacturer location and date.
[00128] Labels or inserts can include information on a condition, disorder, disease or symptom for which a kit component may be used. Labels or inserts can include instructions for the clinician or for a subject for using one or more of the kit components in a method, treatment protocol or therapeutic regimen. Instructions can include dosage amounts, frequency or duration, and instructions for practicing any of the methods, treatment protocols or therapeutic regimes set forth herein. Kits of the invention therefore can additionally include labels or instructions for practicing any of the methods and uses of the invention described herein.
[00129] Labels or inserts can include information on any benefit that a component may provide, such as a prophylactic or therapeutic benefit. Labels or inserts can include information on potential adverse side effects, such as warnings to the subject or clinician regarding situations where it would not be appropriate to use a particular composition. Adverse side effects could also occur when the subject has, will be or is currently taking one or more other medications that may be incompatible with the composition, or the subject has, will be or is currently undergoing another treatment protocol or therapeutic regimen which would be incompatible with the composition and, therefore, instructions could include information regarding such incompatibilities.
[00130] Kits can additionally include other components. Each component of the kit can be enclosed within an individual container and all of the various containers can be within a single package. Invention kits can be designed for cold storage. [00131] The invention has been described herein using specific embodiments for the purposes of illustration only. It will be readily apparent to one of ordinary skill in the art, however, that the principles of the invention can be embodied in other ways. Therefore, the invention should not be regarded as being limited in scope to the specific embodiments disclosed.
EXAMPLES
Example 1
[00132] Effective immunity against a pathogen entails (i) the development of highly specific, neutralizing, and non-self reactive antibodies and T cells and (ii) results in the development of immune memory cells that patrol the body and provide lifelong protection. Immune memory cells are uniquely positioned to rapidly and robustly expand and neutralize a specific pathogen if repeat exposure occurs. Vaccines provide a mechanism to promote safe development of long-lived immune memory against a pathogen(s) by avoiding the deleterious effects of actual pathogen infection. The inventor approach activates and redirects the long-lived immune memory derived from vaccines to fight cancer.
[00133] Peptide antigens are used in combination with other cancer and cancer- immune-based therapies to increase efficacy of patient treatment. Peptide antigens disclosed herein are delivered in combination with viral vectors including oncolytic viruses like T-Vec (Imlygic). T-Vec is the first oncolytic virus approved in the US and has been shown of utility in the treatment of patients with recurrent melanoma.
[00134] Peptide antigens disclosed herein are cloned into different viruses, including the herpes simplex virus type 1 (HSV1) virus, adenovirus (AdV), adeno-associated virus (AAV) and vaccinia virus, for expression in tumor tissue to combine oncolytic potential with increased tumor immunogenicity. Adenovirus (AdV), adeno-associated virus (AAV) and vaccinia virus are in advanced clinical development. These viruses have potential for selective tumor-mediated killing through direct tumor injection or engineered tumor specificity after systemic delivery. [00135] CTLA-4 (Ipilumimab), PDL-1 and PD1 (Nivolumab / Pembrolizumab) are two presently approved targets of antibody-based immune checkpoint inhibitors. These antibodies non-specifically block inhibitory receptors on T cells that maintain T cells in an activated state. Recent publications highlight the efficacy of these therapies may be tied to the mutational burden of the specific tumor and in tumors with low mutational burden, checkpoint inhibitors may be of limited utility. Peptide antigens disclosed herein are delivered to the tumor in combination with checkpoint inhibitors to bring immune memory derived from childhood vaccines to more robustly kill cancerous and precancerous tissue.
[00136] Chimeric Antigen receptor T cell therapies are presently in advanced clinical trials for various cancers. These therapies entail the isolation of patient T cells and re- engineering of T-cell specificity to the target of choosing. While these therapies have shown therapeutic promise in single-cell type tumors such as B cell lymphomas, solid tumors that are under immune-suppression and lack a single marker for targeting are presently intractable. In particular instances, the introduction of peptide antigens disclosed herein and simultaneous engineering of T cells with specificity for the delivered epitopes enable robust and targeted killing of epitope expressing cells.
[00137] Several cytokine and TNF-super family based therapies are in advanced clinical development for the treatment of cancer. Specifically, tumor specific expression of cytokines like IL12 and TNF superfamily members such as OX40 reduce tumor immune suppression and promote inflammation. In fact, oncolytic virus therapies are in development in which IL12 and OX40 are expressed by the therapeutic virus. In particular instances, combination with peptide antigens disclosed herein increase T cell infiltration and activation in the tumor tissue.
[00138] Rituximab represents a particularly successful candidate in the treatment of B cell-based tumors such as non-Hodgkin's lymphoma. Therapeutic antibodies recognize surface antigens of the tumor tissue and induce immune responses through phagocytosis, complement deposition, and cell cytotoxicity. In particular instances, combined delivery of peptide antigens described herein with mAbs increase the immune potential of the tumor leading to increased killing of tumor tissue by the immune system. [00139] Popular developing therapies in clinical development utilize cancer-specific mutations to enable generation or expansion of tumor-specific immunity within a patient. In brief, patient cancer-specific mutations are identified by a variety of sequencing methods and mutation gene products are identified and assessed for the ability to be bound by HLA molecules or the existence of mutation-specific T cells within the donor. Vaccination of these peptides/antigens in combination with adjuvant and checkpoint blockers are then used to expand mutation specific T cells. Various tumors have lower mutational burden and therefore cancer vaccine approaches are of limited practical utility. In particular instances, combination of administration of patient mutation specific peptides with childhood vaccine peptide antigens improve the therapeutic potential against the tumor.
[00140] While not standardly considered an immune-based therapy, increasing evidence of an immune response against the cancer is seen upon tumor killing by radiation or chemotherapy approaches. This abscopal effect is tied to the presence of cancer-specific mutations that can generate adaptive immune responses in likeness to those used in cancer vaccines. In particular instances, selected delivery of vaccine-derived tumor tissue and co-treatment with radiation/chemo therapy lead to a broader more immunogenic based cell death that leads to improved cancer treatment.
[00141] In other approaches, T cells are brought into closer proximity to malignant tissue by fusion of two antibodies or generation of bi-specific antibodies. Such an approach has shown early clinical potential by combined target of CD 19 and T-cell markers such as CD3 to bring T cells in proximity to the cellular target and promote T- cell-mediated killing. In particular instances, delivery of peptide antigens described herein to target tissue and co-administration of BiTE to promote T-cell infiltration into the target tissue improve the frequency or concentration of T cells with potential to specifically kill the tumor tissue. Example 2
[00142] Methods for prediction of human HLA class I alleles were adapted from
published and validated methods from the La Jolla Institute for Allergy and Immunology. All 9- and 10-mer peptides were predicted for their binding affinity to 27 MHC class I molecules. A panel of 16 HLA A (A*01 :01, A*26:01, A*32:01, A*02:01, A*02:03, A*02:06, A*68:02, A*2301, A*24:02, A*03 :01, A* l l :01, A*30:01, A*31 :01, A*33 :01, and A*68:01) and 11 HLA B alleles (B*40:01, B*44:02, B*44:03, B*57:01, B*58:01, B* 15:01 B*07:02, B*35:01, B*51 :01, B*53 :01, and B*08:01) were selected,
which account for 97% of HLA A and B allelic variants in most ethnicities. Binding predictions were performed using the consensus prediction tool available on the IEDB web site (Accessed online at <URL:http:/www.iedb.org/>). For each allele and length combination, peptides from each included vaccine strain were selected if they were in the top 1% of binders. Redundant sequences across various vaccine strains were removed and non-redundant sequences are listed in Table 1.
[00143] The binding affinity of 15-mer peptides derived from childhood vaccines were predicted using the panel of 27 HLA class II DRB1, 3/4/5, DQA1/DQB1, and DPB 1 molecules that provide broad ethnic and geographic population coverage using a consensus prediction approach. Peptides with predicted binding scores in the top 20% for a given allele were considered potential binders. Peptides predicted to bind 13 or more HLA molecules at this threshold were considered promiscuous binders.
Example 3
[00144] The mechanism by which viruses infect their host is well understood in the art.
Following viral infection, viral genes can become incorporated into the host genome. This viral transformation event can lead to uncontrolled proliferation and the development of various diseases and disorders including cancer. Viral, parasitic and bacterial transformation or infection of healthy cells has been identified as a common underlying cause of a great number of human cancers. [00145] In cancers that have been linked to viral gene transformation, specific viral genes are incorporated into the genome of cancers cells and maintain expression within the cancerous cells throughout the lifecycle of the cancer cell. For example, in cervical cancer, which has been linked to infection by human papillomaviruses, transformation of viral genes E6 and E7 has been widely established.
[00146] Today, it is now known that about 15%-20% of cancers have a viral cause, including Burkitt's lymphoma and Kaposi sarcoma (Epstein-Barr virus); cervical cancer (human papillomavirus); liver cancer (hepatitis B and C viruses) and Merkel cell cancer (Merkel cell polyomavirus), which represent a few notable cancers caused by viral transformation with high incidence and impact to society.
[00147] Similarly, chronic inflammation triggered by bacteria, can lead to the production of DNA-damaging molecules that cause mutations; this, combined with signals to produce new cells and grow new blood vessels— hallmarks of wound healing— can create a fertile ground for the emergence and growth of cancer. For example, a clear link between bacterial infection and cancer was established in the early 2000s when it was shown that the bacterium Helicobacter pylorus is a major cause of gastric cancer. In addition, parasitic infections have been linked to certain types of cancer including bladder cancer.
[00148] In particular instances, specific expression of the peptide antigens in target cells is driven by viral or bacterial factors present only in the target cells.
Example 4
[00149] The measurement of pre-existing immune responses derived from childhood vaccines are performed to assess and quantify the presence of circulating antibodies and/or frequency of vaccine-specific immune cells in the individual. The proteins and/or peptides listed in Table 1 are used as part of a kit to specifically measure B cell response by antibody ELISA and/or the CD4 and CD8 T-cell response by cytokine ELISA or ELISPOT to inform the frequency and functionality of vaccine immune memory of the patient. The results obtained from use of the diagnostic kit inform whether a patient has sufficient immune memory for therapeutic application or would benefit from receipt of a booster vaccination prior to administration of the therapeutic to increase the frequency of vaccine-specific immune cells that recognize and promote an anti-tumor immune response.
TABLES
Table 1 . 642 Class I non-redundant predicted peptides derived from ORF22 of the Varicella zoster vaccine Oka strain (Merck). Peptides are 9 and 10 mer in length.
Figure imgf000046_0001
Varicella-zoster virus (strain Oka) ORF22 224 233 EEALSSAVTL
Varicella-zoster virus (strain Oka) ORF22 226 234 ALSSAVTLI
Varicella-zoster virus (strain Oka) ORF22 226 235 ALSSAVTLIY
Varicella-zoster virus (strain Oka) ORF22 227 235 LSSAVTLIY
Varicella-zoster virus (strain Oka) ORF22 233 241 LIYGSCDTY
Varicella-zoster virus (strain Oka) ORF22 233 242 LIYGSCDTYF
Varicella-zoster virus (strain Oka) ORF22 234 242 IYGSCDTYF
Varicella-zoster virus (strain Oka) ORF22 239 247 DTYFTDEQY
Varicella-zoster virus (strain Oka) ORF22 242 250 FTDEQYCEK
Varicella-zoster virus (strain Oka) ORF22 252 260 VTAQHPLLL
Varicella-zoster virus (strain Oka) ORF22 256 264 HPLLLSPPN
Varicella-zoster virus (strain Oka) ORF22 261 270 SPPNSTTIVL
Varicella-zoster virus (strain Oka) ORF22 262 270 PPNSTTIVL
Varicella-zoster virus (strain Oka) ORF22 272 280 KSSIVPLHQ
Varicella-zoster virus (strain Oka) ORF22 272 281 KSSIVPLHQN
Varicella-zoster virus (strain Oka) ORF22 283 292 GESVSLEATL
Varicella-zoster virus (strain Oka) ORF22 288 296 LEATLHSTL
Varicella-zoster virus (strain Oka) ORF22 291 300 TLHSTLTNTV
Varicella-zoster virus (strain Oka) ORF22 294 302 STLTNTVAL
Varicella-zoster virus (strain Oka) ORF22 296 305 LTNTVALDPR
Varicella-zoster virus (strain Oka) ORF22 299 308 TVALDPRCSY
Varicella-zoster virus (strain Oka) ORF22 303 311 DPRCSYSEV
Varicella-zoster virus (strain Oka) ORF22 305 314 RCSYSEVDPW
Varicella-zoster virus (strain Oka) ORF22 306 314 CSYSEVDPW
Varicella-zoster virus (strain Oka) ORF22 309 317 SEVDPWHAV
Varicella-zoster virus (strain Oka) ORF22 309 318 SEVDP HAVL
Varicella-zoster virus (strain Oka) ORF22 319 327 ETTSTGSGV
Varicella-zoster virus (strain Oka) ORF22 319 328 ETTSTGSGVL
Varicella-zoster virus (strain Oka) ORF22 326 335 GVLDCRRRRR
Varicella-zoster virus (strain Oka) ORF22 330 338 CRRRRRPSW
Varicella-zoster virus (strain Oka) ORF22 360 369 STDNLHKPA
Varicella-zoster virus (strain Oka) ORF22 366 374 KPAKKVLKF
Varicella-zoster virus (strain Oka) ORF22 383 392 KTQVAHVLPR
Varicella-zoster virus (strain Oka) ORF22 385 394 QVAHVLPRLR
Varicella-zoster virus (strain Oka) ORF22 394 402 REVANTPDV
Varicella-zoster virus (strain Oka) ORF22 395 403 EVANTPDVV
Varicella-zoster virus (strain Oka) ORF22 398 406 NTPDVVLNV
Varicella-zoster virus (strain Oka) ORF22 410 418 DTPESSPTF
Varicella-zoster virus (strain Oka) ORF22 415 424 SPTFSRN NV
Varicella-zoster virus (strain Oka) ORF22 419 428 SRNMNVGSSL
Varicella-zoster virus (strain Oka) ORF22 420 429 RNMNVGSSLK
Varicella-zoster virus (strain Oka) ORF22 421 429 N NVGSSLK
Varicella-zoster virus (strain Oka) ORF22 427 435 SLKDRKPFL
Varicella-zoster virus (strain Oka) ORF22 434 442 FLFEQSGDV
Varicella-zoster virus (strain Oka) ORF22 448 456 KLLQHGHEI
Varicella-zoster virus (strain Oka) ORF22 470 478 TVITGHTNV
Varicella-zoster virus (strain Oka) ORF22 472 480 ITGHTNVPI
Varicella-zoster virus (strain Oka) ORF22 472 481 ITGHTNVPIW
Varicella-zoster virus (strain Oka) ORF22 475 484 HTNVP!VWTR
Varicella-zoster virus (strain Oka) ORF22 476 484 TNVPIWVTR
Varicella-zoster virus (strain Oka) ORF22 478 486 VPIWVTRPL 98 Varicella-zoster virus (strain Oka) ORF22 478 487 VPIWVTRPLV
99 Varicella-zoster virus (strain Oka) ORF22 488 497 MPDEKDPLEL
100 Varicella-zoster virus (strain Oka) ORF22 493 501 DPLELFINL
101 Varicella-zoster virus (strain Oka) ORF22 495 503 LELFINLTI
102 Varicella-zoster virus (strain Oka) ORF22 495 504 LELFINLTIL
103 Varicella-zoster virus (strain Oka) ORF22 496 505 ELFINLTILR
104 Varicella-zoster virus (strain Oka) ORF22 497 505 LFINLTILR
105 Varicella-zoster virus (strain Oka) ORF22 501 509 LTILRLTGF
106 Varicella-zoster virus (strain Oka) ORF22 523 532 S VSDF!GPL
107 Varicella-zoster virus (strain Oka) ORF22 530 539 GPLGEILTGF
108 Varicella-zoster virus (strain Oka) ORF22 539 547 FPSAAELIR
109 Varicella-zoster virus (strain Oka) ORF22 539 548 FPSAAELIRV
1 10 Varicella-zoster virus (strain Oka) ORF22 543 551 AELIRVTSL
1 11 Varicella-zoster virus (strain Oka) ORF22 543 552 AEURVTSLi
1 12 Varicella-zoster virus (strain Oka) ORF22 544 552 ELIRVTSLI
1 13 Varicella-zoster virus (strain Oka) ORF22 552 561 ILTN PGAEY
1 14 Varicella-zoster virus (strain Oka) ORF22 553 561 LTN PGAEY
1 15 Varicella-zoster virus (strain Oka) ORF22 559 567 AEYAIKTVL
1 16 Varicella-zoster virus (strain Oka) ORF22 559 568 AEYAIKTVLR
1 17 Varicella-zoster virus (strain Oka) ORF22 560 568 EYAIKTVLR
1 18 Varicella-zoster virus (strain Oka) ORF22 571 580 CTiG LNAK
1 19 Varicella-zoster virus (strain Oka) ORF22 572 580 TIG LIIAK
120 Varicella-zoster virus (strain Oka) ORF22 575 583 LIIAKFGL
121 Varicella-zoster virus (strain Oka) ORF22 575 584 MLIIAKFGLV
122 Varicella-zoster virus (strain Oka) ORF22 576 584 LIIAKFGLV
123 Varicella-zoster virus (strain Oka) ORF22 587 595 RVQDTTGAL
124 Varicella-zoster virus (strain Oka) ORF22 597 606 AELDVLEADL
125 Varicella-zoster virus (strain Oka) ORF22 610 618 SPIDLYSRL
126 Varicella-zoster virus (strain Oka) ORF22 614 623 LYSRLSTGLI
127 Varicella-zoster virus (strain Oka) ORF22 617 625 RLSTGLISI
128 Varicella-zoster virus (strain Oka) ORF22 621 630 GLISILNSPI
129 Varicella-zoster virus (strain Oka) ORF22 628 636 SPIISHPGL
130 Varicella-zoster virus (strain Oka) ORF22 628 637 SPilSHPGLF
131 Varicella-zoster virus (strain Oka) ORF22 633 641 HPGLFAELI
132 Varicella-zoster virus (strain Oka) ORF22 638 646 AELIPTRTG
133 Varicella-zoster virus (strain Oka) ORF22 647 655 SLSERIRLL
134 Varicella-zoster virus (strain Oka) ORF22 649 658 SER!RLLCEL
135 Varicella-zoster virus (strain Oka) ORF22 653 662 RLLGELVSAR
136 Varicella-zoster virus (strain Oka) ORF22 657 665 ELVSARETR
137 Varicella-zoster virus (strain Oka) ORF22 657 666 ELVSARETRY
138 Varicella-zoster virus (strain Oka) ORF22 660 668 SARETRY R
139 Varicella-zoster virus (strain Oka) ORF22 664 673 TRYMREHTAL
140 Varicella-zoster virus (strain Oka) ORF22 665 673 RY REHTAL
141 Varicella-zoster virus (strain Oka) ORF22 665 674 RYMREHTALV
142 Varicella-zoster virus (strain Oka) ORF22 666 674 Y REHTALV
143 Varicella-zoster virus (strain Oka) ORF22 668 677 REHTALVSSV
144 Varicella-zoster virus (strain Oka) ORF22 670 678 HTALVSSVK
145 Varicella-zoster virus (strain Oka) ORF22 676 685 SVKALENALR
146 Varicella-zoster virus (strain Oka) ORF22 705 714 EETD!PPEEL
147 Varicella-zoster virus (strain Oka) ORF22 712 721 EELiRRVYEI
148 Varicella-zoster virus (strain Oka) ORF22 713 721 ELIRRVYEI 149 Varicella-zoster virus (strain Oka) ORF22 713 722 EL!RRVYE!R
150 Varicella-zoster virus (strain Oka) ORF22 717 725 RVYEIRSEV
151 Varicella-zoster virus (strain Oka) ORF22 719 727 YEIRSEVT
152 Varicella-zoster virus (strain Oka) ORF22 719 728 YEIRSEVTML
153 Varicella-zoster virus (strain Oka) ORF22 720 729 EIRSEVTMLL
154 Varicella-zoster virus (strain Oka) ORF22 723 731 SEVT LLTS
155 Varicella-zoster virus (strain Oka) ORF22 723 732 SEVTMLLTSA
156 Varicella-zoster virus (strain Oka) ORF22 724 733 EVTMLLTSAV
157 Varicella-zoster virus (strain Oka) ORF22 725 733 VT LLTSAV
158 Varicella-zoster virus (strain Oka) ORF22 727 736 MLLTSAVTEY
159 Varicella-zoster virus (strain Oka) ORF22 729 737 LTSAVTEYF
160 Varicella-zoster virus (strain Oka) ORF22 730 739 TSAVTEYFTR
161 Varicella-zoster virus (strain Oka) ORF22 731 739 SAVTEYFTR
162 Varicella-zoster virus (strain Oka) ORF22 734 742 TEYFTRGVL
163 Varicella-zoster virus (strain Oka) ORF22 734 743 TEYFTRGVLY
164 Varicella-zoster virus (strain Oka) ORF22 737 746 FTRGVLYSTR
165 Varicella-zoster virus (strain Oka) ORF22 750 759 AEQSPRRFRV
166 Varicella-zoster virus (strain Oka) ORF22 753 761 SPRRFRVAT
167 Varicella-zoster virus (strain Oka) ORF22 753 762 SPRRFRVATA
168 Varicella-zoster virus (strain Oka) ORF22 756 764 RFRVATAST
169 Varicella-zoster virus (strain Oka) ORF22 756 765 RFRVATASTA
170 Varicella-zoster virus (strain Oka) ORF22 758 767 RVATASTAPI
171 Varicella-zoster virus (strain Oka) ORF22 759 767 VATASTAPI
172 Varicella-zoster virus (strain Oka) ORF22 760 769 ATASTAP!QR
173 Varicella-zoster virus (strain Oka) ORF22 761 769 TASTAPIQR
174 Varicella-zoster virus (strain Oka) ORF22 763 771 STAPIQRLL
175 Varicella-zoster virus (strain Oka) ORF22 765 774 APiQRLLDSL
176 Varicella-zoster virus (strain Oka) ORF22 769 777 RLLDSLPEF
177 Varicella-zoster virus (strain Oka) ORF22 780 788 KLTAIISSL
178 Varicella-zoster virus (strain Oka) ORF22 782 790 TAIISSLSI
179 Varicella-zoster virus (strain Oka) ORF22 795 803 ETIQNLPVV
180 Varicella-zoster virus (strain Oka) ORF22 800 809 LPWSLLKEL
181 Varicella-zoster virus (strain Oka) ORF22 802 811 WSLLKELIK
182 Varicella-zoster virus (strain Oka) ORF22 813 822 GEDLNTDTAL
183 Varicella-zoster virus (strain Oka) ORF22 817 825 NTDTALVSW
184 Varicella-zoster virus (strain Oka) ORF22 821 829 ALVSWLSVV
185 Varicella-zoster virus (strain Oka) ORF22 830 838 GEAQTAGYL
186 Varicella-zoster virus (strain Oka) ORF22 833 841 QTAGYLSRR
187 Varicella-zoster virus (strain Oka) ORF22 841 850 REFDELSRTI
188 Varicella-zoster virus (strain Oka) ORF22 848 856 RTIKTINTR
189 Varicella-zoster virus (strain Oka) ORF22 851 860 KTINTRATQR
190 Varicella-zoster virus (strain Oka) ORF22 865 873 AELSCFNTL
191 Varicella-zoster virus (strain Oka) ORF22 865 874 AELSCFNTLS
192 Varicella-zoster virus (strain Oka) ORF22 884 892 YETYNNGEV
193 Varicella-zoster virus (strain Oka) ORF22 885 893 ETYNNGEVK
194 Varicella-zoster virus (strain Oka) ORF22 885 894 ETYNNGEVKY
195 Varicella-zoster virus (strain Oka) ORF22 891 899 EVKYPEITR
196 Varicella-zoster virus (strain Oka) ORF22 894 902 YPEITRDDL
197 Varicella-zoster virus (strain Oka) ORF22 894 903 YPEITRDDLL
198 Varicella-zoster virus (strain Oka) ORF22 909 918 ATDDLVRQIK
199 Varicella-zoster virus (strain Oka) ORF22 932 940 SIKRRLETR 200 Varicella-zoster virus (strain Oka) ORF22 934 942 KRRLETRLK
201 Varicella-zoster virus (strain Oka) ORF22 938 947 ETRLKEVQTY
202 Varicella-zoster virus (strain Oka) ORF22 940 948 RLKEVQTYA
203 Varicella-zoster virus (strain Oka) ORF22 943 952 EVQTYANEAR
204 Varicella-zoster virus (strain Oka) ORF22 953 961 TTQDTIKSR
205 Varicella-zoster virus (strain Oka) ORF22 953 962 TTQDTIKSRK
206 Varicella-zoster virus (strain Oka) ORF22 957 965 TIKSRKQAA
207 Varicella-zoster virus (strain Oka) ORF22 959 967 KSRKQAAYN
208 Varicella-zoster virus (strain Oka) ORF22 959 968 KSRKQAAYNK
209 Varicella-zoster virus (strain Oka) ORF22 965 974 AYNKLGGLLR
210 Varicella-zoster virus (strain Oka) ORF22 966 974 YNKLGGLLR
211 Varicella-zoster virus (strain Oka) ORF22 968 976 KLGGLLRPV
212 Varicella-zoster virus (strain Oka) ORF22 971 980 GLLRPVTGFV
213 Varicella-zoster virus (strain Oka) ORF22 972 980 LLRPVTGFV
214 Varicella-zoster virus (strain Oka) ORF22 974 982 RPVTGFVGL
215 Varicella-zoster virus (strain Oka) ORF22 994 1002 SELDVQGAL
216 Varicella-zoster virus (strain Oka) ORF22 997 006 DVQGALVNLR
217 Varicella-zoster virus (strain Oka) ORF22 1004 1012 NLRTKVLEA
218 Varicella-zoster virus (strain Oka) ORF22 1006 1014 RTKVLEAPV
219 Varicella-zoster virus (strain Oka) ORF22 1008 1016 KVLEAPVEI
220 Varicella-zoster virus (strain Oka) ORF22 1012 1020 APVEIRSQL
221 Varicella-zoster virus (strain Oka) ORF22 015 1024 EIRSQLTGDF
222 Varicella-zoster virus (strain Oka) ORF22 1016 1025 IRSQLTGDFW
223 Varicella-zoster virus (strain Oka) ORF22 1017 1025 RSQLTGDFW
224 Varicella-zoster virus (strain Oka) ORF22 1018 1026 SQLTGDFWA
225 Varicella-zoster virus (strain Oka) ORF22 1018 027 SQLTGDFWAL
226 Varicella-zoster virus (strain Oka) ORF22 1019 1027 QLTGDFWAL
227 Varicella-zoster virus (strain Oka) ORF22 1019 028 QLTGDFWALF
228 Varicella-zoster virus (strain Oka) ORF22 1020 1028 LTGDFWALF
229 Varicella-zoster virus (strain Oka) ORF22 023 032 DFWALFNQYR
230 Varicella-zoster virus (strain Oka) ORF22 1024 1032 FWALFNQYR
231 Varicella-zoster virus (strain Oka) ORF22 033 042 DiLEHPGNAR
232 Varicella-zoster virus (strain Oka) ORF22 1037 1045 HPGNARTSV
233 Varicella-zoster virus (strain Oka) ORF22 1037 046 HPGNARTSVL
234 Varicella-zoster virus (strain Oka) ORF22 1044 053 SVLGGLGACF
235 Varicella-zoster virus (strain Oka) ORF22 1053 062 FTA!iEiVP!
236 Varicella-zoster virus (strain Oka) ORF22 1057 066 iE!VP!PTEY
237 Varicella-zoster virus (strain Oka) ORF22 1058 1066 EIVPIPTEY
238 Varicella-zoster virus (strain Oka) ORF22 1058 067 EIVP!PTEYR
239 Varicella-zoster virus (strain Oka) ORF22 1062 1070 IPTEYRPSL
240 Varicella-zoster virus (strain Oka) ORF22 064 1073 TEYRPSLLAF
241 Varicella-zoster virus (strain Oka) ORF22 1065 1073 EYRPSLLAF
242 Varicella-zoster virus (strain Oka) ORF22 065 1074 EYRPSLLAFF
243 Varicella-zoster virus (strain Oka) ORF22 1080 1088 VLASDIATV
244 Varicella-zoster virus (strain Oka) ORF22 1093 1 101 ESESAINAV
245 Varicella-zoster virus (strain Oka) ORF22 093 1 102 ESESAINAW
246 Varicella-zoster virus (strain Oka) ORF22 1094 1 102 SESAINAVV
247 Varicella-zoster virus (strain Oka) ORF22 1 113 1 121 STVPALSFV
248 Varicella-zoster virus (strain Oka) ORF22 1 113 122 STVPALSFVL
249 Varicella-zoster virus (strain Oka) ORF22 1 115 124 VPALSFVLSL
250 Varicella-zoster virus (strain Oka) ORF22 1 116 1 125 PALSFVLSLY 251 Varicella-zoster virus (strain Oka) ORF22 1 117 1 125 ALSFVLSLY
252 Varicella-zoster virus (strain Oka) ORF22 1 117 26 ALSFVLSLYK
253 Varicella-zoster virus (strain Oka) ORF22 1 118 1 126 LSFVLSLYK
254 Varicella-zoster virus (strain Oka) ORF22 1 118 1 127 LSFVLSLYKK
255 Varicella-zoster virus (strain Oka) ORF22 1 122 1 131 LSLYKKYQAL
256 Varicella-zoster virus (strain Oka) ORF22 1 123 1 131 SLYKKYQAL
257 Varicella-zoster virus (strain Oka) ORF22 1 127 1 135 KYQALQQEI
258 Varicella-zoster virus (strain Oka) ORF22 1 133 1 141 QEITNTHKL
259 Varicella-zoster virus (strain Oka) ORF22 1 144 1 152 LQKQLGDDF
260 Varicella-zoster virus (strain Oka) ORF22 1 146 55 KQLGDDFSTL
261 Varicella-zoster virus (strain Oka) ORF22 1 154 1 163 TLAVSSGHLK
262 Varicella-zoster virus (strain Oka) ORF22 1 161 1 70 HLKFISSSNV
263 Varicella-zoster virus (strain Oka) ORF22 164 1 173 FiSSSNVDDY
264 Varicella-zoster virus (strain Oka) ORF22 1 165 1 173 ISSSNVDDY
265 Varicella-zoster virus (strain Oka) ORF22 1 165 74 ISSSNVDDYE
266 Varicella-zoster virus (strain Oka) ORF22 1 173 1 182 YEiNDAiLSI
267 Varicella-zoster virus (strain Oka) ORF22 1 174 1 182 EINDAILSI
268 Varicella-zoster virus (strain Oka) ORF22 1 185 1 193 NVHAL DTV
269 Varicella-zoster virus (strain Oka) ORF22 1 188 1 196 AL DTVKLV
270 Varicella-zoster virus (strain Oka) ORF22 1 194 1202 KLVEVELQK
271 Varicella-zoster virus (strain Oka) ORF22 207 1216 CIAGTSTLSR
272 Varicella-zoster virus (strain Oka) ORF22 1210 1219 GTSTLSRWK
273 Varicella-zoster virus (strain Oka) ORF22 1223 1231 KLVT AHEK
274 Varicella-zoster virus (strain Oka) ORF22 223 1232 KLVTMAHEKK
275 Varicella-zoster virus (strain Oka) ORF22 1236 1244 KVLITDCER
276 Varicella-zoster virus (strain Oka) ORF22 1244 1252 RAHKQQTTR
277 Varicella-zoster virus (strain Oka) ORF22 1247 1255 KQQTTRVLY
278 Varicella-zoster virus (strain Oka) ORF22 1249 1257 QTTRVLYER
279 Varicella-zoster virus (strain Oka) ORF22 249 258 QTTRVLYERW
280 Varicella-zoster virus (strain Oka) ORF22 1250 1258 TTRVLYERW
281 Varicella-zoster virus (strain Oka) ORF22 1252 1260 RVLYERWTR
282 Varicella-zoster virus (strain Oka) ORF22 1254 1262 LYERWTRDI
283 Varicella-zoster virus (strain Oka) ORF22 1261 1269 DIIACLEA
284 Varicella-zoster virus (strain Oka) ORF22 266 275 LEAMETRHVF
285 Varicella-zoster virus (strain Oka) ORF22 1267 1275 EA ETRHVF
286 Varicella-zoster virus (strain Oka) ORF22 1273 1281 HVFNGTELA
287 Varicella-zoster virus (strain Oka) ORF22 1273 282 HVFNGTELAR
288 Varicella-zoster virus (strain Oka) ORF22 1278 1286 TELARLRD
289 Varicella-zoster virus (strain Oka) ORF22 1280 1288 LARLRD AA
290 Varicella-zoster virus (strain Oka) ORF22 1286 1294 AAAGGFDI
291 Varicella-zoster virus (strain Oka) ORF22 293 302 DIHAVYPQAR
292 Varicella-zoster virus (strain Oka) ORF22 1298 1306 YPQARQVVA
293 Varicella-zoster virus (strain Oka) ORF22 29S 307 YPQARQWAA
294 Varicella-zoster virus (strain Oka) ORF22 1308 1316 CETTAVTAL
295 Varicella-zoster virus (strain Oka) ORF22 1310 3 9 TTAVTALDTV
296 Varicella-zoster virus (strain Oka) ORF22 131 1 1320 TAVTALDTVF
297 Varicella-zoster virus (strain Oka) ORF22 1312 321 AVTALDTVFR
298 Varicella-zoster virus (strain Oka) ORF22 1313 1321 VTALDTVFR
299 Varicella-zoster virus (strain Oka) ORF22 1331 340 NiPPPLALLR
300 Varicella-zoster virus (strain Oka) ORF22 1336 1344 LALLRGLTW
301 Varicella-zoster virus (strain Oka) ORF22 1342 1350 LTWFDDFSI 302 Varicella-zoster virus (strain Oka) ORF22 1346 1354 DDFSITAPV
303 Varicella-zoster virus (strain Oka) ORF22 348 357 FSITAPVFTV
304 Varicella-zoster virus (strain Oka) ORF22 1350 1358 ITAPVFTV
305 Varicella-zoster virus (strain Oka) ORF22 355 364 FTVMFPGVSI
306 Varicella-zoster virus (strain Oka) ORF22 1356 1364 TV FPGVSI
307 Varicella-zoster virus (strain Oka) ORF22 1359 1367 FPGVSIEGL
308 Varicella-zoster virus (strain Oka) ORF22 1359 368 FPGVSIEGLL
309 Varicella-zoster virus (strain Oka) ORF22 1368 1376 LLL RIRAV
310 Varicella-zoster virus (strain Oka) ORF22 36S 1377 LLLMRIRAW
311 Varicella-zoster virus (strain Oka) ORF22 1369 1377 LL RIRAVV
312 Varicella-zoster virus (strain Oka) ORF22 369 1378 LL RIRAWL
313 Varicella-zoster virus (strain Oka) ORF22 1370 1378 L RIRAVVL
314 Varicella-zoster virus (strain Oka) ORF22 370 379 LMRIRAWLL
315 Varicella-zoster virus (strain Oka) ORF22 1372 1380 RIRAVVLLS
316 Varicella-zoster virus (strain Oka) ORF22 382 391 DTSINGIPNY
317 Varicella-zoster virus (strain Oka) ORF22 1383 1391 TSINGIPNY
318 Varicella-zoster virus (strain Oka) ORF22 1383 392 TSINGIPNYR
319 Varicella-zoster virus (strain Oka) ORF22 1384 1392 SINGIPNYR
320 Varicella-zoster virus (strain Oka) ORF22 1388 1396 IPNYRD IL
321 Varicella-zoster virus (strain Oka) ORF22 1393 02 DMILRTSGDL
322 Varicella-zoster virus (strain Oka) ORF22 1397 1405 RTSGDLLQI
323 Varicella-zoster virus (strain Oka) ORF22 402 141 1 LLQIPALAGY
324 Varicella-zoster virus (strain Oka) ORF22 1403 141 1 LQIPALAGY
325 Varicella-zoster virus (strain Oka) ORF22 403 1412 LQiPALAGYV
326 Varicella-zoster virus (strain Oka) ORF22 405 41 PALAGYVDF
327 Varicella-zoster virus (strain Oka) ORF22 1407 1415 ALAGYVDFY
328 Varicella-zoster virus (strain Oka) ORF22 407 416 ALAGYVDFYT
329 Varicella-zoster virus (strain Oka) ORF22 1409 1417 AGYVDFYTR
330 Varicella-zoster virus (strain Oka) ORF22 141 1 1419 YVDFYTRSY
331 Varicella-zoster virus (strain Oka) ORF22 1414 1422 FYTRSYDQF
332 Varicella-zoster virus (strain Oka) ORF22 4 423 FYTRSYDQFI
333 Varicella-zoster virus (strain Oka) ORF22 1424 1432 TESVTLSEL
334 Varicella-zoster virus (strain Oka) ORF22 1425 1433 ESVTLSELR
335 Varicella-zoster virus (strain Oka) ORF22 1446 1454 EANKALEEV
336 Varicella-zoster virus (strain Oka) ORF22 1449 1457 KALEEVTHV
337 Varicella-zoster virus (strain Oka) ORF22 1452 1460 EEVTHVRAH
338 Varicella-zoster virus (strain Oka) ORF22 1456 1464 HVRAHETAK
339 Varicella-zoster virus (strain Oka) ORF22 1456 465 HVRAHETAKL
340 Varicella-zoster virus (strain Oka) ORF22 1465 1473 LALKEGVFI
341 Varicella-zoster virus (strain Oka) ORF22 1473 1481 ITLPSEGLL
342 Varicella-zoster virus (strain Oka) ORF22 1477 1485 SEGLLIRAI
343 Varicella-zoster virus (strain Oka) ORF22 1480 1488 LLIRAIEYF
344 Varicella-zoster virus (strain Oka) ORF22 1483 1491 RAIEYFTTF
345 Varicella-zoster virus (strain Oka) ORF22 486 95 EYFTTFDHKR
346 Varicella-zoster virus (strain Oka) ORF22 1487 1495 YFTTFDHKR
347 Varicella-zoster virus (strain Oka) ORF22 1495 1503 RFIGTAYER
348 Varicella-zoster virus (strain Oka) ORF22 1500 1508 AYERVLQT
349 Varicella-zoster virus (strain Oka) ORF22 1503 151 1 RVLQT VDR
350 Varicella-zoster virus (strain Oka) ORF22 1506 1514 QT VDRDLK
351 Varicella-zoster virus (strain Oka) ORF22 1514 523 KEANAELAQF
352 Varicella-zoster virus (strain Oka) ORF22 1515 1523 EANAELAQF 353 Varicella-zoster virus (strain Oka) ORF22 1518 1526 AELAQFR V
354 Varicella-zoster virus (strain Oka) ORF22 5 8 527 AELAQFRMVC
355 Varicella-zoster virus (strain Oka) ORF22 1524 533 RMVCQATKNR
356 Varicella-zoster virus (strain Oka) ORF22 1525 1533 VCQATKNR
357 Varicella-zoster virus (strain Oka) ORF22 1533 1541 RAIQILQNI
358 Varicella-zoster virus (strain Oka) ORF22 1548 1556 TEQQEDVDF
359 Varicella-zoster virus (strain Oka) ORF22 1551 1559 QEDVDFTNL
360 Varicella-zoster virus (strain Oka) ORF22 1556 1564 FTNLKTLLK
361 Varicella-zoster virus (strain Oka) ORF22 560 569 KTLLKLTPPP
362 Varicella-zoster virus (strain Oka) ORF22 561 1570 TLLKLTPPPK
363 Varicella-zoster virus (strain Oka) ORF22 1562 1570 LLKLTPPPK
364 Varicella-zoster virus (strain Oka) ORF22 1566 1574 TPPPKTIAL
365 Varicella-zoster virus (strain Oka) ORF22 573 582 ALAIDRSTSV
366 Varicella-zoster virus (strain Oka) ORF22 1574 1582 LAIDRSTSV
367 Varicella-zoster virus (strain Oka) ORF22 1584 1592 DIVTQFALL
368 Varicella-zoster virus (strain Oka) ORF22 1604 1612 DIQAVDWMY
369 Varicella-zoster virus (strain Oka) ORF22 1629 1637 GTGPLHTYK
370 Varicella-zoster virus (strain Oka) ORF22 1634 1642 HTYKDRVDK
371 Varicella-zoster virus (strain Oka) ORF22 1638 647 DRVDKLYALR
372 Varicella-zoster virus (strain Oka) ORF22 1639 1647 RVDKLYALR
373 Varicella-zoster virus (strain Oka) ORF22 1642 1650 KLYALRTKL
374 Varicella-zoster virus (strain Oka) ORF22 643 1652 LYALRTKLDL
375 Varicella-zoster virus (strain Oka) ORF22 1647 1655 RTKLDLLRR
376 Varicella-zoster virus (strain Oka) ORF22 647 1656 RTKLDLLRRR
377 Varicella-zoster virus (strain Oka) ORF22 1652 1660 LLRRRIETG
378 Varicella-zoster virus (strain Oka) ORF22 1656 1664 RIETGEVTW
379 Varicella-zoster virus (strain Oka) ORF22 1660 1668 GEVTWDDAW
380 Varicella-zoster virus (strain Oka) ORF22 662 1671 VT DDAWTTF
381 Varicella-zoster virus (strain Oka) ORF22 1663 1671 TWDDAWTTF
382 Varicella-zoster virus (strain Oka) ORF22 1665 1673 DDAWTTFKR
383 Varicella-zoster virus (strain Oka) ORF22 1678 1686 LASGDTYA
384 Varicella-zoster virus (strain Oka) ORF22 1684 1692 TYATSVDSI
385 Varicella-zoster virus (strain Oka) ORF22 1685 1693 YATSVDSIK
386 Varicella-zoster virus (strain Oka) ORF22 1695 704 LQASASWDM
387 Varicella-zoster virus (strain Oka) ORF22 1704 1712 LCSEPEFF
388 Varicella-zoster virus (strain Oka) ORF22 1704 713 MLCSEPEFFL
389 Varicella-zoster virus (strain Oka) ORF22 1706 1714 CSEPEFFLL
390 Varicella-zoster virus (strain Oka) ORF22 1707 17 6 SEPEFFLLPV
391 Varicella-zoster virus (strain Oka) ORF22 1721 1729 RLQKKQQER
392 Varicella-zoster virus (strain Oka) ORF22 1730 739 KTALDVVLQK
393 Varicella-zoster virus (strain Oka) ORF22 1731 1739 TALDVVLQK
394 Varicella-zoster virus (strain Oka) ORF22 1741 1749 RQFEETASR
395 Varicella-zoster virus (strain Oka) ORF22 44 1 753 EETASRLRAL
396 Varicella-zoster virus (strain Oka) ORF22 1745 1753 ETASRLRAL
397 Varicella-zoster virus (strain Oka) ORF22 745 54 E1ASRLRALI
398 Varicella-zoster virus (strain Oka) ORF22 747 56 ASRLRALIER
399 Varicella-zoster virus (strain Oka) ORF22 1760 1768 ESDHDVLR
400 Varicella-zoster virus (strain Oka) ORF22 1768 1776 LLHDFDQF
401 Varicella-zoster virus (strain Oka) ORF22 1773 782 FDQFTHLPIW
402 Varicella-zoster virus (strain Oka) ORF22 1779 1787 LPIWIKTQY
403 Varicella-zoster virus (strain Oka) ORF22 1779 788 LPIWIKTQYM 404 Varicella-zoster virus (strain Oka) ORF22 781 790 IWIKTQYMTF
405 Varicella-zoster virus (strain Oka) ORF22 782 791 WIKTQYMTFR
406 Varicella-zoster virus (strain Oka) ORF22 1786 1794 QY TFRNLL
407 Varicella-zoster virus (strain Oka) ORF22 786 795 QYMTFRNLLM
408 Varicella-zoster virus (strain Oka) ORF22 1787 796 YMTFRNLLMV
409 Varicella-zoster virus (strain Oka) ORF22 1788 1796 TFRNLL V
410 Varicella-zoster virus (strain Oka) ORF22 1788 797 MTFRNLLMVR
411 Varicella-zoster virus (strain Oka) ORF22 1789 1797 TFRNLL VR
412 Varicella-zoster virus (strain Oka) ORF22 792 S01 NLL VRLGLY
413 Varicella-zoster virus (strain Oka) ORF22 1793 1801 LL VRLGLY
414 Varicella-zoster virus (strain Oka) ORF22 793 S02 LL VRLGLYA
415 Varicella-zoster virus (strain Oka) ORF22 795 S04 MVRLGLYASY
416 Varicella-zoster virus (strain Oka) ORF22 1799 1807 GLYASYAEI
417 Varicella-zoster virus (strain Oka) ORF22 1800 1808 LYASYAEIF
418 Varicella-zoster virus (strain Oka) ORF22 802 81 ASYAEiFPPA
419 Varicella-zoster virus (strain Oka) ORF22 805 814 AEiFPPASPhi
420 Varicella-zoster virus (strain Oka) ORF22 1808 1816 FPPASPNGV
421 Varicella-zoster virus (strain Oka) ORF22 1808 817 FPPASPNGVF
422 Varicella-zoster virus (strain Oka) ORF22 1809 1817 PPASPNGVF
423 Varicella-zoster virus (strain Oka) ORF22 1812 1820 SPNGVFAPI
424 Varicella-zoster virus (strain Oka) ORF22 817 1826 FAPIPAMSGV
425 Varicella-zoster virus (strain Oka) ORF22 1818 1826 APIPA SGV
426 Varicella-zoster virus (strain Oka) ORF22 1820 1828 IPA SGVCL
427 Varicella-zoster virus (strain Oka) ORF22 833 1842 RC!RARVAAF
428 Varicella-zoster virus (strain Oka) ORF22 1834 1842 CIRARVAAF
429 Varicella-zoster virus (strain Oka) ORF22 834 843 CIRARVAAFM
430 Varicella-zoster virus (strain Oka) ORF22 1844 1852 GEASVVQTF
431 Varicella-zoster virus (strain Oka) ORF22 1845 1853 EASVVQTFR
432 Varicella-zoster virus (strain Oka) ORF22 847 856 SVVQTFREAR
433 Varicella-zoster virus (strain Oka) ORF22 1848 1856 VVQTFREAR
434 Varicella-zoster virus (strain Oka) ORF22 853 862 REARSSIDAL
435 Varicella-zoster virus (strain Oka) ORF22 856 865 RSSIDALFGK
436 Varicella-zoster virus (strain Oka) ORF22 1857 1865 SSIDALFGK
437 Varicella-zoster virus (strain Oka) ORF22 1861 1869 ALFGKNLTF
438 Varicella-zoster virus (strain Oka) ORF22 861 870 ALFGKNLTFY
439 Varicella-zoster virus (strain Oka) ORF22 867 876 LTFYLDTDGV
440 Varicella-zoster virus (strain Oka) ORF22 1869 878 FYLDTDGVPL
441 Varicella-zoster virus (strain Oka) ORF22 1870 1878 YLDTDGVPL
442 Varicella-zoster virus (strain Oka) ORF22 1872 1880 DTDGVPLRY
443 Varicella-zoster virus (strain Oka) ORF22 1872 881 DTDGVPLRYR
444 Varicella-zoster virus (strain Oka) ORF22 1877 1885 PLRYRVCYK
445 Varicella-zoster virus (strain Oka) ORF22 1879 1887 RYRVCYKSV
446 Varicella-zoster virus (strain Oka) ORF22 881 1890 RVCYKSVGVK
447 Varicella-zoster virus (strain Oka) ORF22 1882 1890 VCYKSVGVK
448 Varicella-zoster virus (strain Oka) ORF22 1904 1912 RPALPDEGI
449 Varicella-zoster virus (strain Oka) ORF22 904 913 RPALPDEGIV
450 Varicella-zoster virus (strain Oka) ORF22 1913 1921 VEETTLSAL
451 Varicella-zoster virus (strain Oka) ORF22 1915 1923 ETTLSALRV
452 Varicella-zoster virus (strain Oka) ORF22 1922 931 RVANEVNELR
453 Varicella-zoster virus (strain Oka) ORF22 1923 1931 VANEVNELR
454 Varicella-zoster virus (strain Oka) ORF22 1925 934 NEVNELRIEY 455 Varicella-zoster virus (strain Oka) ORF22 1926 1934 EVNELRIEY
456 Varicella-zoster virus (strain Oka) ORF22 1933 942 EYESAIKSGF
457 Varicella-zoster virus (strain Oka) ORF22 1934 1942 YESAIKSGF
458 Varicella-zoster virus (strain Oka) ORF22 1937 1945 AIKSGFSAF
459 Varicella-zoster virus (strain Oka) ORF22 1940 1948 SGFSAFSTF
460 Varicella-zoster virus (strain Oka) ORF22 1941 950 GFSAFSTFVR
461 Varicella-zoster virus (strain Oka) ORF22 1942 1950 FSAFSTFVR
462 Varicella-zoster virus (strain Oka) ORF22 1943 952 SAFSTFVRHR
463 Varicella-zoster virus (strain Oka) ORF22 1944 1952 AFSTFVRHR
464 Varicella-zoster virus (strain Oka) ORF22 1948 1956 FVRHRHAEW
465 Varicella-zoster virus (strain Oka) ORF22 1950 1958 RHRHAEWGK
466 Varicella-zoster virus (strain Oka) ORF22 1955 1963 EWGKTNARR
467 Varicella-zoster virus (strain Oka) ORF22 1958 1966 KTNARRAIA
468 Varicella-zoster virus (strain Oka) ORF22 1965 1973 IAEIYAGLI
469 Varicella-zoster virus (strain Oka) ORF22 1966 1974 AEIYAGLIT
470 Varicella-zoster virus (strain Oka) ORF22 968 977 IYAGLITTTL
471 Varicella-zoster virus (strain Oka) ORF22 1974 983 TTTLTRQYGV
472 Varicella-zoster virus (strain Oka) ORF22 1975 1983 TTLTRQYGV
473 Varicella-zoster virus (strain Oka) ORF22 1976 985 TLTRQYGVHW
474 Varicella-zoster virus (strain Oka) ORF22 1977 1985 LTRQYGVHW
475 Varicella-zoster virus (strain Oka) ORF22 1979 1987 RQYGVHWDK
476 Varicella-zoster virus (strain Oka) ORF22 981 1990 YGVHWDKLIY
477 Varicella-zoster virus (strain Oka) ORF22 983 1992 VHWDKLIYSF
478 Varicella-zoster virus (strain Oka) ORF22 1984 1992 HWDKLIYSF
479 Varicella-zoster virus (strain Oka) ORF22 1989 1997 IYSFEKHHL
480 Varicella-zoster virus (strain Oka) ORF22 998 2007 TSV GNGLTK
481 Varicella-zoster virus (strain Oka) ORF22 1999 2007 SV GNGLTK
482 Varicella-zoster virus (strain Oka) ORF22 2007 2015 KPIQRRGDV
483 Varicella-zoster virus (strain Oka) ORF22 2020 2028 LTLSDIVTI
484 Varicella-zoster virus (strain Oka) ORF22 2021 2029 TLSDIVTIL
485 Varicella-zoster virus (strain Oka) ORF22 2021 2030 TLSD!VT!LV
486 Varicella-zoster virus (strain Oka) ORF22 2022 2030 LSDIVTILV
487 Varicella-zoster virus (strain Oka) ORF22 2042 2051 RLDLIKQHEY
488 Varicella-zoster virus (strain Oka) ORF22 2045 2054 LIKQHEYMAR
489 Varicella-zoster virus (strain Oka) ORF22 2047 2056 KQHEYMARTL
490 Varicella-zoster virus (strain Oka) ORF22 2049 2057 HEY ARTLR
491 Varicella-zoster virus (strain Oka) ORF22 2051 2059 Y ARTLRPV
492 Varicella-zoster virus (strain Oka) ORF22 2052 2060 ARTLRPVI
493 Varicella-zoster virus (strain Oka) ORF22 2054 2062 RTLRPVIEA
494 Varicella-zoster virus (strain Oka) ORF22 2055 2064 TLRPVIEAAF
495 Varicella-zoster virus (strain Oka) ORF22 2060 2068 IEAAFRGRL
496 Varicella-zoster virus (strain Oka) ORF22 2060 2069 IEAAFRGRLL
497 Varicella-zoster virus (strain Oka) ORF22 2063 2071 AFRGRLLVR
498 Varicella-zoster virus (strain Oka) ORF22 2064 2073 FRGRLLVRSL
499 Varicella-zoster virus (strain Oka) ORF22 2078 2087 KGNARAFFNA
500 Varicella-zoster virus (strain Oka) ORF22 2080 2088 NARAFFNAA
501 Varicella-zoster virus (strain Oka) ORF22 2082 2091 RAFFNAAPSK
502 Varicella-zoster virus (strain Oka) ORF22 2083 2091 AFFNAAPSK
503 Varicella-zoster virus (strain Oka) ORF22 2088 2096 APSKHKLPL
504 Varicella-zoster virus (strain Oka) ORF22 2104 2113 PTGGRIFAFR
505 Varicella-zoster virus (strain Oka) ORF22 2105 2113 TGGRIFAFR 506 Varicella-zoster virus (strain Oka) ORF22 2109 2117 IFAFR ADW
507 Varicella-zoster virus (strain Oka) ORF22 2110 2118 FAFR ADWK
508 Varicella-zoster virus (strain Oka) ORF22 2113 2121 R ADWKLVK
509 Varicella-zoster virus (strain Oka) ORF22 2122 2130 PQKITDPF
510 Varicella-zoster virus (strain Oka) ORF22 2122 2131 MPQKITDPFA
511 Varicella-zoster virus (strain Oka) ORF22 2125 2133 KITDPFAPW
512 Varicella-zoster virus (strain Oka) ORF22 2134 2142 QLSPPPGVK
513 Varicella-zoster virus (strain Oka) ORF22 2142 2150 KANVDAVTR
514 Varicella-zoster virus (strain Oka) ORF22 2151 2160 IMATDRLATI
515 Varicella-zoster virus (strain Oka) ORF22 2152 2160 ATDRLATI
516 Varicella-zoster virus (strain Oka) ORF22 2156 2165 RLAT!TVLGR
517 Varicella-zoster virus (strain Oka) ORF22 2158 2166 ATITVLGR
518 Varicella-zoster virus (strain Oka) ORF22 2165 2 74 R CLPPISLV
519 Varicella-zoster virus (strain Oka) ORF22 2168 2176 LPPISLVS
520 Varicella-zoster virus (strain Oka) ORF22 2168 2177 LPPISLVSMW
521 Varicella-zoster virus (strain Oka) ORF22 2169 2177 PPISLVS W
522 Varicella-zoster virus (strain Oka) ORF22 2171 2180 ISLVSMWNTL
523 Varicella-zoster virus (strain Oka) ORF22 2176 2185 M TLQPEEF
524 Varicella-zoster virus (strain Oka) ORF22 2179 2187 TLQPEEFAY
525 Varicella-zoster virus (strain Oka) ORF22 2185 2193 FAYRTQDDV
526 Varicella-zoster virus (strain Oka) ORF22 2189 2197 TQDDVDIIV
527 Varicella-zoster virus (strain Oka) ORF22 2202 2210 DLSSTLNAR
528 Varicella-zoster virus (strain Oka) ORF22 2203 221 1 LSSTLNARF
529 Varicella-zoster virus (strain Oka) ORF22 2213 2222 TAPSNTTLEW
530 Varicella-zoster virus (strain Oka) ORF22 2214 2222 APSNTTLEW
531 Varicella-zoster virus (strain Oka) ORF22 2218 2226 TTLEWNTDR
532 Varicella-zoster virus (strain Oka) ORF22 22 8 2227 TTLEWNTDRK
533 Varicella-zoster virus (strain Oka) ORF22 2233 2241 YIQTGATTV
534 Varicella-zoster virus (strain Oka) ORF22 2233 2242 YIQTGATTVF
535 Varicella-zoster virus (strain Oka) ORF22 2234 2242 IQTGATTVF
536 Varicella-zoster virus (strain Oka) ORF22 2239 2247 TTVFTVTGA
537 Varicella-zoster virus (strain Oka) ORF22 2239 2248 TTVFTVTGAA
538 Varicella-zoster virus (strain Oka) ORF22 2240 2248 TVFTVTGAA
539 Varicella-zoster virus (strain Oka) ORF22 2258 2267 FDIATTAILF
540 Varicella-zoster virus (strain Oka) ORF22 2259 2267 DIATTAILF
541 Varicella-zoster virus (strain Oka) ORF22 2262 2271 TTA!LFGAPL
542 Varicella-zoster virus (strain Oka) ORF22 2264 2272 AILFGAPLV
543 Varicella-zoster virus (strain Oka) ORF22 2265 2273 ILFGAPLVI
544 Varicella-zoster virus (strain Oka) ORF22 2267 2275 FGAPLVIA
545 Varicella-zoster virus (strain Oka) ORF22 2269 2277 APLVIA EL
546 Varicella-zoster virus (strain Oka) ORF22 2271 2280 LVIAMELTSV
547 Varicella-zoster virus (strain Oka) ORF22 2272 2280 VIA ELTSV
548 Varicella-zoster virus (strain Oka) ORF22 2273 2281 IA ELTSVF
549 Varicella-zoster virus (strain Oka) ORF22 2300 2308 ATDSGISSA
550 Varicella-zoster virus (strain Oka) ORF22 2305 2313 ISSAVSPDI
551 Varicella-zoster virus (strain Oka) ORF22 2307 23 6 SAVSPDIVSW
552 Varicella-zoster virus (strain Oka) ORF22 2308 2316 AVSPDIVSW
553 Varicella-zoster virus (strain Oka) ORF22 23 9 2328 RLLHMDPHPI
554 Varicella-zoster virus (strain Oka) ORF22 2320 2328 LLH DPHPI
555 Varicella-zoster virus (strain Oka) ORF22 2326 2334 HPIENACLI
556 Varicella-zoster virus (strain Oka) ORF22 2326 2335 HPIENACLIV 557 Varicella-zoster virus (strain Oka) ORF22 2328 2337 IENACLIVQL
558 Varicella-zoster virus (strain Oka) ORF22 2334 2343 IVQLEKLSAL
559 Varicella-zoster virus (strain Oka) ORF22 2335 2343 VQLEKLSAL
560 Varicella-zoster virus (strain Oka) ORF22 2339 2347 KLSALIANK
561 Varicella-zoster virus (strain Oka) ORF22 2358 2367 LLDEHMNPSY
562 Varicella-zoster virus (strain Oka) ORF22 2360 2369 DEHMNPSYVL
563 Varicella-zoster virus (strain Oka) ORF22 2361 2370 EHMNPSYVLW
564 Varicella-zoster virus (strain Oka) ORF22 2362 2370 H NPSYVLW
565 Varicella-zoster virus (strain Oka) ORF22 2363 2372 MNPSYVLWER
566 Varicella-zoster virus (strain Oka) ORF22 2364 2372 NPSYVLWER
567 Varicella-zoster virus (strain Oka) ORF22 2372 2381 RKDS!PAPDY
568 Varicella-zoster virus (strain Oka) ORF22 2374 2382 DSIPAPDYV
569 Varicella-zoster virus (strain Oka) ORF22 2376 2384 IPAPDYVVF
570 Varicella-zoster virus (strain Oka) ORF22 2376 2385 IPAPDYWFW
571 Varicella-zoster virus (strain Oka) ORF22 2386 2395 GPESLIDLPY
572 Varicella-zoster virus (strain Oka) ORF22 2387 2395 PESLIDLPY
573 Varicella-zoster virus (strain Oka) ORF22 2394 2403 PYiDSDEDSF
574 Varicella-zoster virus (strain Oka) ORF22 2403 241 1 FPSCPDDPF
575 Varicella-zoster virus (strain Oka) ORF22 2403 24 2 FPSCPDDPFY
576 Varicella-zoster virus (strain Oka) ORF22 2404 2412 PSCPDDPFY
577 Varicella-zoster virus (strain Oka) ORF22 2406 24 5 CPDDPFYSQI
578 Varicella-zoster virus (strain Oka) ORF22 241 1 2419 FYSQIIAGY
579 Varicella-zoster virus (strain Oka) ORF22 2430 2439 TDFYPTEPLF
580 Varicella-zoster virus (strain Oka) ORF22 2431 2439 DFYPTEPLF
581 Varicella-zoster virus (strain Oka) ORF22 2438 2447 LFKSPVQWR
582 Varicella-zoster virus (strain Oka) ORF22 2445 2453 VVRSSKCKK
583 Varicella-zoster virus (strain Oka) ORF22 2448 2456 SSKCKK PV
584 Varicella-zoster virus (strain Oka) ORF22 2454 2462 PVQPVQPA
585 Varicella-zoster virus (strain Oka) ORF22 2454 2463 MPVQPVQPAQ
586 Varicella-zoster virus (strain Oka) ORF22 2466 2474 QPAQPAQPV
587 Varicella-zoster virus (strain Oka) ORF22 2472 2480 QPVQPAQPI
588 Varicella-zoster virus (strain Oka) ORF22 2478 2486 QPIEPGTQI
589 Varicella-zoster virus (strain Oka) ORF22 2478 2487 QPiEPGTQIV
590 Varicella-zoster virus (strain Oka) ORF22 2483 2492 GTQIVVQNFK
591 Varicella-zoster virus (strain Oka) ORF22 2484 2492 TQIVVQNFK
592 Varicella-zoster virus (strain Oka) ORF22 2484 2493 TQIWQNFKK
593 Varicella-zoster virus (strain Oka) ORF22 2493 2501 KPQSVKTTL
594 Varicella-zoster virus (strain Oka) ORF22 2496 2504 SVKTTLSQK
595 Varicella-zoster virus (strain Oka) ORF22 2500 2509 TLSQKD!PLY
596 Varicella-zoster virus (strain Oka) ORF22 2501 2509 LSQKDIPLY
597 Varicella-zoster virus (strain Oka) ORF22 25 0 25 9 VETESETAVL
598 Varicella-zoster virus (strain Oka) ORF22 2512 2520 TESETAVLI
599 Varicella-zoster virus (strain Oka) ORF22 2520 2528 IPKQLTTSI
600 Varicella-zoster virus (strain Oka) ORF22 2525 2534 TTSIKTTVCK
601 Varicella-zoster virus (strain Oka) ORF22 2526 2534 TSIKTTVCK
602 Varicella-zoster virus (strain Oka) ORF22 2537 2546 TPPNNQLSDW
603 Varicella-zoster virus (strain Oka) ORF22 2538 2546 PPNNQLSDW
604 Varicella-zoster virus (strain Oka) ORF22 2551 2560 QGNQTLNQAF
605 Varicella-zoster virus (strain Oka) ORF22 2554 2562 QTLNQAFNK
606 Varicella-zoster virus (strain Oka) ORF22 2562 2570 KPILEITSI
607 Varicella-zoster virus (strain Oka) ORF22 2569 2578 SIPTDDSiSY 608 Varicella-zoster virus (strain Oka) ORF22 2570 2578 IPTDDSISY
609 Varicella-zoster virus (strain Oka) ORF22 2573 2581 DDSISYRTW
610 Varicella-zoster virus (strain Oka) ORF22 2575 2584 SISYRTWiEK
611 Varicella-zoster virus (strain Oka) ORF22 2576 2584 ISYRTWIEK
612 Varicella-zoster virus (strain Oka) ORF22 2597 2605 R YNSKTVF
613 Varicella-zoster virus (strain Oka) ORF22 2597 2606 RMYNSKTVFH
614 Varicella-zoster virus (strain Oka) ORF22 2600 2608 NSKTVFHPV
615 Varicella-zoster virus (strain Oka) ORF22 2606 2615 HPVNNQLPSW
616 Varicella-zoster virus (strain Oka) ORF22 2612 2620 LPSWVDTAA
617 Varicella-zoster virus (strain Oka) ORF22 2622 2631 APQTDLLTNY
618 Varicella-zoster virus (strain Oka) ORF22 2624 2632 QTDLLTNYK
619 Varicella-zoster virus (strain Oka) ORF22 2632 2640 KTRQPSPNF
620 Varicella-zoster virus (strain Oka) ORF22 2634 2642 RQPSPNFPR
621 Varicella-zoster virus (strain Oka) ORF22 2639 2647 NFPRDVHTW
622 Varicella-zoster virus (strain Oka) ORF22 2640 2649 FPRDVHTWGV
623 Varicella-zoster virus (strain Oka) ORF22 2646 2654 TWGVSSNPF
624 Varicella-zoster virus (strain Oka) ORF22 2650 2659 SSNPFNSPNR
625 Varicella-zoster virus (strain Oka) ORF22 2675 2683 SESENSIVL
626 Varicella-zoster virus (strain Oka) ORF22 2677 2685 SENSIVLSL
627 Varicella-zoster virus (strain Oka) ORF22 2677 2686 SE SIVLSLD
628 Varicella-zoster virus (strain Oka) ORF22 2687 2695 EHRSCRVPR
629 Varicella-zoster virus (strain Oka) ORF22 2689 2698 RSCRVPRHVR
630 Varicella-zoster virus (strain Oka) ORF22 2696 2704 HVRVVNADV
631 Varicella-zoster virus (strain Oka) ORF22 2701 27 0 NADVVTGRRY
632 Varicella-zoster virus (strain Oka) ORF22 2703 2712 DVVTGRRYVR
633 Varicella-zoster virus (strain Oka) ORF22 2704 2712 VVTGRRYVR
634 Varicella-zoster virus (strain Oka) ORF22 2728 2737 R iDNVRYTR
635 Varicella-zoster virus (strain Oka) ORF22 2729 2737 IDNVRYTR
636 Varicella-zoster virus (strain Oka) ORF22 2729 2738 MIDNVRYTRK
637 Varicella-zoster virus (strain Oka) ORF22 2739 2747 LL DHTEDI
638 Varicella-zoster virus (strain Oka) ORF22 2744 2753 TEDiFQGLGY
639 Varicella-zoster virus (strain Oka) ORF22 2745 2753 EDIFQGLGY
640 Varicella-zoster virus (strain Oka) ORF22 2746 2754 DIFQGLGYV
641 Varicella-zoster virus (strain Oka) ORF22 2746 2755 DiFGGLGYVK
642 Varicella-zoster virus (strain Oka) ORF22 2753 2762 YVKLLLDGTY

Claims

CLAIMS What is claimed is:
1. A method of treating cancer comprising:
a) providing a subject having or suspected of having a cancer; and b) administering a vector to the subject, wherein the vector directs expression of one or more peptide antigens on a target cell of the subject, wherein a memory or recall immune response is elicited against the one or more peptide antigens.
2. A method of treating cancer comprising:
a) providing a subject having or suspected of having a cancer; and b) administering a composition to the subject, wherein the composition comprises one or more peptide antigens,
wherein a memory or recall immune response is elicited against the one or more peptide antigens.
3. The method of claim 1 or 2, wherein from 1 day to 100 years prior to (b), the subject was vaccinated against a pathogen.
4. The method of claim 3, wherein prior to (b), the subject was vaccinated against chickenpox, measles, mumps, rubella, polio, or shingles/varicella zoster; a commercially available vaccine against another pathogen; one or more peptides listed in Table 1; or one or more peptides derived from a pathogen.
5. The method of any one of claims 1 to 4, wherein the administering elicits a recall immune response directed against the target cell presenting said peptide antigens, against the one or more peptide antigens, against chickenpox, measles, mumps, rubella, polio, or shingles/varicella zoster or against one or more peptide antigens derived from chickenpox, measles, mumps, rubella, polio, or shingles/varicella zoster.
6. The method of any one of claims 1 to 5, wherein the subject is further administered one or more anti-cancer therapies prior to, concurrently with, or after the administering of (b).
7. The method of claim 6, wherein the anti-cancer therapy comprises surgical resection, administering radiation therapy, chemotherapy or an immunotherapy which can be administered separately or in combination.
8. The method of claim 7, wherein the radiation therapy comprises external beam radiation, proton therapy, or internal radiation therapy.
9. The method of claim 7, wherein the immunotherapy comprises (i) chimeric antigen receptor (CAR)-T immunotherapy and optionally (ii) wherein the CAR-T immunotherapy comprises administering CAR T-cells configured to specifically bind to and kill the target cell.
10. The method of any one of claims 1 to 9, wherein the one or more peptide antigens bind to MHC class I or MHC class II and recall CD8 T-cell or CD4 T-cell responses.
11. The method of claim 7, wherein the immunotherapy comprises administering an antibody or antibody fragment that specifically binds to a tumor marker.
12. The method of claim 7 or 11, wherein the immunotherapy comprises administering an antibody or antibody fragment that specifically binds to CD20, CD30, CD33, CD47 (SIRP-1 alpha soluble receptor), CD52, CD138, CD274, CD279, PD-L1, CTLA-4, programmed cell death 1 (PD-1), CD33, EGFR, ERBB2 (CD340), ERBB3, VEGF, VEGFR, GM-2, gp-100, EPCAM, CD19, CD3, CEA, gpA33, mucins, TAG-72, CAIX, PSMA, folate-binding protein, GD2, GD3, Integrin _V_3, Integrin 5 1, IGF1R, EPHA3, TRAILRl, TRAILR2, RANKL, FAP, Tenascin, and/or administering cytokines, modified cytokines, chemokines, or toll-like receptors agonists, wherein the cytokines may optionally be selected from 11-2, 11-12, and IL- 15.
13. The method of claim 7, 11 or 12, wherein the immunotherapy comprises
administering cytokines, modified cytokines, chemokines, or toll-like receptors agonists, alone or in combination with other anti-cancer therapies, and wherein the cytokines may optionally be selected from 11-2, 11-12, and IL-15.
14. The method of claim 7, wherein the immunotherapy comprises administering an antibody or T cell that binds an immune checkpoint protein.
15. The method of any one of claims 3 to 14, wherein the peptide antigen is derived from the pathogen.
16. The method of any one of claims 3 to 15, wherein the pathogen is a human pathogen or non-human pathogen, or wherein the peptide antigen is conserved between human and another species, or wherein the peptide antigen cross-reacts between human and another species.
17. The method of any one of claims 3 to 16, wherein the pathogen is selected from a virus, bacteria, parasite or protozoa.
18. The method of any one of claims 3 to 17, wherein the pathogen is a virus selected from the order Flaviviridae, Retroviridae, Orthomyxoviridae, Arenaviridae, and
Mononegavirales.
19. The method of any one of claims 3 to 18, wherein the pathogen is a virus selected from the family Togaviridae, Reoviridae, picornaviridae, herpesviridae and Bunyaviridae.
20. The method of any one of claims 3 to 19, wherein the pathogen is a virus from the family Reoviridae.
21. The method of any one of claims 1 to 20, wherein the target cell is a cancer cell or neoplastic cell.
22. The method of any one of claims 1 to 21, wherein the target cell is a metastatic cancerous cell.
23. The method of claim 21 or 22, wherein the cancer cell or neoplastic cell is selected from a carcinoma, sarcoma, neuroblastoma, cervical cancer cell, hepatocellular cancer cell, bladder cancer cell, mesothelioma, glioblastoma, myeloma, lymphoma, neoplastic
lymphocyte, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, lymphosarcoma, liposarcoma, osteosarcoma, chondrosarcoma, leiomyosarcoma, rhabdomyosarcoma or fibrosarcoma.
24. The method of claim 23, wherein the cancer cell or neoplastic cell is a carcinoma or sarcoma.
25. The method of any one of claims 1 to 24, wherein the target cell is a cell that is or has been infected with a virus, bacteria, parasite or protozoa.
26. The method of any one of claims 1 to 25, wherein the one or more peptide antigens are selected from a peptide listed in Table 1, or a subsequence, homologue, variant or derivative thereof.
27. The method of any one of claims 1 to 26, wherein the subject is a mammal,
28. The method of any one of claims 1 to 26, wherein the subject is a non-human mammal.
29. The method of any one of claims 1 to 26, wherein the subject is a human.
30. The method of any one of claims 2 or 29, wherein prior to b), the subject was infected with the pathogen.
31. The method of any one of claims 1 to 30, wherein the cancer is a neoplasia derived from lung, thyroid, head or neck, nasopharynx, throat, nose, sinus, brain, spine, breast, adrenal gland, pituitary gland, thyroid, lymph, mouth, esophagus, stomach, duodenum, ileum, jejunum intestine, colon, rectum, uterus, urethra, ovary, cervix, endometrium, bladder, testicle, penis, prostate, kidney, pancreas, liver, bone, bone marrow, lymph, blood, muscle, or skin.
32. The method of any one of claims 1 to 30, wherein the cancer is selected from carcinoma, sarcoma, neuroblastoma, hepatocellular cancer, mesothelioma, glioblastoma, myeloma, lymphoma, myeloma, leukemia, adenoma, adenocarcinoma, glioma, glioblastoma, retinoblastoma, astrocytoma, oligodendrocytoma, meningioma, melanoma, cervical cancer, breast cancer, liver cancer, pancreatic cancer, brain cancer, lung cancer, skin cancer, ovarian cancer, and testicular cancer.
33. The method of any one of claims 1 to 30, wherein the cancer is an infection-induced cancer selected from hepatocellular carcinoma, cervical cancer, head and neck squamous cell carcinoma, adult T-cell leukemia/lymphoma, Burkitt lymphoma, Merkel cell carcinoma, Kaposi sarcoma, bladder cancer and gastric cancer.
34. The method of any one of claims 1 to 30, wherein the cancer comprises a low mutational burden.
35. The method of claim 34, wherein the cancer of low mutational burden comprises one or more of pilocytic astrocytoma, ALL, medulloblastoma, kidney chromophobe, thyroid, CLL, neuroblastoma, glioblastoma, and pancreatic cancer.
36. The method of claim 1, wherein the vector selectively directs expression of the one or more peptide antigens on the target cell.
37. The method of claim 1 or 36, wherein the vector directs expression of the one or more peptide antigens on the cell surface of the target cell.
38. The method of any one of claims 1 to 37, wherein the one or more peptide antigens are expressed or presented on the cell surface of the target cell.
39. The method of claim 2, wherein the vector comprises a nucleic acid encoding the one or more peptide antigens.
40. The method of claim 2 or 39, wherein the vector comprises a promoter operatively linked to the nucleic acid encoding the one or more peptides, and/or the vector is a replicating or non-replicating vector.
41. The method of claim 40, wherein the promoter is inducible by a factor.
42. The method of claim 41, wherein the factor is derived from the pathogen.
43. The method of claim 41, wherein the factor is a mutated human protein.
44. The method of claim 41, wherein the factor is one or more viral proteins.
45. The method of any one of claims 41 to 44, wherein the factor is a viral transcription factor.
46. The method of any one of claims 1 to 46, wherein the administering of (b) comprises administering nano-carriers, micro-carriers, microparticles, nanoparticles, liposomes, exosomes or recombinant virus to the subject, wherein the recombinant virus is a replicating or non-replicating virus.
47. The method of claim 46, wherein the microparticles, nanoparticles, liposomes, exosomes or recombinant virus comprise the vector.
48. The method of claim 46 or 47, wherein the administering of (b) comprises selectively delivering the nano-carriers, micro-carriers, microparticles, nanoparticles, liposomes, exosomes or recombinant virus to the target cell.
49. The method of claim 1, wherein the administering of (b) comprises administering a nucleic acid to the subject, wherein the nucleic acid comprises the vector.
50. The method of claim 1, wherein the vector is a plasmid.
51. The method of claim 1, wherein the vector is a linear nucleic acid molecule.
52. The method of claim 1, wherein the vector comprises a CRISPR element.
53. The method of claim 1, wherein the vector is configured to express (CRISPR- associated protein-9 nuclease) CAS9.
54. The method of claims 1, wherein the vector is integrated into the target cell by a method comprising CRISPR and (CRISPR-associated protein-9 nuclease) CAS9.
55. The method of any one of claims 1 to 54, wherein the administering comprises direct intra-tumoral injection, peri -tumoral, intravesical (bladder), or stereotactic (glioblastoma) administration, and wherein the intravesical administration is optionally intravesical administration comprising use of a urethral catheter.
56. The method of any one of claims 1 to 54, wherein the administering comprises intravenous injection, intraperitoneal injection, subcutaneous injection, intramuscular injection, bolus injection or slow perfusion.
57. The method of any one of claims 1 to 56, further comprising, concomitant with, or after b):
c) initiating, eliciting, promoting, or enhancing an immune response in the subject.
58. The method of claim 57, wherein the immune response is directed against the target cell and/or target antigen.
59. The method of claim 57 or 58, wherein the eliciting of (c) comprises initiating, eliciting, promoting or enhancing a recall response in the subject to the one or more peptide antigens.
60. The method of any one of claims 57 to 59, wherein the eliciting of (c) comprises administering a vaccine to the subject alone or in combination with immunostimulants.
61. The method of claim 60, wherein the vaccine is selected from a vaccine for chickenpox, measles, mumps, rubella, and polio.
62. The method of any one of claims 57 to 61, wherein the eliciting of (c) comprises administering a pharmaceutical composition to the subject, wherein the pharmaceutical composition comprises at least one of the one or more of the peptide antigens.
63. The method of claim 62, wherein the pharmaceutical composition comprises an adjuvant, or formulation with liposome, nano- or micro-particles or any other pharmaceutically acceptable compositions for administration.
64. The method of any one of claims 57 to 63, wherein the eliciting of (c) comprises administering an agent that modulates an immune response in the subject.
65. The method of claim 64, wherein the agent increases, enhances, promotes or elicits an innate immune response.
66. The method of claim 64 or 65, wherein the agent comprises a checkpoint blocker.
67. The method of any one of claims 64 or 66, wherein the agent increases, enhances, promotes or elicits a recall immune response.
68. The method of any one of claims 64 to 67, wherein the agent comprises an antibody.
69. The method of claim 68, wherein the antibody specifically binds to the one or more peptide antigens.
70. The method of any one of claims 64 to 69, wherein the agent comprises a nucleic acid, synthetic chemical, or small molecule.
71. The method of any one of claims 1 to 70, further comprising administering a checkpoint blocker.
72. The method of any one of claims 1 to 70, further comprising administering an agent to improve or abet antigen presentation through phagocytosis, endoplasmic reticulum based protein processing, peptide:MHC loading, and MHC surface expression.
73. The method of any one of claims 1 to 72, further comprising administering TAP to the target cell, wherein the TAP promotes peptide:MHC loading and expression.
74. The method of claim 2, wherein the composition comprises an adjuvant.
75. The method of claim 2, wherein the composition comprises exosomes.
76. The method of claim 2, wherein the composition comprises a nano-carrier, micro- carrier, microparticle, nanoparticle, or liposome.
77. The method of claim 76, wherein the microparticle, nanoparticle, or liposome configured to selectively deliver the one or more peptides to the target cell.
78. A pharmaceutical composition comprising one or more of a peptide listed in Table 1.
79. The composition of claim 78, further comprising an adjuvant.
80. A pharmaceutical composition comprising a vector configured to direct expression of one or more peptide antigens on a target cell of a subject, wherein the one or more peptide antigens is selected from a peptide listed in Table 1.
81. A diagnostic tool for measuring immune response to a vaccine or recall immunity to a target cell comprising measuring an immune response to one or more peptides listed Table 1.
82. The diagnostic tool of claim 81, wherein the measurement of recall immunity is prior to administration of a peptide listed in Table 1.
83. The diagnostic tool of claim 81, wherein the measurement of recall immunity is prior to administration of a vector configured to expresses at least one peptide listed in Table 1.
84. The method of claim 1 or 2 wherein the subject is vaccinated prior to, concurrently with, or after the administering of (b).
85. The method of claim 84, wherein the vaccination is a booster vaccination.
86. The method of claim 85, wherein the booster vaccination comprises at least one peptide listed in Table 1.
87. The method of claim 85, wherein the booster vaccination is a booster vaccination against chickenpox, measles, mumps, rubella, or polio.
88. The method of any one of claims 6 to 13, wherein the anti-cancer therapy comprises administering combinations of one or more small molecules, immune checkpoint inhibitors optionally selected from IDO/TDO inhibitors, and epigenetic regulators such as HDAC inhibitors.
89. The method of any one of claims 6 to 13, wherein the anti-cancer therapy comprises administering, alone or in combination, one or more cancer vaccines optionally based on cancer-associated tumor antigens, or neo-antigens.
90. The method of claim 2, wherein one or more of the one or more peptide antigens are linked to a targeting antibody, antibody fragment or nanobody that is internalized by the target cell.
91. The method of any one of claims 1 to 77, wherein pre-existing T cell memory is measured prior to the administering of (b).
92. The method of any one of claims 1 to 78, and 91, wherein a few days to weeks prior to the administering of (b), the method comprises boosting immune memory in the subject by administering a vaccine or antigen that stimulates a recall immune response against the one or more peptide antigens.
93. The method of any one of claims 1 to 78 and 91 or 92, wherein recall and targeting of a pre-existing immune response (CD8/CD4) from childhood vaccines leads to tumor cell killing and optionally a subsequent 'abscopal' effect with systemic, cancer-specific immune responses.
94. The method of any one of claims 2 to 78, and 91 to 93, wherein the one or more peptide antigens comprise full antigens or complete proteins derived from the pathogens.
PCT/US2017/026719 2016-04-09 2017-04-07 Leveraging immune memory from common childhood vaccines to fight disease WO2017177204A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662320488P 2016-04-09 2016-04-09
US62/320,488 2016-04-09
USPCT/US2016/068147 2016-12-21
PCT/US2016/068147 WO2017112830A1 (en) 2015-12-21 2016-12-21 Leveraging immune memory from common childhood vaccines to fight disease

Publications (1)

Publication Number Publication Date
WO2017177204A1 true WO2017177204A1 (en) 2017-10-12

Family

ID=60001452

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/026719 WO2017177204A1 (en) 2016-04-09 2017-04-07 Leveraging immune memory from common childhood vaccines to fight disease

Country Status (1)

Country Link
WO (1) WO2017177204A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019090304A1 (en) * 2017-11-06 2019-05-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cancer treatment utilizing pre-existing microbial immunity
CN110856751A (en) * 2018-08-24 2020-03-03 合成免疫股份有限公司 Therapeutic agents comprising nucleic acids and TCR-modified immune cells and uses thereof
CN110856724A (en) * 2018-08-24 2020-03-03 杭州优善生物科技有限公司 Therapeutic agents comprising nucleic acids and CAR-modified immune cells and uses thereof
US10881730B2 (en) 2017-02-01 2021-01-05 Modernatx, Inc. Immunomodulatory therapeutic MRNA compositions encoding activating oncogene mutation peptides
WO2021130377A1 (en) * 2019-12-24 2021-07-01 The Queen's University Of Belfast Polymeric nanoparticles for enhanced cancer treatment
EP3661550A4 (en) * 2017-08-03 2021-08-04 Regents of the University of Minnesota Activation of resident memory t cells for the treatment of cancer
WO2021197430A1 (en) * 2020-04-01 2021-10-07 Nanjing Legend Biotech Co., Ltd. Compositions and methods for reducing graft rejection in allogeneic cell therapy
US11285203B2 (en) 2017-06-23 2022-03-29 Verimmune Inc. Chimeric virus-like particles and uses thereof as antigen-specific redirectors of immune responses
US11560408B2 (en) 2018-12-27 2023-01-24 Verimmune Inc. Conjugated virus-like particles and uses thereof as anti-tumor immune redirectors
US11642401B2 (en) 2015-11-06 2023-05-09 Regents Of The University Of Minnesota Activation of resident memory T cells for cancer immunotherapy
US11858964B2 (en) 2020-10-19 2024-01-02 Verimmune Inc. Virus-inspired compositions and methods of redirecting preexisting immune responses using the same for treatment of cancer

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0192902A2 (en) * 1984-08-21 1986-09-03 Merck & Co. Inc. DNA sequences from Varicella-zoster virus, vectors and hosts containing them, the corresponding polypeptides and the compositions containing them
WO1999003499A1 (en) * 1997-07-16 1999-01-28 Institut National De La Sante Et De La Recherche Medicale Cellular vesicle called ''exosome'', preparation and use thereof in immune stimulation
US20050226846A1 (en) * 1999-11-03 2005-10-13 Powderject Research Limited Nucleic acid vaccine compositions having a mammalian CD80/CD86 gene promoter driving antigen expression
US20060263389A1 (en) * 2003-07-08 2006-11-23 Stacy Sue C Methods and compositions to enhance immune responses via recall antigens

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0192902A2 (en) * 1984-08-21 1986-09-03 Merck & Co. Inc. DNA sequences from Varicella-zoster virus, vectors and hosts containing them, the corresponding polypeptides and the compositions containing them
WO1999003499A1 (en) * 1997-07-16 1999-01-28 Institut National De La Sante Et De La Recherche Medicale Cellular vesicle called ''exosome'', preparation and use thereof in immune stimulation
US20050226846A1 (en) * 1999-11-03 2005-10-13 Powderject Research Limited Nucleic acid vaccine compositions having a mammalian CD80/CD86 gene promoter driving antigen expression
US20060263389A1 (en) * 2003-07-08 2006-11-23 Stacy Sue C Methods and compositions to enhance immune responses via recall antigens

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BERGEN RANDY E. ET AL.: "The Immunogenicity of the Oka/Merck Varicella Vaccine in Relation to Infectious Varicella-Zoster Virus and Relative Viral Antigen Content", THE JOURNAL OF INFECTIOUS DISEASES, vol. 162, 1990, pages 1049 - 1054, XP001056706 *
DAKAPPAGARI ET AL.: "Internalizing antibodies to the C-Type lectins, L-SIGN and DC-SIGN, inhibit viral glycoprotein binding and deliver antigen to human dendritic cells for the induction of T- cell responses", THE JOURNAL OF IMMUNOLOGY, vol. 176, 2006, pages 426 - 440, XP002385270 *
DATABASE DATABASE UniProtKB [O] 1 July 1989 (1989-07-01), XP055431189, Database accession no. P09278.1 *
SENIS E. ET AL.: "CRISPR/Cas9-mediated genome engineering: An adeno-associated viral (AAV) vector toolbox", BIOTECHNOLOGY JOURNAL, 3 September 2014 (2014-09-03), pages 1 - 32, XP002737562 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11642401B2 (en) 2015-11-06 2023-05-09 Regents Of The University Of Minnesota Activation of resident memory T cells for cancer immunotherapy
US10881730B2 (en) 2017-02-01 2021-01-05 Modernatx, Inc. Immunomodulatory therapeutic MRNA compositions encoding activating oncogene mutation peptides
US11944677B2 (en) 2017-06-23 2024-04-02 Verimmune Inc. Chimeric virus-like particles and uses thereof as antigen-specific redirectors of immune responses
US11285203B2 (en) 2017-06-23 2022-03-29 Verimmune Inc. Chimeric virus-like particles and uses thereof as antigen-specific redirectors of immune responses
EP3661550A4 (en) * 2017-08-03 2021-08-04 Regents of the University of Minnesota Activation of resident memory t cells for the treatment of cancer
JP2021502355A (en) * 2017-11-06 2021-01-28 ザ・ユナイテッド・ステイツ・オブ・アメリカ・アズ・リプリゼンテッド・バイ・ザ・セクレタリー・デパートメント・オブ・ヘルス・アンド・ヒューマン・サービシーズThe United States of America,as represented by the Secretary,Department of Health and Human Services Cancer treatment using existing microbial immunity
WO2019090304A1 (en) * 2017-11-06 2019-05-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cancer treatment utilizing pre-existing microbial immunity
CN110856724A (en) * 2018-08-24 2020-03-03 杭州优善生物科技有限公司 Therapeutic agents comprising nucleic acids and CAR-modified immune cells and uses thereof
CN112584849A (en) * 2018-08-24 2021-03-30 杭州优善生物科技有限公司 Therapeutic agents comprising nucleic acids and CAR-modified immune cells and uses thereof
US11786552B2 (en) 2018-08-24 2023-10-17 Hangzhou Convero Co., Ltd. Therapeutic compositions and applications that comprise nucleic acids and adoptively transferred immune cell
US11819519B2 (en) 2018-08-24 2023-11-21 Hangzhou Converd Co., Ltd. Therapeutic agents comprising nucleic acids and TCR modified immune cells and uses thereof
JP2021536435A (en) * 2018-08-24 2021-12-27 杭州康万達医薬科技有限公司Hangzhou Converd Co., Ltd. Therapeutic agents containing nucleic acids and CAR-modified immune cells and their use
EP3842051A4 (en) * 2018-08-24 2022-06-29 Hangzhou Converd Co., Ltd. Therapeutic agent comprising nucleic acid and car-modified immune cell and application thereof
CN110856751A (en) * 2018-08-24 2020-03-03 合成免疫股份有限公司 Therapeutic agents comprising nucleic acids and TCR-modified immune cells and uses thereof
US11560408B2 (en) 2018-12-27 2023-01-24 Verimmune Inc. Conjugated virus-like particles and uses thereof as anti-tumor immune redirectors
WO2021130377A1 (en) * 2019-12-24 2021-07-01 The Queen's University Of Belfast Polymeric nanoparticles for enhanced cancer treatment
WO2021197430A1 (en) * 2020-04-01 2021-10-07 Nanjing Legend Biotech Co., Ltd. Compositions and methods for reducing graft rejection in allogeneic cell therapy
US11858964B2 (en) 2020-10-19 2024-01-02 Verimmune Inc. Virus-inspired compositions and methods of redirecting preexisting immune responses using the same for treatment of cancer

Similar Documents

Publication Publication Date Title
WO2017177204A1 (en) Leveraging immune memory from common childhood vaccines to fight disease
WO2017112830A1 (en) Leveraging immune memory from common childhood vaccines to fight disease
JP6903120B2 (en) Compositions and Methods for Alpha Virus Vaccination
TWI731095B (en) Compositions and methods for tumor vaccination using prostate cancer-associated antigens
TW201805013A (en) Compositions and methods for tumor vaccination and immunotherapy involving HER2/NEU
US20220016234A1 (en) Anti covid-19 therapies using nucleocapsid and spike proteins
US20220168406A1 (en) Cancer vaccines against mucosal antigens and methods of making and using the same
Haug et al. Photochemical internalization of peptide antigens provides a novel strategy to realize therapeutic cancer vaccination
US20110081317A1 (en) Enhancing Gene Transfer
AU2022201142A1 (en) Vaccine compositions and methods for restoring NKG2D pathway function against cancers
WO2023184861A1 (en) Hpv epitope and identification method therefor, and application thereof
WO2021183665A1 (en) Treatment of covid-19 and methods therefor
Ayesha et al. Physiopathology and effectiveness of therapeutic vaccines against human papillomavirus
Tseng et al. Albumin and interferon-β fusion protein serves as an effective vaccine adjuvant to enhance antigen-specific CD8+ T cell-mediated antitumor immunity
Liu et al. Advances in cancer vaccine research
Zhang et al. Recombinant cell-penetrating trichosanthin synergizes anti-PD-1 therapy in colorectal tumor
ES2687189T3 (en) Compositions and methods for the treatment of type 1 diabetes
ES2809508T3 (en) Artificial structures of polyepitopes for use in immunotherapy
EP1613731A2 (en) Modified adenoviral e1a constructs and methods of use thereof
Jadav et al. Improved immune response by ID-pVAC: a secretory DNA vaccine construct delivered by PLG micro particles against foot and mouth disease in guinea pigs
AU2021236141B2 (en) Treatment of covid-19 and methods therefor
US20230321230A1 (en) Compositions and Methods for Adjuvanted Vaccines
US11857620B2 (en) Method of inducing immunity against SARS-CoV-2 using spike (s) and nucleocapsid (N)-ETSD immunogens delivered by a replication-defective adenovirus
Wang et al. The Role and Development of Peptide Vaccines in Cervical Cancer Treatment
EP4118210A1 (en) Treatment of covid-19 and methods therefor

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17779958

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17779958

Country of ref document: EP

Kind code of ref document: A1